1
|
Lu N, Liu L, Cao Y, Zhang R, Zhai W, Chen X, Ma Q, Yang D, Pang A, Wei J, He Y, Feng S, Han M, Jiang E. Efficacy and safety of mitoxantrone hydrochloride liposome-containing regimens in treating refractory/relapsed acute myeloid leukemia. Discov Oncol 2025; 16:727. [PMID: 40353947 PMCID: PMC12069160 DOI: 10.1007/s12672-025-02526-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/28/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Despite the poor prognosis for patients with relapsed/refractory (R/R) acute myeloid leukemia (AML), an optimal treatment strategy remains undefined. Mitoxantrone (MIT) is widely used to treat R/R AML. METHODS This prospective, single-center, open-label study assessed the efficacy and toxicity of mitoxantrone hydrochloride liposome (Lipo-MIT)-containing regimen therapy, intensified by adding cytarabine (Ara-C), cyclophosphamide (CTX) or other agents. The primary endpoint was composite complete remission (CRc), including complete remission (CR), complete remission with incomplete count recovery (CRi), and morphologic leukemia-free state (MLFS). The secondary endpoints included overall response rate (ORR), event-free survival (EFS), overall survival (OS), and safety. RESULTS We enrolled 20 patients (median, 38.50 years; range, 20.00-53.00 years) and treated them with a Lipo-MIT-containing regimen from April 29, 2022, to July 11, 2023. Twelve patients (60.00%) achieved CRc after one course of induction therapy, of which MAC (Lipo-MIT, Ara-C, CTX)-based regimen was the most commonly used (12/20, 60.00%) with a CRc rate of 66.67% (8/12). Additionally, 13 patients relapsed after allogeneic stem cell transplantation (allo-HSCT) with a CRc of 69.20% (9/13). The median follow-up time was 6.64 months, with a median OS of 9.99 months (range, 1.64-19.61; 95% confidence interval [CI], 2.05-17.92). Moreover, 95% patients experienced grade 3/4 hematologic treatment-related adverse events (TRAEs), including anemia (60.0%), thrombocytopenia (60.0%), leukopenia (65.0%), and neutropenia (55.0%). All patients experienced nonhematologic TRAEs, with 14 patients showing grade 3/4 toxicity. CONCLUSION The Lipo-MIT-based regimens demonstrate preliminary efficacy in R/R AML, particularly in those who relapsed after allo-HSCT, though hematologic toxicity warrants careful monitoring. CLINICAL TRIAL REGISTRATION Clinicaltrials.gov Identifier: NCT04645199 in November 27, 2020.
Collapse
Affiliation(s)
- Ni Lu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 300020, China
| | - Li Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 300020, China
| | - Yigeng Cao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 300020, China
| | - Rongli Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 300020, China
| | - Weihua Zhai
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 300020, China
| | - Xin Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 300020, China
| | - Qiaoling Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 300020, China
| | - Donglin Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 300020, China
| | - Aiming Pang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 300020, China
| | - Jialin Wei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 300020, China
| | - Yi He
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 300020, China
| | - Sizhou Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 300020, China
| | - Mingzhe Han
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 300020, China
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Hematopoietic Stem Cell Transplantation Center, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin 300020, China.
| |
Collapse
|
2
|
Ghahremanloo A, Erfani B, Asgharzadeh F, Mansoori S, Gheybi F, Hashemy SI. Reducing toxicity and enhancing efficacy of doxorubicin by liposomal doxorubicin and aprepitant in breast cancer. Sci Rep 2025; 15:9798. [PMID: 40118925 PMCID: PMC11928494 DOI: 10.1038/s41598-025-94291-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/12/2025] [Indexed: 03/24/2025] Open
Abstract
This study investigates the efficacy and toxicity profiles of pegylated liposomal doxorubicin (Doxil) compared to conventional doxorubicin. Additionally, it evaluates the potential of combination therapy involving Doxil and doxorubicin with aprepitant, an FDA-approved agent for the management of chemotherapy-induced nausea and vomiting. Using a mouse model induced with 4T1 breast cancer cells, tumor size, and weight were assessed following treatment with either single doses or combination therapies. The study also examined oxidative and antioxidant stress markers in tumor, liver, and cardiac tissues, complemented by histopathological analysis of these tissues using hematoxylin and eosin staining. Results indicated that prepared liposomal doxorubicin significantly enhanced antitumor efficacy, as evidenced by decreased tumor size and weight. Moreover, it positively influenced oxidative stress markers, promoting apoptosis in tumor tissues. Notably, Doxil also reduced adverse effects compared to standard doxorubicin, as indicated by lower oxidative stress levels and increased antioxidant activity in both cardiac and liver tissues. The combined administration of doxorubicin and aprepitant further improved therapeutic efficacy and reduced side effects. Consequently, the formulation of doxorubicin in liposomes and aprepitant-based combination therapy represents a promising strategy for enhancing treatment effectiveness while minimizing adverse effects in breast cancer management.
Collapse
Affiliation(s)
- Atefeh Ghahremanloo
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahareh Erfani
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fereshteh Asgharzadeh
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeide Mansoori
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Gheybi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Liu Z, Liu Y, Kang X, Li L, Xiang Y. Subcellular Organelle Targeting as a Novel Approach to Combat Tumor Metastasis. Pharmaceutics 2025; 17:198. [PMID: 40006565 PMCID: PMC11859411 DOI: 10.3390/pharmaceutics17020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/28/2025] [Accepted: 02/02/2025] [Indexed: 02/27/2025] Open
Abstract
Tumor metastasis, the spread of cancer cells from the primary site to distant organs, remains a formidable challenge in oncology. Central to this process is the involvement of subcellular organelles, which undergo significant functional and structural changes during metastasis. Targeting these specific organelles offers a promising avenue for enhanced drug delivery and metastasis therapeutic efficacy. This precision increases the potency and reduces potential off-target effects. Moreover, by understanding the role of each organelle in metastasis, treatments can be designed to disrupt the metastatic process at multiple stages, from cell migration to the establishment of secondary tumors. This review delves deeply into tumor metastasis processes and their connection with subcellular organelles. In order to target these organelles, biomembranes, cell-penetrating peptides, localization signal peptides, aptamers, specific small molecules, and various other strategies have been developed. In this review, we will elucidate targeting delivery strategies for each subcellular organelle and look forward to prospects in this domain.
Collapse
Affiliation(s)
- Zefan Liu
- Department of General Surgery, First People‘s Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China; (Z.L.); (Y.L.)
| | - Yang Liu
- Department of General Surgery, First People‘s Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China; (Z.L.); (Y.L.)
| | - Xin Kang
- Department of General Surgery, First People‘s Hospital of Shuangliu District (West China Airport Hospital of Sichuan University), Chengdu 610200, China; (Z.L.); (Y.L.)
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China;
| | - Yucheng Xiang
- School of Pharmacy, Chengdu Medical College, Chengdu 610500, China
| |
Collapse
|
4
|
Buchalska B, Kamińska K, Kowara M, Sobiborowicz-Sadowska A, Cudnoch-Jędrzejewska A. Doxorubicin or Epirubicin Versus Liposomal Doxorubicin Therapy-Differences in Cardiotoxicity. Cardiovasc Toxicol 2025; 25:248-268. [PMID: 39810066 DOI: 10.1007/s12012-024-09952-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025]
Abstract
Doxorubicin (DOX) is an important drug used in the treatment of many malignancies. Unfortunately DOX causes various side effects, with cardiotoxicity being the most characteristic. Risk factors for DOX induced cardiotoxicity (DIC) include cumulative dose of DOX, preexisting cardiovascular diseases, dyslipidemia, diabetes, smoking, along with the use of other cardiotoxic agents. Development of DIC is associated with many pathological phenomena - increased oxidative stress, as well as upregulation of ferroptosis, apoptosis, necrosis, and autophagy. In DIC expression of many microRNAs is also deregulated. In order to avoid cardiotoxicity and still use DOX effectively DOX derivatives such as epirubicin were synthesized. Nowadays a new liposomal form of DOX (L-DOX) appeared as an alternative to conventional treatment with greatly reduced cardiotoxicity. L-DOX can be divided into two groups of substances - pegylated (PLD) with increased solubility and stability, and non-pegylated (NLPD). Many metaanalyses, clinical along with preclinical studies have shown L-DOX treatment is associated with a smaller decrease of left ventricular ejection fraction (LVEF) and other heart functions, but efficacy of this treatment is comparable to the use of convenctional DOX.
Collapse
Affiliation(s)
- Barbara Buchalska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| | - Katarzyna Kamińska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland.
| | - Michał Kowara
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| | - Aleksandra Sobiborowicz-Sadowska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, 02-097, Warsaw, Poland
| |
Collapse
|
5
|
Palvia AR, Damera AR, Nandi AR, Magar S, Patidar S, Kasarla S, Ghantasala V, Shah MK, Goyal M. Cardio-Oncology's Modern Approaches to Prevent Doxorubicin-Induced Cardiotoxicity: A Systematic Review. Cureus 2024; 16:e66215. [PMID: 39238705 PMCID: PMC11375109 DOI: 10.7759/cureus.66215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
Advances in the field of oncology have led to the advent of doxorubicin (DOX), an anthracycline chemotherapeutic agent, through which cancer survival rates have remarkably improved. There has, however, been a rise in adverse effects from the use of DOX, most notably cardiotoxicity. DOX-induced cardiotoxicity is thought to arise through the generation of reactive oxygen species (ROS), causing mitochondrial dysfunction in the cardiomyocytes. This systematic review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) standards and focused on cancer patients undergoing DOX therapy. The research question addressed interventions aimed at preventing DOX-induced cardiotoxicity. Google Scholar, PubMed, and ScienceDirect databases were used to conduct a systematic search. Next, screening was carried out by reviewing the title and abstract of various articles to exclude irrelevant studies, followed by the retrieval of full-text articles. Scale for the assessment of narrative review articles 2 (SANRA 2) for narrative reviews, a measurement tool to assess systematic reviews (AMSTAR) checklist for systematic reviews, and the Cochrane risk of bias tool for randomized controlled trials (RCTs) were the tools employed for quality assessment. This systematic review provides convincing evidence about preventive interventions to counteract DOX-induced cardiotoxicity. Primary prevention strategies against DOX-induced cardiotoxicity include pharmacological and non-pharmacological measures. Dexrazoxane reduces cardiotoxicity without therapeutic compromise. Beta-blockers showed mixed results in preserving cardiac function. The research on renin-angiotensin-aldosterone system (RAAS) inhibitors suggests that most of these agents can reduce the risk of DOX-induced cardiotoxicity. The liposomal formulation of DOX decreases cardiotoxicity without sacrificing effectiveness. Chemotherapy regimens should be supplemented with cardioprotective medications to increase therapeutic efficacy and lower cardiac risks. Exercise is an essential non-pharmacological strategy for decreasing DOX-induced cardiotoxicity. It acts by lowering oxidative stress, maintaining mitochondrial function, and averting apoptosis. Other non-pharmacological interventions through antioxidative, anti-apoptotic, and mitochondrial protective mechanisms, such as resveratrol, vitamin E, curcumin, and visnagin, show promise in lowering DOX-induced cardiotoxicity and may be useful as supplementary therapy during cancer treatment. In conclusion, this review highlights the need for a multimodal strategy that incorporates different tactics, as well as the need for additional research and strong clinical trials, with the ultimate goal of protecting cardiac health in patients receiving chemotherapy with DOX.
