1
|
Zheng S, Gu Y, Gu Y, Zhao Y, Li L, Wang M, Jiang R, Yu X, Chen T, Li J. Machine learning-enabled virtual screening indicates the anti-tuberculosis activity of aldoxorubicin and quarfloxin with verification by molecular docking, molecular dynamics simulations, and biological evaluations. Brief Bioinform 2024; 26:bbae696. [PMID: 39737570 DOI: 10.1093/bib/bbae696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/16/2024] [Accepted: 12/17/2024] [Indexed: 01/01/2025] Open
Abstract
Drug resistance in Mycobacterium tuberculosis (Mtb) is a significant challenge in the control and treatment of tuberculosis, making efforts to combat the spread of this global health burden more difficult. To accelerate anti-tuberculosis drug discovery, repurposing clinically approved or investigational drugs for the treatment of tuberculosis by computational methods has become an attractive strategy. In this study, we developed a virtual screening workflow that combines multiple machine learning and deep learning models, and 11 576 compounds extracted from the DrugBank database were screened against Mtb. Our screening method produced satisfactory predictions on three data-splitting settings, with the top predicted bioactive compounds all known antibacterial or anti-TB drugs. To further identify and evaluate drugs with repurposing potential in TB therapy, 15 screened potential compounds were selected for subsequent computational and experimental evaluations, out of which aldoxorubicin and quarfloxin showed potent inhibition of Mtb strain H37Rv, with minimal inhibitory concentrations of 4.16 and 20.67 μM/mL, respectively. More inspiringly, these two compounds also showed antibacterial activity against multidrug-resistant TB isolates and exhibited strong antimicrobial activity against Mtb. Furthermore, molecular docking, molecular dynamics simulation, and the surface plasmon resonance experiments validated the direct binding of the two compounds to Mtb DNA gyrase. In summary, our effective comprehensive virtual screening workflow successfully repurposed two novel drugs (aldoxorubicin and quarfloxin) as promising anti-Mtb candidates. The verification results provide useful information for the further development and clinical verification of anti-TB drugs.
Collapse
Affiliation(s)
- Si Zheng
- Institute for Artificial Intelligence & Department of Computer Science and Technology, Tsinghua University, Haidian District, Beijing 100084, China
- Institute of Medical Information, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing 100020, China
| | - Yaowen Gu
- Department of Chemistry, New York University, New York, NY 10027, United States
| | - Yuzhen Gu
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory on Drug-Resistant Tuberculosis, Beijing Chest Hospital, Capital Medical University, Tongzhou District, Beijing 101149, China
| | - Yelin Zhao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Xicheng District, Beijing 100050, China
| | - Liang Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Xicheng District, Beijing 100050, China
| | - Min Wang
- Institute of Medical Information, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing 100020, China
| | - Rui Jiang
- Department of Automation, Tsinghua University, Haidian District, Beijing 100084, China
| | - Xia Yu
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory on Drug-Resistant Tuberculosis, Beijing Chest Hospital, Capital Medical University, Tongzhou District, Beijing 101149, China
| | - Ting Chen
- Institute for Artificial Intelligence & Department of Computer Science and Technology, Tsinghua University, Haidian District, Beijing 100084, China
| | - Jiao Li
- Institute of Medical Information, Chinese Academy of Medical Sciences and Peking Union Medical College, Chaoyang District, Beijing 100020, China
| |
Collapse
|
2
|
Nazli A, Irshad Khan MZ, Rácz Á, Béni S. Acid-sensitive prodrugs; a promising approach for site-specific and targeted drug release. Eur J Med Chem 2024; 276:116699. [PMID: 39089000 DOI: 10.1016/j.ejmech.2024.116699] [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: 02/26/2024] [Revised: 07/02/2024] [Accepted: 07/18/2024] [Indexed: 08/03/2024]
Abstract
Drugs administered through conventional formulations are devoid of targeting and often spread to various undesired sites, leading to sub-lethal concentrations at the site of action and the emergence of undesired effects. Hence, therapeutic agents should be delivered in a controlled manner at target sites. Currently, stimuli-based drug delivery systems have demonstrated a remarkable potential for the site-specific delivery of therapeutic moieties. pH is one of the widely exploited stimuli for drug delivery as several pathogenic conditions such as tumor cells, infectious and inflammatory sites are characterized by a low pH environment. This review article aims to demonstrate various strategies employed in the design of acid-sensitive prodrugs, providing an overview of commercially available acid-sensitive prodrugs. Furthermore, we have compiled the progress made for the development of new acid-sensitive prodrugs currently undergoing clinical trials. These prodrugs include albumin-binding prodrugs (Aldoxorubicin and DK049), polymeric micelle (NC-6300), polymer conjugates (ProLindac™), and an immunoconjugate (IMMU-110). The article encompasses a broad spectrum of studies focused on the development of acid-sensitive prodrugs for anticancer, antibacterial, and anti-inflammatory agents. Finally, the challenges associated with the acid-sensitive prodrug strategy are discussed, along with future directions.
Collapse
Affiliation(s)
- Adila Nazli
- Department of Pharmacognosy, Semmelweis University, 1085, Budapest, Hungary.
| | | | - Ákos Rácz
- Department of Pharmacognosy, Semmelweis University, 1085, Budapest, Hungary.
| | - Szabolcs Béni
- Integrative Health and Environmental Analysis Research Laboratory, Department of Analytical Chemistry, Institute of Chemistry, Eötvös Loránd University, 1117, Budapest, Hungary.
| |
Collapse
|
3
|
Adhikari S, Nath S, Kansız S, Balidya N, Paul AK, Dege N, Sahin O, Mahmoudi G, Verma AK, Safin DA. Zinc(II) coordination compound with N'-(pyridin-2-ylmethylene)nicotinohydrazide: Synthesis, crystal structure, computational and cytotoxicity studies. J Inorg Biochem 2024; 257:112598. [PMID: 38763101 DOI: 10.1016/j.jinorgbio.2024.112598] [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: 04/02/2024] [Accepted: 05/06/2024] [Indexed: 05/21/2024]
Abstract
In this work, we report on the synthesis of a novel zinc(II) coordination compound [ZnL2] (1), which was readily obtained from the reaction of Zn(OAc)·2H2O and N'-(pyridin-2-ylmethylene)nicotinohydrazide (HL) in methanol. Recrystallization of 1 from dimethylformamide under ambient conditions allowed to produce yellow block-like crystals of 1·H2O. Complex 1·H2O was characterized by FT-IR and 1H NMR spectroscopy, while its optical properties were studied by UV-vis and spectrofluorimetry in methanol. The crystal structure of the title complex was revealed by single crystal X-ray diffraction and further explored in detail by the Hirshfeld surface analysis. Theoretical investigations based on the DFT calculations have also been applied to show the electronic properties of complex 1. The antitumor activities of the parent ligand HL and complex 1 were studied using Dalton's lymphoma malignant cancer model. Both compounds were found to induce concentration-dependent cytotoxicity and apoptotic cell death, leading to a decrease in cell viability, body weight, and tumor volume in mice with the superior activity of complex 1 over HL. Mice treated with complex 1 demonstrated an increase in life span with a survival period of 23 days. Finally, using a molecular docking approach, we have probed complex 1 to inhibit the recombinant mouse tumor-necrosis factor alpha (mTNF).
Collapse
Affiliation(s)
- Suman Adhikari
- Department of Chemistry, Govt. Degree College, Dharmanagar, Tripura(N) 799253, India.
| | - Sourav Nath
- Department of Chemistry, Govt. Degree College, Dharmanagar, Tripura(N) 799253, India
| | - Sevgi Kansız
- Samsun University, Faculty of Engineering, Department of Fundamental Sciences, 55420 Samsun, Turkey
| | - Nabajyoti Balidya
- Department of Chemistry, Milki High School, Milki, Malda, 732209, India
| | - Anirban Kumar Paul
- Department of Zoology, Cell & Biochemical Technology Laboratory, Cotton University, Guwahati 781001, India
| | - Necmi Dege
- Ondokuz Mayıs University, Faculty of Arts and Sciences, Department of Physics, 55139 Samsun, Turkey
| | - Onur Sahin
- Sinop University, Scientific and Technological Research Application and Research Center, 57000 Sinop, Turkey
| | - Ghodrat Mahmoudi
- Department of Chemistry, Faculty of Science, University of Maragheh, P.O. Box 55181-83111, Maragheh, Iran; Chemistry Department, Faculty of Engineering and Natural Sciences, Istinye University, Sarıyer, Istanbul 34396, Turkey.
| | - Akalesh Kumar Verma
- Department of Zoology, Cell & Biochemical Technology Laboratory, Cotton University, Guwahati 781001, India.
| | - Damir A Safin
- University of Tyumen, Volodarskogo Str. 6, 625003 Tyumen, Russian Federation; Scientific and Educational and Innovation Center for Chemical and Pharmaceutical Technologies, Ural Federal University named after the First President of Russia B.N. Yeltsin, Ekaterinburg 620002, Russian Federation.
| |
Collapse
|
4
|
Aalhate M, Mahajan S, Singh H, Guru SK, Singh PK. Nanomedicine in therapeutic warfront against estrogen receptor-positive breast cancer. Drug Deliv Transl Res 2023; 13:1621-1653. [PMID: 36795198 DOI: 10.1007/s13346-023-01299-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2023] [Indexed: 02/17/2023]
Abstract
Breast cancer (BC) is the most frequently diagnosed malignancy in women worldwide. Almost 70-80% of cases of BC are curable at the early non-metastatic stage. BC is a heterogeneous disease with different molecular subtypes. Around 70% of breast tumors exhibit estrogen-receptor (ER) expression and endocrine therapy is used for the treatment of these patients. However, there are high chances of recurrence in the endocrine therapy regimen. Though chemotherapy and radiation therapy have substantially improved survival rates and treatment outcomes in BC patients, there is an increased possibility of the development of resistance and dose-limiting toxicities. Conventional treatment approaches often suffer from low bioavailability, adverse effects due to the non-specific action of chemotherapeutics, and low antitumor efficacy. Nanomedicine has emerged as a conspicuous strategy for delivering anticancer therapeutics in BC management. It has revolutionized the area of cancer therapy by increasing the bioavailability of the therapeutics and improving their anticancer efficacy with reduced toxicities on healthy tissues. In this article, we have highlighted various mechanisms and pathways involved in the progression of ER-positive BC. Further, different nanocarriers delivering drugs, genes, and natural therapeutic agents for surmounting BC are the spotlights of this article.
Collapse
Affiliation(s)
- Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India
| | - Hoshiyar Singh
- Department of Biological Science, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Santosh Kumar Guru
- Department of Biological Science, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, 500037, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500037, India.
| |
Collapse
|
5
|
Srinivasan S, Yee NA, Zakharian M, Alečković M, Mahmoodi A, Nguyen TH, Mejía Oneto JM. SQ3370, the first clinical click chemistry-activated cancer therapeutic, shows safety in humans and translatability across species. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.28.534654. [PMID: 37034617 PMCID: PMC10081183 DOI: 10.1101/2023.03.28.534654] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
BACKGROUND SQ3370 is the first demonstration of the Click Activated Protodrugs Against Cancer (CAPAC™) platform that uses click chemistry to activate drugs directly at tumor sites, maximizing therapeutic exposure. SQ3370 consists of a tumor-localizing biopolymer (SQL70) and a chemically-attenuated doxorubicin (Dox) protodrug SQP33; the protodrug is activated upon clicking with the biopolymer at tumor sites. Here, we present data from preclinical studies and a Phase 1 dose-escalation clinical trial in adult patients with advanced solid tumors ( NCT04106492 ) demonstrating SQ3370's activation at tumor sites, safety, systemic pharmacokinetics (PK), and immunological activity. METHODS Treatment cycles consisting of an intratumoral or subcutaneous injection of SQL70 biopolymer followed by 5 daily intravenous doses of SQP33 protodrug were evaluated in tumor-bearing mice, healthy dogs, and adult patients with solid tumors. RESULTS SQL70 effectively activated SQP33 at tumor sites, resulting in high Dox concentrations that were well tolerated and unachievable by conventional treatment. SQ3370 was safely administered at 8.9x the veterinary Dox dose in dogs and 12x the conventional Dox dose in patients, with no dose-limiting toxicity reported to date. SQ3370's safety, toxicology, and PK profiles were highly translatable across species. SQ3370 increased cytotoxic CD3 + and CD8 + T-cells in patient tumors indicating T-cell-dependent immune activation in the tumor microenvironment. CONCLUSIONS SQ3370, the initial demonstration of click chemistry in humans, enhances the safety of Dox at unprecedented doses and has the potential to increase therapeutic index. Consistent safety, toxicology, PK, and immune activation results observed with SQ3370 across species highlight the translatability of the click chemistry approach in drug development. TRIAL REGISTRATION NCT04106492; 7 September 2019.
