1
|
Li X, Jin K, Liao YC, Lee WJ, Chen LC, Cheng TC, Ho YS, Guo P. RNA Nanotechnology for Codelivering High-Payload Nucleoside Analogs to Cancer with a Synergetic Effect. Mol Pharm 2024; 21:5690-5702. [PMID: 39388598 DOI: 10.1021/acs.molpharmaceut.4c00674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Nucleoside analogs are potent inhibitors for cancer treatment, but the main obstacles to their application in humans are their toxicity, nonspecificity, and lack of targeted delivery tools. Here, we report the use of RNA four-way junctions (4WJs) to deliver two nucleoside analogs, floxuridine (FUDR) and gemcitabine (GEM), with high payloads through routine and simple solid-state RNA synthesis and nanoparticle assembly. The design of RNA nanotechnology for the co-delivery of nucleoside analogs and the chemotherapeutic drug paclitaxel (PTX) resulted in synergistic effects and high efficacy in the treatment of Triple-Negative Breast Cancer (TNBC). The 4WJ-drug complexes were confirmed to have efficient tumor spontaneous targeting and no toxicity because the motility of RNA nanoparticles has been previously shown to enable these RNA-drug complexes to spontaneously accumulate in tumor blood vessels. The negative charge of RNA enables those RNA complexes that are not targeted to tumor vasculature to circulate in the blood and enter the urine through the kidney glomerulus, without accumulating in organs, therefore being nontoxic. Drug incorporation into RNA 4WJ can be precisely controlled with a defined loading amount, location, and ratio. The incorporation of nucleoside analogs into 4WJ only requires one step using nucleoside analogue phosphoramidites during solid-phase RNA synthesis, without the need for additional conjugation and purification processes.
Collapse
Affiliation(s)
| | | | - You-Cheng Liao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | | | - Li-Ching Chen
- Department of Biological Science & Technology, China Medical University, Taichung 40402, Taiwan
| | - Tzu-Chun Cheng
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung 40402, Taiwan
| | - Yuan-Soon Ho
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung 40402, Taiwan
| | | |
Collapse
|
2
|
Lee DJ, Cao Y, Juvekar V, Sauraj, Noh CK, Shin SJ, Liu Z, Kim HM. Development of a small molecule-based two-photon photosensitizer for targeting cancer cells. J Mater Chem B 2024. [PMID: 39469993 DOI: 10.1039/d4tb01706d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Photodynamic therapy (PDT) employing two-photon (TP) excitation is increasingly recognized to induce cell damage selectively in targeted areas, underscoring the importance of developing TP photosensitizers (TP-PSs). In this study, we developed BSe-B, a novel PS that combines a selenium containing dye with biotin, a cancer-selective ligand, and is optimized for TP excitation. BSe-B demonstrated enhanced cancer selectivity, efficient generation of type-I based reactive oxygen species (ROS), low dark toxicity, and excellent cell-staining capability. Evaluation across diverse cell lines (HeLa, A549, OVCAR-3, WI-38, and L-929) demonstrated that BSe-B differentiated and targeted cancer cells while sparing normal cells. BSe-B displayed excellent in vivo biocompatibility. In cancer models such as three-dimensional spheroids and actual colon cancer tissues, BSe-B selectively induced ROS production and cell death under TP irradiation, demonstrating precise spatial control. These findings highlight the potential of BSe-B for imaging-guided PDT and its capability for micro treatment within tissues. Thus, BSe-B demonstrates robust TP-PDT capabilities, making it a promising dual-purpose tool for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Dong Joon Lee
- Department of Energy Systems Research and Department of Chemistry, Ajou University, Suwon 16499, Korea.
| | - Yu Cao
- College of Health Science and Engineering, Hubei University, Wuhan 430062, China.
| | - Vinayak Juvekar
- Department of Energy Systems Research and Department of Chemistry, Ajou University, Suwon 16499, Korea.
| | - Sauraj
- Department of Energy Systems Research and Department of Chemistry, Ajou University, Suwon 16499, Korea.
| | - Choong-Kyun Noh
- Department of Gastroenterology, Ajou University School of Medicine, Suwon 16499, Korea.
| | - Sung Jae Shin
- Department of Gastroenterology, Ajou University School of Medicine, Suwon 16499, Korea.
| | - Zhihong Liu
- College of Health Science and Engineering, Hubei University, Wuhan 430062, China.
| | - Hwan Myung Kim
- Department of Energy Systems Research and Department of Chemistry, Ajou University, Suwon 16499, Korea.
| |
Collapse
|
3
|
Farias FFS, Mittersteiner M, Kieling AM, Lima PSV, Weimer GH, Bonacorso HG, Zanatta N, Martins MAP. The Persistence of Hydrogen Bonds in Pyrimidinones: From Solution to Crystal. ACS ORGANIC & INORGANIC AU 2024; 4:557-570. [PMID: 39371326 PMCID: PMC11450830 DOI: 10.1021/acsorginorgau.4c00057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 10/08/2024]
Abstract
Pyrimidinone scaffolds are present in a wide array of molecules with synthetic and pharmacological utility. The inherent properties of these compounds may be attributed to intermolecular interactions analogous to the interactions that molecules tend to establish with active sites. Pyrimidinones and their fused derivatives have garnered significant interest due to their structural features, which resemble nitrogenous bases, the foundational building blocks of DNA and RNA. Similarly, pyrimidinones are predisposed to forming N-H···O hydrogen bonds akin to nitrogenous bases. Given this context, this study explored the supramolecular features and the predisposition to form hydrogen bonds in a series of 18 substituted 4-(trihalomethyl)-2(1H)-pyrimidinones. The formation of hydrogen bonds was observed in solution via nuclear magnetic resonance (NMR) spectroscopy experiments, and subsequently confirmed in the crystalline solid state. Hence, the 18 compounds were crystallized through crystallization assays by slow solvent evaporation, followed by single-crystal X-ray diffraction (SC-XRD). The supramolecular cluster demarcation was employed to evaluate all intermolecular interactions, and all crystalline structures exhibited robust hydrogen bonds, with an average energy of approximately -21.64 kcal mol-1 (∼19% of the total stabilization energy of the supramolecular clusters), irrespective of the substituents at positions 4, 5, or 6 of the pyrimidinone core. To elucidate the nature of these hydrogen bonds, an analysis based on the quantum theory of atoms in molecules (QTAIM) revealed that the predominant intermolecular interactions are N-H···O (average of -16.55 kcal mol-1) and C-H···O (average of -6.48 kcal mol-1). Through proposing crystallization mechanisms based on molecular stabilization energy data and contact areas between molecules and employing the supramolecular cluster and retrocrystallization concepts, it was determined that altering the halogen (F/Cl) at position 4 of the pyrimidinone nucleus modifies the crystallization mechanism pathway. Notably, the hydrogen bonds present in the initial proposed steps were confirmed by 1H NMR experiments using concentration-dependent techniques.
Collapse
Affiliation(s)
- Fellipe F. S. Farias
- Núcleo de Química
de Heterociclos (NUQUIMHE), Department of Chemistry, Federal University of Santa Maria (UFSM), 97105-900 Santa Maria, Rio Grande do Sul, Brazil
| | - Mateus Mittersteiner
- Núcleo de Química
de Heterociclos (NUQUIMHE), Department of Chemistry, Federal University of Santa Maria (UFSM), 97105-900 Santa Maria, Rio Grande do Sul, Brazil
| | - Amanda M. Kieling
- Núcleo de Química
de Heterociclos (NUQUIMHE), Department of Chemistry, Federal University of Santa Maria (UFSM), 97105-900 Santa Maria, Rio Grande do Sul, Brazil
| | - Priscila S. V. Lima
- Núcleo de Química
de Heterociclos (NUQUIMHE), Department of Chemistry, Federal University of Santa Maria (UFSM), 97105-900 Santa Maria, Rio Grande do Sul, Brazil
| | - Gustavo H. Weimer
- Núcleo de Química
de Heterociclos (NUQUIMHE), Department of Chemistry, Federal University of Santa Maria (UFSM), 97105-900 Santa Maria, Rio Grande do Sul, Brazil
| | - Helio G. Bonacorso
- Núcleo de Química
de Heterociclos (NUQUIMHE), Department of Chemistry, Federal University of Santa Maria (UFSM), 97105-900 Santa Maria, Rio Grande do Sul, Brazil
| | - Nilo Zanatta
- Núcleo de Química
de Heterociclos (NUQUIMHE), Department of Chemistry, Federal University of Santa Maria (UFSM), 97105-900 Santa Maria, Rio Grande do Sul, Brazil
| | - Marcos A. P. Martins
- Núcleo de Química
de Heterociclos (NUQUIMHE), Department of Chemistry, Federal University of Santa Maria (UFSM), 97105-900 Santa Maria, Rio Grande do Sul, Brazil
| |
Collapse
|
4
|
Vasvani S, Vasukutty A, Bardhan R, Park IK, Uthaman S. Reactive oxygen species driven prodrug-based nanoscale carriers for transformative therapies. Biomater Sci 2024; 12:4335-4353. [PMID: 39041781 DOI: 10.1039/d4bm00647j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Reactive oxygen species (ROS) drive processes in various pathological conditions serving as an attractive target for therapeutic strategies. This review highlights the development and use of ROS-dependent prodrug-based nanoscale carriers that has transformed many biomedical applications. Incorporating prodrugs into nanoscale carriers not only improves their stability and solubility but also enables site-specific drug delivery ultimately enhancing the therapeutic effectiveness of the nanoscale carriers. We critically examine recent advances in ROS-responsive nanoparticulate platforms, encompassing liposomes, polymeric nanoparticles, and inorganic nanocarriers. These platforms facilitate precise control over drug release upon encountering elevated ROS levels at disease sites, thereby minimizing off-target effects and maximizing therapeutic efficiency. Furthermore, we investigate the potential of combination therapies in which ROS-activated prodrugs are combined with other therapeutic agents and underscore their synergistic potential for treating multifaceted diseases. This comprehensive review highlights the immense potential of ROS-dependent prodrug-based nanoparticulate systems in revolutionizing biomedical applications; such nanoparticulate systems can facilitate selective and controlled drug delivery, reduce toxicity, and improve therapeutic outcomes for ROS-associated diseases.
Collapse
Affiliation(s)
- Shyam Vasvani
- Department of Biomedical Sciences and BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju 61469, Republic of Korea.
- DR Cure Inc., Hwasun 58128, Republic of Korea
| | - Arathy Vasukutty
- Department of Biomedical Sciences and BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju 61469, Republic of Korea.
| | - Rizia Bardhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa, 50011, USA
| | - In-Kyu Park
- Department of Biomedical Sciences and BioMedical Sciences Graduate Program (BMSGP), Chonnam National University Medical School, Gwangju 61469, Republic of Korea.
- DR Cure Inc., Hwasun 58128, Republic of Korea
- Center for Global Future Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Hwasun 58128, Republic of Korea
| | - Saji Uthaman
- Smart Materials and Devices (SMAD) Division, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India.
| |
Collapse
|
5
|
Anichina K, Lumov N, Bakov V, Yancheva D, Georgiev N. Recent Advances in the Application of Nitro(het)aromatic Compounds for Treating and/or Fluorescent Imaging of Tumor Hypoxia. Molecules 2024; 29:3475. [PMID: 39124883 PMCID: PMC11314162 DOI: 10.3390/molecules29153475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
This review delves into recent advancements in the field of nitro(het)aromatic bioreductive agents tailored for hypoxic environments. These compounds are designed to exploit the low-oxygen conditions typically found in solid tumors, making them promising candidates for targeted cancer therapies. Initially, this review focused on their role as gene-directed enzyme prodrugs, which are inert until activated by specific enzymes within tumor cells. Upon activation, these prodrugs undergo chemical transformations that convert them into potent cytotoxic agents, selectively targeting cancerous tissue while sparing healthy cells. Additionally, this review discusses recent developments in prodrug conjugates containing nitro(het)aromatic moieties, designed to activate under low-oxygen conditions within tumors. This approach enhances their efficacy and specificity in cancer treatment. Furthermore, this review covers innovative research on using nitro(het)aromatic compounds as fluorescent probes for imaging hypoxic tumors. These probes enable non-invasive visualization of low-oxygen regions within tumors, providing valuable insights for the diagnosis, treatment planning, and monitoring of therapeutic responses. We hope this review will inspire researchers to design and synthesize improved compounds for selective cancer treatment and early diagnostics.