Collapse
Affiliation(s)
- Aadi R Palvia
- Internal Medicine, Kharghar Medicity Hospital, Navi Mumbai, IND
| | - Abhiram Rao Damera
- Internal Medicine, MediCiti Institute of Medical Sciences, Hyderabad, IND
| | | | - Shikha Magar
- Internal Medicine, Kempegowda Institute of Medical Sciences, Bengaluru, IND
| | - Saloni Patidar
- Internal Medicine, Bharati Vidyapeeth Medical College, Pune, IND
| | - Sachin Kasarla
- Internal Medicine, Gitam Institute of Medical Sciences and Research, Visakhapatnam, IND
| | | | - Mishank K Shah
- Internal Medicine, Gujarat Medical Education and Research Society (GMERS) Medical College, Vadodara, IND
| | | |
Collapse
|
6
|
Kadry MO, Abdel-Megeed RM. CRISPR-Cas9 genome and long non-coding RNAs as a novel diagnostic index for prostate cancer therapy via liposomal-coated compounds. PLoS One 2024; 19:e0302264. [PMID: 38723038 PMCID: PMC11081254 DOI: 10.1371/journal.pone.0302264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 03/31/2024] [Indexed: 05/13/2024] Open
Abstract
CRISPR/Cas9 is a recently discovered genomic editing technique that altered scientist's sight in studying genes function. Cas9 is controlled via guide (g) RNAs, which match the DNA targeted in cleavage to modify the respective gene. The development in prostate cancer (PC) modeling directed not only to novel resources for recognizing the signaling pathways overriding prostate cell carcinoma, but it has also created a vast reservoir for complementary tools to examine therapies counteracting this type of cancer. Various cultured somatic rat models for prostate cancer have been developed that nearly mimic human prostate cancer. Nano-medicine can passively target cancer cells via increasing bioavailability and conjugation via specific legend, contributing to reduced systemic side-effects and increased efficacy. This article highlights liposomal loaded Nano-medicine as a potential treatment for prostate cancer and clarifies the CRISPR/Cas9 variation accompanied with prostate cancer. PC is induced experimentally in western rat model via ethinyl estradiol for 4 weeks and SC. dose of 3, 2'- dimethyl-4-aminobiphenyl estradiol (DAE) (50mg/kg) followed by treatment via targeted liposomal-coated compounds such as liposomal dexamethasone (DXM), liposomal doxorubicin (DOX) and liposomal Turmeric (TUR) (3mg/kg IP) for four weeks in a comparative study to their non-targeted analogue dexamethasone, doxorubicin and Turmeric. 3, 2'- dimethyl-4-aminobiphenylestradiol elicit prostate cancer in western rats within 5 months. Simultaneous supplementations with these liposomal compounds influence on prostate cancer; tumor markers were investigated via prostate-specific antigen (PSA), Nitric oxide (NOX) and CRISPR/Cas9 gene editing. Several long non-coding RNAs were reported to be deregulated in prostate cell carcinoma, including MALAT1. On the other hand, gene expression of apoptotic biomarkers focal adhesion kinase (AKT-1), phosphatidylinistol kinase (PI3K) and glycogen synthase kinase-3 (GSK-3) was also investigated and further confirming these results via histopathological examination. Liposomal loaded dexamethasone; doxorubicin and Turmeric can be considered as promising therapeutic agents for prostate cancer via modulating CRISPR/Cas9 gene editing and long non coding gene MALAT1.
Collapse
Affiliation(s)
- Mai O. Kadry
- Therapeutic Chemistry Department, National Research Center, Al Bhoouth Street, Cairo, Egypt
| | - Rehab M. Abdel-Megeed
- Therapeutic Chemistry Department, National Research Center, Al Bhoouth Street, Cairo, Egypt
| |
Collapse
|
7
|
Maciel e Silva AT, Maia ALC, Silva JDO, Miranda SEM, Cantini TS, de Barros ALB, Soares DCF, de Magalhães MTQ, Alves RJ, Ramaldes GA. In Vitro and Preclinical Antitumor Evaluation of Doxorubicin Liposomes Coated with a Cholesterol-Based Trimeric β-D-Glucopyranosyltriazole. Pharmaceutics 2023; 15:2751. [PMID: 38140092 PMCID: PMC10747952 DOI: 10.3390/pharmaceutics15122751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
The coating of liposomes with polyethyleneglycol (PEG) has been extensively discussed over the years as a strategy for enhancing the in vivo and in vitro stability of nanostructures, including doxorubicin-loaded liposomes. However, studies have shown some important disadvantages of the PEG molecule as a long-circulation agent, including the immunogenic role of PEG, which limits its clinical use in repeated doses. In this context, hydrophilic molecules as carbohydrates have been proposed as an alternative to coating liposomes. Thus, this work studied the cytotoxicity and preclinical antitumor activity of liposomes coated with a glycosyl triazole glucose (GlcL-DOX) derivative as a potential strategy against breast cancer. The glucose-coating of liposomes enhanced the storage stability compared to PEG-coated liposomes, with the suitable retention of DOX encapsulation. The antitumor activity, using a 4T1 breast cancer mouse model, shows that GlcL-DOX controlled the tumor growth in 58.5% versus 35.3% for PEG-coated liposomes (PegL-DOX). Additionally, in the preliminary analysis of the GlcL-DOX systemic toxicity, the glucose-coating liposomes reduced the body weight loss and hepatotoxicity compared to other DOX-treated groups. Therefore, GlcL-DOX could be a promising alternative for treating breast tumors. Further studies are required to elucidate the complete GlcL-DOX safety profile.
Collapse
Affiliation(s)
- Aline Teixeira Maciel e Silva
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (A.T.M.e.S.); (A.L.C.M.); (J.d.O.S.); (S.E.M.M.); (T.S.C.)
| | - Ana Luiza Chaves Maia
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (A.T.M.e.S.); (A.L.C.M.); (J.d.O.S.); (S.E.M.M.); (T.S.C.)
| | - Juliana de Oliveira Silva
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (A.T.M.e.S.); (A.L.C.M.); (J.d.O.S.); (S.E.M.M.); (T.S.C.)
| | - Sued Eustáquio Mendes Miranda
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (A.T.M.e.S.); (A.L.C.M.); (J.d.O.S.); (S.E.M.M.); (T.S.C.)
| | - Talia Silva Cantini
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (A.T.M.e.S.); (A.L.C.M.); (J.d.O.S.); (S.E.M.M.); (T.S.C.)
| | - Andre Luis Branco de Barros
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil
| | - Daniel Crístian Ferreira Soares
- Laboratório de Bioengenharia, Universidade Federal de Itajubá, Rua Irmã Ivone Drumond, 200, Distrito Industrial II, Itabira 35903-087, MG, Brazil;
| | - Mariana Torquato Quezado de Magalhães
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil;
| | - Ricardo José Alves
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (A.T.M.e.S.); (A.L.C.M.); (J.d.O.S.); (S.E.M.M.); (T.S.C.)
| | - Gilson Andrade Ramaldes
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Belo Horizonte 31270-901, MG, Brazil; (A.T.M.e.S.); (A.L.C.M.); (J.d.O.S.); (S.E.M.M.); (T.S.C.)
| |
Collapse
|
8
|
Abukwaik R, Vera-Siguenza E, Tennant DA, Spill F. Interplay of p53 and XIAP protein dynamics orchestrates cell fate in response to chemotherapy. J Theor Biol 2023; 572:111562. [PMID: 37348784 DOI: 10.1016/j.jtbi.2023.111562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 04/06/2023] [Accepted: 06/16/2023] [Indexed: 06/24/2023]
Abstract
Chemotherapeutic drugs are used to treat almost all types of cancer, but the intended response, i.e., elimination, is often incomplete, with a subset of cancer cells resisting treatment. Two critical factors play a role in chemoresistance: the p53 tumour suppressor gene and the X-linked inhibitor of apoptosis (XIAP). These proteins have been shown to act synergistically to elicit cellular responses upon DNA damage induced by chemotherapy, yet, the mechanism is poorly understood. This study introduces a mathematical model characterising the apoptosis pathway activation by p53 before and after mitochondrial outer membrane permeabilisation upon treatment with the chemotherapy Doxorubicin (Dox). "In-silico" simulations show that the p53 dynamics change dose-dependently. Under medium to high doses of Dox, p53 concentration ultimately stabilises to a high level regardless of XIAP concentrations. However, caspase-3 activation may be triggered or not depending on the XIAP induction rate, ultimately determining whether the cell will perish or resist. Consequently, the model predicts that failure to activate apoptosis in some cancer cells expressing wild-type p53 might be due to heterogeneity between cells in upregulating the XIAP protein, rather than due to the p53 protein concentration. Our model suggests that the interplay of the p53 dynamics and the XIAP induction rate is critical to determine the cancer cells' therapeutic response.