Collapse
|
6
|
Yang J, Wang X, Wang B, Park K, Wooley K, Zhang S. Challenging the fundamental conjectures in nanoparticle drug delivery for chemotherapy treatment of solid cancers. Adv Drug Deliv Rev 2022; 190:114525. [PMID: 36100142 DOI: 10.1016/j.addr.2022.114525] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 01/24/2023]
Abstract
Nanomedicines for cancer treatment have been studied extensively over the last few decades. Yet, only five anticancer nanomedicines have received approvals from the United States Food and Drug Administration (FDA) for treating solid tumors. This drastic mismatch between effort and return calls into question the basic understanding of this field. Various viewpoints on nanomedicines have been presented regarding their potentials and inefficiencies. However, the underlying logics of nanomedicine research and its inadequate translation to the successful use in the clinic have not been thoroughly examined. Tumor-targeted drug delivery was used to understand the shortfalls of the nanomedicine field in general. The concept of tumor-targeted drug delivery by nanomedicine has been based on two conjectures: (i) increased drug delivery to tumors provides better efficacy, and (ii) decreased drug delivery to healthy organs results in fewer side effects. The clinical evidence gathered from the literature indicates that nanomedicines bearing classic chemotherapeutic drugs, such as Dox, cis-Pt, CPT and PTX, have already reached the maximum drug delivery limit to solid tumors in humans. Still, the anticancer efficacy and safety remain unchanged despite the increased tumor accumulation. Thus, it is understandable to see few nanomedicine-based formulations approved by the FDA. The examination of FDA-approved nanomedicine formulations indicates that their approvals were not based on the improved delivery to tumors but mostly on changes in dose-limiting toxicity unique to each drug. This comprehensive analysis of the fundamentals of anticancer nanomedicines is designed to provide an accurate picture of the field's underlying false conjectures, hopefully, thereby accelerating the future clinical translations of many formulations under research.
Collapse
Affiliation(s)
- Juanjuan Yang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| | - Xiaojin Wang
- Department of Biostatistics, Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, 227 South Chongqing Road, Shanghai 200025, PR China
| | - Bingshun Wang
- Department of Biostatistics, Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, 227 South Chongqing Road, Shanghai 200025, PR China
| | - Kinam Park
- Weldon School of Biomedical Engineering, and Department of Pharmaceutics, Purdue University, West Lafayette, IN 47907, USA
| | - Karen Wooley
- Departments of Chemistry, Materials Science & Engineering and Chemical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Shiyi Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, PR China
| |
Collapse
|
7
|
Paul M, Itoo AM, Ghosh B, Biswas S. Current trends in the use of human serum albumin for drug delivery in cancer. Expert Opin Drug Deliv 2022; 19:1449-1470. [PMID: 36253957 DOI: 10.1080/17425247.2022.2134341] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Human serum albumin is the most abundant transport protein in plasma, which has recently been extensively utilized to form nanoparticles for drug delivery in cancer. The primary reason for selecting albumin protein as drug delivery cargo is its excellent biocompatibility, biodegradability, and non-immunogenicity. Moreover, the albumin structure containing three homologous domains constituted of a single polypeptide (585 amino acid) incorporates various hydrophobic drugs by non-covalent interactions. Albumin shows active tumor targeting via their interaction with gp60 and SPARC proteins abundant in the tumor-associated endothelial cells and the tumor microenvironment. AREAS COVERED The review discusses the importance of albumin as a drug-carrier system, general procedures to prepare albumin NPs, and the current trends in using albumin-based nanomedicines to deliver various chemotherapeutic agents. The various applications of albumin in the nanomedicines, such as NPs surface modifier and fabrication of hybrid/active-tumor targeted NPs, are delineated based on current trends. EXPERT OPINION Nanomedicines have the potential to revolutionize cancer treatment. However, clinical translation is limited majorly due to the lack of suitable nanomaterials offering systemic stability, optimum drug encapsulation, tumor-targeted delivery, sustained drug release, and biocompatibility. The potential of albumin could be explored in nanomedicines fabrication for superior treatment outcomes in cancer.
Collapse
Affiliation(s)
- Milan Paul
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad-500078, India
| | - Asif Mohd Itoo
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad-500078, India
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad-500078, India
| | - Swati Biswas
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad-500078, India
| |
Collapse
|
8
|
Van Renterghem B, Wozniak A, Tarantola L, Casazza A, Wellens J, Nysen M, Vanleeuw U, Lee CJ, Reyns G, Sciot R, Kindt N, Schöffski P. Enhanced Antitumor Efficacy of PhAc-ALGP-Dox, an Enzyme-Activated Doxorubicin Prodrug, in a Panel of THOP1-Expressing Patient-Derived Xenografts of Soft Tissue Sarcoma. Biomedicines 2022; 10:biomedicines10040862. [PMID: 35453612 PMCID: PMC9025547 DOI: 10.3390/biomedicines10040862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/29/2022] [Accepted: 04/04/2022] [Indexed: 12/10/2022] Open
Abstract
Despite poor response rates and dose-limiting cardiotoxicity, doxorubicin (doxo) remains the standard-of-care for patients with advanced soft tissue sarcoma. We evaluated the efficacy of two tetrapeptidic doxo prodrugs (PhAc-ALGP-Dox or CBR-049 and CBR-050) that are locally activated by enzymes expressed in the tumor environment, in ten sarcoma patient-derived xenografts. Xenograft models were selected based on expression of the main activating enzyme, i.e., thimet oligopeptidase (THOP1). Mice were either randomized to vehicle, doxo, CBR-049 and CBR-050 or control, doxo, aldoxorubicin (aldoxo) and CBR-049. Treatment efficacy was assessed by tumor volume measurement and histological assessment of ex-mouse tumors. CBR-049 showed significant tumor growth delay compared to control in all xenografts investigated and was superior compared to doxo in all but one. At the same time, CBR-049 showed comparable efficacy to aldoxo but the latter was found to have a complex safety profile in mice. CBR-050 demonstrated tumor growth delay compared to control in one xenograft but was not superior to doxo. For both experimental prodrugs, strong immunostaining for THOP1 was found to predict better antitumor efficacy. The prodrugs were well tolerated without any adverse events, even though molar doses were 17-fold higher than those administered and tolerated for doxo.
Collapse
Affiliation(s)
- Britt Van Renterghem
- Laboratory of Experimental Oncology, Catholic University of Leuven, 3000 Leuven, Belgium; (B.V.R.); (A.W.); (L.T.); (J.W.); (M.N.); (U.V.); (C.-J.L.)
| | - Agnieszka Wozniak
- Laboratory of Experimental Oncology, Catholic University of Leuven, 3000 Leuven, Belgium; (B.V.R.); (A.W.); (L.T.); (J.W.); (M.N.); (U.V.); (C.-J.L.)
| | - Ludovica Tarantola
- Laboratory of Experimental Oncology, Catholic University of Leuven, 3000 Leuven, Belgium; (B.V.R.); (A.W.); (L.T.); (J.W.); (M.N.); (U.V.); (C.-J.L.)
| | - Andrea Casazza
- CoBioRes, Campus Gasthuisberg, Catholic University of Leuven, 3000 Leuven, Belgium; (A.C.); (G.R.); (N.K.)
| | - Jasmien Wellens
- Laboratory of Experimental Oncology, Catholic University of Leuven, 3000 Leuven, Belgium; (B.V.R.); (A.W.); (L.T.); (J.W.); (M.N.); (U.V.); (C.-J.L.)
| | - Madita Nysen
- Laboratory of Experimental Oncology, Catholic University of Leuven, 3000 Leuven, Belgium; (B.V.R.); (A.W.); (L.T.); (J.W.); (M.N.); (U.V.); (C.-J.L.)
| | - Ulla Vanleeuw
- Laboratory of Experimental Oncology, Catholic University of Leuven, 3000 Leuven, Belgium; (B.V.R.); (A.W.); (L.T.); (J.W.); (M.N.); (U.V.); (C.-J.L.)
| | - Che-Jui Lee
- Laboratory of Experimental Oncology, Catholic University of Leuven, 3000 Leuven, Belgium; (B.V.R.); (A.W.); (L.T.); (J.W.); (M.N.); (U.V.); (C.-J.L.)
| | - Geert Reyns
- CoBioRes, Campus Gasthuisberg, Catholic University of Leuven, 3000 Leuven, Belgium; (A.C.); (G.R.); (N.K.)
| | - Raf Sciot
- Department of Pathology, University Hospitals Leuven, 3000 Leuven, Belgium;
| | - Nele Kindt
- CoBioRes, Campus Gasthuisberg, Catholic University of Leuven, 3000 Leuven, Belgium; (A.C.); (G.R.); (N.K.)
| | - Patrick Schöffski
- Laboratory of Experimental Oncology, Catholic University of Leuven, 3000 Leuven, Belgium; (B.V.R.); (A.W.); (L.T.); (J.W.); (M.N.); (U.V.); (C.-J.L.)
- Department of General Medical Oncology, University Hospitals Leuven, 3000 Leuven, Belgium
- Correspondence:
| |
Collapse
|
9
|
Veselov VV, Nosyrev AE, Jicsinszky L, Alyautdin RN, Cravotto G. Targeted Delivery Methods for Anticancer Drugs. Cancers (Basel) 2022; 14:622. [PMID: 35158888 PMCID: PMC8833699 DOI: 10.3390/cancers14030622] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 02/07/2023] Open
Abstract
Several drug-delivery systems have been reported on and often successfully applied in cancer therapy. Cell-targeted delivery can reduce the overall toxicity of cytotoxic drugs and increase their effectiveness and selectivity. Besides traditional liposomal and micellar formulations, various nanocarrier systems have recently become the focus of developmental interest. This review discusses the preparation and targeting techniques as well as the properties of several liposome-, micelle-, solid-lipid nanoparticle-, dendrimer-, gold-, and magnetic-nanoparticle-based delivery systems. Approaches for targeted drug delivery and systems for drug release under a range of stimuli are also discussed.