Collapse
Affiliation(s)
- Kameliya Anichina
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria; (K.A.); (N.L.); (V.B.); (D.Y.)
| | - Nikolay Lumov
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria; (K.A.); (N.L.); (V.B.); (D.Y.)
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev str. Bl. 9, 1113 Sofia, Bulgaria
| | - Ventsislav Bakov
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria; (K.A.); (N.L.); (V.B.); (D.Y.)
| | - Denitsa Yancheva
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria; (K.A.); (N.L.); (V.B.); (D.Y.)
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev str. Bl. 9, 1113 Sofia, Bulgaria
| | - Nikolai Georgiev
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria; (K.A.); (N.L.); (V.B.); (D.Y.)
| |
Collapse
|
6
|
Xu X, Zhang J, Wang T, Li J, Rong Y, Wang Y, Bai C, Yan Q, Ran X, Wang Y, Zhang T, Sun J, Jiang Q. Emerging non-antibody‒drug conjugates (non-ADCs) therapeutics of toxins for cancer treatment. Acta Pharm Sin B 2024; 14:1542-1559. [PMID: 38572098 PMCID: PMC10985036 DOI: 10.1016/j.apsb.2023.11.029] [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: 09/04/2023] [Revised: 10/31/2023] [Accepted: 11/23/2023] [Indexed: 04/05/2024] Open
Abstract
The non-selective cytotoxicity of toxins limits the clinical relevance of the toxins. In recent years, toxins have been widely used as warheads for antibody‒drug conjugates (ADCs) due to their efficient killing activity against various cancer cells. Although ADCs confer certain targeting properties to the toxins, low drug loading capacity, possible immunogenicity, and other drawbacks also limit the potential application of ADCs. Recently, non-ADC delivery strategies for toxins have been extensively investigated. To further understand the application of toxins in anti-tumor, this paper provided an overview of prodrugs, nanodrug delivery systems, and biomimetic drug delivery systems. In addition, toxins and their combination strategies with other therapies were discussed. Finally, the prospect and challenge of toxins in cancer treatment were also summarized.
Collapse
Affiliation(s)
- Xiaolan Xu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiaming Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tao Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jing Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yukang Rong
- School of Education, University of Nottingham, Nottingham NG7 2RD, UK
| | - Yanfang Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chenxia Bai
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qing Yan
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaohua Ran
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yingli Wang
- Department of Pharmacy, Linyi People's Hospital, Shandong University, Linyi 276000, China
| | - Tianhong Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qikun Jiang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100871, China
| |
Collapse
|
7
|
Ibrahim AA, Nsairat H, Al-Sulaibi M, El-Tanani M, Jaber AM, Lafi Z, Barakat R, Abuarqoub DA, Mahmoud IS, Obare SO, Aljabali AAA, Alkilany AM, Alshaer W. Doxorubicin conjugates: a practical approach for its cardiotoxicity alleviation. Expert Opin Drug Deliv 2024; 21:399-422. [PMID: 38623735 DOI: 10.1080/17425247.2024.2343882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/29/2024] [Indexed: 04/17/2024]
Abstract
INTRODUCTION Doxorubicin (DOX) emerges as a cornerstone in the arsenal of potent chemotherapeutic agents. Yet, the clinical deployment of DOX is tarnished by its proclivity to induce severe cardiotoxic effects, culminating in heart failure and other consequential morbidities. In response, a panoply of strategies has undergone rigorous exploration over recent decades, all aimed at attenuating DOX's cardiotoxic impact. The advent of encapsulating DOX within lipidic or polymeric nanocarriers has yielded a dual triumph, augmenting DOX's therapeutic efficacy while mitigating its deleterious side effects. AREAS COVERED Recent strides have spotlighted the emergence of DOX conjugates as particularly auspicious avenues for ameliorating DOX-induced cardiotoxicity. These conjugates entail the fusion of DOX through physical or chemical bonds with diminutive natural or synthetic moieties, polymers, biomolecules, and nanoparticles. This spectrum encompasses interventions that impinge upon DOX's cardiotoxic mechanism, modulate cellular uptake and localization, confer antioxidative properties, or refine cellular targeting. EXPERT OPINION The endorsement of DOX conjugates as a compelling stratagem to mitigate DOX-induced cardiotoxicity resounds from this exegesis, amplifying safety margins and the therapeutic profile of this venerated chemotherapeutic agent. Within this ambit, DOX conjugates stand as a beacon of promise in the perpetual pursuit of refining chemotherapy-induced cardiac compromise.
Collapse
Affiliation(s)
- Abed Alqader Ibrahim
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, NC, USA
| | - Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Mazen Al-Sulaibi
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Mohamed El-Tanani
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Areej M Jaber
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Zainab Lafi
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Rahmeh Barakat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Duaa Azmi Abuarqoub
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Ismail Sami Mahmoud
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, Zarqa, Jordan
| | - Sherine O Obare
- Department of Nanoscience, Joint School of Nanoscience and Nanoengineering, University of North Carolina at Greensboro, Greensboro, NC, USA
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Alaa A A Aljabali
- Faculty of Pharmacy, Department of Pharmaceutical Sciences, Yarmouk University, Irbid, Jordan
| | | | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| |
Collapse
|
8
|
Tantra T, Singh Y, Patekar R, Kulkarni S, Kumar P, Thareja S. Phosphate Prodrugs: An Approach to Improve the Bioavailability of Clinically Approved Drugs. Curr Med Chem 2024; 31:336-357. [PMID: 36757029 DOI: 10.2174/0929867330666230209094738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/17/2022] [Accepted: 11/23/2022] [Indexed: 02/10/2023]
Abstract
The phosphate prodrug approach has emerged as a viable option for increasing the bioavailability of a drug candidate with low hydrophilicity and poor cell membrane permeability. When a phosphoric acid moiety is attached to the parent drug, it results in a several-fold elevation in aqueous solubility which helps to achieve desired bioavailability of the pharmaceutically active parental molecule. The neutral phosphate prodrugs have rapid diffusion ability through the plasma membrane as compared to their charged counterpart. The presence of phosphate mono ester breaking alkaline phosphatase (ALP) enzyme throughout the whole human body, is the main consideration behind the development of phosphate prodrug strategy. The popularity of this phosphate prodrug strategy is increasing nowadays due to the fulfillment of different desired pharmacokinetic characteristics required to get pharmaceutical and therapeutic responses without showing any serious adverse drug reactions (ADR). This review article mainly focuses on various phosphate prodrugs synthesized within the last decade to get an improved pharmacological response of the parent moiety along with various preclinical and clinical challenges associated with this approach. Emphasis is also given to the chemical mechanism to release the parent moiety from the prodrug.
Collapse
Affiliation(s)
- Tanmoy Tantra
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Yogesh Singh
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Rohan Patekar
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Swanand Kulkarni
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Pradeep Kumar
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| |
Collapse
|
9
|
Husain A, Monga J, Narwal S, Singh G, Rashid M, Afzal O, Alatawi A, Almadani NM. Prodrug Rewards in Medicinal Chemistry: An Advance and Challenges Approach for Drug Designing. Chem Biodivers 2023; 20:e202301169. [PMID: 37833241 DOI: 10.1002/cbdv.202301169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 10/15/2023]
Abstract
This article emphasizes the importance of prodrugs and their diverse spectrum of effects in the field of developing novel drugs for a variety of biological applications. Prodrugs are chemicals that are supplied inactively, but then go through enzymatic and chemical transformation in vivo to release the active parent medication that can have the desired pharmacological effect. By adding an inactive chemical moiety, prodrugs are improved in a number of ways that contribute to their potency and durability. For the purpose of illustrating the usefulness of the prodrug approach, this review covers examples of prodrugs that have been made available or are now undergoing human trials. Additionally, it included lists of the most common functional groups, carrier linkers, and reactive chemicals that can be used to create prodrugs. The current study also provides a brief introduction, several chemical methods and modifications for creating prodrugs and mutual prodrugs, as well as an explanation of recent advancements and difficulties in the field of prodrug design. The primary chemical carriers employed in the creation of prodrugs, such as esters, amides, imides, NH-acidic carriers, amines, alcohols, carbonyl, carboxylic, and azo-linkages, are also discussed. This review also discusses glycosidic and triglyceride mutually activated prodrugs, which aim to deliver the drugs after bioconversion at the intended site of action. The article also discusses the extensive chemistry and wide variety of applications of recently approved prodrugs, such as antibacterial, anti-inflammatory, cardiovascular, antiplatelet, antihypertensive, atherosclerotic, antiviral, etc. In order to illustrate the prodrug and mutual drug concept's various applications and highlight its many triumphs in overcoming the formulation and delivery of problematic pharmaceuticals, this work represents a thorough guide that includes the synthetic moiety for the reader.
Collapse
Affiliation(s)
- Asif Husain
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110064, India
| | - Jyoti Monga
- Ch. Devi Lal College of Pharmacy, Jagadhri, 135003, Haryana, India
| | - Smita Narwal
- Global Research Institute of Pharmacy, Nachraun, Radaur, 135133, Haryana, India
| | - Gurvirender Singh
- Institute of Pharmaceutical Sciences, Kurukshetra University Kurukshetra-136119, Haryana, India
| | - Mohammad Rashid
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Dentistry and Pharmacy, Buraydah Private Colleges, Buraydah, 51418, Saudi Arabia
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj, 11942, Saudi Arabia
| | - Abdurahhman Alatawi
- Clinical Pharmacist, Pharmaceutical Care Department, King Fahad Specialized Hospital, Tabuk, 47717, Saudi Arabia
| | - Norah M Almadani
- Biochemistry Department, Faculty of Science, University of Tabuk, Tabuk, 47914, Saudi Arabia
| |
Collapse
|
10
|
Kumari M, Acharya A, Krishnamurthy PT. Antibody-conjugated nanoparticles for target-specific drug delivery of chemotherapeutics. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2023; 14:912-926. [PMID: 37701520 PMCID: PMC10494237 DOI: 10.3762/bjnano.14.75] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/22/2023] [Indexed: 09/14/2023]
Abstract
Nanotechnology provides effective methods for precisely delivering chemotherapeutics to cancer cells, thereby improving efficacy and reducing off-target side effects. The targeted delivery of nanoscale chemotherapeutics is accomplished by two different approaches, namely the exploitation of leaky tumor vasculature (EPR effect) and the surface modification of nanoparticles (NPs) with various tumor-homing peptides, aptamers, oligonucleotides, and monoclonal antibodies (mAbs). Because of higher binding affinity and specificity, mAbs have received a lot of attention for the detection of selective cancer biomarkers and also for the treatment of various types of cancer. Antibody-conjugated nanoparticles (ACNPs) are an effective targeted therapy for the efficient delivery of chemotherapeutics specifically to the targeted cancer cells. ACNPs combine the benefits of NPs and mAbs to provide high drug loads at the tumor site with better selectivity and delivery efficiency. The mAbs on the NP surfaces recognize their specific receptors expressed on the target cells and release the chemotherapeutic agent in a controlled manner. Appropriately designed and synthesized ACNPs are essential to fully realize their therapeutic benefits. In blood stream, ACNPs instantly interact with biological molecules, and a protein corona is formed. Protein corona formation triggers an immune response and affects the targeting ability of the nanoformulation. In this review, we provide recent findings to highlight several antibody conjugation methods such as adsorption, covalent conjugation, and biotin-avidin interaction. This review also provides an overview of the many effects of the protein corona and the theranostic applications of ACNPs for the treatment of cancer.