Collapse
Affiliation(s)
- Roba Abukwaik
- Mathematics Department, Faculty of Science and Arts, King Abdulaziz University, Rabigh, Saudi Arabia; School of Mathematics, University of Birmingham, B15 2TS, United Kingdom.
| | - Elias Vera-Siguenza
- School of Mathematics, University of Birmingham, B15 2TS, United Kingdom; Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, United Kingdom.
| | - Daniel A Tennant
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, B15 2TT, United Kingdom
| | - Fabian Spill
- School of Mathematics, University of Birmingham, B15 2TS, United Kingdom.
| |
Collapse
|
9
|
Akhlaq A, Ashraf M, Omer MO, Altaf I. Carvacrol-Fabricated Chitosan Nanoparticle Synergistic Potential with Topoisomerase Inhibitors on Breast and Cervical Cancer Cells. ACS OMEGA 2023; 8:31826-31838. [PMID: 37692253 PMCID: PMC10483689 DOI: 10.1021/acsomega.3c03337] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 08/15/2023] [Indexed: 09/12/2023]
Abstract
Breast and cervical cancers are the most common heterogeneous malignancies in women. Chemotherapy with conventional drug delivery systems having several limitations along with development of multidrug resistance compelled us to seek out targeted therapeutics. Nanoparticles are suitable substitutes to circumvent multidrug resistance for the targeted treatment of cancer. The current study was aimed to investigate the anticancer effect of carvacrol-loaded chitosan nanoparticles with topoisomerase inhibitors. The average size of carvacrol-loaded chitosan nanoparticles was found to be 80 nm with 24.7 mV ζ-potential, and maximum absorbance was observed at 275 nm. Among all drug combinations, the carvacrol nanoparticles with the doxorubicin combination group exerted greater dose-dependent growth inhibition of both MCF-7 and HeLa cells as compared to single carvacrol nanoparticles and doxorubicin. Combination index values of carvacrol nanoparticles and the doxorubicin combination group showed a strong synergistic effect as they were found to be between 0.2 and 0.4, 0.31 for MCF-7 and 0.34 for HeLa cells. The carvacrol nanoparticles in combination with doxorubicin on MCF-7 cells reduced the dose 16.32-fold for carvacrol nanoparticles and 4.09-fold for doxorubicin at 6.23 μg/mL IC50, while on HeLa cells, this combination reduced the dose 13.18-fold for carvacrol nanoparticles and 3.83-fold for doxorubicin at 9.33 μg/mL IC50. As the dose reduction values were greater than 1, they indicated favorable dose reduction. It was concluded that the combination of carvacrol-loaded chitosan nanoparticles with topoisomerase inhibitors may represent an innovative and promising strategy to improve the efficacy, resistance, and targeted delivery of chemotherapeutics in cancer.
Collapse
Affiliation(s)
- Amina Akhlaq
- Department
of Pharmacology and Toxicology, University
of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Muhammad Ashraf
- Department
of Pharmacology and Toxicology, University
of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Muhammad Ovais Omer
- Department
of Pharmacology and Toxicology, University
of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Imran Altaf
- Institute
of Microbiology, University of Veterinary
and Animal Sciences, Lahore 54000, Pakistan
| |
Collapse
|
10
|
Ou X, Zhang Z, Lin L, Du Y, Tang Y, Wang Y, Zou J. Tumor-homing bacterium-adsorbed liposomes encapsulating perfluorohexane/doxorubicin enhance pulsed-focused ultrasound for tumor therapy. RSC Adv 2023; 13:19065-19078. [PMID: 37362333 PMCID: PMC10288177 DOI: 10.1039/d3ra01876h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Objective: To make up for the insufficient ultrasound ablation of tumors, the energy output or synergist is increased but faces the big challenge of normal tissue damage. In this study, we report a tumor-homing bacterium, Bifidobacterium bifidum (B. bifidum), adsorbing liposomes that encapsulate perfluorohexane (PFH) and doxorubicin (DOX) to enhance the pulsed-focused ultrasound (PFUS) for tumor therapy, so as to improve the efficacy, safety and controllability of ultrasound treatment. Methods: The PFH and DOX co-loaded cationic liposomal nanoparticles (CL-PFH-DOX-NPs) were prepared for ultrasound (US) imaging, cell-killing, and B. bifidum adsorption for the reactive oxygen species (ROS) testing. The aggregation of B. bifidum and CL-PFH-DOX-NPs is called tumor-homing aggregation (B. bifidum@CL-PFH-DOX-NPs) in this study, and the synergistic effects of B. bifidum@CL-PFH-DOX-NPs were analyzed in vivo. Results: Comprehensive studies validated that CL-PFH-DOX-NPs can enhance US imaging and cell-killing and B. bifidum can promote ROS, and B. bifidum@CL-PFH-DOX-NPs achieve PFUS synergism in vivo. Importantly, active homing of B. bifidum facilitated the delivery and retention of CL-PFH-DOX-NPs in tumors, reducing dispersion in normal tissues, achieving the targeting ability of B. bifidum@CL-PFH-DOX-NPs. The best sonication time was chosen according to the distribution of CL-PFH-DOX-NPs in vivo to achieve efficient therapy. Especially, B. bifidum@CL-PFH-DOX-NPs amplified cavitation and the immune-boosting effects. Conclusion: Multifunctional B. bifidum@CL-PFH-DOX-NPs were successfully constructed with well targeting, which not only realized US imaging monitoring, strong cavitation and complementary killing during PFUS, but also achieved immunity enhancement after PFUS. The combination of PFUS, B. bifidum and CL-PFH-DOX-NPs provides a new idea for the potential application of ultrasound therapy in solid tumors.
Collapse
Affiliation(s)
- Xia Ou
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 People's Republic of China +86-13708302390
| | - Zhong Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 People's Republic of China +86-13708302390
| | - Li Lin
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 People's Republic of China +86-13708302390
| | - Yan Du
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 People's Republic of China +86-13708302390
| | - Yu Tang
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 People's Republic of China +86-13708302390
| | - Yaotai Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 People's Republic of China +86-13708302390
| | - Jianzhong Zou
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University Chongqing 400016 People's Republic of China +86-13708302390
| |
Collapse
|
11
|
Al-hussaniy HA, Alburghaif AH, alkhafaje Z, AL-Zobaidy MAHJ, Alkuraishy HM, Mostafa-Hedeab G, Azam F, Al-Samydai AM, Al-tameemi ZS, Naji MA. Chemotherapy-induced cardiotoxicity: a new perspective on the role of Digoxin, ATG7 activators, Resveratrol, and herbal drugs. J Med Life 2023; 16:491-500. [PMID: 37305823 PMCID: PMC10251384 DOI: 10.25122/jml-2022-0322] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 12/22/2022] [Indexed: 06/13/2023] Open
Abstract
Cancer is a major public health problem, and chemotherapy plays a significant role in the management of neoplastic diseases. However, chemotherapy-induced cardiotoxicity is a serious side effect secondary to cardiac damage caused by antineoplastic's direct and indirect toxicity. Currently, there are no reliable and approved methods for preventing or treating chemotherapy-induced cardiotoxicity. Understanding the mechanisms of chemotherapy-induced cardiotoxicity may be vital to improving survival. The independent risk factors for developing cardiotoxicity must be considered to prevent myocardial damage without decreasing the therapeutic efficacy of cancer treatment. This systematic review aimed to identify and analyze the evidence on chemotherapy-induced cardiotoxicity, associated risk factors, and methods to decrease or prevent it. We conducted a comprehensive search on PubMed, Google Scholar, and Directory of Open Access Journals (DOAJ) using the following keywords: "doxorubicin cardiotoxicity", "anthracycline cardiotoxicity", "chemotherapy", "digoxin decrease cardiotoxicity", "ATG7 activators", retrieving 59 articles fulfilling the inclusion criteria. Therapeutic schemes can be changed by choosing prolonged infusion application over boluses. In addition, some agents like Dexrazoxane can reduce chemotherapy-induced cardiotoxicity in high-risk groups. Recent research found that Digoxin, ATG7 activators, Resveratrol, and other medical substances or herbal compounds have a comparable effect on Dexrazoxane in anthracycline-induced cardiotoxicity.
Collapse
Affiliation(s)
- Hany Akeel Al-hussaniy
- Department of Pharmacy, Bilad Alrafidain University College, Diyala, Iraq
- Dr. Hany Akeel Institute, Iraqi Medical Research Center, Baghdad, Iraq
| | | | - Zahraa alkhafaje
- Department of Pharmacy, Alfarahidi University College, Baghdad, Iraq
| | | | - Hayder Mutair Alkuraishy
- Department of Clinical Pharmacology, College of Medicine, Almustansria University, Baghdad, Iraq
| | - Gomaa Mostafa-Hedeab
- Pharmacology Department & Health Research Unit, Medical College, Jouf University, Jouf, Saudi Arabia
- Pharmacology Department, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Faizul Azam
- Department of Pharmaceutical Chemistry and Pharmacognosy, Unaizah College of Pharmacy, Qassim University, Uniazah, Saudi Arabia
| | - Ali Mahmoud Al-Samydai
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Zahraa Salam Al-tameemi
- Department of Pharmacy, Bilad Alrafidain University College, Diyala, Iraq
- Dr. Hany Akeel Institute, Iraqi Medical Research Center, Baghdad, Iraq
| | - Meena Akeel Naji
- Dr. Hany Akeel Institute, Iraqi Medical Research Center, Baghdad, Iraq
| |
Collapse
|
12
|
El Khoury R, Ramirez SP, Loyola CD, Joddar B. Demonstration of doxorubicin's cardiotoxicity and screening using a 3D bioprinted spheroidal droplet-based system. RSC Adv 2023; 13:8338-8351. [PMID: 36922946 PMCID: PMC10010162 DOI: 10.1039/d3ra00421j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 03/03/2023] [Indexed: 03/16/2023] Open
Abstract
Doxorubicin (DOX) is a highly effective anthracycline chemotherapy agent effective in treating a broad range of life-threatening malignancies but it causes cardiotoxicity in many subjects. While the mechanism of its cardiotoxic effects remains elusive, DOX-related cardiotoxicity can lead to heart failure in patients. In this study, we investigated the effects of DOX-induced cardiotoxicity on human cardiomyocytes (CMs) using a three-dimensional (3D) bioprinted cardiac spheroidal droplet based-system in comparison with the traditional two-dimensional cell (2D) culture model. The effects of DOX were alleviated with the addition of N-acetylcysteine (NAC) and Tiron. Caspase-3 activity was quantified, and reactive oxygen species (ROS) production was measured using dihydroethidium (DHE) staining. Application of varying concentrations of DOX (0.4 μM-1 μM) to CMs revealed a dose-specific response, with 1 μM concentration imposing maximum cytotoxicity and 0.22 ± 0.11% of viable cells in 3D samples versus 1.02 ± 0.28% viable cells in 2D cultures, after 5 days of culture. Moreover, a flow cytometric analysis study was conducted to study CMs proliferation in the presence of DOX and antioxidants. Our data support the use of a 3D bioprinted cardiac spheroidal droplet as a robust and high-throughput screening model for drug toxicity. In the future, this 3D spheroidal droplet model can be adopted as a human-derived tissue-engineered equivalent to address challenges in other various aspects of biomedical pre-clinical research.