Collapse
Affiliation(s)
- Valery V. Veselov
- Center of Bioanalytical Investigation and Molecular Design, Sechenov First Moscow State Medical University, 8 Trubetskaya ul, 119991 Moscow, Russia; (V.V.V.); (A.E.N.)
| | - Alexander E. Nosyrev
- Center of Bioanalytical Investigation and Molecular Design, Sechenov First Moscow State Medical University, 8 Trubetskaya ul, 119991 Moscow, Russia; (V.V.V.); (A.E.N.)
| | - László Jicsinszky
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Turin, Italy;
| | - Renad N. Alyautdin
- Department of Pharmacology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia;
| | - Giancarlo Cravotto
- Department of Drug Science and Technology, University of Turin, Via P. Giuria 9, 10125 Turin, Italy;
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, 8 Trubetskaya ul, 119991 Moscow, Russia
| |
Collapse
|
10
|
Jiang Y, Jiang Z, Wang M, Ma L. Current understandings and clinical translation of nanomedicines for breast cancer therapy. Adv Drug Deliv Rev 2022; 180:114034. [PMID: 34736986 DOI: 10.1016/j.addr.2021.114034] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/20/2021] [Accepted: 10/28/2021] [Indexed: 02/08/2023]
Abstract
Breast cancer is one of the most frequently diagnosed cancers that is threatening women's life. Current clinical treatment regimens for breast cancer often involve neoadjuvant and adjuvant systemic therapies, which somewhat are associated with unfavorable features. Also, the heterogeneous nature of breast cancers requires precision medicine that cannot be fulfilled by a single type of systemically administered drug. Taking advantage of the nanocarriers, nanomedicines emerge as promising therapeutic agents for breast cancer that could resolve the defects of drugs and achieve precise drug delivery to almost all sites of primary and metastatic breast tumors (e.g. tumor vasculature, tumor stroma components, breast cancer cells, and some immune cells). Seven nanomedicines as represented by Doxil® have been approved for breast cancer clinical treatment so far. More nanomedicines including both non-targeting and active targeting nanomedicines are being evaluated in the clinical trials. However, we have to realize that the translation of nanomedicines, particularly the active targeting nanomedicines is not as successful as people have expected. This review provides a comprehensive landscape of the nanomedicines for breast cancer treatment, from laboratory investigations to clinical applications. We also highlight the key advances in the understanding of the biological fate and the targeting strategies of breast cancer nanomedicine and the implications to clinical translation.
Collapse
|
11
|
Liu SY, Zhang QZ, Hu MQ, Li FX, Fu JM, Zhu ZD, Li QK, Yang Z, Quan JM. Targeting Extracellular Cyclophilin A via an Albumin-Binding Cyclosporine A Analogue. ChemMedChem 2021; 16:3649-3652. [PMID: 34595834 DOI: 10.1002/cmdc.202100526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/29/2021] [Indexed: 11/09/2022]
Abstract
An albumin-binding CsA analogue 4MCsA was achieved by attachment of a thiol-reactive maleimide group at the side-chain of P4 position of CsA derivative. 4MCsA was semi-synthesized from CsA, and the cell-impermeability of albumin-4MCsA was detected by mass spectrometry and a competitive flow cytometry. 4MCsA exhibits inhibition of chemotaxis activity and inflammation by targeting extracellular CypA without immunosuppressive effect and cellular toxicity. These combined results suggested that 4MCsA can be restricted extracellularly through covalently binding to Cys34 of albumin with its maleimide group, and regulate the functions of cyclophilin A extracellularly.
Collapse
Affiliation(s)
- Si-Yu Liu
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | | | - Min-Qiang Hu
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Feng-Xia Li
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Jia-Miao Fu
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | | | - Qin-Kai Li
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Zhen Yang
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Jun-Min Quan
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| |
Collapse
|
12
|
Sung YS, Wu W, Ewbank MA, Utterback RD, Marty MT, Tomat E. Albumin Conjugates of Thiosemicarbazone and Imidazole-2-thione Prochelators: Iron Coordination and Antiproliferative Activity. ChemMedChem 2021; 16:2764-2768. [PMID: 33974730 PMCID: PMC8448912 DOI: 10.1002/cmdc.202100278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Indexed: 12/19/2022]
Abstract
The central role of iron in tumor progression and metastasis motivates the development of iron-binding approaches in cancer chemotherapy. Disulfide-based prochelators are reductively activated upon cellular uptake to liberate thiol chelators responsible for iron sequestration. Herein, a trimethyl thiosemicarbazone moiety and the imidazole-2-thione heterocycle are incorporated in this prochelator design. Iron binding of the corresponding tridentate chelators leads to the stabilization of a low-spin ferric center in 2 : 1 ligand-to-metal complexes. Native mass spectrometry experiments show that the prochelators form stable disulfide conjugates with bovine serum albumin, thus affording novel bioconjugate prochelator systems. Antiproliferative activities at sub-micromolar levels are recorded in a panel of breast, ovarian and colorectal cancer cells, along with significantly lower activity in normal fibroblasts.
Collapse
Affiliation(s)
- Yu-Shien Sung
- Department of Chemistry and Biochemistry, The University of Arizona, 1306 E. University Blvd., Tucson, AZ 85721-0041, USA
| | - Wangbin Wu
- Department of Chemistry and Biochemistry, The University of Arizona, 1306 E. University Blvd., Tucson, AZ 85721-0041, USA
| | - Megan A Ewbank
- Department of Chemistry and Biochemistry, The University of Arizona, 1306 E. University Blvd., Tucson, AZ 85721-0041, USA
| | - Rachel D Utterback
- Department of Chemistry and Biochemistry, The University of Arizona, 1306 E. University Blvd., Tucson, AZ 85721-0041, USA
| | - Michael T Marty
- Department of Chemistry and Biochemistry, The University of Arizona, 1306 E. University Blvd., Tucson, AZ 85721-0041, USA
| | - Elisa Tomat
- Department of Chemistry and Biochemistry, The University of Arizona, 1306 E. University Blvd., Tucson, AZ 85721-0041, USA
| |
Collapse
|
13
|
Serrano R, Martinez-Argudo I, Fernandez-Sanchez M, Pacheco-Liñan PJ, Bravo I, Cohen B, Calero R, Ruiz MJ. New titanocene derivative with improved stability and binding ability to albumin exhibits high anticancer activity. J Inorg Biochem 2021; 223:111562. [PMID: 34364140 DOI: 10.1016/j.jinorgbio.2021.111562] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 07/23/2021] [Accepted: 07/30/2021] [Indexed: 11/15/2022]
Abstract
Titanium-based therapies have emerged as a promising alternative for the treatment of cancer patients, particularly those with cisplatin resistant tumors. Unfortunately, some titanium compounds show stability and solubility problems that have hindered their use in clinical practice. Here, we designed and synthesized a new titanium complex containing a titanocene fragment, a tridentate ligand to improve its stability in water, and a long aliphatic chain, designed to facilitate a non-covalent interaction with albumin, the most abundant protein in human serum. The stability and human serum albumin affinity of the resulting titanium complex was investigated by UV-Vis absorption and fluorescence spectroscopy techniques. Complex [TiCp2{(OOC)2py-O-myr}] (3) (myr = C14H29, py = pyridine) and its analogous [TiCp2{(OOC)2py-OH}] (4), lacking the aliphatic chain, showed improved stability in phosphate saline buffer compared with [TiCp2Cl2] (1). 3 showed a strong interaction with human serum albumin in a 1:1 stoichiometry. The cytotoxic effect of 3 was higher compared to [TiCp2Cl2] in tumor cell lines and showed potential tumor selectivity when assayed in non-tumor human epithelial cells. Finally, 3 showed an antiproliferative effect on cancer cells, decreasing the population in the S phase, and increasing apoptotic cells in a significant manner. All this makes the novel Ti(IV) compound 3 a firm candidate to continue further studies of its therapeutic potential in vitro and in vivo.
Collapse
Affiliation(s)
- Rosario Serrano
- Universidad de Castilla-La Mancha, Facultad de Ciencias Ambientales y Bioquímica, 45071 Toledo, Spain; Universidad de Castilla-La Mancha, Departamento de Química Orgánica, Inorgánica y Bioquímica, Spain
| | - Isabel Martinez-Argudo
- Universidad de Castilla-La Mancha, Facultad de Ciencias Ambientales y Bioquímica, 45071 Toledo, Spain; Universidad de Castilla-La Mancha, Departamento de Ciencia y Tecnología Agroforestal y Genética, Spain
| | - Miguel Fernandez-Sanchez
- Universidad de Castilla-La Mancha, Facultad de Ciencias Ambientales y Bioquímica, 45071 Toledo, Spain
| | - Pedro J Pacheco-Liñan
- Universidad de Castilla-La Mancha, Facultad de Farmacia, 02071 Albacete, Spain; Universidad de Castilla-La Mancha, Departamento de Química Física, Spain
| | - Ivan Bravo
- Universidad de Castilla-La Mancha, Facultad de Farmacia, 02071 Albacete, Spain; Universidad de Castilla-La Mancha, Departamento de Química Física, Spain
| | - Boiko Cohen
- Universidad de Castilla-La Mancha, Facultad de Ciencias Ambientales y Bioquímica, 45071 Toledo, Spain; Universidad de Castilla-La Mancha, Departamento de Química Física, Spain; Universidad de Castilla-La Mancha, INAMOL, 45071 Toledo, Spain
| | - Raul Calero
- Universidad de Castilla-La Mancha, Facultad de Ciencias Ambientales y Bioquímica, 45071 Toledo, Spain; Universidad de Castilla-La Mancha, Departamento de Química Orgánica, Inorgánica y Bioquímica, Spain.
| | - Maria Jose Ruiz
- Universidad de Castilla-La Mancha, Facultad de Ciencias Ambientales y Bioquímica, 45071 Toledo, Spain; Universidad de Castilla-La Mancha, Departamento de Química Orgánica, Inorgánica y Bioquímica, Spain
| |
Collapse
|
14
|
Evaluation of the antitumor activity of a series of the pincer-type metallocomplexes produced from isonicotinohydrazide derivative. J Inorg Biochem 2021; 223:111525. [PMID: 34237626 DOI: 10.1016/j.jinorgbio.2021.111525] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/18/2021] [Accepted: 06/25/2021] [Indexed: 01/30/2023]
Abstract
In this work we report on the antitumor properties of a series of pincer-type metallocomplexes [Hg2(HL-keto)Cl4]n (1), [Hg(HL-keto)I2] (2) and [Mn(HL-zwitterion)Cl2]∙MeOH (3∙MeOH), derived from N'-(1-(pyridin-2-yl)ethylidene)isonicotinohydrazide (HL) and corresponding metal salts. The Hg(II) and Mn(II) salts are chelated by the keto (HL-keto) or zwitterionic (HL-zwitterion) form of HL, respectively. The cytotoxic effects of these compounds have been accessed against lung adenocarcinoma (A549) and hepatocellular carcinoma (HepG2 and Huh7) cell lines. Complexes 1 and 2 were found to be most efficient against the cell line Huh7 with IC50 value of 2.56 and 9.90 μM, respectively, while they exhibit moderate activity towards cell lines A549 and HepG2, as evidenced from IC50 values in the range 27.98-56.99 μM. Complex 3∙MeOH is less efficient towards all the three cell lines with relatively high IC50 values. The mechanisms of the metallocomplexes killing the aforementioned cells were elucidated by flow cytometry, colony formation and polymerase chain reaction (PCR) analysis of apoptosis related expression of the genes. The results of the cytotoxic effects and antitumor activity on different cell lines are affected by the metal nature and the presence of the coordinated halide.
Collapse
|
15
|
Spada A, Emami J, Tuszynski JA, Lavasanifar A. The Uniqueness of Albumin as a Carrier in Nanodrug Delivery. Mol Pharm 2021; 18:1862-1894. [PMID: 33787270 DOI: 10.1021/acs.molpharmaceut.1c00046] [Citation(s) in RCA: 243] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Albumin is an appealing carrier in nanomedicine because of its unique features. First, it is the most abundant protein in plasma, endowing high biocompatibility, biodegradability, nonimmunogenicity, and safety for its clinical application. Second, albumin chemical structure and conformation allows interaction with many different drugs, potentially protecting them from elimination and metabolism in vivo, thus improving their pharmacokinetic properties. Finally, albumin can interact with receptors overexpressed in many diseased tissues and cells, providing a unique feature for active targeting of the disease site without the addition of specific ligands to the nanocarrier. For this reason, albumin, characterized by an extended serum half-life of around 19 days, has the potential of promoting half-life extension and targeted delivery of drugs. Therefore, this article focuses on the importance of albumin as a nanodrug delivery carrier for hydrophobic drugs, taking advantage of the passive as well as active targeting potential of this nanocarrier. Particular attention is paid to the breakthrough NAB-Technology, with emphasis on the advantages of Nab-Paclitaxel (Abraxane), compared to the solvent-based formulations of Paclitaxel, i.e., CrEL-paclitaxel (Taxol) in a clinical setting. Finally, the role of albumin in carrying anticancer compounds is depicted, with a particular focus on the albumin-based formulations that are currently undergoing clinical trials. The article sheds light on the power of an endogenous substance, such as albumin, as a drug delivery system, signifies the importance of the drug vehicle in drug performance in the biological systems, and highlights the possible future trends in the use of this drug delivery system.