Collapse
Affiliation(s)
- Mamta Kumari
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - Amitabha Acharya
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur (H.P.) 176061, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Praveen Thaggikuppe Krishnamurthy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, The Nilgiris, Tamil Nadu, India
| |
Collapse
|
11
|
Ramsay E, Lajunen T, Bhattacharya M, Reinisalo M, Rilla K, Kidron H, Terasaki T, Urtti A. Selective drug delivery to the retinal cells: Biological barriers and avenues. J Control Release 2023; 361:1-19. [PMID: 37481214 DOI: 10.1016/j.jconrel.2023.07.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 06/09/2023] [Accepted: 07/16/2023] [Indexed: 07/24/2023]
Abstract
Retinal drug delivery is a challenging, but important task, because most retinal diseases are still without any proper therapy. Drug delivery to the retina is hampered by the anatomical and physiological barriers resulting in minimal bioavailability after topical ocular and systemic administrations. Intravitreal injections are current method-of-choice in retinal delivery, but these injections show short duration of action for small molecules and low target bioavailability for many protein, gene based drugs and nanomedicines. State-of-art delivery systems are based on prolonged retention, controlled drug release and physical features (e.g. size and charge). However, drug delivery to the retina is not cell-specific and these approaches do not facilitate intracellular delivery of modern biological drugs (e.g. intracellular proteins, RNA based medicines, gene editing). In this focused review we highlight biological factors and mechanisms that form the basis for the selective retinal drug delivery systems in the future. Therefore, we are presenting current knowledge related to retinal membrane transporters, receptors and targeting ligands in relation to nanomedicines, conjugates, extracellular vesicles, and melanin binding. These issues are discussed in the light of retinal structure and cell types as well as future prospects in the field. Unlike in some other fields of targeted drug delivery (e.g. cancer research), selective delivery technologies have been rarely studied, even though cell targeted delivery may be even more feasible after local administration into the eye.
Collapse
Affiliation(s)
- Eva Ramsay
- Drug Research Programme, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 University of Helsinki, Finland
| | - Tatu Lajunen
- Drug Research Programme, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 University of Helsinki, Finland; School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70211 Kuopio, Finland
| | - Madhushree Bhattacharya
- Drug Research Programme, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 University of Helsinki, Finland
| | - Mika Reinisalo
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70211 Kuopio, Finland
| | - Kirsi Rilla
- School of Medicine, University of Eastern Finland, Yliopistonranta 1 C, 70211 Kuopio, Finland
| | - Heidi Kidron
- Drug Research Programme, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 University of Helsinki, Finland
| | - Tetsuya Terasaki
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70211 Kuopio, Finland
| | - Arto Urtti
- Drug Research Programme, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, 00014 University of Helsinki, Finland; School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, 70211 Kuopio, Finland.
| |
Collapse
|
12
|
Mahmood T, Shahbaz A, Hussain N, Ali R, Bashir H, Rizwan K. Recent advancements in fusion protein technologies in oncotherapy: A review. Int J Biol Macromol 2023; 230:123161. [PMID: 36610574 DOI: 10.1016/j.ijbiomac.2023.123161] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/01/2023] [Accepted: 01/02/2023] [Indexed: 01/05/2023]
Abstract
Cancer is a complicated, adaptable, and heterogeneous disease caused by a wide variety of genetic changes that might impair ability of cells to function normally. The majority of the tumors can only be shrunk using conventional oncology therapies like chemotherapy, radiation, and surgical resection, and the tumor often recurs. The inability of conventional cancer therapies to completely destroy the Cancer Stem Cells (CSCs) that otherwise lead to therapy resistance is thus addressed by therapeutic approaches that concentrate on targeting CSCs and their micro-environmental niche. In this review, we summarize approaches that are used for the development of fusion proteins and their therapeutic applications for treating cancer. The main purpose of making advancements towards the fusion technology instead of using conventional treatment methods is to achieve a prolonged half-life of the therapeutic drugs. The fusion of drugs to the immune response enhancing cytokines or the fusion of antibody and cytokines not only increases half-life but also increase the stability of the anti-tumor drug. Several molecules including different fragments of antibodies, cytokines, Human Serum Albumin, transferrin, XTEN polymers, Elastin-like polypeptides (ELPs) can be employed as a fusion partner and the resulting fusion proteins are reported to show enhanced anti-tumor response.
Collapse
Affiliation(s)
- Tehreem Mahmood
- Department of Biotechnology, Quaid-i-azam University, Islamabad, Pakistan
| | - Areej Shahbaz
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Nazim Hussain
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan.
| | - Rahat Ali
- Department of Chemistry, University of Agriculture Faisalabad, Pakistan
| | - Hamid Bashir
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Komal Rizwan
- Department of Chemistry, University of Sahiwal, Sahiwal 57000, Pakistan.
| |
Collapse
|
13
|
Ioele G, Chieffallo M, Occhiuzzi MA, De Luca M, Garofalo A, Ragno G, Grande F. Anticancer Drugs: Recent Strategies to Improve Stability Profile, Pharmacokinetic and Pharmacodynamic Properties. Molecules 2022; 27:molecules27175436. [PMID: 36080203 PMCID: PMC9457551 DOI: 10.3390/molecules27175436] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 12/20/2022] Open
Abstract
In past decades, anticancer research has led to remarkable results despite many of the approved drugs still being characterized by high systemic toxicity mainly due to the lack of tumor selectivity and present pharmacokinetic drawbacks, including low water solubility, that negatively affect the drug circulation time and bioavailability. The stability studies, performed in mild conditions during their development or under stressing exposure to high temperature, hydrolytic medium or light source, have demonstrated the sensitivity of anticancer drugs to many parameters. For this reason, the formation of degradation products is assessed both in pharmaceutical formulations and in the environment as hospital waste. To date, numerous formulations have been developed for achieving tissue-specific drug targeting and reducing toxic side effects, as well as for improving drug stability. The development of prodrugs represents a promising strategy in targeted cancer therapy for improving the selectivity, efficacy and stability of active compounds. Recent studies show that the incorporation of anticancer drugs into vesicular systems, such as polymeric micelles or cyclodextrins, or the use of nanocarriers containing chemotherapeutics that conjugate to monoclonal antibodies can improve solubility, pharmacokinetics, cellular absorption and stability. In this study, we summarize the latest advances in knowledge regarding the development of effective highly stable anticancer drugs formulated as stable prodrugs or entrapped in nanosystems.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fedora Grande
- Correspondence: (G.I.); (F.G.); Tel.: +39-0984-493268 (G.I.)
| |
Collapse
|
14
|
Yuan Y, Shi C, Wu X, Li W, Huang C, Liang L, Chen J, Wang Y, Liu Y. Synthesis and anticancer activity in vitro and in vivo evaluation of iridium(III) complexes on mouse melanoma B16 cells. J Inorg Biochem 2022; 232:111820. [DOI: 10.1016/j.jinorgbio.2022.111820] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/22/2022] [Accepted: 04/02/2022] [Indexed: 02/06/2023]
|
15
|
Hwang SR, Chakraborty K, An JM, Mondal J, Yoon HY, Lee YK. Pharmaceutical Aspects of Nanocarriers for Smart Anticancer Therapy. Pharmaceutics 2021; 13:pharmaceutics13111875. [PMID: 34834290 PMCID: PMC8619450 DOI: 10.3390/pharmaceutics13111875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/28/2021] [Accepted: 11/03/2021] [Indexed: 12/13/2022] Open
Abstract
Drug delivery to tumor sites using nanotechnology has been demonstrated to overcome the drawbacks of conventional anticancer drugs. Altering the surface shape and geometry of nanocomposites alters their chemical properties, which can confer multiple attributes to nanocarriers for the treatment of cancer and their use as imaging agents for cancer diagnosis. However, heterogeneity and blood flow in human cancer limit the distribution of nanoparticles at the site of tumor tisues. For targeted delivery and controlled release of drug molecules in harsh tumor microenvironments, smart nanocarriers combined with various stimuli-responsive materials have been developed. In this review, we describe nanomaterials for smart anticancer therapy as well as their pharmaceutical aspects including pharmaceutical process, formulation, controlled drug release, drug targetability, and pharmacokinetic or pharmacodynamic profiles of smart nanocarriers. Inorganic or organic-inorganic hybrid nanoplatforms and the electrospinning process have also been briefly described here.
Collapse
Affiliation(s)
- Seung Rim Hwang
- College of Pharmacy, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju 61452, Korea;
| | - Kushal Chakraborty
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju 27469, Korea;
| | - Jeong Man An
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Korea;
| | - Jagannath Mondal
- Department of Green Bio Engineering, Graduate School, Korea National University of Transportation, Chungju 27469, Korea;
- 4D Convergence Technology Institute, Korea National University of Transportation, Jeungpyeong 27909, Korea
| | - Hong Yeol Yoon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea;
| | - Yong-kyu Lee
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju 27469, Korea;
- Department of Green Bio Engineering, Graduate School, Korea National University of Transportation, Chungju 27469, Korea;
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju 27469, Korea
- Correspondence: ; Tel.: +82-43-841-5224
| |
Collapse
|
16
|
Maslah H, Skarbek C, Gourson C, Plamont MA, Pethe S, Jullien L, Le Saux T, Labruère R. In-Cell Generation of Anticancer Phenanthridine Through Bioorthogonal Cyclization in Antitumor Prodrug Development. Angew Chem Int Ed Engl 2021; 60:24043-24047. [PMID: 34487611 DOI: 10.1002/anie.202110041] [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: 07/27/2021] [Revised: 08/26/2021] [Indexed: 01/06/2023]
Abstract
Pharmacological inactivation of antitumor drugs toward healthy cells is a critical factor in prodrug development. Typically, pharmaceutical chemists graft temporary moieties to existing antitumor drugs to reduce their pharmacological activity. Here, we report a platform able to generate the cytotoxic agent by intramolecular cyclization. Using phenanthridines as cytotoxic model compounds, we designed ring-opened biaryl precursors that generated the phenanthridines through bioorthogonal irreversible imination. This reaction was triggered by reactive oxygen species, commonly overproduced in cancer cells, able to convert a vinyl boronate ester function into a ketone that subsequently reacted with a pendant aniline. An inactive precursor was shown to engender a cytotoxic phenanthridine against KB cancer cells. Moreover, the kinetic of cyclization of this prodrug was extremely rapid inside living cells of KB cancer spheroids so as to circumvent drug action.