Collapse
Affiliation(s)
- Raven El Khoury
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso El Paso TX 79968 USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso 500 W. University Avenue El Paso TX 79968 USA
| | - Salma P Ramirez
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso El Paso TX 79968 USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso 500 W. University Avenue El Paso TX 79968 USA
| | - Carla D Loyola
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso El Paso TX 79968 USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso 500 W. University Avenue El Paso TX 79968 USA
| | - Binata Joddar
- Inspired Materials & Stem-Cell Based Tissue Engineering Laboratory (IMSTEL), The University of Texas at El Paso El Paso TX 79968 USA
- Department of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso 500 W. University Avenue El Paso TX 79968 USA
- Border Biomedical Research Center, The University of Texas at El Paso 500 W. University Avenue El Paso TX 79968 USA
| |
Collapse
|
13
|
Li Z, Zhang S, Liu M, Zhong T, Li H, Wang J, Zhao H, Tian Y, Wang H, Wang J, Xu M, Wang S, Zhang X. Antitumor Activity of the Zinc Oxide Nanoparticles Coated with Low-Molecular-Weight Heparin and Doxorubicin Complex In Vitro and In Vivo. Mol Pharm 2022; 19:4179-4190. [PMID: 36223494 DOI: 10.1021/acs.molpharmaceut.2c00553] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Various metal oxide nanomaterials have been widely used as carriers to prepare pH-sensitive nanomedicines to respond to the acidic tumor microenvironment promoting antitumor efficiency. Herein, we used zinc oxide nanoparticles (ZnO NPs) as metal oxide nanomaterial coated with low-molecular-weight heparin (LMHP) and doxorubicin (DOX) complex (LMHP-DOX) to prepare ZnO-LD NPs for controllable pH-triggered DOX release on the targeted site. Our results indicated that the released DOX from ZnO-LD NPs was pH-sensitive. The oxygen produced by ZnO-LD NPs in H2O2 solution was observed in in vitro experiment. The ZnO-LD NPs entered into both PC-3M and 4T1 tumor cells via clathrin-mediated endocytosis and micropinocytosis pathway. The intracellular reactive oxygen species (ROS) generated by ZnO-LD NPs could significantly increase the caspase 3/7 level, leading to tumor cell apoptosis. The in vitro and in vivo antitumor activity was confirmed in PC-3M and 4T1 cell lines or tumor-bearing mice models. The in vivo and in vitro tumor images via second-order nonlinearity of ZnO-LD NPs indicated that ZnO-LD NPs could penetrate deep into the tumor tissues. Therefore, the ZnO-LD NPs developed in our study could provide an efficient approach for the preparation of pH-sensitive nano delivery systems suitable for tumor therapy and imaging.
Collapse
Affiliation(s)
- Zhuoyue Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Shuang Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Man Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Ting Zhong
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Hui Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Jingru Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Heng Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Yubo Tian
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Hui Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Jingwen Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Meiqi Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Shumin Wang
- Department of Ultrasound, Peking University Third Hospital, Peking University, Xueyuan Road 38, Beijing 100191, China
| | - Xuan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Xueyuan Road 38, Beijing 100191, China
| |
Collapse
|
14
|
Rao X, Zhang C, Luo H, Zhang J, Zhuang Z, Liang Z, Wu X. Targeting Gastric Cancer Stem Cells to Enhance Treatment Response. Cells 2022; 11:cells11182828. [PMID: 36139403 PMCID: PMC9496718 DOI: 10.3390/cells11182828] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
Gastric cancer (GC) was the fourth deadliest cancer in the world in 2020, and about 770,000 people died from GC that year. The death of patients with GC is mainly caused by the metastasis, recurrence, and chemotherapy resistance of GC cells. The cancer stem cell theory defines cancer stem cells (CSCs) as a key factor in the metastasis, recurrence, and chemotherapy resistance of cancer. It considers targeting gastric cancer stem cells (GCSCs) to be an effective method for the treatment of GC. For GCSCs, genes or noncoding RNAs are important regulatory factors. Many experimental studies have found that some drugs can target the stemness of gastric cancer by regulating these genes or noncoding RNAs, which may bring new directions for the clinical treatment of gastric cancer. Therefore, this review mainly discusses related genes or noncoding RNAs in GCSCs and drugs that target its stemness, thereby providing some information for the treatment of GC.
Collapse
|
15
|
Plasma Pharmacokinetics and Tissue Distribution of Doxorubicin in Rats following Treatment with Astragali Radix. Pharmaceuticals (Basel) 2022; 15:ph15091104. [PMID: 36145325 PMCID: PMC9505068 DOI: 10.3390/ph15091104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Doxorubicin (DOX) is an essential component in chemotherapy, and Astragali Radix (AR) is a widely used tonic herbal medicine. The combination of DOX and AR offers widespread, well-documented advantages in treating cancer, e.g., reducing the risk of adverse effects. This study mainly aims to uncover the impact of AR on DOX disposition in vivo. Rats received a single intravenous dose of 5 mg/kg DOX following a single-dose co-treatment or multiple-dose pre-treatment of AR (10 g/kg × 1 or × 10). The concentrations of DOX in rat plasma and six tissues, including heart, liver, lung, kidney, spleen, and skeletal muscle, were determined by a fully validated LC-MS/MS method. A network-based approach was further employed to quantify the relationships between enzymes that metabolize and transport DOX and the targets of nine representative AR components in the human protein−protein interactome. We found that short-term (≤10 d) AR administration was ineffective in changing the plasma pharmacokinetics of DOX in terms of the area under the concentration−time curve (AUC, 1303.35 ± 271.74 μg/L*h versus 1208.74 ± 145.35 μg/L*h, p > 0.46), peak concentrations (Cmax, 1351.21 ± 364.86 μg/L versus 1411.01 ± 368.38 μg/L, p > 0.78), and half-life (t1/2, 31.79 ± 5.12 h versus 32.05 ± 6.95 h, p > 0.94), etc. Compared to the isotype control group, DOX concentrations in six tissues slightly decreased under AR pre-administration but only showed statistical significance (p < 0.05) in the liver. Using network analysis, we showed that five of the nine representative AR components were not localized to the vicinity of the DOX disposition-associated module. These findings suggest that AR may mitigate DOX-induced toxicity by affecting drug targets rather than drug disposition.
Collapse
|
16
|
de Souza Guimarães M, Cachumba JJM, Bueno CZ, Torres-Obreque KM, Lara GVR, Monteiro G, Barbosa LRS, Pessoa A, Rangel-Yagui CDO. Peg-Grafted Liposomes for L-Asparaginase Encapsulation. Pharmaceutics 2022; 14:1819. [PMID: 36145567 PMCID: PMC9503594 DOI: 10.3390/pharmaceutics14091819] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
L-asparaginase (ASNase) is an important biological drug used to treat Acute Lymphoblastic Leukemia (ALL). It catalyzes the hydrolysis of L-asparagine (Asn) in the bloodstream and, since ALL cells cannot synthesize Asn, protein synthesis is impaired leading to apoptosis. Despite its therapeutic importance, ASNase treatment is associated to side effects, mainly hypersensitivity and immunogenicity. Furthermore, degradation by plasma proteases and immunogenicity shortens the enzyme half-life. Encapsulation of ASNase in liposomes, nanostructures formed by the self-aggregation of phospholipids, is an attractive alternative to protect the enzyme from plasma proteases and enhance pharmacokinetics profile. In addition, PEGylation might prolong the in vivo circulation of liposomes owing to the spherical shielding conferred by the polyethylene (PEG) corona around the nanostructures. In this paper, ASNase was encapsulated in liposomal formulations composed by 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) or 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) containing or not different concentrations of 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N [methoxy (polyethylene glycol)-2000] (DSPE-PEG). Nanostructures of approximately 142-202 nm of diameter and polydispersity index (PDI) of 0.069 to 0.190 were obtained and the vesicular shape confirmed by Transmission Electron Microscopy (TEM and cryo-TEM). The encapsulation efficiency (%EE) varied from 10% to 16%. All formulations presented activity in contact with ASNase substrate, indicating the liposomes permeability to Asn and/or enzyme adsorption at the nanostructures' surface; the highest activity was observed for DMPC/DSPE-PEG 10%. Finally, we investigated the activity against the Molt 4 leukemic cell line and found a lower IC50 for the DMPC/DSPE-PEG 10% formulation in comparison to the free enzyme, indicating our system could provide in vivo activity while protecting the enzyme from immune system recognition and proteases degradation.