Collapse
Affiliation(s)
- Alessandra Spada
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada.,DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, Turin 10129, Italy.,Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Jaber Emami
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada.,Department of Pharmaceutics, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Jack A Tuszynski
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada.,DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, Turin 10129, Italy
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| |
Collapse
|
16
|
Martins-Teixeira MB, Carvalho I. Antitumour Anthracyclines: Progress and Perspectives. ChemMedChem 2020; 15:933-948. [PMID: 32314528 DOI: 10.1002/cmdc.202000131] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Indexed: 12/31/2022]
Abstract
Anthracyclines are ranked among the most effective chemotherapeutics against cancer. They are glycoside drugs comprising the amino sugar daunosamine linked to a hydroxy anthraquinone aglycone, and act by DNA intercalation, oxidative stress generation and topoisomerase II poisoning. Regardless of their therapeutic value, multidrug resistance and severe cardiotoxicity are important limitations of anthracycline treatment that have prompted the discovery of novel analogues. This review covers the most clinically relevant anthracyclines and their development over decades, since the first discovered natural prototypes to recent semisynthetic and synthetic derivatives. These include registered drugs, drug candidates undergoing clinical trials, and compounds under pre-clinical investigation. The impact of the structural modifications on antitumour activity, toxicity and resistance profile is addressed.
Collapse
Affiliation(s)
- Maristela B Martins-Teixeira
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo Avenida do Café s/n Monte Alegre, Ribeirão Preto, 14040903, Brazil
| | - Ivone Carvalho
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo Avenida do Café s/n Monte Alegre, Ribeirão Preto, 14040903, Brazil
| |
Collapse
|
17
|
Najjar A, Najjar A, Karaman R. Newly Developed Prodrugs and Prodrugs in Development; an Insight of the Recent Years. Molecules 2020; 25:E884. [PMID: 32079289 PMCID: PMC7070911 DOI: 10.3390/molecules25040884] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/06/2020] [Accepted: 02/10/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The design and development of prodrugs is the most common and effective strategy to overcome pharmacokinetic and pharmacodynamic drawbacks of active drugs. A respected number of prodrugs have been reached the drugs market throughout history and the recent years have witnessed a significant increase in the use of prodrugs as a replacement of their parent drugs for an efficient treatment of various ailment. METHODS A Scan conducted to find recent approved prodrugs and prodrugs in development. RESULTS Selected prodrugs were reported and categorized in accordance to their target systems. CONCLUSIONS the prodrug approach has shown many successes and still remains a viable and effective approach to deliver new active agents. This conclusion is supported by the recent approved prodrugs and the scan of clinical trials conducted between 2013-2018.
Collapse
Affiliation(s)
- Anas Najjar
- Faculty of Pharmacy, Department of Bioorganic & Pharmaceutical Chemistry, Al-Quds University, Jerusalem P.O. Box 20002, Palestine;
| | - Abderrahman Najjar
- Institute of Pathology, Rabin Medical Centre, PetachTikva 49100, Israel;
| | - Rafik Karaman
- Faculty of Pharmacy, Department of Bioorganic & Pharmaceutical Chemistry, Al-Quds University, Jerusalem P.O. Box 20002, Palestine;
| |
Collapse
|
18
|
Liu H, Bolleddula J, Nichols A, Tang L, Zhao Z, Prakash C. Metabolism of bioconjugate therapeutics: why, when, and how? Drug Metab Rev 2020; 52:66-124. [PMID: 32045530 DOI: 10.1080/03602532.2020.1716784] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Bioconjugation of therapeutic agents has been used as a selective drug delivery platform for many therapeutic areas. Bioconjugates are prepared by the covalent linkage of active compounds (small or large molecule) to a carrier molecule (lipids, proteins, peptides, carbohydrates, and polymers) through a chemical linker. The linkage of the active component to a carrier molecule enhances the therapeutic window through a targeted delivery and by reducing toxicity. Bioconjugates also possess improved pharmacokinetic properties such as a long half-life, increased stability, and cleavage by intracellular enzymes/environment. However, premature cleavage of the bioconjugates and the resulting metabolites/catabolites may produce undesirable toxic effects and, hence, it is critical to understand cleavage mechanisms, metabolism of bioconjugates, and translatability to human in the discovery stages. This article provides a comprehensive overview of linker cleavage pathways and catabolism/metabolism of antibody-drug conjugates, glycoconjugates, polymer-drug conjugates, lipid-drug conjugates, folate-targeted small molecule-drug conjugates, and drug-drug conjugates.
Collapse
Affiliation(s)
- Hanlan Liu
- KSQ Therapeutics Inc., Cambridge, MA, USA
| | | | | | | | | | | |
Collapse
|
19
|
Chamberlain FE, Jones RL, Chawla SP. Aldoxorubicin in soft tissue sarcomas. Future Oncol 2019; 15:1429-1435. [DOI: 10.2217/fon-2018-0922] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aldoxorubicin is a prodrug formulation of doxorubicin currently under investigation for the treatment of soft tissue sarcomas. Early studies have demonstrated a promising reduction in the cardiotoxicity of aldoxorubicin compared with equivalent doses of doxorubicin leading to an increase in the equivalent cumulative dose of aldoxorubicin. The current clinical and pharmacological data available for aldoxorubicin are extremely promising for its use in the treatment of advanced and metastatic soft tissue sarcomas compared with equivalent doses of doxorubicin although Phase III data are lacking. We review aldoxorubicin for the treatment of advanced and metastatic soft tissue sarcomas and discuss the impact it may have in the future.
Collapse
Affiliation(s)
- Florence E Chamberlain
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, Fulham Road, London, SW3 6JJ, UK
- The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
- Sarcoma Oncology Center, 2811 Wilshire Blvd #414, Santa Monica, CA 90403, USA
| | - Robin L Jones
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, Fulham Road, London, SW3 6JJ, UK
- The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
- Sarcoma Oncology Center, 2811 Wilshire Blvd #414, Santa Monica, CA 90403, USA
| | - Sant P Chawla
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, Fulham Road, London, SW3 6JJ, UK
- The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
- Sarcoma Oncology Center, 2811 Wilshire Blvd #414, Santa Monica, CA 90403, USA
| |
Collapse
|
20
|
Cranmer LD. Spotlight on aldoxorubicin (INNO-206) and its potential in the treatment of soft tissue sarcomas: evidence to date. Onco Targets Ther 2019; 12:2047-2062. [PMID: 30936721 PMCID: PMC6430065 DOI: 10.2147/ott.s145539] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Anthracyclines, and doxorubicin in particular, remain a mainstay of sarcoma therapy. Despite modest activity and significant toxicities, no cytotoxic monotherapy has yet yielded superior overall survival over doxorubicin for therapy of advanced soft tissue sarcomas in a randomized trial. Similarly, combination regimens have also been unable to overcome doxorubicin in terms of overall survival. Strategies to ameliorate the most prominent side effect of doxorubicin, cardiotoxicity, are available, but their use in sarcoma patients has been limited. Aldoxorubicin is a prodrug consisting of doxorubicin with a covalent linker. It binds rapidly after intravenous infusion to cysteine-34 of human serum albumin. The drug-albumin conjugate is preferentially retained in tumor tissue, with uptake into tumoral cells. At physiologic pH, the complex is stable. Hydrolysis can occur under the acidic conditions of the endocytic lysosome, releasing doxorubicin. Doxorubicin then distributes to various cellular compartments, including Golgi, mitochondrion, and nucleus, with subsequent cytotoxic effects. Aldoxorubicin has demonstrated in vitro and in vivo activities in both cancer model systems and human xenografts. Preclinical models also support its decreased cardiac effects vs doxorubicin, although such promising results require formal comparison at efficacy equivalent doses of the two drugs. Phase I studies confirmed the tolerability of aldoxorubicin in humans. Clinical cardiotoxicity was not observed, but molecular and subclinical cardiac effects could be demonstrated. A Phase II study in treatment-naïve, advanced sarcoma patients demonstrated improved progression-free survival and response rate over doxorubicin, although no survival benefit was evident. A Phase III study of aldoxorubicin vs investigator's choice from a panel of chemotherapy regimens in the salvage setting was unable to demonstrate a benefit in progression-free or overall survival in the entire population. Progression-free survival in L-sarcomas (leiomyosarcomas and liposarcomas) was documented. While evidence of subclinical cardiac effects was seen in a small proportion of aldoxorubicin-treated patients, data from both the Phase II and III studies indicated a favorable cardiotoxicity profile vs doxorubicin. Despite the negative results from this Phase III study, the importance of anthracycline therapy in sarcoma management merits further investigation of the potential role of aldoxorubicin in this indication. Other avenues for progress include identification of sensitive histologies and biomarkers of activity, exploration of clinical niches without proven standard therapies, and exploration of alternate dosing strategies.
Collapse
Affiliation(s)
- Lee D Cranmer
- Division of Medical Oncology, University of Washington, Seattle, WA, USA,
| |
Collapse
|
21
|
Coyne CP, Narayanan L. Anti-neoplastic cytotoxicity by complementary simultaneous selective “targeted” delivery for pulmonary adenocarcinoma: fludarabine-(5′-phosphoramidate)-[anti-IGF-1R] in dual-combination with dexamethasone-(C21-phosphoramidate)-[anti-EGFR]. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2018. [DOI: 10.1007/s40005-018-0401-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
22
|
Zhang H, Sun Z, Wang K, Li N, Chen H, Tan X, Li L, He Z, Sun J. Multifunctional Tumor-Targeting Cathepsin B-Sensitive Gemcitabine Prodrug Covalently Targets Albumin in Situ and Improves Cancer Therapy. Bioconjug Chem 2018; 29:1852-1858. [DOI: 10.1021/acs.bioconjchem.8b00223] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Huicong Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, No. 59 Mailbox, No. 103 Wenhua Road, Shenyang, Liaoning 110016, P. R. China
| | - Zhisu Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, No. 59 Mailbox, No. 103 Wenhua Road, Shenyang, Liaoning 110016, P. R. China
| | - Kuanglei Wang
- Wuyi University, Jiangmen, Guangdong 529020, P. R. China
- International Healthcare Innovation Institute (Jiangmen), Jiangmen, Guangdong 529080, P. R. China
| | - Na Li
- Clinical Pharmacy, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Hongxiang Chen
- Center for Drug Evaluation, China Food and Drug Administration, Beijing 100022, P. R. China
| | - Xiao Tan
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
| | - Lingxiao Li
- School of Pharmacy, Queen’s University of Belfast, 97 Lisburn Road, Belfast BT9 7BL, Northern Ireland, U.K
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110013, P. R. China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, No. 59 Mailbox, No. 103 Wenhua Road, Shenyang, Liaoning 110016, P. R. China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, No. 59 Mailbox, No. 103 Wenhua Road, Shenyang, Liaoning 110016, P. R. China
| |
Collapse
|
23
|
Hoogenboezem EN, Duvall CL. Harnessing albumin as a carrier for cancer therapies. Adv Drug Deliv Rev 2018; 130:73-89. [PMID: 30012492 PMCID: PMC6200408 DOI: 10.1016/j.addr.2018.07.011] [Citation(s) in RCA: 373] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 07/10/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022]
Abstract
Serum albumin, a natural ligand carrier that is highly concentrated and long-circulating in the blood, has shown remarkable promise as a carrier for anti-cancer agents. Albumin is able to prolong the circulation half-life of otherwise rapidly cleared drugs and, importantly, promote their accumulation within tumors. The applications for using albumin as a cancer drug carrier are broad and include both traditional cancer chemotherapeutics and new classes of biologics. Strategies for leveraging albumin for drug delivery can be classified broadly into exogenous and in situ binding formulations that utilize covalent attachment, non-covalent association, or encapsulation in albumin-based nanoparticles. These methods have shown remarkable preclinical and clinical successes that are examined in this review.