Collapse
Affiliation(s)
- Hichem Maslah
- Université Paris-Saclay, CNRS, Institut de chimie moléculaire et des matériaux d'Orsay, 91405, Orsay, France
| | - Charles Skarbek
- Université Paris-Saclay, CNRS, Institut de chimie moléculaire et des matériaux d'Orsay, 91405, Orsay, France
| | - Catherine Gourson
- Université Paris-Saclay, CNRS, Institut de chimie moléculaire et des matériaux d'Orsay, 91405, Orsay, France
| | - Marie-Aude Plamont
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, 24, rue Lhomond, 75005, Paris, France
| | - Stéphanie Pethe
- Université Paris-Saclay, CNRS, Institut de chimie moléculaire et des matériaux d'Orsay, 91405, Orsay, France
| | - Ludovic Jullien
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, 24, rue Lhomond, 75005, Paris, France
| | - Thomas Le Saux
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, 24, rue Lhomond, 75005, Paris, France
| | - Raphaël Labruère
- Université Paris-Saclay, CNRS, Institut de chimie moléculaire et des matériaux d'Orsay, 91405, Orsay, France
| |
Collapse
|
17
|
van der Westhuizen D, Bezuidenhout DI, Munro OQ. Cancer molecular biology and strategies for the design of cytotoxic gold(I) and gold(III) complexes: a tutorial review. Dalton Trans 2021; 50:17413-17437. [PMID: 34693422 DOI: 10.1039/d1dt02783b] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This tutorial review highlights key principles underpinning the design of selected metallodrugs to target specific biological macromolecules (DNA and proteins). The review commences with a descriptive overview of the eukaryotic cell cycle and the molecular biology of cancer, particularly apoptosis, which is provided as a necessary foundation for the discovery, design, and targeting of metal-based anticancer agents. Drugs which target DNA have been highlighted and clinically approved metallodrugs discussed. A brief history of the development of mainly gold-based metallodrugs is presented prior to addressing ligand systems for stabilizing and adding functionality to bio-active gold(I) and gold(III) complexes, particularly in the burgeoning field of anticancer metallodrugs. Concepts such as multi-modal and selective cytotoxic agents are covered where necessary for selected compounds. The emerging role of carbenes as the ligand system of choice to achieve these goals for gold-based metallodrug candidates is highlighted prior to closing the review with comments on some future directions that this research field might follow. The latter section ultimately emphasizes the importance of understanding the fate of metal complexes in cells to garner key mechanistic insights.
Collapse
Affiliation(s)
- Danielle van der Westhuizen
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, Johannesburg 2050, South Africa.
| | - Daniela I Bezuidenhout
- Laboratory of Inorganic Chemistry, Environmental and Chemical Engineering, University of Oulu, P. O. Box 3000, 90014 Oulu, Finland.
| | - Orde Q Munro
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, Johannesburg 2050, South Africa.
| |
Collapse
|
18
|
Maslah H, Skarbek C, Gourson C, Plamont M, Pethe S, Jullien L, Le Saux T, Labruère R. In‐Cell Generation of Anticancer Phenanthridine Through Bioorthogonal Cyclization in Antitumor Prodrug Development. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202110041] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Hichem Maslah
- Université Paris-Saclay CNRS Institut de chimie moléculaire et des matériaux d'Orsay 91405 Orsay France
| | - Charles Skarbek
- Université Paris-Saclay CNRS Institut de chimie moléculaire et des matériaux d'Orsay 91405 Orsay France
| | - Catherine Gourson
- Université Paris-Saclay CNRS Institut de chimie moléculaire et des matériaux d'Orsay 91405 Orsay France
| | - Marie‐Aude Plamont
- PASTEUR Département de chimie École normale supérieure PSL University Sorbonne Université CNRS 24, rue Lhomond 75005 Paris France
| | - Stéphanie Pethe
- Université Paris-Saclay CNRS Institut de chimie moléculaire et des matériaux d'Orsay 91405 Orsay France
| | - Ludovic Jullien
- PASTEUR Département de chimie École normale supérieure PSL University Sorbonne Université CNRS 24, rue Lhomond 75005 Paris France
| | - Thomas Le Saux
- PASTEUR Département de chimie École normale supérieure PSL University Sorbonne Université CNRS 24, rue Lhomond 75005 Paris France
| | - Raphaël Labruère
- Université Paris-Saclay CNRS Institut de chimie moléculaire et des matériaux d'Orsay 91405 Orsay France
| |
Collapse
|
19
|
Humer D, Spadiut O. Enzyme prodrug therapy: cytotoxic potential of paracetamol turnover with recombinant horseradish peroxidase. MONATSHEFTE FUR CHEMIE 2021; 152:1389-1397. [PMID: 34759433 PMCID: PMC8542555 DOI: 10.1007/s00706-021-02848-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 09/06/2021] [Indexed: 11/05/2022]
Abstract
Targeted cancer treatment is a promising, less invasive alternative to chemotherapy as it is precisely directed against tumor cells whilst leaving healthy tissue unaffected. The plant-derived enzyme horseradish peroxidase (HRP) can be used for enzyme prodrug cancer therapy with indole-3-acetic acid or the analgesic paracetamol (acetaminophen). Oxidation of paracetamol by HRP in the presence of hydrogen peroxide leads to N-acetyl-p-benzoquinone imine and polymer formation via a radical reaction mechanism. N-acetyl-p-benzoquinone imine binds to DNA and proteins, resulting in severe cytotoxicity. However, plant HRP is not suitable for this application since the foreign glycosylation pattern is recognized by the human immune system, causing rapid clearance from the body. Furthermore, plant-derived HRP is a mixture of isoenzymes with a heterogeneous composition. Here, we investigated the reaction of paracetamol with defined recombinant HRP variants produced in E. coli, as well as plant HRP, and found that they are equally effective in paracetamol oxidation at a concentration ≥ 400 µM. At low paracetamol concentrations, however, recombinant HRP seems to be more efficient in paracetamol oxidation. Yet upon treatment of HCT-116 colon carcinoma and FaDu squamous carcinoma cells with HRP-paracetamol no cytotoxic effect was observed, neither in the presence nor absence of hydrogen peroxide. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s00706-021-02848-x.
Collapse
Affiliation(s)
- Diana Humer
- TU Wien, Institute of Chemical, Environmental and Bioscience Engineering, Research Area Biochemical Engineering, Gumpendorfer Straße 1a, 1060 Vienna, Austria
| | - Oliver Spadiut
- TU Wien, Institute of Chemical, Environmental and Bioscience Engineering, Research Area Biochemical Engineering, Gumpendorfer Straße 1a, 1060 Vienna, Austria
| |
Collapse
|
20
|
Chis AA, Dobrea CM, Rus LL, Frum A, Morgovan C, Butuca A, Totan M, Juncan AM, Gligor FG, Arseniu AM. Dendrimers as Non-Viral Vectors in Gene-Directed Enzyme Prodrug Therapy. Molecules 2021; 26:5976. [PMID: 34641519 PMCID: PMC8512881 DOI: 10.3390/molecules26195976] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/19/2021] [Accepted: 09/29/2021] [Indexed: 01/02/2023] Open
Abstract
Gene-directed enzyme prodrug therapy (GDEPT) has been intensively studied as a promising new strategy of prodrug delivery, with its main advantages being represented by an enhanced efficacy and a reduced off-target toxicity of the active drug. In recent years, numerous therapeutic systems based on GDEPT strategy have entered clinical trials. In order to deliver the desired gene at a specific site of action, this therapeutic approach uses vectors divided in two major categories, viral vectors and non-viral vectors, with the latter being represented by chemical delivery agents. There is considerable interest in the development of non-viral vectors due to their decreased immunogenicity, higher specificity, ease of synthesis and greater flexibility for subsequent modulations. Dendrimers used as delivery vehicles offer many advantages, such as: nanoscale size, precise molecular weight, increased solubility, high load capacity, high bioavailability and low immunogenicity. The aim of the present work was to provide a comprehensive overview of the recent advances regarding the use of dendrimers as non-viral carriers in the GDEPT therapy.
Collapse
Affiliation(s)
| | | | | | - Adina Frum
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (A.A.C.); (C.M.D.); (L.-L.R.); (A.B.); (M.T.); (A.M.J.); (F.G.G.); (A.M.A.)
| | - Claudiu Morgovan
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (A.A.C.); (C.M.D.); (L.-L.R.); (A.B.); (M.T.); (A.M.J.); (F.G.G.); (A.M.A.)
| | | | | | | | | | | |
Collapse
|
21
|
Li H, Lee CH, Shin I. A triple-targeting delivery system carrying two anticancer agents. Org Biomol Chem 2021; 19:8009-8013. [PMID: 34236071 DOI: 10.1039/d1ob01089a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
To improve tumor selectivity, a triple-targeting delivery system (Oct-FK(PBA-Az)-Dox) carrying two anticancer agents (apoptozole (Az) and doxorubicin (Dox)) was designed and synthesized. The results showed that both anticancer agents in Oct-FK(PBA-Az)-Dox are liberated in the presence of both H2O2 and cathepsin B, which are normally present at high levels in tumors.
Collapse
Affiliation(s)
- Hui Li
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea.
| | - Chang-Hee Lee
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea.
| | - Injae Shin
- Department of Chemistry, Yonsei University, Seoul 03722, Republic of Korea.
| |
Collapse
|
22
|
Alemayehu YA, Ilhami FB, Manayia AH, Cheng CC. Mercury-containing supramolecular micelles with highly sensitive pH-responsiveness for selective cancer therapy. Acta Biomater 2021; 129:235-244. [PMID: 34087441 DOI: 10.1016/j.actbio.2021.05.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 02/07/2023]
Abstract
Construction and manipulation of metal-based supramolecular polymers-which are based on a combination of nucleobase hydrogen bonding interactions and functional metal ions-to obtain the desired physicochemical properties and achieve the efficacy and safety required for biomedical applications remain extremely challenging. We successfully designed and synthesized a new mercury-based supramolecular polymer, Hg-BU-PPG, containing an oligomeric polypropylene glycol backbone and pH-sensitive uracil-mercury-uracil (U-Hg-U) linkages. This multifunctional metallo-supramolecular material spontaneously self-organizes into nanosized spherical micelles in aqueous solution. The micelles possess several attractive properties, including desired long-term structural stability in serum-rich conditions, unique fluorescence behavior and highly sensitive, well-controlled pH-responsiveness. Interestingly, Hg-BU-PPG micelles exhibited strong, selective cytotoxic effects towards cancer cells in vitro, without harming normal cells. The highly selective cytotoxicity can be attributed to rapid dissociation of the U-Hg-U complexes within the micelles in the mildly acidic intracellular pH of cancer cells, followed by release of inherently toxic mercury ions. Importantly, fluorescence microscopy and flow cytometry clearly demonstrated that Hg-BU-PPG selectively entered the cancer cells via endocytosis and rapidly promoted massive apoptotic cell death. In contrast, internalization of Hg-BU-PPG by normal cells was limited, resulting in high biocompatibility and no cytotoxic effects. Thus, this newly discovered 'cytotoxicity-concealing' supramolecular system could represent a viable route to enhance the safety and efficacy of cancer therapy and bioimaging via a strategy that does not require incorporation of anticancer drugs and fluorescent probes. STATEMENT OF SIGNIFICANCE: We report a significant breakthrough in the construction of mercury-containing supramolecular polymers, namely the creation of multifunctional micelles with unique chemical and physical properties conferred by pH-sensitive uracil-mercury-uracil (U-Hg-U) linkages and tunable structural and dynamical features due to the presence of hydrogen-bonded uracil moieties. Importantly, in vitro experiments clearly demonstrated that introduction of the U-Hg-U complexes into the micelles not only improved the efficiency of selective uptake via endocytosis into cancer cells, but also accelerated the induction of massive apoptotic cell death. Thus, this work provides crucial new insight for the development of metallo-supramolecular polymeric micelles that may substantially enhance the safety and efficacy of cancer therapy and bioimaging without requiring incorporation of anticancer drugs or fluorescent probes.