Collapse
Affiliation(s)
- Marina de Souza Guimarães
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Jorge Javier Muso Cachumba
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Cecilia Zorzi Bueno
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Karin Mariana Torres-Obreque
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Grace Verónica Ruiz Lara
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Gisele Monteiro
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Leandro Ramos Souza Barbosa
- Department of General Physics, Institute of Physics, University of São Paulo, São Paulo 05508-000, SP, Brazil
- Brazilian Synchrotron Light Laboratory (LNLS), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas 13083-100, SP, Brazil
| | - Adalberto Pessoa
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
| | - Carlota de Oliveira Rangel-Yagui
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo 05508-000, SP, Brazil
| |
Collapse
|
17
|
Al Bostami RD, Abuwatfa WH, Husseini GA. Recent Advances in Nanoparticle-Based Co-Delivery Systems for Cancer Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2672. [PMID: 35957103 PMCID: PMC9370272 DOI: 10.3390/nano12152672] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 12/20/2022]
Abstract
Cancer therapies have advanced tremendously throughout the last decade, yet multiple factors still hinder the success of the different cancer therapeutics. The traditional therapeutic approach has been proven insufficient and lacking in the suppression of tumor growth. The simultaneous delivery of multiple small-molecule chemotherapeutic drugs and genes improves the effectiveness of each treatment, thus optimizing efficacy and improving synergistic effects. Nanomedicines integrating inorganic, lipid, and polymeric-based nanoparticles have been designed to regulate the spatiotemporal release of the encapsulated drugs. Multidrug-loaded nanocarriers are a potential strategy to fight cancer and the incorporation of co-delivery systems as a feasible treatment method has projected synergistic benefits and limited undesirable effects. Moreover, the development of co-delivery systems for maximum therapeutic impact necessitates better knowledge of the appropriate therapeutic agent ratio as well as the inherent heterogeneity of the cancer cells. Co-delivery systems can simplify clinical processes and increase patient quality of life, even though such systems are more difficult to prepare than single drug delivery systems. This review highlights the progress attained in the development and design of nano carrier-based co-delivery systems and discusses the limitations, challenges, and future perspectives in the design and fabrication of co-delivery systems.
Collapse
Affiliation(s)
- Rouba D. Al Bostami
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Waad H. Abuwatfa
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| | - Ghaleb A. Husseini
- Department of Chemical Engineering, College of Engineering, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
- Materials Science and Engineering Program, College of Arts and Sciences, American University of Sharjah, Sharjah P.O. Box 26666, United Arab Emirates
| |
Collapse
|
18
|
Ogunleye AZ, Piyawajanusorn C, Gonçalves A, Ghislat G, Ballester PJ. Interpretable Machine Learning Models to Predict the Resistance of Breast Cancer Patients to Doxorubicin from Their microRNA Profiles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2201501. [PMID: 35785523 PMCID: PMC9403644 DOI: 10.1002/advs.202201501] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/02/2022] [Indexed: 05/05/2023]
Abstract
Doxorubicin is a common treatment for breast cancer. However, not all patients respond to this drug, which sometimes causes life-threatening side effects. Accurately anticipating doxorubicin-resistant patients would therefore permit to spare them this risk while considering alternative treatments without delay. Stratifying patients based on molecular markers in their pretreatment tumors is a promising approach to advance toward this ambitious goal, but single-gene gene markers such as HER2 expression have not shown to be sufficiently predictive. The recent availability of matched doxorubicin-response and diverse molecular profiles across breast cancer patients permits now analysis at a much larger scale. 16 machine learning algorithms and 8 molecular profiles are systematically evaluated on the same cohort of patients. Only 2 of the 128 resulting models are substantially predictive, showing that they can be easily missed by a standard-scale analysis. The best model is classification and regression tree (CART) nonlinearly combining 4 selected miRNA isoforms to predict doxorubicin response (median Matthew correlation coefficient (MCC) and area under the curve (AUC) of 0.56 and 0.80, respectively). By contrast, HER2 expression is significantly less predictive (median MCC and AUC of 0.14 and 0.57, respectively). As the predictive accuracy of this CART model increases with larger training sets, its update with future data should result in even better accuracy.
Collapse
Affiliation(s)
- Adeolu Z. Ogunleye
- Cancer Research Center of Marseille (CRCM)INSERM U1068MarseilleF‐13009France
- Cancer Research Center of Marseille (CRCM)Institut Paoli‐CalmettesMarseilleF‐13009France
- Cancer Research Center of Marseille (CRCM)Aix‐Marseille UniversitéMarseilleF‐13284France
- Cancer Research Center of Marseille (CRCM)CNRS UMR7258MarseilleF‐13009France
| | - Chayanit Piyawajanusorn
- Cancer Research Center of Marseille (CRCM)INSERM U1068MarseilleF‐13009France
- Cancer Research Center of Marseille (CRCM)Institut Paoli‐CalmettesMarseilleF‐13009France
- Cancer Research Center of Marseille (CRCM)Aix‐Marseille UniversitéMarseilleF‐13284France
- Cancer Research Center of Marseille (CRCM)CNRS UMR7258MarseilleF‐13009France
| | - Anthony Gonçalves
- Cancer Research Center of Marseille (CRCM)INSERM U1068MarseilleF‐13009France
- Cancer Research Center of Marseille (CRCM)Institut Paoli‐CalmettesMarseilleF‐13009France
- Cancer Research Center of Marseille (CRCM)Aix‐Marseille UniversitéMarseilleF‐13284France
- Cancer Research Center of Marseille (CRCM)CNRS UMR7258MarseilleF‐13009France
| | - Ghita Ghislat
- Cancer Research Center of Marseille (CRCM)INSERM U1068MarseilleF‐13009France
- Cancer Research Center of Marseille (CRCM)Institut Paoli‐CalmettesMarseilleF‐13009France
- Cancer Research Center of Marseille (CRCM)Aix‐Marseille UniversitéMarseilleF‐13284France
- Cancer Research Center of Marseille (CRCM)CNRS UMR7258MarseilleF‐13009France
| | - Pedro J. Ballester
- Cancer Research Center of Marseille (CRCM)INSERM U1068MarseilleF‐13009France
- Cancer Research Center of Marseille (CRCM)Institut Paoli‐CalmettesMarseilleF‐13009France
- Cancer Research Center of Marseille (CRCM)Aix‐Marseille UniversitéMarseilleF‐13284France
- Cancer Research Center of Marseille (CRCM)CNRS UMR7258MarseilleF‐13009France
- Department of BioengineeringImperial College LondonLondonSW7 2AZUK
| |
Collapse
|
19
|
Ultrasound responsive Gd-DOTA/doxorubicin-loaded nanodroplet as a theranostic agent for magnetic resonance image-guided controlled release drug delivery of melanoma cancer. Eur J Pharm Sci 2022; 174:106207. [DOI: 10.1016/j.ejps.2022.106207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 05/08/2022] [Accepted: 05/11/2022] [Indexed: 11/24/2022]
|
20
|
Yang R, Wang L, Wu Z, Yin Y, Jiang SW. How Nanotechniques Could Vitalize the O-GlcNAcylation-Targeting Approach for Cancer Therapy. Int J Nanomedicine 2022; 17:1829-1841. [PMID: 35498390 PMCID: PMC9049135 DOI: 10.2147/ijn.s360488] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/11/2022] [Indexed: 12/11/2022] Open
Abstract
Accumulated data indicated that many types of cancers have increased protein O-GlcNAcylation at cell surface and inside cells. The aberrant O-GlcNAcylation is considered a potential therapeutic target. Although several types of compounds capable of inhibiting O-GlcNAcylation have been developed, their low solubility, poor permeability and delivery efficiency have impeded the application for in vivo and pre-clinical studies. Nanocarriers have the advantages of controllable drug release and active cancer-targeting capability. Moreover, nanoparticles can improve drug delivery efficiency and reduce the non-specific distribution in normal tissues by the enhanced permeability and retention (EPR) effect in cancer. Taking the advantage of O-GlcNAc-specific antibodies or lectins, nanoparticles could further improve their cancer-targeting capability. Although nanocarriers targeting the canonical N- and O-linked glycosylation have been extensively investigated for cancer detection and therapy, application of nanotechniques for the specific targeting of O-GlcNAcylation has not been actively pursued. This review summarizes the general features of GlcNAcylation and its alterations in cancers. Analyses are focused on the following areas: How the nanocarriers may improve the solubility and/or cell permeability of O-GlcNAc transferase (OGT) inhibitors; The modification of nanocarriers with lectins or antibodies for active targeting of O-GlcNAc; The nanocarriers-mediated co-delivery of OGT inhibitors and conventional drugs, which may lead to synergistic effects. Unsolved issues impeding the research progression on O-GlcNAcylation-targeting scheme are also discussed.
Collapse
Affiliation(s)
- Rui Yang
- Center of Reproductive Medicine, State Key Laboratory of Reproductive Medicine, Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, 214002, Jiangsu, People’s Republic of China
| | - Leilei Wang
- Department of Medical Genetics, Lianyungang Maternal and Child Health Hospital Affiliated to Yangzhou University, Lianyungang, 222000, Jiangsu, People’s Republic of China
| | - Zhifeng Wu
- Department of Ophthalmology, The Affiliated Wuxi Clinical College of Nantong University, Wuxi, 214002, Jiangsu, People’s Republic of China
| | - Yongxiang Yin
- Department of Pathology, The Affiliated Maternity and Child Health Hospital of Nanjing Medical University, Wuxi, 214002, Jiangsu, People’s Republic of China
| | - Shi-Wen Jiang
- Center of Reproductive Medicine, State Key Laboratory of Reproductive Medicine, Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, 214002, Jiangsu, People’s Republic of China
| |
Collapse
|
21
|
Pial MMH, Tomitaka A, Pala N, Roy U. Implantable Devices for the Treatment of Breast Cancer. JOURNAL OF NANOTHERANOSTICS 2022; 3:19-38. [PMID: 37600442 PMCID: PMC10438892 DOI: 10.3390/jnt3010003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023] Open
Abstract
Breast cancer is one of the leading causes of death in the female population worldwide. Standard treatments such as chemotherapy show noticeable results. However, along with killing cancer cells, it causes systemic toxicity and apoptosis of the nearby healthy cells, therefore patients must endure side effects during the treatment process. Implantable drug delivery devices that enhance therapeutic efficacy by allowing localized therapy with programmed or controlled drug release can overcome the shortcomings of conventional treatments. An implantable device can be composed of biopolymer materials, nanocomposite materials, or a combination of both. This review summarizes the recent research and current state-of-the art in these types of implantable devices and gives perspective for future directions.