Collapse
Affiliation(s)
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN.
| |
Collapse
|
24
|
Albumin-covered lipid nanocapsules exhibit enhanced uptake performance by breast-tumor cells. Colloids Surf B Biointerfaces 2018; 165:103-110. [DOI: 10.1016/j.colsurfb.2018.02.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/06/2018] [Accepted: 02/11/2018] [Indexed: 01/01/2023]
|
25
|
Gong J, Yan J, Forscher C, Hendifar A. Aldoxorubicin: a tumor-targeted doxorubicin conjugate for relapsed or refractory soft tissue sarcomas. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:777-786. [PMID: 29670334 PMCID: PMC5896668 DOI: 10.2147/dddt.s140638] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite available therapies after initial systemic therapy, prognosis remains poor in relapsed or refractory soft tissue sarcomas (STS). The rational and clinical development of novel agents to improve outcomes in this area of high unmet need is desperately warranted. Aldoxorubicin is a prodrug of doxorubicin that binds to serum albumin immediately after administration through an acid-sensitive hydrazone linker and is subsequently transported to tumor tissues where the acidic environment cleaves the linker and facilitates delivery of a tumor-targeted drug payload. In clinical studies to date, there has been evidence of efficacy and mitigated cardiac toxicity. In this review, we comprehensively detail the clinical development of aldoxorubicin in STS to date. Specifically, we highlight available data on the pharmacokinetics and efficacy from Phase I, Phase II, and Phase III trials in advanced or metastatic STS. We conclude with considerations for future directions of investigation for this promising antitumor agent.
Collapse
Affiliation(s)
- Jun Gong
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Jessica Yan
- Department of Hematology and Oncology, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Charles Forscher
- Sarcoma Program, Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Andrew Hendifar
- Gastrointestinal and Neuroendocrine Malignancies, Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
26
|
Zhang Y, Sun T, Jiang C. Biomacromolecules as carriers in drug delivery and tissue engineering. Acta Pharm Sin B 2018; 8:34-50. [PMID: 29872621 PMCID: PMC5985630 DOI: 10.1016/j.apsb.2017.11.005] [Citation(s) in RCA: 258] [Impact Index Per Article: 36.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/05/2017] [Accepted: 10/07/2017] [Indexed: 12/14/2022] Open
Abstract
Natural biomacromolecules have attracted increased attention as carriers in biomedicine in recent years because of their inherent biochemical and biophysical properties including renewability, nontoxicity, biocompatibility, biodegradability, long blood circulation time and targeting ability. Recent advances in our understanding of the biological functions of natural-origin biomacromolecules and the progress in the study of biological drug carriers indicate that such carriers may have advantages over synthetic material-based carriers in terms of half-life, stability, safety and ease of manufacture. In this review, we give a brief introduction to the biochemical properties of the widely used biomacromolecule-based carriers such as albumin, lipoproteins and polysaccharides. Then examples from the clinic and in recent laboratory development are summarized. Finally the current challenges and future prospects of present biological carriers are discussed.
Collapse
Key Words
- ABD, albumin binding domain
- ACM, aclacinomycin
- ACS, absorbable collagen sponge
- ADH, adipic dihydrazide
- ART, artemisinin
- ASF, Antheraea mylitta silk fibroin
- ATRA, all-trans retinoic acid
- ATS, artesunate
- BCEC, brain capillary endothelial cells
- BMP-2, bone morphogenetic protein-2
- BSA, bovine serum albumin
- BSF, Bombyx mori silk fibroin
- Biomacromolecule
- CC-HAM, core-crosslinked polymeric micelle based hyaluronic acid
- CD, cyclodextrin
- CD-NPs, amphiphilic MMA–tBA β-CD star copolymers that are capable of forming nanoparticles
- CD-g-CS, chitosan grafted with β-cyclodextrin
- CD/BP, cyclodextrin–bisphosphonate complexes
- CIA, collagen-induced arthritis
- CM, collagen matrices
- CMD-ChNP, carboxylmethyl dextran chitosan nanoparticle
- DHA, dihydroartesunate
- DOXO-EMCH, (6-maleimidocaproyl)hydrazone derivative of doxorubicin
- DOX–TRF, doxorubincin–transferrin conjugate
- DTX-HPLGA, HA coated PLGA nanoparticulate docetaxel
- Drug delivery
- ECM, extracellular matrix
- EMT, epithelial mesenchymal transition
- EPR, enhanced permeability and retention
- FcRn, neonatal Fc receptor
- GAG, glycosaminoglycan
- GC-DOX, glycol–chitosan–doxorubicin conjugate
- GDNF, glial-derived neurotrophic factor
- GO, grapheme oxide
- GSH, glutathione
- Gd, gadolinium
- HA, hyaluronic acid
- HA-CA, catechol-modified hyaluronic acid
- HCF, heparin-conjugated fibrin
- HDL, high density lipoprotein
- HEK, human embryonic kidney
- HSA, human serum albumin
- IDL, intermediate density lipoprotein
- INF, interferon
- LDL, low density lipoprotein
- LDLR, low density lipoprotein receptor
- LDV, leucine–aspartic acid–valine
- LMWH, low molecular weight heparin
- MSA, mouse serum albumin
- MTX–HSA, methotrexate–albumin conjugate
- NIR, near-infrared
- NSCLC, non-small cell lung cancer
- OP-Gel-NS, oxidized pectin-gelatin-nanosliver
- PEC, polyelectrolyte
- PTX, paclitaxel
- Polysaccharide
- Protein
- RES, reticuloendothelial system
- RGD, Arg–Gly–Asp peptide
- SF, silk fibroin
- SF-CSNP, silk fibroin modified chitosan nanoparticle
- SFNP, silk fibroin nanoparticle
- SPARC, secreted protein acidic and rich in cysteine
- TRAIL, tumor-necrosis factor-related apoptosis-inducing ligand
- Tf, transferrin
- TfR, transferrin receptor
- Tissue engineering
- VEGF, vascular endothelial growth factor
- VLDL, very low density lipoprotein
- pDNA, plasmid DNA
- rHDL, recombinant HDL
- rhEGF-2/HA, recombinant human fibroblast growth factor type 2 in a hyaluronic acid carrier
Collapse
Affiliation(s)
| | | | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 200032, China
| |
Collapse
|
27
|
Sachdev E, Sachdev D, Mita M. Aldoxorubicin for the treatment of soft tissue sarcoma. Expert Opin Investig Drugs 2017; 26:1175-1179. [DOI: 10.1080/13543784.2017.1371134] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Esha Sachdev
- Cedars-Sinai Medical Center, Samuel Oschin Comprehensive Cancer Institute, Medicine, Los Angeles, CA, USA
| | - Divesh Sachdev
- Cedars-Sinai Medical Center, Samuel Oschin Comprehensive Cancer Institute, Medicine, Los Angeles, CA, USA
| | - Monica Mita
- Cedars-Sinai Medical Center, Samuel Oschin Comprehensive Cancer Institute, Medicine, Los Angeles, CA, USA
| |
Collapse
|
28
|
Liu J, Yan F, Chen H, Wang W, Liu W, Hao K, Wang G, Zhou F, Zhang J. A novel individual-cell-based mathematical model based on multicellular tumour spheroids for evaluating doxorubicin-related delivery in avascular regions. Br J Pharmacol 2017; 174:2862-2879. [PMID: 28608595 DOI: 10.1111/bph.13909] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 05/25/2017] [Accepted: 06/05/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Effective drug delivery in the avascular regions of tumours, which is crucial for the promising antitumour activity of doxorubicin-related therapy, is governed by two inseparable processes: intercellular diffusion and intracellular retention. To accurately evaluate doxorubicin-related delivery in the avascular regions, these two processes should be assessed together. Here we describe a new approach to such an assessment. EXPERIMENTAL APPROACH An individual-cell-based mathematical model based on multicellular tumour spheroids was developed that describes the different intercellular diffusion and intracellular retention kinetics of doxorubicin in each cell layer. The different effects of a P-glycoprotein inhibitor (LY335979) and a hypoxia inhibitor (YC-1) were quantitatively evaluated and compared, in vitro (tumour spheroids) and in vivo (HepG2 tumours in mice). This approach was further tested by evaluating in these models, an experimental doxorubicin derivative, INNO 206, which is in Phase II clinical trials. KEY RESULTS Inhomogeneous, hypoxia-induced, P-glycoprotein expression compromised active transport of doxorubicin in the central area, that is, far from the vasculature. LY335979 inhibited efflux due to P-glycoprotein but limited levels of doxorubicin outside the inner cells, whereas YC-1 co-administration specifically increased doxorubicin accumulation in the inner cells without affecting the extracellular levels. INNO 206 exhibited a more effective distribution profile than doxorubicin. CONCLUSIONS AND IMPLICATIONS The individual-cell-based mathematical model accurately evaluated and predicted doxorubicin-related delivery and regulation in the avascular regions of tumours. The described framework provides a mechanistic basis for the proper development of doxorubicin-related drug co-administration profiles and nanoparticle development and could avoid unnecessary clinical trials.
Collapse
Affiliation(s)
- Jiali Liu
- Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Fangrong Yan
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Hongzhu Chen
- Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wenjie Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wenyue Liu
- Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Kun Hao
- Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Guangji Wang
- Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China.,Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Fang Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jingwei Zhang
- Key Lab of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
29
|
Burade V, Bhowmick S, Maiti K, Zalawadia R, Ruan H, Thennati R. Lipodox® (generic doxorubicin hydrochloride liposome injection): in vivo efficacy and bioequivalence versus Caelyx® (doxorubicin hydrochloride liposome injection) in human mammary carcinoma (MX-1) xenograft and syngeneic fibrosarcoma (WEHI 164) mouse models. BMC Cancer 2017; 17:405. [PMID: 28587612 PMCID: PMC5461687 DOI: 10.1186/s12885-017-3377-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 05/19/2017] [Indexed: 11/14/2022] Open
Abstract
Background Doxorubicin (DXR) hydrochloride (HCl) liposome injection is an important part of the treatment armamentarium for a number of cancers. With growing needs for affordable and effective anticancer treatments, the development of generics is becoming increasingly important to facilitate patient access to vital medications. We conducted studies in relevant mouse models of cancer to compare the preclinical antitumour efficacy and plasma pharmacokinetic profile of a proposed generic DXR HCl liposome injection developed by Sun Pharmaceutical Industries Ltd. (SPIL DXR HCl liposome injection) with Caelyx® (reference DXR HCl liposome injection). Methods Syngeneic fibrosarcoma (WEHI 164)-bearing BALB/c mice and athymic nude mice transplanted with MX-1 human mammary carcinoma xenografts were treated with SPIL DXR HCl liposome injection, reference DXR HCl liposome injection or placebo, to compare tumour volume, antitumour activity (percentage test/control [%T/C] ratio, tumour regression, and specific tumour growth delay) and toxicity (survival and weight changes) in response to treatment. The pharmacokinetic profile of the SPIL and reference product was also studied in syngeneic fibrosarcoma-bearing mice. Results Treatment with either SPIL or reference DXR HCl liposome injection resulted in significant reduction in tumour volume from baseline in both models at all doses tested. High antitumour activity (%T/C ≤ 10) was seen from Day 21 and Day 14 onwards in SPIL and reference DXR HCl liposome injection–treated syngeneic fibrosarcoma-bearing mice, respectively, at 9 mg/kg. Moderate antitumour activity (%T/C ≤ 20) was seen from Day 17 and Day 24 onwards in SPIL and reference DXR HCl liposome injection–treated MX-1-bearing mice, respectively, at 6 mg/kg. No significant differences in tumour volume and %T/C were observed between SPIL and reference DXR HCl liposome injection–treated groups at any dose (p ≥ 0.05). Toxicity profiles were considered to be generally comparable. Evaluation of test/reference (A/B) ratios and 90% confidence intervals (CIs) for peak serum concentration (Cmax) and area under the curve (AUC0-t, and AUC0-∞) demonstrated bioequivalence of SPIL and reference DXR HCl liposome injections. Conclusions Establishing similarity is of critical importance during the development of generic treatments. SPIL and reference DXR HCl liposome injections were shown to be comparable with regards to antitumour activity, toxicity and pharmacokinetics.