Collapse
|
23
|
Ashique S, Sandhu NK, Chawla V, Chawla PA. Targeted Drug Delivery: Trends and Perspectives. Curr Drug Deliv 2021; 18:1435-1455. [PMID: 34151759 DOI: 10.2174/1567201818666210609161301] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 03/03/2021] [Accepted: 04/07/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Due to various limitations in conventional drug delivery system, it is important to focus on the target-specific drug delivery system where we can deliver the drug without any degradation. Among various challenges faced by a formulation scientist, delivering the drug to its right site, in its right dose, is also an important aim. A focused drug transport aims to extend, localize, target and have a safe drug interaction with the diseased tissue. OBJECTIVE The aim of targeted drug delivery is to make the required amount of the drug available at its desired site of action. Drug targeting can be accomplished in a number ways that include enzyme mediation, pH-dependent release, use of special vehicles, receptor targeting among other mechanisms. Intelligently designed targeted drug delivery systems also offer the advantages of a low dose of the drug along with reduced side effects which ultimately improves patient compliance. Incidences of dose dumping and dosage form failure are negligible. A focused drug transport aims to have a safe drug interaction with the diseased tissue. CONCLUSION This review focuses on the available targeting techniques for delivery to the colon, brain and other sites of interest. Overall, the article should make an excellent read for the researchers in this area. Newer drug targets may be identified and exploited for successful drug targeting.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutics, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Navjot Kaur Sandhu
- Department of Pharmaceutical Analysis, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Viney Chawla
- University Institute of Pharmaceutical Sciences and Research, Baba Farid University of Health Sciences, Faridkot, Punjab, India
| | - Pooja A Chawla
- Department of Pharmaceutical Analysis, ISF College of Pharmacy, Moga-142001, Punjab, India
| |
Collapse
|
24
|
Bulanadi JC, Xue A, Gong X, Bean PA, Julovi SM, de Campo L, Smith RC, Moghaddam MJ. Biomimetic Gemcitabine-Lipid Prodrug Nanoparticles for Pancreatic Cancer. Chempluschem 2021; 85:1283-1291. [PMID: 32543086 DOI: 10.1002/cplu.202000253] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/15/2020] [Indexed: 12/18/2022]
Abstract
Gemcitabine (Gem) is a key drug for pancreatic cancer, yet limited by high systemic toxicity, low bioavailability and poor pharmacokinetic profiles. To overcome these limitations, Gem prodrug amphiphiles were synthesised with oleyl, linoleyl and phytanyl chains. Self-assembly and lyotropic mesophase behaviour of these amphiphiles were examined using polarised optical microscopy and Synchrotron SAXS (SSAXS). Gem-phytanyl was found to form liquid crystalline inverse cubic mesophase. This prodrug was combined with phospholipids and cholesterol to create biomimetic Gem-lipid prodrug nanoparticles (Gem-LPNP), verified by SSAXS and cryo-TEM to form liposomes. In vitro testing of the Gem-LPNP in several pancreatic cancer cell lines showed lower toxicity than Gem. However, in a cell line-derived pancreatic cancer mouse model Gem-LPNP displayed greater tumour growth inhibition than Gem using a fraction (<6 %) of the clinical dose and without any systemic toxicity. The easy production, improved efficacy and low toxicity of Gem-LPNP represents a promising new nanomedicine for pancreatic cancer.
Collapse
Affiliation(s)
- Jerikho C Bulanadi
- CSIRO Manufacturing, P.O. BOX 52, North Ryde, NSW, 1670, Australia.,Cancer Surgery and Metabolism Group, University of Sydney, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia
| | - Aiqun Xue
- Cancer Surgery and Metabolism Group, University of Sydney, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia
| | - Xiaojuan Gong
- CSIRO Manufacturing, P.O. BOX 52, North Ryde, NSW, 1670, Australia.,NanoMed Pty Ltd., 2/11-13 Orion Road, Lane Cove West, NSW, 2066, Australia
| | - Penelope A Bean
- CSIRO Manufacturing, P.O. BOX 52, North Ryde, NSW, 1670, Australia.,NanoMed Pty Ltd., 2/11-13 Orion Road, Lane Cove West, NSW, 2066, Australia
| | - Sohel M Julovi
- Cancer Surgery and Metabolism Group, University of Sydney, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia
| | | | - Ross C Smith
- Cancer Surgery and Metabolism Group, University of Sydney, Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia.,NanoMed Pty Ltd., 2/11-13 Orion Road, Lane Cove West, NSW, 2066, Australia
| | - Minoo J Moghaddam
- CSIRO Manufacturing, P.O. BOX 52, North Ryde, NSW, 1670, Australia.,NanoMed Pty Ltd., 2/11-13 Orion Road, Lane Cove West, NSW, 2066, Australia
| |
Collapse
|
25
|
Baroud M, Lepeltier E, Thepot S, El-Makhour Y, Duval O. The evolution of nucleosidic analogues: self-assembly of prodrugs into nanoparticles for cancer drug delivery. NANOSCALE ADVANCES 2021; 3:2157-2179. [PMID: 36133769 PMCID: PMC9418958 DOI: 10.1039/d0na01084g] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/20/2021] [Indexed: 05/12/2023]
Abstract
Nucleoside and nucleotide analogs are essential tools in our limited arsenal in the fight against cancer. However, these structures face severe drawbacks such as rapid plasma degradation or hydrophilicity, limiting their clinical application. Here, different aspects of nucleoside and nucleotide analogs have been exposed, while providing their shortcomings. Aiming to improve their fate in the body and combating their drawbacks, two different approaches have been discussed, the prodrug and nanocarrier technologies. Finally, a novel approach called "PUFAylation" based on both the prodrug and nanocarrier technologies has been introduced, promising to be the supreme method to create a novel nucleoside or nucleotide analog based formulation, with enhanced efficacy and highly reduced toxicity.
Collapse
Affiliation(s)
- Milad Baroud
- Micro et Nanomédecines Translationnelles, MINT, UNIV Angers, UMR INSERM 1066, UMR CNRS 6021 Angers France
| | - Elise Lepeltier
- Micro et Nanomédecines Translationnelles, MINT, UNIV Angers, UMR INSERM 1066, UMR CNRS 6021 Angers France
| | - Sylvain Thepot
- University Hospital of Angers, Hematology 49933 Angers France
- Université d'Angers, Inserm, CRCINA 49000 Angers France
- Fédération Hospitalo-Universitaire 'Grand Ouest Against Leukemia' (FHU GOAL) France
| | - Yolla El-Makhour
- Environmental Health Research Lab (EHRL), Faculty of Sciences V, Lebanese University Nabatieh Lebanon
| | - Olivier Duval
- Micro et Nanomédecines Translationnelles, MINT, UNIV Angers, UMR INSERM 1066, UMR CNRS 6021 Angers France
- University Hospital of Angers, Hematology 49933 Angers France
| |
Collapse
|
26
|
Xu ZY, Liu HK, Wu Y, Zhang YC, Zhou W, Wang H, Zhang DW, Ma D, Li ZT. Flexible Organic Framework-Based Anthracycline Prodrugs for Enhanced Tumor Growth Inhibition. ACS APPLIED BIO MATERIALS 2021; 4:4591-4597. [DOI: 10.1021/acsabm.1c00316] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Zi-Yue Xu
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200438, China
| | - Hong-Kun Liu
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200438, China
| | - Yan Wu
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200438, China
| | - Yun-Chang Zhang
- Department of Inorganic Chemistry, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Wei Zhou
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200438, China
| | - Hui Wang
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200438, China
| | - Dan-Wei Zhang
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200438, China
| | - Da Ma
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200438, China
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| | - Zhan-Ting Li
- Department of Chemistry, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200438, China
| |
Collapse
|
27
|
Wang K, Shang F, Chen D, Cao T, Wang X, Jiao J, He S, Liang X. Protein liposomes-mediated targeted acetylcholinesterase gene delivery for effective liver cancer therapy. J Nanobiotechnology 2021; 19:31. [PMID: 33482834 PMCID: PMC7821407 DOI: 10.1186/s12951-021-00777-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 01/13/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Effective methods to deliver therapeutic genes to solid tumors and improve their bioavailability are the main challenges of current medical research on gene therapy. The development of efficient non-viral gene vector with tumor-targeting has very important application value in the field of cancer therapy. Proteolipid integrated with tumor-targeting potential of functional protein and excellent gene delivery performance has shown potential for targeted gene therapy. RESULTS Herein, we prepared transferrin-modified liposomes (Tf-PL) for the targeted delivery of acetylcholinesterase (AChE) therapeutic gene to liver cancer. We found that the derived Tf-PL/AChE liposomes exhibited much higher transfection efficiency than the commercial product Lipo 2000 and shown premium targeting efficacy to liver cancer SMMC-7721 cells in vitro. In vivo, the Tf-PL/AChE could effectively target liver cancer, and significantly inhibit the growth of liver cancer xenografts grafted in nude mice by subcutaneous administration. CONCLUSIONS This study proposed a transferrin-modified proteolipid-mediated gene delivery strategy for targeted liver cancer treatment, which has a promising potential for precise personalized cancer therapy.
Collapse
Affiliation(s)
- Kai Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, 200032, People's Republic of China
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Fusheng Shang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, People's Republic of China
| | - Dagui Chen
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, People's Republic of China
| | - Tieliu Cao
- Department of Hepatobiliary-pancreatic and Integrative Oncology, Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, 200240, People's Republic of China
| | - Xiaowei Wang
- Department of traditional Chinese medicine, Changzheng Hospital, Shanghai, 200001, People's Republic of China
| | - Jianpeng Jiao
- Department of traditional Chinese medicine, Changzheng Hospital, Shanghai, 200001, People's Republic of China
| | - Shengli He
- Department of Hepatobiliary-pancreatic and Integrative Oncology, Minhang Branch, Fudan University Shanghai Cancer Center, Shanghai, 200240, People's Republic of China.
| | - Xiaofei Liang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
28
|
Metcalf KJ, Kimmel BR, Sykora DJ, Modica JA, Parker KA, Berens E, Dai R, Dravid VP, Werb Z, Mrksich M. Synthetic Tuning of Domain Stoichiometry in Nanobody-Enzyme Megamolecules. Bioconjug Chem 2020; 32:143-152. [PMID: 33301672 DOI: 10.1021/acs.bioconjchem.0c00578] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This paper presents a method to synthetically tune atomically precise megamolecule nanobody-enzyme conjugates for prodrug cancer therapy. Previous efforts to create heterobifunctional protein conjugates suffered from heterogeneity in domain stoichiometry, which in part led to the failure of antibody-enzyme conjugates in clinical trials. We used the megamolecule approach to synthesize anti-HER2 nanobody-cytosine deaminase conjugates with tunable numbers of nanobody and enzyme domains in a single, covalent molecule. Linking two nanobody domains to one enzyme domain improved avidity to a human cancer cell line by 4-fold but did not increase cytotoxicity significantly due to lowered enzyme activity. In contrast, a megamolecule composed of one nanobody and two enzyme domains resulted in an 8-fold improvement in the catalytic efficiency and increased the cytotoxic effect by over 5-fold in spheroid culture, indicating that the multimeric structure allowed for an increase in local drug activation. Our work demonstrates that the megamolecule strategy can be used to study structure-function relationships of protein conjugate therapeutics with synthetic control of protein domain stoichiometry.