Collapse
Affiliation(s)
| | - Asahi Tomitaka
- Department of Electrical and Computer Engineering, Florida International University, Miami, FL 33174, USA
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
- Department of Computer Science, University of Houston-Victoria, Victoria, TX 77901, USA
| | - Nezih Pala
- Department of Electrical and Computer Engineering, Florida International University, Miami, FL 33174, USA
| | - Upal Roy
- Department of Health and Biomedical Sciences, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| |
Collapse
|
22
|
Brickler M, Raskin A, Ryan TD. Current State of Pediatric Cardio-Oncology: A Review. CHILDREN (BASEL, SWITZERLAND) 2022; 9:127. [PMID: 35204848 PMCID: PMC8870613 DOI: 10.3390/children9020127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 01/03/2023]
Abstract
The landscape of pediatric oncology has dramatically changed over the course of the past several decades with five-year survival rates surpassing 80%. Anthracycline therapy has been the cornerstone of many chemotherapy regimens for pediatric patients since its introduction in the 1960s, and recent improved survival has been in large part due to advancements in chemotherapy, refinement of supportive care treatments, and development of novel therapeutics such as small molecule inhibitors, chimeric antigen receptor T-cell therapy, and immune checkpoint inhibitors. Unfortunately, many cancer-targeted therapies can lead to acute and chronic cardiovascular pathologies. The range of cardiotoxicity can vary but includes symptomatic or asymptotic heart failure, arrhythmias, coronary artery disease, valvar disease, pericardial disease, hypertension, and peripheral vascular disease. There is lack of data guiding primary prevention and treatment strategies in the pediatric population, which leads to substantial practice variability. Several important future research directions have been identified, including as they relate to cardiac disease, prevention strategies, management of cardiovascular risk factors, risk prediction, early detection, and the role of genetic susceptibility in development of cardiotoxicity. Continued collaborative research will be key in advancing the field. The ideal model for pediatric cardio-oncology is a proactive partnership between pediatric cardiologists and oncologists in order to better understand, treat, and ideally prevent cardiac disease in pediatric oncology patients.
Collapse
Affiliation(s)
| | | | - Thomas D. Ryan
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
| |
Collapse
|
23
|
Application of Optical Methods for Determination of Concentration of Doxorubicin in Blood and Plasma. Pharmaceuticals (Basel) 2022; 15:ph15020112. [PMID: 35215225 PMCID: PMC8880482 DOI: 10.3390/ph15020112] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 02/04/2023] Open
Abstract
The aim of presented research is to develop a simple and quick method of spectrophotometric detection for the determination of doxorubicin hydrochloride in blood and plasma. Anthracycline antibiotics are among the most effective antineoplastic agents. However, despite their high efficacy in the treatment of various types of cancer, their administration is limited primarily because they exhibit myocardial toxicity. This may be a limiting factor in the dosage of medications; nevertheless, drugs exhibiting this mechanism of action constitute a very important group of chemotherapeutics. One of the more widely studied antibiotics from the anthracycline group is doxorubicin. It exhibits the highest antineoplastic activity from among a number of derivative compounds. Because of the adverse effects of doxorubicin, especially cardiotoxicity, it is important to maintain control of its concentration in body fluids. The method in the study consists of extraction doxorubicin from the plasma or blood and measurements of the absorbance of light in the visible light range in a DOX solution with respect to a reference sample. The research used blood and plasma samples spiked with doxorubicin to give concentrations in the range of 0.2–10 µg/mL. Obtained LODs were 1.6 µg/mL and 1.2 µg/mL, respectively.
Collapse
|
24
|
Development of Pharmaceutical Nanomedicines: From the Bench to the Market. Pharmaceutics 2022; 14:pharmaceutics14010106. [PMID: 35057002 PMCID: PMC8777701 DOI: 10.3390/pharmaceutics14010106] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/15/2021] [Accepted: 12/30/2021] [Indexed: 12/13/2022] Open
Abstract
Nanotechnology plays a significant role in the field of medicine and in drug delivery, mainly due to the major limitations affecting the conventional pharmaceutical agents, and older formulations and delivery systems. The effect of nanotechnology on healthcare is already being felt, as various nanotechnology applications have been developed, and several nanotechnology-based medicines are now on the market. Across many parts of the world, nanotechnology draws increasing investment from public authorities and the private sector. Most conventional drug-delivery systems (CDDSs) have an immediate, high drug release after administration, leading to increased administration frequency. Thus, many studies have been carried out worldwide focusing on the development of pharmaceutical nanomedicines for translation into products manufactured by local pharmaceutical companies. Pharmaceutical nanomedicine products are projected to play a major role in the global pharmaceutical market and healthcare system. Our objectives were to examine the nanomedicines approved by the Food and Drug Administration (FDA) and the European Medicines Agency (EMA) in the global market, to briefly cover the challenges faced during their development, and to look at future perspectives. Additionally, the importance of nanotechnology in developing pharmaceutical products, the ideal properties of nanocarriers, the reasons behind the failure of some nanomedicines, and the important considerations in the development of nanomedicines will be discussed in brief.
Collapse
|
25
|
Flörkemeier I, Steinhauer TN, Hedemann N, Weimer JP, Rogmans C, van Mackelenbergh MT, Maass N, Clement B, Bauerschlag DO. High Antitumor Activity of the Dual Topoisomerase Inhibitor P8-D6 in Breast Cancer. Cancers (Basel) 2021; 14:2. [PMID: 35008166 PMCID: PMC8750241 DOI: 10.3390/cancers14010002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/17/2021] [Accepted: 12/18/2021] [Indexed: 12/12/2022] Open
Abstract
Breast cancer constitutes the leading cause of cancer deaths among females. However, numerous shortcomings, including low bioavailability, resistance and significant side effects, are responsible for insufficient treatment. The ultimate goal, therefore, is to improve the success rates and, thus, the range available treatment options for breast cancer. Consequently, the identification, development and evaluation of potential novel drugs such as P8-D6 with seminal antitumor capacities have a high clinical need. P8-D6 effectively induces apoptosis by acting as a dual topoisomerase I/II inhibitor. This study provides an overview of the effectiveness of P8-D6 in breast cancer with both 2D monolayers and 3D spheroids compared to standard therapeutic agents. For this drug effectiveness review, cell lines and ex vivo primary cells were used and cytotoxicity, apoptosis rates and membrane integrity were examined. This study provides evidence for a significant P8-D6-induced increase in apoptosis and cytotoxicity in breast cancer cells compared to the efficacy of standard therapeutic drugs. To sum up, P8-D6 is a fast and powerful inductor of apoptosis and might become a new and suitable therapeutic option for breast cancer in the future.
Collapse
Affiliation(s)
- Inken Flörkemeier
- Department of Gynaecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, 24105 Kiel, Germany; (I.F.); (N.H.); (J.P.W.); (C.R.); (M.T.v.M.); (N.M.)
- Pharmaceutical Institute, Department of Pharmaceutical and Medicinal Chemistry, Christian-Albrechts-University Kiel, 24118 Kiel, Germany; (T.N.S.); (B.C.)
| | - Tamara N. Steinhauer
- Pharmaceutical Institute, Department of Pharmaceutical and Medicinal Chemistry, Christian-Albrechts-University Kiel, 24118 Kiel, Germany; (T.N.S.); (B.C.)
| | - Nina Hedemann
- Department of Gynaecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, 24105 Kiel, Germany; (I.F.); (N.H.); (J.P.W.); (C.R.); (M.T.v.M.); (N.M.)
| | - Jörg Paul Weimer
- Department of Gynaecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, 24105 Kiel, Germany; (I.F.); (N.H.); (J.P.W.); (C.R.); (M.T.v.M.); (N.M.)
| | - Christoph Rogmans
- Department of Gynaecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, 24105 Kiel, Germany; (I.F.); (N.H.); (J.P.W.); (C.R.); (M.T.v.M.); (N.M.)
| | - Marion T. van Mackelenbergh
- Department of Gynaecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, 24105 Kiel, Germany; (I.F.); (N.H.); (J.P.W.); (C.R.); (M.T.v.M.); (N.M.)
| | - Nicolai Maass
- Department of Gynaecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, 24105 Kiel, Germany; (I.F.); (N.H.); (J.P.W.); (C.R.); (M.T.v.M.); (N.M.)
| | - Bernd Clement
- Pharmaceutical Institute, Department of Pharmaceutical and Medicinal Chemistry, Christian-Albrechts-University Kiel, 24118 Kiel, Germany; (T.N.S.); (B.C.)
| | - Dirk O. Bauerschlag
- Department of Gynaecology and Obstetrics, Christian-Albrechts-University Kiel and University Medical Center Schleswig-Holstein Campus Kiel, 24105 Kiel, Germany; (I.F.); (N.H.); (J.P.W.); (C.R.); (M.T.v.M.); (N.M.)
| |
Collapse
|
26
|
Dellapasqua S, Trillo Aliaga P, Munzone E, Bagnardi V, Pagan E, Montagna E, Cancello G, Ghisini R, Sangalli C, Negri M, Mazza M, Iorfida M, Cardillo A, Sciandivasci A, Bianco N, De Maio AP, Milano M, Campennì GM, Sansonno L, Viale G, Morra A, Leonardi MC, Galimberti V, Veronesi P, Colleoni M. Pegylated Liposomal Doxorubicin (Caelyx®) as Adjuvant Treatment in Early-Stage Luminal B-like Breast Cancer: A Feasibility Phase II Trial. Curr Oncol 2021; 28:5167-5178. [PMID: 34940072 PMCID: PMC8700739 DOI: 10.3390/curroncol28060433] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/19/2021] [Accepted: 11/30/2021] [Indexed: 11/28/2022] Open
Abstract
Background: Adjuvant chemotherapy for Luminal B-like breast cancers usually includes anthracycline-based regimens. However, some patients are reluctant to receive chemotherapy because of side-effects, especially alopecia, and ask for a “less intensive” or personalized approach. Patients and methods: We conducted a phase II feasibility trial to evaluate pegylated liposomal doxorubicin (PLD, Caelyx®) as adjuvant chemotherapy. Patients who received surgery for pT1–3, any N, and luminal B-like early-stage breast cancer (EBC) candidates for adjuvant chemotherapy were included. PLD was administered intravenously at 20 mg/m2 biweekly for eight courses. Endocrine therapy was given according to menopausal status. Trastuzumab was administered in HER2-positive disease. The primary endpoint was to evaluate the feasibility of this regimen, defined as the ability of a patient to achieve a relative dose intensity (RDI) of at least 85% of the eight cycles of treatment. Secondary endpoints included adverse events (AEs), tolerability, breast cancer-free survival, disease-free survival, and overall survival. Results: From March 2016 to July 2018, 63 patients were included in the trial. Median age was 49 years (range: 33–76), with mostly pre- and peri-menopausal (65%) and stage I–II (94%). Only 5% of patients had HER2-positive EBC. Median RDI was 100% (range: 12.5–100%; interquartile range, IQR: 87.5–100%). The proportion of patients meeting the primary endpoint was 84% (95% confidence interval, CI: 73–92%). Overall, 55 out of 63 enrolled patients completed treatment (87%, 95% CI: 77–94%). Most common AEs were palmar-plantar erythrodysesthesia (12.2%), fatigue (10.4%), and mucositis (8.5%). Only 13% of patients had G3 AEs. None had alopecia. After a median follow-up of 3.9 years (range: 0.3–4.7) two distant events were observed, and all patients were alive at the date of last visit. Conclusions: The trial successfully met its primary endpoint: the regimen was feasible and well tolerated and could be considered for further evaluation as a treatment option for patients with contraindications to standard anthracyclines or requiring a personalized, less intensive approach.