Collapse
Affiliation(s)
- Vinod Burade
- Sun Pharma Advanced Research Company Ltd., 17 B Mahal Industrial Estate, Mahakali Caves Road, Andheri (East), Mumbai, Maharashtra, 400 093, India. .,Sun Pharma Advanced Research Centre (SPARC), Tandalja, Vadodara, Gujarat, 390 020, India.
| | - Subhas Bhowmick
- Sun Pharmaceutical Industries Ltd., Sun Pharma Advanced Research Centre (SPARC), Tandalja, Vadodara, Gujarat, 390 020, India
| | - Kuntal Maiti
- Sun Pharma Advanced Research Company Ltd., 17 B Mahal Industrial Estate, Mahakali Caves Road, Andheri (East), Mumbai, Maharashtra, 400 093, India
| | - Rishit Zalawadia
- Sun Pharma Advanced Research Company Ltd., 17 B Mahal Industrial Estate, Mahakali Caves Road, Andheri (East), Mumbai, Maharashtra, 400 093, India
| | - Harry Ruan
- Sun Pharma Advanced Research Company Ltd., 17 B Mahal Industrial Estate, Mahakali Caves Road, Andheri (East), Mumbai, Maharashtra, 400 093, India
| | - Rajamannar Thennati
- Sun Pharmaceutical Industries Ltd., Sun Pharma Advanced Research Centre (SPARC), Tandalja, Vadodara, Gujarat, 390 020, India
| |
Collapse
|
30
|
Hrynchak I, Sousa E, Pinto M, Costa VM. The importance of drug metabolites synthesis: the case-study of cardiotoxic anticancer drugs. Drug Metab Rev 2017; 49:158-196. [DOI: 10.1080/03602532.2017.1316285] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ivanna Hrynchak
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
| | - Emília Sousa
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
- CIIMAR – Centro Interdisciplinar de Investigação Marinha e Ambiental, Matosinhos, Portugal
| | - Madalena Pinto
- Laboratório de Química Orgânica e Farmacêutica, Departamento de Ciências Químicas, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
- CIIMAR – Centro Interdisciplinar de Investigação Marinha e Ambiental, Matosinhos, Portugal
| | - Vera Marisa Costa
- Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, UCIBIO, REQUIMTE (Rede de Química e Tecnologia), Universidade do Porto, Porto, Portugal
| |
Collapse
|
31
|
Burade V, Bhowmick S, Maiti K, Zalawadia R, Jain D, Rajamannar T. Comparative plasma and tissue distribution of Sun Pharma's generic doxorubicin HCl liposome injection versus Caelyx ® (doxorubicin HCl liposome injection) in syngeneic fibrosarcoma-bearing BALB/c mice and Sprague-Dawley rats. Cancer Chemother Pharmacol 2017; 79:899-913. [PMID: 28349166 PMCID: PMC5403873 DOI: 10.1007/s00280-017-3278-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 03/01/2017] [Indexed: 11/26/2022]
Abstract
Purpose The liposomal formulation of doxorubicin [doxorubicin (DXR) hydrochloride (HCl) liposome injection, Caelyx®] alters the tissue distribution of DXR as compared with nonliposomal DXR, resulting in an improved benefit-risk profile. We conducted studies in murine models to compare the plasma and tissue distribution of a proposed generic DXR HCl liposome injection developed by Sun Pharmaceuticals Industries Limited (SPIL DXR HCl liposome injection) with Caelyx®. Methods The plasma and tissue distributions of the SPIL and reference DXR HCl liposome injections were compared in syngeneic fibrosarcoma-bearing BALB/c mice and Sprague–Dawley rats. Different batches and different lots of the same batch of the reference product were also compared with each other. Results The SPIL and reference DXR HCl liposome injections exhibited generally comparable plasma and tissue distribution profiles in both models. While minor differences were observed between the two products in some tissues, different batches and lots of the reference product also showed some differences in the distribution of various analytes in some tissues. The ratios of estimated free to encapsulated DXR for plasma and tissue were generally comparable between the SPIL and reference DXR HCl liposome injections in both models, indicating similar extents of absorption into the tissues and similar rates of drug release from liposomes. Conclusions The plasma and tissue distribution profiles of the SPIL and reference DXR HCl liposome injections were shown to be generally comparable. Inconsistencies between the products observed in some tissues were thought to be due to biological variation.
Collapse
Affiliation(s)
- Vinod Burade
- Sun Pharma Advanced Research Company Ltd., 17 B Mahal Industrial Estate, Mahakali Caves Road, Andheri (East), Mumbai, Maharashtra 400 093 India
| | - Subhas Bhowmick
- Sun Pharmaceutical Industries Ltd., Sun Pharma Advanced Research Centre (SPARC), Tandalja, Vadodara, Gujarat 390 020 India
| | - Kuntal Maiti
- Sun Pharma Advanced Research Company Ltd., 17 B Mahal Industrial Estate, Mahakali Caves Road, Andheri (East), Mumbai, Maharashtra 400 093 India
| | - Rishit Zalawadia
- Sun Pharma Advanced Research Company Ltd., 17 B Mahal Industrial Estate, Mahakali Caves Road, Andheri (East), Mumbai, Maharashtra 400 093 India
| | - Deepak Jain
- Sun Pharmaceutical Industries Ltd., Sun Pharma Advanced Research Centre (SPARC), Tandalja, Vadodara, Gujarat 390 020 India
| | - Thennati Rajamannar
- Sun Pharmaceutical Industries Ltd., Sun Pharma Advanced Research Centre (SPARC), Tandalja, Vadodara, Gujarat 390 020 India
| |
Collapse
|
32
|
Coyne CP, Narayanan L. Gemcitabine-(5'-phosphoramidate)-[anti-IGF-1R]: molecular design, synthetic organic chemistry reactions, and antineoplastic cytotoxic potency in populations of pulmonary adenocarcinoma (A549). Chem Biol Drug Des 2017; 89:379-399. [PMID: 27561602 PMCID: PMC5396302 DOI: 10.1111/cbdd.12845] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 08/12/2016] [Accepted: 08/19/2016] [Indexed: 02/06/2023]
Abstract
One molecular-based approach that increases potency and reduces dose-limited sequela is the implementation of selective 'targeted' delivery strategies for conventional small molecular weight chemotherapeutic agents. Descriptions of the molecular design and organic chemistry reactions that are applicable for synthesis of covalent gemcitabine-monophosphate immunochemotherapeutics have to date not been reported. The covalent immunopharmaceutical, gemcitabine-(5'-phosphoramidate)-[anti-IGF-1R] was synthesized by reacting gemcitabine with a carbodiimide reagent to form a gemcitabine carbodiimide phosphate ester intermediate which was subsequently reacted with imidazole to create amine-reactive gemcitabine-(5'-phosphorylimidazolide) intermediate. Monoclonal anti-IGF-1R immunoglobulin was combined with gemcitabine-(5'-phosphorylimidazolide) resulting in the synthetic formation of gemcitabine-(5'-phosphoramidate)-[anti-IGF-1R]. The gemcitabine molar incorporation index for gemcitabine-(5'-phosphoramidate)-[anti-IGF-R1] was 2.67:1. Cytotoxicity Analysis - dramatic increases in antineoplastic cytotoxicity were observed at and between the gemcitabine-equivalent concentrations of 10-9 M and 10-7 M where lethal cancer cell death increased from 0.0% to a 93.1% maximum (100.% to 6.93% residual survival), respectively. Advantages of the organic chemistry reactions in the multistage synthesis scheme for gemcitabine-(5'-phosphoramidate)-[anti-IGF-1R] include their capacity to achieve high chemotherapeutic molar incorporation ratios; option of producing an amine-reactive chemotherapeutic intermediate that can be preserved for future synthesis applications; and non-dedicated organic chemistry reaction scheme that allows substitutions of either or both therapeutic moieties, and molecular delivery platforms.
Collapse
Affiliation(s)
- Cody P. Coyne
- Department of Basic SciencesCollege of Veterinary MedicineWise CenterMississippi State UniversityMississippi StateMSUSA
- College of Veterinary MedicineWise CenterMississippi State UniversityMississippi StateMSUSA
| | - Lakshmi Narayanan
- Department of Basic SciencesCollege of Veterinary MedicineWise CenterMississippi State UniversityMississippi StateMSUSA
- College of Veterinary MedicineWise CenterMississippi State UniversityMississippi StateMSUSA
- Present address: Fishery and Wildlife Research CenterMississippi State UniversityLocksley Way 201Mississippi StateMSUSA
| |
Collapse
|
33
|
Hattinger CM, Fanelli M, Tavanti E, Vella S, Riganti C, Picci P, Serra M. Doxorubicin-resistant osteosarcoma: novel therapeutic approaches in sight? Future Oncol 2017; 13:673-677. [PMID: 28183198 DOI: 10.2217/fon-2016-0519] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Claudia M Hattinger
- Pharmacogenomics & Pharmacogenetics Research Unit, Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Marilù Fanelli
- Pharmacogenomics & Pharmacogenetics Research Unit, Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Elisa Tavanti
- Pharmacogenomics & Pharmacogenetics Research Unit, Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Serena Vella
- Pharmacogenomics & Pharmacogenetics Research Unit, Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Chiara Riganti
- Department of Oncology & Center for Experimental Research & Medical Studies (CeRMS), University of Torino, via Santena, 5/bis, 10126, Torino, Italy
| | - Piero Picci
- Pharmacogenomics & Pharmacogenetics Research Unit, Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Massimo Serra
- Pharmacogenomics & Pharmacogenetics Research Unit, Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, via di Barbiano 1/10, 40136 Bologna, Italy
| |
Collapse
|
34
|
Coyne CP, Narayanan L. Dexamethasone-(C21-phosphoramide)-[anti-EGFR]: molecular design, synthetic organic chemistry reactions, and antineoplastic cytotoxic potency against pulmonary adenocarcinoma (A549). Drug Des Devel Ther 2016; 10:2575-97. [PMID: 27574398 PMCID: PMC4990379 DOI: 10.2147/dddt.s102075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
PURPOSE Corticosteroids are effective in the management of a variety of disease states, such as several forms of neoplasia (leukemia and lymphoma), autoimmune conditions, and severe inflammatory responses. Molecular strategies that selectively "target" delivery of corticosteroids minimize or prevents large amounts of the pharmaceutical moiety from passively diffusing into normal healthy cell populations residing within tissues and organ systems. MATERIALS AND METHODS The covalent immunopharmaceutical, dexamethasone-(C21-phosphoramide)-[anti-EGFR] was synthesized by reacting dexamethasone-21-monophosphate with a carbodiimide reagent to form a dexamethasone phosphate carbodiimide ester that was subsequently reacted with imidazole to create an amine-reactive dexamethasone-(C21-phosphorylimidazolide) intermediate. Monoclonal anti-EGFR immunoglobulin was combined with the amine-reactive dexamethasone-(C21-phosphorylimidazolide) intermediate, resulting in the synthesis of the covalent immunopharmaceutical, dexamethasone-(C21-phosphoramide)-[anti-EGFR]. Following spectrophotometric analysis and validation of retained epidermal growth factor receptor type 1 (EGFR)-binding avidity by cell-ELISA, the selective anti-neoplasic cytotoxic potency of dexamethasone-(C21-phosphoramide)-[anti-EGFR] was established by MTT-based vitality stain methodology using adherent monolayer populations of human pulmonary adenocarcinoma (A549) known to overexpress the tropic membrane receptors EGFR and insulin-like growth factor receptor type 1. RESULTS The dexamethasone:IgG molar-incorporation-index for dexamethasone-(C21-phosphoramide)-[anti-EGFR] was 6.95:1 following exhaustive serial microfiltration. Cytotoxicity analysis: covalent bonding of dexamethasone to monoclonal anti-EGFR immunoglobulin did not significantly modify the ex vivo antineoplastic cytotoxicity of dexamethasone against pulmonary adenocarcinoma at and between the standardized dexamethasone equivalent concentrations of 10(-9) M and 10(-5) M. Rapid increases in antineoplastic cytotoxicity were observed at and between the dexamethasone equivalent concentrations of 10(-9) M and 10(-7) M where cancer cell death increased from 7.7% to a maximum of 64.9% (92.3%-35.1% residual survival), respectively, which closely paralleled values for "free" noncovalently bound dexamethasone. DISCUSSION Organic chemistry reaction regimens were optimized to develop a multiphase synthesis regimen for dexamethasone-(C21-phosphoramide)-[anti-EGFR]. Attributes of dexamethasone-(C21-phosphoramide)-[anti-EGFR] include a high dexamethasone molar incorporation-index, lack of extraneous chemical group introduction, retained EGFR-binding avidity ("targeted" delivery properties), and potential to enhance long-term pharmaceutical moiety effectiveness.