Collapse
Affiliation(s)
- Kevin J Metcalf
- Department of Anatomy, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, California 94143, United States.,Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Blaise R Kimmel
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Daniel J Sykora
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Justin A Modica
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Kelly A Parker
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Eric Berens
- Department of Anatomy, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, California 94143, United States
| | - Raymond Dai
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Vinayak P Dravid
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Zena Werb
- Department of Anatomy, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, California 94143, United States
| | - Milan Mrksich
- Department of Biomedical Engineering, Northwestern University, Evanston, Illinois 60208, United States.,Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|
29
|
Torrens-Mas M, Roca P. Phytoestrogens for Cancer Prevention and Treatment. BIOLOGY 2020; 9:E427. [PMID: 33261116 PMCID: PMC7759898 DOI: 10.3390/biology9120427] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/21/2020] [Accepted: 11/25/2020] [Indexed: 12/17/2022]
Abstract
Phytoestrogens are a large group of natural compounds found in more than 300 plants. They have a close structural similarity to estrogens, which allow them to bind to both estrogen receptors (ER), ERα and ERβ, presenting a weak estrogenic activity. Phytoestrogens have been described as antioxidant, anti-inflammatory, anti-thrombotic, anti-allergic, and anti-tumoral agents. Their role in cancer prevention has been well documented, although their impact on treatment efficiency is controversial. Several reports suggest that phytoestrogens may interfere with the effect of anti-cancer drugs through the regulation of oxidative stress and other mechanisms. Furthermore, some phytoestrogens could exert a protective effect on healthy cells, thus reducing the secondary effects of cancer treatment. In this review, we have studied the recent research in this area to find evidence for the role of phytoestrogens in cancer prevention and therapy efficacy.
Collapse
Affiliation(s)
- Margalida Torrens-Mas
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut, Universitat de les Illes Balears, 07122 Palma, Spain;
- Instituto de Investigación Sanitaria Illes Balears, 07010 Palma, Spain
| | - Pilar Roca
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut, Universitat de les Illes Balears, 07122 Palma, Spain;
- Instituto de Investigación Sanitaria Illes Balears, 07010 Palma, Spain
- Ciber Fisiopatología Obesidad y Nutrición (CB06/03), Instituto Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
30
|
Mansoori B, Mohammadi A, Abedi-Gaballu F, Abbaspour S, Ghasabi M, Yekta R, Shirjang S, Dehghan G, Hamblin MR, Baradaran B. Hyaluronic acid-decorated liposomal nanoparticles for targeted delivery of 5-fluorouracil into HT-29 colorectal cancer cells. J Cell Physiol 2020; 235:6817-6830. [PMID: 31989649 PMCID: PMC7384933 DOI: 10.1002/jcp.29576] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 01/10/2020] [Indexed: 12/14/2022]
Abstract
The use of liposomes as drug carriers improves the therapeutic efficacy of anticancer drugs, while at the same time reducing side effects. Hyaluronic acid (HA) is recognized by the CD44 receptor, which is overexpressed in many cancer cells. In this study, we developed HA-modified liposomes encapsulating 5-fluorouracil (5-FU) and tested them against a CD44 expressing colorectal cell line (HT29) and a non-CD44 expressing hepatoma cell line. The average size of 5-FU-lipo and 5-FU-lipo-HA nanoparticles were 112 ± 28 and 144 ± 77 nm, respectively. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT) assay showed selective cancer cell death depending on the CD44 expression in a time-dependent manner. Apoptosis assays and cell-cycle analysis indicated that G0/G1 arrest occurred. The colony formation study revealed that cells treated with 5-FU-lipo and 5-FU-lipo-HA had reduced colony formation. Quantitative reverse-transcription polymerase chain reaction study showed that the oncogenic messenger RNA and microRNA levels were significantly reduced in the 5-FU-lipo-HA-treated group, while tumor suppressors were increased in that group. We suggest that optimal targeted delivery and release of 5-FU into colorectal cancer cells, renders them susceptible to apoptosis, cell-cycle arrest, and decreased colony formation.
Collapse
Affiliation(s)
- Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Fereydoon Abedi-Gaballu
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Soheil Abbaspour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehri Ghasabi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Yekta
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Solmaz Shirjang
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gholamreza Dehghan
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Michael R. Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
31
|
Molecular targeted therapy: novel therapeutic approach for head and neck cancer. Ther Deliv 2020; 11:637-651. [DOI: 10.4155/tde-2020-0028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cancer is a major public health burden worldwide, affecting millions of people each year. One of the major hallmarks of cancer is rapid growth and progression by evasion of host immune responses. Tumor resistance to conventional anticancer drugs by several mechanisms, such as drug inactivation, efflux pumps and enhanced toxicity to normal cells decreases their clinical efficacy. These limitations resulted in the development of new targeted agents, such as monoclonal antibodies and small molecule inhibitors that have high tumor specificity. This paper discusses the therapeutic applications of novel molecular targeted agents and immunotherapy as an alternative treatment option for head and neck cancers, as well as provides insight into future therapeutic approaches for advanced head and neck cancers.
Collapse
|
32
|
Zhou W. Cholic Acid-Functionalized Mesoporous Silica Nanoparticles Loaded With Ruthenium Pro-drug Delivery to Cervical Cancer Therapy. J Inorg Organomet Polym Mater 2020. [DOI: 10.1007/s10904-020-01710-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
33
|
Herrlinger E, Hau M, Redhaber DM, Greve G, Willmann D, Steimle S, Müller M, Lübbert M, Miething CC, Schüle R, Jung M. Nitroreductase-Mediated Release of Inhibitors of Lysine-Specific Demethylase 1 (LSD1) from Prodrugs in Transfected Acute Myeloid Leukaemia Cells. Chembiochem 2020; 21:2329-2347. [PMID: 32227662 PMCID: PMC7497180 DOI: 10.1002/cbic.202000138] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/29/2020] [Indexed: 12/14/2022]
Abstract
Lysine-specific demethylase 1 (LSD1) has evolved as a promising therapeutic target for cancer treatment, especially in acute myeloid leukaemia (AML). To approach the challenge of site-specific LSD1 inhibition, we developed an enzyme-prodrug system with the bacterial nitroreductase NfsB (NTR) that was expressed in the virally transfected AML cell line THP1-NTR+ . The cellular activity of the NTR was proven with a new luminescent NTR probe. We synthesised a diverse set of nitroaromatic prodrugs that by design do not affect LSD1 and are reduced by the NTR to release an active LSD1 inhibitor. The emerging side products were differentially analysed using negative controls, thereby revealing cytotoxic effects. The 2-nitroimidazolyl prodrug of a potent LSD1 inhibitor emerged as one of the best prodrug candidates with a pronounced selectivity window between wild-type and transfected THP1 cells. Our prodrugs are selectively activated and release the LSD1 inhibitor locally, proving their suitability for future targeting approaches.
Collapse
Affiliation(s)
- Eva‐Maria Herrlinger
- Department of Chemistry and Pharmacy, University of FreiburgInstitute of Pharmaceutical SciencesAlbertstrasse 2579104FreiburgGermany
| | - Mirjam Hau
- Department of Chemistry and Pharmacy, University of FreiburgInstitute of Pharmaceutical SciencesAlbertstrasse 2579104FreiburgGermany
- CIBSS – Centre for Integrative Biological Signalling StudiesUniversity of FreiburgSchänzlestrasse 1879104FreiburgGermany
| | - Desiree Melanie Redhaber
- Division of Hematology, Oncology and Stem Cell TransplantationUniversity of Freiburg Medical CenterHugstetter Strasse 5579106FreiburgGermany
| | - Gabriele Greve
- Division of Hematology, Oncology and Stem Cell TransplantationUniversity of Freiburg Medical CenterHugstetter Strasse 5579106FreiburgGermany
| | - Dominica Willmann
- Department of Urology and Center for Clinical ResearchUniversity of Freiburg Medical CenterBreisacher Strasse 6679106FreiburgGermany
| | - Simon Steimle
- Department of Chemistry and Pharmacy, University of FreiburgInstitute of Pharmaceutical SciencesAlbertstrasse 2579104FreiburgGermany
| | - Michael Müller
- Department of Chemistry and Pharmacy, University of FreiburgInstitute of Pharmaceutical SciencesAlbertstrasse 2579104FreiburgGermany
| | - Michael Lübbert
- Division of Hematology, Oncology and Stem Cell TransplantationUniversity of Freiburg Medical CenterHugstetter Strasse 5579106FreiburgGermany
- German Cancer Consortium (DKTK)FreiburgGermany
- German Cancer Research Center (DKFZ)
| | - Christoph Cornelius Miething
- Division of Hematology, Oncology and Stem Cell TransplantationUniversity of Freiburg Medical CenterHugstetter Strasse 5579106FreiburgGermany
| | - Roland Schüle
- CIBSS – Centre for Integrative Biological Signalling StudiesUniversity of FreiburgSchänzlestrasse 1879104FreiburgGermany
- Department of Urology and Center for Clinical ResearchUniversity of Freiburg Medical CenterBreisacher Strasse 6679106FreiburgGermany
| | - Manfred Jung
- Department of Chemistry and Pharmacy, University of FreiburgInstitute of Pharmaceutical SciencesAlbertstrasse 2579104FreiburgGermany
- CIBSS – Centre for Integrative Biological Signalling StudiesUniversity of FreiburgSchänzlestrasse 1879104FreiburgGermany
- German Cancer Consortium (DKTK)FreiburgGermany
- German Cancer Research Center (DKFZ)
| |
Collapse
|
34
|
Li Q, Li X, Zhao C. Strategies to Obtain Encapsulation and Controlled Release of Small Hydrophilic Molecules. Front Bioeng Biotechnol 2020; 8:437. [PMID: 32478055 PMCID: PMC7237580 DOI: 10.3389/fbioe.2020.00437] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 04/16/2020] [Indexed: 12/03/2022] Open
Abstract
The therapeutic effect of small hydrophilic molecules is limited by the rapid clearance from the systemic circulation or a local site of administration. The unsuitable pharmacokinetics and biodistribution can be improved by encapsulating them in drug delivery systems. However, the high-water solubility, very hydrophilic nature, and low molecular weight make it difficult to encapsulate small hydrophilic molecules in many drug delivery systems. In this mini-review, we highlight three strategies to efficiently encapsulate small hydrophilic molecules and achieve controlled release: physical encapsulation in micro/nanocapsules, physical adsorption via electronic interactions, and covalent conjugation. The principles, advantages, and disadvantages of each strategy are discussed. This review paper could be a guide for scientists, engineers, and medical doctors who want to improve the therapeutic efficacy of small hydrophilic drugs.
Collapse
Affiliation(s)
| | | | - Chao Zhao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, United States
| |
Collapse
|
35
|
Poreba M. Recent advances in the development of legumain-selective chemical probes and peptide prodrugs. Biol Chem 2020; 400:1529-1550. [PMID: 31021817 DOI: 10.1515/hsz-2019-0135] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 04/18/2019] [Indexed: 12/13/2022]
Abstract
Legumain, which is also known as vacuolar processing enzyme (VPE) or asparaginyl endopeptidase (AEP), is a cysteine protease that was first discovered and characterized in the leguminous seeds of the moth bean in the early 1990s. Later, this enzyme was also detected in higher organisms, including eukaryotes. This pH-dependent protease displays the highest activity in acidic endolysosomal compartments; however, legumain also displays nuclear, cytosolic and extracellular activity when stabilized by other proteins or intramolecular complexes. Based on the results from over 25 years of research, this protease is involved in multiple cellular events, including protein degradation and antigen presentation. Moreover, when dysregulated, this protease contributes to the progression of several diseases, with cancer being the well-studied example. Research on legumain biology was undoubtedly facilitated by the use of small molecule chemical tools. Therefore, in this review, I present the historical perspectives and most current strategies for the development of small molecule substrates, inhibitors and activity-based probes for legumain. These tools are of paramount importance in elucidating the roles of legumain in multiple biological processes. Finally, as this enzyme appears to be a promising molecular target for anticancer therapies, the development of legumain-activated prodrugs is also described.