Collapse
Affiliation(s)
- Silvia Dellapasqua
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
- Correspondence: ; Tel.: +39-02-57-489-502
| | - Pamela Trillo Aliaga
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
| | - Elisabetta Munzone
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
| | - Vincenzo Bagnardi
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, 20126 Milan, Italy; (V.B.); (E.P.)
| | - Eleonora Pagan
- Department of Statistics and Quantitative Methods, University of Milan-Bicocca, 20126 Milan, Italy; (V.B.); (E.P.)
| | - Emilia Montagna
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
| | - Giuseppe Cancello
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
| | - Raffaella Ghisini
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
| | - Claudia Sangalli
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
| | - Mara Negri
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
| | - Manuelita Mazza
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
| | - Monica Iorfida
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
| | - Anna Cardillo
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
| | - Angela Sciandivasci
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
| | - Nadia Bianco
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
| | - Ana Paula De Maio
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
| | - Monica Milano
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
| | - Giuseppe Maria Campennì
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
| | - Loredana Sansonno
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
| | - Giuseppe Viale
- Department of Pathology, European Institute of Oncology IRCCS and University of Milan, 20141 Milan, Italy;
| | - Anna Morra
- Division of Radiotherapy, European Institute of Oncology IRCCS, 20141 Milan, Italy; (A.M.); (M.C.L.)
| | - Maria Cristina Leonardi
- Division of Radiotherapy, European Institute of Oncology IRCCS, 20141 Milan, Italy; (A.M.); (M.C.L.)
| | - Viviana Galimberti
- Division of Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (V.G.); (P.V.)
| | - Paolo Veronesi
- Division of Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (V.G.); (P.V.)
| | - Marco Colleoni
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (P.T.A.); (E.M.); (E.M.); (G.C.); (R.G.); (C.S.); (M.N.); (M.M.); (M.I.); (A.C.); (A.S.); (N.B.); (A.P.D.M.); (M.M.); (G.M.C.); (L.S.); (M.C.)
| |
Collapse
|
27
|
Zaid ASA, Aleissawy AE, Yahia IS, Yassien MA, Hassouna NA, Aboshanab KM. Streptomyces griseus KJ623766: A Natural Producer of Two Anthracycline Cytotoxic Metabolites β- and γ-Rhodomycinone. Molecules 2021; 26:molecules26134009. [PMID: 34209170 PMCID: PMC8271628 DOI: 10.3390/molecules26134009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Background: This study aimed to produce, purify, structurally elucidate, and explore the biological activities of metabolites produced by Streptomyces (S.) griseus isolate KJ623766, a recovered soil bacterium previously screened in our lab that showed promising cytotoxic activities against various cancer cell lines. Methods: Production of cytotoxic metabolites from S. griseus isolate KJ623766 was carried out in a 14L laboratory fermenter under specified optimum conditions. Using a 3-(4,5-dimethylthazol-2-yl)-2,5-diphenyl tetrazolium-bromide assay, the cytotoxic activity of the ethyl acetate extract against Caco2 and Hela cancer cell lines was determined. Bioassay-guided fractionation of the ethyl acetate extract using different chromatographic techniques was used for cytotoxic metabolite purification. Chemical structures of the purified metabolites were identified using mass, 1D, and 2D NMR spectroscopic analysis. Results: Bioassay-guided fractionation of the ethyl acetate extract led to the purification of two cytotoxic metabolites, R1 and R2, of reproducible amounts of 5 and 1.5 mg/L, respectively. The structures of R1 and R2 metabolites were identified as β- and γ-rhodomycinone with CD50 of 6.3, 9.45, 64.8 and 9.11, 9.35, 67.3 µg/mL against Caco2, Hela and Vero cell lines, respectively. Values were comparable to those of the positive control doxorubicin. Conclusions: This is the first report about the production of β- and γ-rhodomycinone, two important scaffolds for synthesis of anticancer drugs, from S. griseus.
Collapse
Affiliation(s)
- Ahmed S. Abu Zaid
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Organization of African Unity St, Abbassia, Cairo P.O. Box 11566, Egypt; (A.S.A.Z.); (M.A.Y.); (N.A.H.)
| | - Ahmed E. Aleissawy
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Organization of African Unity St, Abbassia, Cairo P.O. Box 11566, Egypt;
| | - Ibrahim S. Yahia
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha P.O. Box 9004, Saudi Arabia;
- Advanced Functional Materials & Optoelectronic Laboratory (AFMOL), Department of Physics, Faculty of Science, King Khalid University, Abha P.O. Box 9004, Saudi Arabia
- Nanoscience Laboratory for Environmental and Bio-Medical Applications (NLEBA), Semiconductor Lab., Physics Department, Faculty of Education, Ain Shams University, Cairo P.O. Box 11757, Egypt
| | - Mahmoud A. Yassien
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Organization of African Unity St, Abbassia, Cairo P.O. Box 11566, Egypt; (A.S.A.Z.); (M.A.Y.); (N.A.H.)
| | - Nadia A. Hassouna
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Organization of African Unity St, Abbassia, Cairo P.O. Box 11566, Egypt; (A.S.A.Z.); (M.A.Y.); (N.A.H.)
| | - Khaled M. Aboshanab
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Organization of African Unity St, Abbassia, Cairo P.O. Box 11566, Egypt; (A.S.A.Z.); (M.A.Y.); (N.A.H.)
- Correspondence: ; Tel.: +20-100-758-2620
| |
Collapse
|
28
|
Role of Calixarene in Chemotherapy Delivery Strategies. Molecules 2021; 26:molecules26133963. [PMID: 34209495 PMCID: PMC8272165 DOI: 10.3390/molecules26133963] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 01/14/2023] Open
Abstract
Since cancer is a multifactorial disease with a high mortality rate, the study of new therapeutic strategies is one of the main objectives in modern research. Numerous chemotherapeutic agents, although widely used, have the disadvantage of being not very soluble in water or selective towards cancerous cells, with consequent side effects. Therefore, in recent years, a greater interest has emerged in innovative drug delivery systems (DDSs) such as calixarene, a third-generation supramolecular compound. Calixarene and its water-soluble derivatives show good biocompatibility and have low cytotoxicity. Thanks to their chemical–physical characteristics, calixarenes can be easily functionalized, and by itself can encapsulate host molecules forming nanostructures capable of releasing drugs in a controlled way. The encapsulation of anticancer drugs in a calixarene derivate improves their bioavailability and efficacy. Thus, the use of calixarenes as carriers of anticancer drugs could reduce their side effects and increase their affinity towards the target. This review summarizes the numerous research advances regarding the development of calixarene nanoparticles capable of encapsulating various anticancer drugs.
Collapse
|
29
|
Anthracycline-related cardiotoxicity in older patients with acute myeloid leukemia: a Young SIOG review paper. Blood Adv 2021; 4:762-775. [PMID: 32097461 DOI: 10.1182/bloodadvances.2019000955] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 01/13/2020] [Indexed: 12/16/2022] Open
Abstract
The incidence of acute myeloid leukemia (AML) increases with age. Intensive induction chemotherapy containing cytarabine and an anthracycline has been part of the upfront and salvage treatment of AML for decades. Anthracyclines are associated with a significant risk of cardiotoxicity (especially anthracycline-related left ventricular dysfunction [ARLVD]). In the older adult population, the higher prevalence of cardiac comorbidities and risk factors may further increase the risk of ARLVD. In this article of the Young International Society of Geriatric Oncology group, we review the prevalence of ARLVD in patients with AML and factors predisposing to ARLVD, focusing on older adults when possible. In addition, we review the assessment of cardiac function and management of ARLVD during and after treatment. It is worth noting that only a minority of clinical trials focus on alternative treatment strategies in patients with mildly declined left ventricular ejection fraction or at a high risk for ARLVD. The limited evidence for preventive strategies to ameliorate ARLVD and alternative strategies to anthracycline use in the setting of cardiac comorbidities are discussed. Based on extrapolation of findings from younger adults and nonrandomized trials, we recommend a comprehensive baseline evaluation of cardiac function by imaging, cardiac risk factors, and symptoms to risk stratify for ARLVD. Anthracyclines remain an appropriate choice for induction although careful risk-stratification based on cardiac disease, risk factors, and predicted chemotherapy-response are warranted. In case of declined left ventricular ejection fraction, alternative strategies should be considered.