Collapse
Affiliation(s)
| | - Lakshmi Narayanan
- Department of Clinical Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS, USA
| |
Collapse
|
35
|
Selective delivery of doxorubicin by novel stimuli-sensitive nano-ferritins overcomes tumor refractoriness. J Control Release 2016; 239:10-8. [PMID: 27524282 DOI: 10.1016/j.jconrel.2016.08.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 08/04/2016] [Accepted: 08/09/2016] [Indexed: 12/12/2022]
Abstract
Human ferritin heavy chain (HFt) has been demonstrated to possess considerable potential for targeted delivery of drugs and diagnostic agents to cancer cells. Here, we report the development of a novel HFt-based genetic construct (HFt-MP-PAS) containing a short peptide linker (MP) between each HFt subunit and an outer shielding polypeptide sequence rich in proline (P), serine (S) and alanine (A) residues (PAS). The peptide linker contains a matrix-metalloproteinases (MMPs) cleavage site that permits the protective PAS shield to be removed by tumor-driven proteolytic cleavage within the tumor microenvironment. For the first time HFt-MP-PAS ability to deliver doxorubicin to cancer cells, subcellular localization, and therapeutic efficacy on a xenogeneic mouse model of a highly refractory to conventional chemotherapeutics type of cancer were evaluated. HFt-MP-PAS-DOXO performance was compared with the novel albumin-based drug delivery system INNO-206, currently in phase III clinical trials. The results of this work provide solid evidence indicating that the stimuli-sensitive, long-circulating HFt-MP-PAS nanocarriers described herein have the potential to be exploited in cancer therapy.
Collapse
|
36
|
Shaikh AB, Li F, Li M, He B, He X, Chen G, Guo B, Li D, Jiang F, Dang L, Zheng S, Liang C, Liu J, Lu C, Liu B, Lu J, Wang L, Lu A, Zhang G. Present Advances and Future Perspectives of Molecular Targeted Therapy for Osteosarcoma. Int J Mol Sci 2016; 17:506. [PMID: 27058531 PMCID: PMC4848962 DOI: 10.3390/ijms17040506] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 03/30/2016] [Indexed: 12/20/2022] Open
Abstract
Osteosarcoma (OS) is a bone cancer mostly occurring in pediatric population. Current treatment regime of surgery and intensive chemotherapy could cure about 60%-75% patients with primary osteosarcoma, however only 15% to 30% can be cured when pulmonary metastasis or relapse has taken place. Hence, novel precise OS-targeting therapies are being developed with the hope of addressing this issue. This review summarizes the current development of molecular mechanisms and targets for osteosarcoma. Therapies that target these mechanisms with updated information on clinical trials are also reviewed. Meanwhile, we further discuss novel therapeutic targets and OS-targeting drug delivery systems. In conclusion, a full insight in OS pathogenesis and OS-targeting strategies would help us explore novel targeted therapies for metastatic osteosarcoma.
Collapse
Affiliation(s)
- Atik Badshah Shaikh
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Fangfei Li
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Min Li
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Department of Orthopaedic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China.
| | - Bing He
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Xiaojuan He
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Guofen Chen
- Orthopaedic Surgery Department, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Baosheng Guo
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Defang Li
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Feng Jiang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Lei Dang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Shaowei Zheng
- Department of Orthopaedic Surgery, the First Hospital of Huizhou, Huizhou 516000, China.
| | - Chao Liang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Jin Liu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Cheng Lu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Biao Liu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Jun Lu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Luyao Wang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Aiping Lu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Ge Zhang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| |
Collapse
|
37
|
Ross PL, Wolfe JL. Physical and Chemical Stability of Antibody Drug Conjugates: Current Status. J Pharm Sci 2016; 105:391-397. [DOI: 10.1016/j.xphs.2015.11.037] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/13/2015] [Accepted: 11/17/2015] [Indexed: 01/24/2023]
|
38
|
Liang C, Xu L, Song G, Liu Z. Emerging nanomedicine approaches fighting tumor metastasis: animal models, metastasis-targeted drug delivery, phototherapy, and immunotherapy. Chem Soc Rev 2016; 45:6250-6269. [DOI: 10.1039/c6cs00458j] [Citation(s) in RCA: 302] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nanomedicine approaches may bring new opportunities for tumor metastasis treatment.
Collapse
Affiliation(s)
- Chao Liang
- Institute of Functional Nano & Soft Materials (FUNSOM)
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices
- Soochow University
- Suzhou
- China
| | - Ligeng Xu
- Institute of Functional Nano & Soft Materials (FUNSOM)
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices
- Soochow University
- Suzhou
- China
| | - Guosheng Song
- Institute of Functional Nano & Soft Materials (FUNSOM)
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices
- Soochow University
- Suzhou
- China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM)
- Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices
- Soochow University
- Suzhou
- China
| |
Collapse
|
39
|
Falvo E, Tremante E, Arcovito A, Papi M, Elad N, Boffi A, Morea V, Conti G, Toffoli G, Fracasso G, Giacomini P, Ceci P. Improved Doxorubicin Encapsulation and Pharmacokinetics of Ferritin-Fusion Protein Nanocarriers Bearing Proline, Serine, and Alanine Elements. Biomacromolecules 2015; 17:514-22. [PMID: 26686226 DOI: 10.1021/acs.biomac.5b01446] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A novel human ferritin-based nanocarrier, composed of 24 modified monomers able to auto-assemble into a modified protein cage, was produced and used as selective carrier of anti-tumor payloads. Each modified monomer derives from the genetic fusion of two distinct modules, namely the heavy chain of human ferritin (HFt) and a stabilizing/protective PAS polypeptide sequence rich in proline (P), serine (S), and alanine (A) residues. Two genetically fused protein constructs containing PAS polymers with 40- and 75-residue lengths, respectively, were compared. They were produced and purified as recombinant proteins in Escherichia coli at high yields. Both preparations were highly soluble and stable in vitro as well as in mouse plasma. Size-exclusion chromatography, dynamic light scattering, and transmission electron microscopy results indicated that PASylated ferritins are fully assembled and highly monodispersed. In addition, yields and stability of encapsulated doxorubicin were significantly better for both HFt-PAS proteins than for wild-type HFt. Importantly, PAS sequences considerably prolonged the half-life of HFt in the mouse bloodstream. Finally, our doxorubicin-loaded nanocages preserved the pharmacological activity of the drug. Taken together, these results indicate that both of the developed HFt-PAS fusion proteins are promising nanocarriers for future applications in cancer therapy.
Collapse
Affiliation(s)
- Elisabetta Falvo
- Institute of Molecular Biology and Pathology CNR, National Research Council of Italy , 00185 Rome, Italy.,Department of Biochemical Sciences "A. Rossi Fanelli", University "Sapienza" , 00185 Rome, Italy
| | - Elisa Tremante
- Regina Elena National Cancer Institute , 00144 Rome, Italy
| | - Alessandro Arcovito
- Istituto di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore , Largo F. Vito 1, 00168 Rome, Italy
| | - Massimiliano Papi
- Istituto di Fisica, Università Cattolica del Sacro Cuore , Largo F. Vito 1, 00168, Rome, Italy
| | - Nadav Elad
- Department of Chemical Research Support, Weizmann Institute of Science , Rehovot 7610001, Israel
| | - Alberto Boffi
- Institute of Molecular Biology and Pathology CNR, National Research Council of Italy , 00185 Rome, Italy.,Department of Biochemical Sciences "A. Rossi Fanelli", University "Sapienza" , 00185 Rome, Italy.,Center for Life Nano Science at Sapienza, Istituto Italiano di Tecnologia (IIT) , 00161 Rome, Italy
| | - Veronica Morea
- Institute of Molecular Biology and Pathology CNR, National Research Council of Italy , 00185 Rome, Italy
| | - Giamaica Conti
- Department of Neurological, Biomedical and Movement Sciences, University of Verona , 37134 Verona, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, CRO Aviano National Cancer Institute , 33081Aviano (Pordenone), Italy
| | - Giulio Fracasso
- Department of Medicine, University of Verona , 37134 Verona, Italy
| | | | - Pierpaolo Ceci
- Institute of Molecular Biology and Pathology CNR, National Research Council of Italy , 00185 Rome, Italy
| |
Collapse
|
40
|
Emerging delivery systems to reduce doxorubicin cardiotoxicity and improve therapeutic index: focus on liposomes. Anticancer Drugs 2015; 26:241-58. [PMID: 25415656 DOI: 10.1097/cad.0000000000000182] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Anthracyclines are powerful anticancer agents and among the most important tools in the chemotherapy armamentarium of medical oncologists. They are approved for use in the treatment of a broad variety of solid and hematologic neoplasms. However, the usefulness of these agents, particularly doxorubicin, the most widely used anthracycline, is limited by considerable toxicity, especially damage to the cardiac muscle, which is cumulative and mostly irreversible, restricting extended use of this drug. In the last 30 years, extensive research with a variety of drug-delivery systems has attempted to overcome this limitation, with clinical success mostly confined to liposome formulations. Liposomal doxorubicin, and particularly pegylated liposomal doxorubicin, has shown significant pharmacologic advantages and an added clinical value over doxorubicin. Here, we review the mechanisms of action and toxicity of doxorubicin, and ways to reduce toxicity, with a focus on liposome-based drug-delivery systems.
Collapse
|
41
|
Hattinger CM, Fanelli M, Tavanti E, Vella S, Ferrari S, Picci P, Serra M. Advances in emerging drugs for osteosarcoma. Expert Opin Emerg Drugs 2015; 20:495-514. [PMID: 26021401 DOI: 10.1517/14728214.2015.1051965] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Osteosarcoma (OS), the most common primary malignant bone tumor, is currently treated with pre- and postoperative chemotherapy in association with the surgical removal of the tumor. Conventional treatments allow to cure about 60 - 65% of patients with primary tumors and only 20 - 25% of patients with recurrent disease. New treatment approaches and drugs are therefore highly warranted to improve prognosis. AREAS COVERED This review focuses on the therapeutic approaches that are under development or clinical evaluation in OS. Information was obtained from different and continuously updated data bases, as well as from literature searches, in which particular relevance was given to reports and reviews on new targeted therapies under clinical investigation in high-grade OS. EXPERT OPINION OS is a heterogeneous tumor, with a great variability in treatment response between patients. It is therefore unlikely that a single therapeutic tool will be uniformly successful for all OS patients. This claims for the validation of new treatment approaches together with biologic/(pharmaco)genetic markers, which may select the most appropriate subgroup of patients for each treatment approach. Since some promising novel agents and treatment strategies are currently tested in Phase I/II/III clinical trials, we may hope that new therapies with superior efficacy and safety profiles will be identified in the next few years.