Collapse
Affiliation(s)
- Marcin Poreba
- Department of Bioorganic Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370, Wroclaw, Poland
| |
Collapse
|
36
|
Mansour M, Ismail S, Abou-Aisha K. Bacterial delivery of the anti-tumor azurin-like protein Laz to glioblastoma cells. AMB Express 2020; 10:59. [PMID: 32221741 PMCID: PMC7099546 DOI: 10.1186/s13568-020-00995-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 03/18/2020] [Indexed: 02/02/2023] Open
Abstract
Salmonella typhimurium VNP-20009 (VNP) is a non-pathogenic attenuated strain, which, as a facultative anaerobe, preferentially accumulates in hypoxic regions of solid tumors. Here, VNP was utilized as a delivery vehicle of the anti-tumor protein Lipidated azurin, Laz, which is produced by the meningitis-causing bacterium Neisseria meningitides. In brain cancer cells, Laz has been demonstrated to induce apoptosis through an interaction with the tumor suppressor protein p53. In this study, the laz gene, including its signal sequence, was cloned downstream of a hypoxia inducible promoter (HIP-1), before being electroporated into VNP. Successful ectopic expression and export of the Laz protein by VNP under hypoxic conditions were confirmed by Western blot analysis of the cell-free culture medium. Effective expression of Laz by VNP was investigated in two glioblastoma cell lines: LN-229 and U-373, with the latter line carrying a mutated version of p53; as well as in the breast cancer line MCF-7. Cytotoxicity of the VNP-Laz was assessed by determining the fluorescence of the apoptotic marker caspases 3/7. Compared to the purified Laz, VNP-Laz, significantly induced apoptosis in MCF-7, LN-229 and, to a much lower extent in U-373 cells, suggesting a p53-linked mechanism. Our results might represent a new approach of targeted gene delivery and suggest a potential application in brain tumor therapy.
Collapse
Affiliation(s)
- Manar Mansour
- Department of Microbiology and Immunology, The German University in Cairo (GUC), Main Entrance Fifth Settlement, Cairo, Egypt.
| | - Shehab Ismail
- The Cancer Research Institute CRUK Beatson Institute, Glasgow, UK
| | - Khaled Abou-Aisha
- Department of Microbiology and Immunology, The German University in Cairo (GUC), Main Entrance Fifth Settlement, Cairo, Egypt
| |
Collapse
|
37
|
Ito K, Tatsumi T, Takahashi K, Shimizu Y, Yamatsugu K, Kanai M. A Stable and Cleavable O-Linked Spacer for Drug Delivery Systems. Chem Pharm Bull (Tokyo) 2020; 68:212-215. [PMID: 31189762 DOI: 10.1248/cpb.c19-00376] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Anti-cancer chemotherapy with good efficacy and fewer side effects is highly desirable. A drug delivery system comprising a cancer-targeting module and a cytotoxic agent connected with a cleavable linker is promising for reducing side effects. The development of a cleavable linker satisfying the requirements of both stability and cleavability, however, is difficult, especially when a carbonate moiety is used for conjugating the linker to a hydroxy group in a drug of interest. We herein report a new stable linker comprising carbamate and ester spacers, which can be introduced on a hydroxy group of a drug. This linker is more stable in aqueous neutral buffer than a corresponding carbonate-type linker, and releases a payload anti-cancer drug, SN-38, through a two-step sequence upon cathepsin B treatment. This linker may have potential use in other drug delivery systems to lower side effects by selectively transporting cytotoxic drugs to tumor cells.
Collapse
Affiliation(s)
- Kei Ito
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | | | - Kazuki Takahashi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Yohei Shimizu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Kenzo Yamatsugu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| | - Motomu Kanai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo
| |
Collapse
|
38
|
Hellinen L, Sato K, Reinisalo M, Kidron H, Rilla K, Tachikawa M, Uchida Y, Terasaki T, Urtti A. Quantitative Protein Expression in the Human Retinal Pigment Epithelium: Comparison Between Apical and Basolateral Plasma Membranes With Emphasis on Transporters. Invest Ophthalmol Vis Sci 2020; 60:5022-5034. [PMID: 31791063 DOI: 10.1167/iovs.19-27328] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Retinal pigment epithelium (RPE) limits the xenobiotic entry from the systemic blood stream to the eye. RPE surface transporters can be important in ocular drug distribution, but it has been unclear whether they are expressed on the apical, basal, or both cellular surfaces. In this paper, we provide quantitative comparison of apical and basolateral RPE surface proteomes. Methods We separated the apical and basolateral membranes of differentiated human fetal RPE (hfRPE) cells by combining apical membrane peeling and sucrose density gradient centrifugation. The membrane fractions were analyzed with quantitative targeted absolute proteomics (QTAP) and sequential window acquisition of all theoretical fragment ion spectra mass spectrometry (SWATH-MS) to reveal the membrane protein localization on the RPE cell surfaces. We quantitated 15 transporters in unfractionated RPE cells and scaled their expression to tissue level. Results Several proteins involved in visual cycle, cell adhesion, and ion and nutrient transport were expressed on the hfRPE plasma membranes. Most drug transporters showed similar abundance on both RPE surfaces, whereas large neutral amino acids transporter 1 (LAT1), p-glycoprotein (P-gp), and monocarboxylate transporter 1 (MCT1) showed modest apical enrichment. Many solute carriers (SLC) that are potential prodrug targets were present on both cellular surfaces, whereas putative sodium-coupled neutral amino acid transporter 7 (SNAT7) and riboflavin transporter (RFT3) were enriched on the basolateral and sodium- and chloride-dependent neutral and basic amino acid transporter (ATB0+) on the apical membrane. Conclusions Comprehensive quantitative information of the RPE surface proteomes was reported for the first time. The scientific community can use the data to further increase understanding of the RPE functions. In addition, we provide insights for transporter protein localization in the human RPE and the significance for ocular pharmacokinetics.
Collapse
Affiliation(s)
- Laura Hellinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Kazuki Sato
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Mika Reinisalo
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.,Institute of Clinical Medicine, Department of Ophthalmology, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Heidi Kidron
- Drug Research Programme, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Kirsi Rilla
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Masanori Tachikawa
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yasuo Uchida
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Tetsuya Terasaki
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Arto Urtti
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland.,Drug Research Programme, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Laboratory of Biohybrid Technologies, Institute of Chemistry, St. Petersburg State University, St. Petersburg, Russian Federation
| |
Collapse
|
39
|
Amani N, Dorkoosh FA, Mobedi H. ADCs, as Novel Revolutionary Weapons for Providing a Step Forward in Targeted Therapy of Malignancies. Curr Drug Deliv 2020; 17:23-51. [DOI: 10.2174/1567201816666191121145109] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 08/01/2019] [Accepted: 10/29/2019] [Indexed: 11/22/2022]
Abstract
:Antibody drug conjugates (ADCs), as potent pharmaceutical trojan horses for cancer treatment, provide superior efficacy and specific targeting along with low risk of adverse reactions compared to traditional chemotherapeutics. In fact, the development of these agents combines the selective targeting capability of monoclonal antibody (mAb) with high cytotoxicity of chemotherapeutics for controlling the neoplastic mass growth. Different ADCs (more than 60 ADCs) in preclinical and clinical trials were introduced in this novel pharmaceutical field. Various design-based factors must be taken into account for improving the functionality of ADC technology, including selection of appropriate target antigen and high binding affinity of fragment (miniaturized ADCs) or full mAbs (preferentially use of humanized or fully human antibodies compared to murine and chimeric ones), use of bispecific antibodies for dual targeting effect, linker engineering and conjugation method efficacy to obtain more controlled drug to antibody ratio (DAR). Challenging issues affecting therapeutic efficacy and safety of ADCs, including bystander effect, on- and off-target toxicities, multi drug resistance (MDR) are also addressed. 4 FDA-approved ADCs in the market, including ADCETRIS ®, MYLOTARG®, BESPONSA ®, KADCYLA®. The goal of the current review is to evaluate the key parameters affecting ADCs development.
Collapse
Affiliation(s)
- Nooshafarin Amani
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Abedin Dorkoosh
- Medical Biomaterial Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Mobedi
- Novel Drug Delivery Systems (NDDS) Department, Iran Polymer and Petrochemical Institute, Tehran, Iran
| |
Collapse
|
40
|
Isaac-Lam MF, Hammonds DM. Synthesis and Photodynamic Activity of Vitamin-Chlorin Conjugates at Nanomolar Concentrations against Prostate Cancer Cells. ACS OMEGA 2019; 4:21712-21723. [PMID: 31891050 PMCID: PMC6933591 DOI: 10.1021/acsomega.9b02394] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 11/06/2019] [Indexed: 05/11/2023]
Abstract
Phototoxicity response of synthesized vitamin-chlorin conjugates and their zinc and indium complexes was determined in the human PC-3 prostate cancer cell line, which was previously demonstrated to overexpress vitamin receptors on the cell surface. Pantothenic acid (Vit B5) and lipoic acid (or thioctic acid) were covalently linked to methyl pheophorbide (a chlorophyll derivative) and subsequently metallated with zinc and indium. Cell survival assay indicated that the vitamin-chlorin conjugates have better photodynamic activity against the PC-3 prostate cancer line at the nanomolar concentration range than the commercially available starting precursor methyl pheophorbide. Fluorescence and transmission electron microscopy studies indicated some formation of apoptotic cells and cytoplasmic vacuoles of photosensitized prostatic cells. Targeting vitamin receptors in prostatic cancer cells can be utilized to enhance specificity of photosensitizers for photodynamic therapy applications.
Collapse
Affiliation(s)
- Meden F. Isaac-Lam
- Department of Chemistry and Physics, Purdue University Northwest, 1401 S US Hwy 421, Westville, Indiana 46391, United
States
| | - Dewana M. Hammonds
- Department of Chemistry and Physics, Purdue University Northwest, 1401 S US Hwy 421, Westville, Indiana 46391, United
States
| |
Collapse
|
41
|
Evans MA, Shields CW, Krishnan V, Wang LL, Zhao Z, Ukidve A, Lewandowski M, Gao Y, Mitragotri S. Macrophage‐Mediated Delivery of Hypoxia‐Activated Prodrug Nanoparticles. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900162] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Michael A. Evans
- John A. Paulson School of Engineering and Applied SciencesHarvard University 29 Oxford St. Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
| | - C. Wyatt Shields
- John A. Paulson School of Engineering and Applied SciencesHarvard University 29 Oxford St. Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
| | - Vinu Krishnan
- John A. Paulson School of Engineering and Applied SciencesHarvard University 29 Oxford St. Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
| | - Lily Li‐Wen Wang
- John A. Paulson School of Engineering and Applied SciencesHarvard University 29 Oxford St. Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
- Harvard‐MIT Division of Health Sciences and TechnologyMassachusetts Institute of Technology Cambridge MA 02139 USA
| | - Zhongmin Zhao
- John A. Paulson School of Engineering and Applied SciencesHarvard University 29 Oxford St. Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
| | - Anvay Ukidve
- John A. Paulson School of Engineering and Applied SciencesHarvard University 29 Oxford St. Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
| | - Michael Lewandowski
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
| | - Yongsheng Gao
- John A. Paulson School of Engineering and Applied SciencesHarvard University 29 Oxford St. Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied SciencesHarvard University 29 Oxford St. Cambridge MA 02138 USA
- Wyss Institute for Biologically Inspired EngineeringHarvard University Cambridge MA 02138 USA
| |
Collapse
|
42
|
Cooper I, Atrakchi D, Walker MD, Horovitz A, Fridkin M, Shechter Y. Converting bleomycin into a prodrug that undergoes spontaneous reactivation under physiological conditions. Toxicol Appl Pharmacol 2019; 384:114782. [PMID: 31655077 DOI: 10.1016/j.taap.2019.114782] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 10/08/2019] [Accepted: 10/10/2019] [Indexed: 11/24/2022]
Abstract
Bleomycin is an anticancer antibiotic effective against a range of human malignancies. Yet its usefulness is limited by serious side effects. In this study, we converted bleomycin into a prodrug by covalently linking 2-sulfo, 9 fluorenylmethoxycarbonyl (FMS) to the primary amino side chain of bleomycin. FMS-bleomycin lost its efficacy to bind transition metal ions and therefore was converted into an inactive derivative. Upon incubation in vitro under physiological conditions, the FMS-moiety undergoes spontaneous hydrolysis, generating native bleomycin possessing full anti-bacterial potency. FMS hydrolysis and reactivation takes place with a t1/2 value of 17 ± 1 h. In silico simulation predicts a narrow therapeutic window in human patients of seven hours, starting 40 min after administration. In mice, close agreement was obtained between the experimental and the simulated pharmacokinetic profiles for FMS-bleomycin. FMS-bleomycin is thus shown to be a classical prodrug: it is inactive at the time of administration and the non-modified (active) bleomycin is released with a desirable pharmacokinetic profile following administration, suggesting it may have therapeutic value in the clinic.