Collapse
|
42
|
Muzammil Afzal S, Naidu VGM, Harishankar N, Kishan V. Albumin anchored docetaxel lipid nanoemulsion for improved targeting efficiency – preparation, characterization, cytotoxic, antitumor and in vivo imaging studies. Drug Deliv 2015; 23:1355-63. [DOI: 10.3109/10717544.2015.1030715] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Syed Muzammil Afzal
- Nanotechnology Research Lab, Department of Pharmaceutics, University College of Pharmaceutical Sciences, Kakatiya University, Warangal, Telangana, India,
| | - V. G. M Naidu
- National Institute of Pharmaceutical Education and Research, Balanagar, Hyderabad, India, and
| | - N. Harishankar
- National Center for Laboratory Animal Sciences, National Institute of Nutrition, Tarnaka, Hyderabad, India
| | - Veerabrahma Kishan
- Nanotechnology Research Lab, Department of Pharmaceutics, University College of Pharmaceutical Sciences, Kakatiya University, Warangal, Telangana, India,
| |
Collapse
|
43
|
Anti-tumor activity of fenretinide complexed with human serum albumin in lung cancer xenograft mouse model. Oncotarget 2015; 5:4811-20. [PMID: 25015569 PMCID: PMC4148101 DOI: 10.18632/oncotarget.2038] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Sufficient knowledge regarding cellular and molecular basis of lung cancer progression and metastasis would help in the development of novel and effective strategies for the treatment of lung cancer. 4HPR is a synthetic retinoid with potential anti-tumor activity but is still limited because of its poor bioavailability. The use of albumin as a complexing agent for a hydrophobic drug is expected to improve the water solubility and consequently their bioavailability.This study investigated the antitumor activity of a novel complex between albumin and 4-HPR in a mouse model of human lung cancer and focuses on role and mechanism of Cav-1 mainly involved in regulating cancer and Acsvl3 mainly connected with tumor growth. Their expressions were assayed by immunohistochemistry and qRT-PCR, to demonstrate the reduction of the tumor growth following the drug treatment. Our results showed a high antitumor activity of 4HPR-HSA by reduction of the volume of tumor mass and the presence of a high level of apoptotic cell by TUNEL assay. The downregulation of Cav-1 and Acsvl3 suggested a reduction of tumor growth. In conclusion, we demonstrated the great potential of 4HPR-HSA in the treatment of lung cancer. More data about the mechanism of drug delivery the 4HPR-HSA are necessary.
Collapse
|
44
|
Sankhala KK, Chawla NS, Chawla SP. Aldoxorubicin for the treatment of advanced soft tissue sarcoma. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1018179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
45
|
Holstein SA, Bigelow JC, Olson RD, Vestal RE, Walsh GM, Hohl RJ. Phase I and pharmacokinetic study of the novel anthracycline derivative 5-imino-13-deoxydoxorubicin (GPX-150) in patients with advanced solid tumors. Invest New Drugs 2015; 33:594-602. [PMID: 25698442 DOI: 10.1007/s10637-015-0220-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 02/10/2015] [Indexed: 10/24/2022]
Abstract
PURPOSE 5-imino-13-deoxydoxorubicin (DIDOX; GPX-150) is a doxorubicin analog modified in two locations to prevent formation of cardiotoxic metabolites and reactive oxygen species. Preclinical studies have demonstrated anti-cancer activity without cardiotoxicity. A phase I study was performed in order to determine the maximum-tolerated dose (MTD) of GPX-150 in patients with metastatic solid tumors. METHODS GPX-150 was administered as an intravenous infusion every 21 days for up to 8 cycles. An accelerated dose escalation was used for the first three treatment groups. The dosing groups were (A) 14 mg/m(2), (B) 28 mg/m(2), (C), 56 mg/m(2), (D) 84 mg/m(2), (E) 112 mg/m(2), (F) 150 mg/m(2), (G) 200 mg/m(2), and (H) 265 mg/m(2). Pharmacokinetic samples were drawn during the first 72 h of cycle 1. RESULTS The MTD was considered to be reached at the highest dosing level of 265 mg/m(2) since dose reduction was required in 5 of 6 patients for neutropenia. The most frequent adverse events were neutropenia, anemia, fatigue, and nausea. No patients experienced cardiotoxicity while on the study. The best overall response was stable disease in four (20 %) patients. Pharmacokinetic analysis revealed an AUC of 8.0 (±2.6) μg · h/mL, a clearance of 607 (±210) mL/min/m(2) and a t1/2β of 13.8 (±4.6) hours. CONCLUSIONS GPX-150 administered every 21 days has an acceptable side effect profile and no cardiotoxicity was observed. Further investigation is needed to determine the efficacy of GPX-150 in anthracycline-sensitive malignancies.
Collapse
Affiliation(s)
- Sarah A Holstein
- Department of Medicine, Roswell Park Cancer Institute, Elm & Carlton Sts, Buffalo, NY, 14263, USA,
| | | | | | | | | | | |
Collapse
|
46
|
Protein– and Peptide–Drug Conjugates. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015; 98:1-55. [DOI: 10.1016/bs.apcsb.2014.11.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
47
|
Pignatta S, Orienti I, Falconi M, Teti G, Arienti C, Medri L, Zanoni M, Carloni S, Zoli W, Amadori D, Tesei A. Albumin nanocapsules containing fenretinide: pre-clinical evaluation of cytotoxic activity in experimental models of human non-small cell lung cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2014; 11:263-73. [PMID: 25461293 DOI: 10.1016/j.nano.2014.10.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 10/16/2014] [Indexed: 11/26/2022]
Abstract
UNLABELLED The present study deals with the preparation of albumin nanocapsules containing fenretinide and their evaluation in experimental models of human non-small cell lung cancer. These nanocapsules showed enhanced antitumor activity with respect to free fenretinide due to the solubilization effect of albumin on the hydrophobic drug, known to improve bioavailability. The high expression of caveolin-1 on the A549 cell surface further enhanced the antitumor activity of the nanoencapsulated fenretinide. Caveolin-1 favored albumin uptake and improved the efficacy of the fenretinide-loaded albumin nanocapsules, especially in 3-D cultures where the densely packed 3-D structures impaired drug diffusibility and severely reduced the activity of the free drug. The efficacy of the fenretinide albumin nanocapsules was further confirmed in tumor xenograft models of A549 by the significant delay in tumor progression observed with respect to control after intravenous administration of the novel formulation. FROM THE CLINICAL EDITOR This study describes the preparation of fenretinide containing albumin nanocapsules and their evaluation in experimental models of non-small cell lung cancer, showing enhanced antitumor activity compared to free fenretinide.
Collapse
Affiliation(s)
- Sara Pignatta
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, (IRST) IRCCS, Meldola, FC, Italy
| | - Isabella Orienti
- FaBiT-Department of Pharmacy and Biotechnology, University of Bologna, Bologna, BO, Italy.
| | - Mirella Falconi
- DIBINEM-Department of Biomedical and Neuromotor Sciences, University of Bologna, BO, Italy
| | - Gabriella Teti
- DIBINEM-Department of Biomedical and Neuromotor Sciences, University of Bologna, BO, Italy
| | - Chiara Arienti
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, (IRST) IRCCS, Meldola, FC, Italy
| | - Laura Medri
- Pathology Unit, Morgagni-Pierantoni Hospital, Forlì, FC, Italy
| | - Michele Zanoni
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, (IRST) IRCCS, Meldola, FC, Italy
| | - Silvia Carloni
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, (IRST) IRCCS, Meldola, FC, Italy
| | - Wainer Zoli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, (IRST) IRCCS, Meldola, FC, Italy
| | - Dino Amadori
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, (IRST) IRCCS, Meldola, FC, Italy
| | - Anna Tesei
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, (IRST) IRCCS, Meldola, FC, Italy
| |
Collapse
|
48
|
Pharmacokinetic study of aldoxorubicin in patients with solid tumors. Invest New Drugs 2014; 33:341-8. [PMID: 25388939 PMCID: PMC4387275 DOI: 10.1007/s10637-014-0183-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 11/03/2014] [Indexed: 11/21/2022]
Abstract
Introduction Aldoxorubicin, a prodrug of doxorubicin, binds covalently to serum albumin in the bloodstream and accumulates in tumors. Aldoxorubicin can be administered at doses several-fold higher than doxorubicin can, without associated acute cardiotoxicity. Purpose This study fully evaluated the pharmacokinetic profile of aldoxorubicin (serum and urine). Methods Eighteen patients with advanced solid tumors received aldoxorubicin 230 or 350 mg/m2 (equivalent in drug load to doxorubicin at doses of 170 or 260 mg/m2, respectively) once every 21 days. Blood samples were taken in cycle 1 before aldoxorubicin infusion, and at 5, 15, 30, and 60 min, and at 2, 4, 8, 12, 16, 24, 48, and 72 h after infusion. Urine samples were taken in cycle 1 at 24, 48, and 72 h after infusion. Limited blood sampling was done in cycle 3, before aldoxorubicin infusion, and at 60 min and at 2, 4, and 8 h after infusion. Results The long mean half-life (20.1–21.1 h), narrow mean volume of distribution (3.96–4.08 L/m2), and slow mean clearance rate (0.136–0.152 L/h/m2) suggest that aldoxorubicin is stable in circulation and does not accumulate readily in body compartments outside of the bloodstream. Very little doxorubicin and its major metabolite doxorubicinol, which has been implicated in doxorubicin-associated cardiotoxicity, are excreted in urine. This might explain the lack of cardiotoxicity observed thus far with aldoxorubicin. Conclusions Our findings support dosing and administration schemas used in an ongoing phase 3 clinical study of aldoxorubicin in soft tissue sarcoma, and phase 2 clinical studies in small cell lung cancer, glioblastoma, and Kaposi’s sarcoma.
Collapse
|
49
|
Therapeutic efficacy of aldoxorubicin in an intracranial xenograft mouse model of human glioblastoma. Neoplasia 2014; 16:874-82. [PMID: 25379024 PMCID: PMC4212249 DOI: 10.1016/j.neo.2014.08.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 08/22/2014] [Accepted: 08/25/2014] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive primary brain tumor with a median survival of 12 to 15 months after diagnosis. Acquired chemoresistance, high systemic toxicity, and low penetration of the blood brain barrier by many anticancer drugs contribute to the failure of anti-GBM therapies. To circumvent some of these obstacles, we tested a novel prodrug approach to evaluate anti-GBM efficacy by utilizing serum albumin-binding doxorubicin (Doxo), aldoxorubicin (Aldoxo), which is less toxic, is released from albumin in an acidic environment and accumulates in tumor tissues. A human GBM cell line that expresses a luciferase reporter (U87-luc) was stereotactically injected into the left striatum of the brain of immunodeficient mice. Following initial tumor growth for 12 days, mice were injected once a week in the tail-vein with Aldoxo [24 mg/kg or 18 mg/kg of doxorubicin equivalents—3/4 maximum tolerated dose (MTD)], Doxo [6 mg/kg (3/4 MTD)], or vehicle. Aldoxo-treated mice demonstrated significantly slower growth of the tumor when compared to vehicle-treated or Doxo-treated mice. Five out of eight Aldoxo-treated mice remained alive more than 60 days with a median survival of 62 days, while the median survival of vehicle- and Doxo-treated mice was only 26 days. Importantly, Aldoxo-treated mice exhibited high levels of Doxo within the tumor tissue, accompanied by low tumor cell proliferation (Ki67) and abundant intratumoral programmed cell death (cleaved caspase-3). Effective accumulation of Aldoxo in brain tumor tissues but not normal brain, its anti-tumor efficacy, and low toxicity, provide a strong rationale for evaluating this novel drug conjugate as a treatment for patients afflicted with GBM.
Collapse
|
50
|
Chawla SP, Chua VS, Hendifar AF, Quon DV, Soman N, Sankhala KK, Wieland DS, Levitt DJ. A phase 1B/2 study of aldoxorubicin in patients with soft tissue sarcoma. Cancer 2014; 121:570-9. [DOI: 10.1002/cncr.29081] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 07/14/2014] [Accepted: 07/16/2014] [Indexed: 11/10/2022]
Affiliation(s)
| | | | | | | | | | - Kamalesh K. Sankhala
- Institute for Drug DevelopmentCancer Therapy and Research CenterUniversity of Texas Health Science CenterSan Antonio Texas
| | | | | |
Collapse
|