Collapse
Affiliation(s)
- Itzik Cooper
- The Joseph Sagol Neuroscience Center, Sheba Medical Center Tel Hashomer, Ramat Gan 52621, Israel.
| | - Dana Atrakchi
- The Joseph Sagol Neuroscience Center, Sheba Medical Center Tel Hashomer, Ramat Gan 52621, Israel
| | - Michael D Walker
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Amnon Horovitz
- Department of Structural Biology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Mati Fridkin
- Department of Organic Chemistry, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yoram Shechter
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
43
|
|
44
|
Delahousse J, Skarbek C, Paci A. Prodrugs as drug delivery system in oncology. Cancer Chemother Pharmacol 2019; 84:937-958. [DOI: 10.1007/s00280-019-03906-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 07/05/2019] [Indexed: 02/07/2023]
|
45
|
Nemutlu E, Eroğlu İ, Eroğlu H, Kır S. In Vitro Release Test of Nano-drug Delivery Systems Based on Analytical and Technological Perspectives. CURR ANAL CHEM 2019. [DOI: 10.2174/1573411014666180912125931] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background:Nanotech products are gaining more attention depending on their advantages for improving drug solubility, maintenance of drug targeting, and attenuation of drug toxicity. In vitro release test is the critical physical parameter to determine the pharmaceutical quality of the product, to monitor formulation design and batch-to-batch variation.Methods:Spectrophotometric and chromatographic methods are mostly used in quantification studies from in vitro release test of nano-drug delivery systems. These techniques have advantages and disadvantages with respect to each other considering dynamic range, selectivity, automation, compatibility with in vitro release media and cost per sample.Results:It is very important to determine the correct kinetic profile of active pharmaceutical substances. At this point, the analytical method used for in vitro release tests has become a very critical parameter to correctly assess the profiles. In this review, we provided an overview of analytical methods applied to the in vitro release assay of various nanopharmaceuticals.Conclusion:This review presents practical direction on analytical method selection for in vitro release test on nanopharmaceuticals. Moreover, precautions on analytical method selection, optimization and validation were discussed.
Collapse
Affiliation(s)
- Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, 06100, Sıhhiye, Ankara, Turkey
| | - İpek Eroğlu
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University, 06100, Sıhhiye, Ankara, Turkey
| | - Hakan Eroğlu
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100, Sıhhiye, Ankara, Turkey
| | - Sedef Kır
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, 06100, Sıhhiye, Ankara, Turkey
| |
Collapse
|
46
|
Citartan M, Kaur H, Presela R, Tang TH. Aptamers as the chaperones (Aptachaperones) of drugs-from siRNAs to DNA nanorobots. Int J Pharm 2019; 567:118483. [PMID: 31260780 DOI: 10.1016/j.ijpharm.2019.118483] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/25/2019] [Accepted: 06/27/2019] [Indexed: 12/16/2022]
Abstract
Aptamers, nucleic acid ligands that are specific against their corresponding targets are increasingly employed in a variety of applications including diagnostics and therapeutics. The specificity of the aptamers against their targets is also used as the basis for the formulation of the aptamer-based drug delivery system. In this review, we aim to provide an overview on the chaperoning roles of aptamers in acting as the cargo or load carriers, delivering contents to the targeted sites via cell surface receptors. Internalization of the aptamer-biomolecule conjugates via receptor-mediated endocytosis and the strategies to augment the rate of endocytosis are underscored. The cargos chaperoned by aptamers, ranging from siRNAs to DNA origami are illuminated. Possible impediments to the aptamer-based drug deliveries such as susceptibility to nuclease resistance, potentiality for immunogenicity activation, tumor heterogeneity are speculated and the corresponding amendment strategies to address these shortcomings are discussed. We prophesy that the future of the aptamer-based drug delivery will take a trajectory towards DNA nanorobot-based assay.
Collapse
Affiliation(s)
- Marimuthu Citartan
- Advanced Medical & Dental Institute (AMDI), Universiti Sains Malaysia, Bertam, 13200 Kepala Batas, Penang, Malaysia.
| | - Harleen Kaur
- Aurobindo Biologics, Biologics R&D Center, Unit-17, Industrial Area, Survey No: 77 & 78, Indrakaran Village, Kandi(Mandal), Sangareddy (District), Hyderabad 502329, India
| | - Ravinderan Presela
- Advanced Medical & Dental Institute (AMDI), Universiti Sains Malaysia, Bertam, 13200 Kepala Batas, Penang, Malaysia
| | - Thean-Hock Tang
- Advanced Medical & Dental Institute (AMDI), Universiti Sains Malaysia, Bertam, 13200 Kepala Batas, Penang, Malaysia.
| |
Collapse
|
47
|
Lu R, Groer C, Kleindl PA, Moulder KR, Huang A, Hunt JR, Cai S, Aires DJ, Berkland C, Forrest ML. Formulation and preclinical evaluation of a toll-like receptor 7/8 agonist as an anti-tumoral immunomodulator. J Control Release 2019; 306:165-176. [PMID: 31173789 DOI: 10.1016/j.jconrel.2019.06.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/21/2019] [Accepted: 06/02/2019] [Indexed: 12/22/2022]
Abstract
The toll-like receptor 7 and 8 (TLR7/8) agonist Resiquimod (R848) has been recognized as a promising immunostimulator for the treatment of cutaneous cancers in multiple clinical trials. However, systemic administration of R848 often results in strong immune-related toxicities while having limited therapeutic effects to the tumor. In the present study, a prodrug-based nanocarrier delivery system was developed that exhibited high therapeutic efficiency. R848 was conjugated to α-tocopherol to constitute an R848-Toco prodrug, followed by formulating with a tocopherol-modified hyaluronic acid (HA-Toco) as a polymeric nano-suspension. In vitro evaluation showed that the delivery system prolonged the release kinetics while maintaining TLR agonist activities. When administered subcutaneously, the nano-suspension formed a depot at the injection site, inducing localized immune responses without systemic expansion. This formulation also suppressed tumor growth and recruited immune cells to the tumor in a murine model of head and neck cancer. In a preclinical canine study of spontaneous mast cell tumors, the treatment led to a 67% response rate (three partial remissions and one complete remission).
Collapse
Affiliation(s)
- Ruolin Lu
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, United States of America
| | - Chad Groer
- HylaPharm LLC, Lawrence, Kansas, United States of America
| | - Peter A Kleindl
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, United States of America
| | - K Ryan Moulder
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, United States of America
| | - Aric Huang
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, United States of America
| | - Jordan R Hunt
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, United States of America
| | - Shuang Cai
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, United States of America; HylaPharm LLC, Lawrence, Kansas, United States of America
| | - Daniel J Aires
- HylaPharm LLC, Lawrence, Kansas, United States of America; Division of Dermatology, School of Medicine, The University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Cory Berkland
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, United States of America; Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, United States of America
| | - M Laird Forrest
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, United States of America; HylaPharm LLC, Lawrence, Kansas, United States of America.
| |
Collapse
|
48
|
Zhang Y, Zhang X, Zeng C, Li B, Zhang C, Li W, Hou X, Dong Y. Targeted delivery of atorvastatin via asialoglycoprotein receptor (ASGPR). Bioorg Med Chem 2019; 27:2187-2191. [PMID: 31005367 PMCID: PMC6535107 DOI: 10.1016/j.bmc.2019.04.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/08/2019] [Accepted: 04/10/2019] [Indexed: 01/26/2023]
Abstract
Targeted drug delivery platforms can increase the concentration of drugs in specific cell populations, reduce adverse effects, and hence improve the therapeutic effect of drugs. Herein, we designed two conjugates by installing the targeting ligand GalNAc (N-acetylgalactosamine) onto atorvastatin (AT). Compared to the parent drug, these two conjugates, termed G2-AT and G2-K-AT, showed increased hepatic cellular uptake. Moreover, both conjugates were able to release atorvastatin, and consequently showed dramatic inhibition of β-hydroxy-β-methylglutaryl-CoA (HMG-CoA) reductase and increased LDL receptors on cell surface.
Collapse
Affiliation(s)
- Youxi Zhang
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States; Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, No. 4, Chongshan Eastern Road, Shenyang 110032, China
| | - Xinfu Zhang
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States; State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, China
| | - Chunxi Zeng
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Bin Li
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Chengxiang Zhang
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Wenqing Li
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Xucheng Hou
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States; Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, United States; The Center for Clinical and Translational Science, The Ohio State University, Columbus, OH 43210, United States; The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, United States; Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, United States; Department of Radiation Oncology, The Ohio State University, Columbus, OH 43210, United States.
| |
Collapse
|
49
|
Abstract
Introduction: Prodrugs have been used to improve the selectivity and efficacy of cancer therapy by targeting unique abnormal markers that are overexpressed by cancer cells and are absent in normal tissues. In this context, different strategies have been exploited and new ones are being developed each year. Areas covered: In this review, an integrated view of the potential use of prodrugs in targeted cancer therapy is provided. Passive and active strategies are discussed in light of the advantages of each one and some successful examples are provided, as well as the clinical status of several prodrugs. Among them, antibody-drug conjugates (ADCs) are the most commonly used. However, several drawbacks, including limited prodrug uptake, poor pharmacokinetics, immunogenicity problems, difficulties in selective targeting and gene expression, and optimized bystander effects limit their clinical applications. Expert opinion: Despite the efforts of different companies and research groups, several drawbacks, such as the lack of relevant in vivo models, complexity of the human metabolism, and economic limitations, have hampered the development of new prodrugs for targeted cancer therapy. As a result, we believe that the combination of prodrugs with cancer nanotechnology and other newly developed approaches, such as aptamer-conjugated nanomaterials, are efficient strategies.
Collapse
Affiliation(s)
- Carla Souza
- a Center of Nanotechnology and Tissue Engineering, Department of Chemistry , School of Philosophy, Sciences and Letters of Ribeirão Preto- USP , Ribeirão Preto , Brazil
| | - Diogo Silva Pellosi
- b Department of Chemistry, Laboratory of Hybrid Materials , Federal University of São Paulo - UNIFESP , Diadema , Brazil
| | - Antonio Claudio Tedesco
- a Center of Nanotechnology and Tissue Engineering, Department of Chemistry , School of Philosophy, Sciences and Letters of Ribeirão Preto- USP , Ribeirão Preto , Brazil
| |
Collapse
|
50
|
Abstract
On the basis of the discovery that the proapoptotic aldehyde 3-hydroxypropanal is a cyclophosphamide metabolite, a novel mechanism of action of oxazaphosphorine cytostatics is presented and confirmed by animal experiments. Furthermore, it is shown that new oxazaphosphorine cytostatics, which are on orders of magnitude more effective than already existing, can be developed on the basis of the new model for the mechanism of action.
Collapse
|