Collapse
|
30
|
Di Francesco M, Celia C, Cristiano MC, d’Avanzo N, Ruozi B, Mircioiu C, Cosco D, Di Marzio L, Fresta M. Doxorubicin Hydrochloride-Loaded Nonionic Surfactant Vesicles to Treat Metastatic and Non-Metastatic Breast Cancer. ACS OMEGA 2021; 6:2973-2989. [PMID: 33553916 PMCID: PMC7860091 DOI: 10.1021/acsomega.0c05350] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/08/2020] [Indexed: 05/06/2023]
Abstract
Doxorubicin hydrochloride (DOX) is currently used to treat orthotropic and metastatic breast cancer. Because of its side effects, the use of DOX in cancer patients is sometimes limited; for this reason, several scientists tried designing drug delivery systems which can improve drug therapeutic efficacy and decrease its side effects. In this study, we designed, prepared, and physiochemically characterized nonionic surfactant vesicles (NSVs) which are obtained by self-assembling different combinations of hydrophilic (Tween 20) and hydrophobic (Span 20) surfactants, with cholesterol. DOX was loaded in NSVs using a passive and pH gradient remote loading procedure, which increased drug loading from ∼1 to ∼45%. NSVs were analyzed in terms of size, shape, size distribution, zeta potential, long-term stability, entrapment efficiency, and release kinetics, and nanocarriers having the best physiochemical parameters were selected for further in vitro tests. NSVs with and without DOX were stable and showed a sustained drug release up to 72 h. In vitro studies, with MCF-7 and MDA MB 468 cells, demonstrated that NSVs, containing Span 20, were better internalized in MCF-7 and MDA MB 468 cells than NSVs with Tween 20. NSVs increased the anticancer effect of DOX in MCF-7 and MDA MB 468 cells, and this effect is time and dose dependent. In vitro studies using metastatic and nonmetastatic breast cancer cells also demonstrated that NSVs, containing Span 20, had higher cytotoxicity than NSVs with Tween 20. The resulting data suggested that DOX-loaded NSVs could be a promising nanocarrier for the potential treatment of metastatic breast cancer.
Collapse
Affiliation(s)
- Martina Di Francesco
- Department
of Health Sciences, University of Catanzaro
“Magna Graecia”, Campus Universitario “S. Venuta” s.n.c., 88100 Catanzaro, Italy
- Laboratory
of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Christian Celia
- Department
of Pharmacy, University of Chieti−Pescara
“G. d’Annunzio”, Via dei Vestini 31, 66100 Chieti, Italy
| | - Maria Chiara Cristiano
- Department
of Clinical and Experimental Medicine, University
of Catanzaro “Magna Graecia”, Campus Universitario “S. Venuta”
s.n.c., 88100 Catanzaro, Italy
| | - Nicola d’Avanzo
- Department
of Health Sciences, University of Catanzaro
“Magna Graecia”, Campus Universitario “S. Venuta” s.n.c., 88100 Catanzaro, Italy
- Department
of Pharmacy, University of Chieti−Pescara
“G. d’Annunzio”, Via dei Vestini 31, 66100 Chieti, Italy
| | - Barbara Ruozi
- Department
of Life Sciences, University of Modena and
Reggio Emilia, Via Campi
183, I-41100 Modena, Italy
| | - Constantin Mircioiu
- Department
of Applied Mathematics and Biostatistics, Faculty of Pharmacy, “Carol Davila” University of Medicine
and Pharmacy, 020956 Bucharest, Romania
| | - Donato Cosco
- Department
of Health Sciences, University of Catanzaro
“Magna Graecia”, Campus Universitario “S. Venuta” s.n.c., 88100 Catanzaro, Italy
| | - Luisa Di Marzio
- Department
of Pharmacy, University of Chieti−Pescara
“G. d’Annunzio”, Via dei Vestini 31, 66100 Chieti, Italy
| | - Massimo Fresta
- Department
of Health Sciences, University of Catanzaro
“Magna Graecia”, Campus Universitario “S. Venuta” s.n.c., 88100 Catanzaro, Italy
| |
Collapse
|
31
|
Levi M, Salaroli R, Parenti F, De Maria R, Zannoni A, Bernardini C, Gola C, Brocco A, Marangio A, Benazzi C, Muscatello LV, Brunetti B, Forni M, Sarli G. Doxorubicin treatment modulates chemoresistance and affects the cell cycle in two canine mammary tumour cell lines. BMC Vet Res 2021; 17:30. [PMID: 33461558 PMCID: PMC7814552 DOI: 10.1186/s12917-020-02709-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Doxorubicin (DOX) is widely used in both human and veterinary oncology although the onset of multidrug resistance (MDR) in neoplastic cells often leads to chemotherapy failure. Better understanding of the cellular mechanisms that circumvent chemotherapy efficacy is paramount. The aim of this study was to investigate the response of two canine mammary tumour cell lines, CIPp from a primary tumour and CIPm, from its lymph node metastasis, to exposure to EC50(20h) DOX at 12, 24 and 48 h of treatment. We assessed the uptake and subcellular distribution of DOX, the expression and function of P-glycoprotein (P-gp) and Breast Cancer Resistance Protein (BCRP), two important MDR mediators. To better understand this phenomenon the effects of DOX on the cell cycle and Ki67 cell proliferation index and the expression of p53 and telomerase reverse transcriptase (TERT) were also evaluated by immunocytochemistry (ICC). RESULTS Both cell lines were able to uptake DOX within the nucleus at 3 h treatment while at 48 h DOX was absent from the intracellular compartment (assessed by fluorescence microscope) in all the surviving cells. CIPm, originated from the metastatic tumour, were more efficient in extruding P-gp substrates. By ICC and qRT-PCR an overall increase in both P-gp and BCRP were observed at 48 h of EC50(20h) DOX treatment in both cell lines and were associated with a striking increase in the percentage of p53 and TERT expressing cells by ICC. The cell proliferation fraction was decreased at 48 h in both cell lines and cell cycle analysis showed a DOX-induced arrest in the S phase for CIPp, while CIPm had an increase in cellular death without arrest. Both cells lines were therefore composed by a fraction of cells sensible to DOX that underwent apoptosis/necrosis. CONCLUSIONS DOX administration results in interlinked modifications in the cellular population including a substantial effect on the cell cycle, in particular arrest in the S phase for CIPp and the selection of a subpopulation of neoplastic cells bearing MDR phenotype characterized by P-gp and BCRP expression, TERT activation, p53 accumulation and decrease in the proliferating fraction. Important information is given for understanding the dynamic and mechanisms of the onset of drug resistance in a neoplastic cell population.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- Animals
- Cell Cycle/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Dogs
- Doxorubicin/pharmacology
- Drug Resistance, Neoplasm/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Mammary Neoplasms, Animal
- Multidrug Resistance-Associated Proteins/genetics
- Multidrug Resistance-Associated Proteins/metabolism
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
Collapse
Affiliation(s)
- Michela Levi
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, Italy
| | - Roberta Salaroli
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, Italy
| | - Federico Parenti
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, Italy
| | - Raffaella De Maria
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Augusta Zannoni
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, Italy
| | - Chiara Bernardini
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, Italy
| | - Cecilia Gola
- Department of Veterinary Sciences, University of Turin, Grugliasco, Italy
| | - Antonio Brocco
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, Italy
| | - Asia Marangio
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, Italy
| | - Cinzia Benazzi
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, Italy
| | - Luisa Vera Muscatello
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, Italy
| | - Barbara Brunetti
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, Italy
| | - Monica Forni
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, Italy
| | - Giuseppe Sarli
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano dell'Emilia, Italy.
| |
Collapse
|
32
|
Schilt Y, Berman T, Wei X, Nativ-Roth E, Barenholz Y, Raviv U. Effect of the ammonium salt anion on the structure of doxorubicin complex and PEGylated liposomal doxorubicin nanodrugs. Biochim Biophys Acta Gen Subj 2021; 1865:129849. [PMID: 33460771 DOI: 10.1016/j.bbagen.2021.129849] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 01/06/2021] [Accepted: 01/11/2021] [Indexed: 10/22/2022]
Abstract
BACKGROUND In Doxil®, PEGylated nanoliposomes are created by hydration of the lipids in ammonium sulfate, and are remotely loaded with doxorubicin by a transmembrane ammonium gradient. The ammonium sulfate is then removed from the external aqueous phase, surrounding the liposomes, and replaced by an isoosmotic sucrose solution in 10 mM histidine buffer at pH 6.5. METHODS We prepared PEGylated liposomal doxorubicin (PLD) with a series of ammonium monovalent salts that after remote loading became the intraliposome doxorubicin counteranions. We analyzed the liposomes by solution X-ray scattering, differential scanning calorimetry, and electron micropscopy. RESULTS PLDs prepared with sulfonic acid derivatives as counteranion exhibited chemical and physical stabilities. We determined the effect of these ammonium salt counteranions on the structure, morphology, and thermotropic behavior of the PEGylated nanoliposomes, formed before and after doxorubicin loading, and the bulk properties of the doxorubicin-counteranion complexes. By comparing the structure of the doxorubicin complexes in the bulk and inside the nanoliposomes, we revealed the effect of confinement on the structure and doxorubicin release rate for each of the derivatives of the ammonium sulfonic acid counteranions. CONCLUSIONS We found that the extent and direction of the doxorubicin confinement effect and its release rate were strongly dependent on the type of counteranion. The counteranions, however, neither affected the structure and thermotropic behavior of the liposome membrane, nor the thickness and density of the liposome PEG layers. In an additional study, it was demonstrated that PLD made with ammonium-methane sulfonate exhibit a much lower Hand and Foot syndrome. GENERAL SIGNIFICANCE The structure, physical state, and pharmacokinetics of doxorubicin in PEGylated nanoliposomes, prepared by transmembrane remote loading using gradients of ammonium salts, strongly depend on the counteranions.
Collapse
Affiliation(s)
- Yaelle Schilt
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 91904, Israel; The Center for Nanoscience and Nanotechnology of the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Tal Berman
- Laboratory of Membrane and Liposome Research, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel; The Center for Nanoscience and Nanotechnology of the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Xiaohui Wei
- Laboratory of Membrane and Liposome Research, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel; The Center for Nanoscience and Nanotechnology of the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Einat Nativ-Roth
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Yechezkel Barenholz
- Laboratory of Membrane and Liposome Research, IMRIC, The Hebrew University-Hadassah Medical School, Jerusalem, Israel; The Center for Nanoscience and Nanotechnology of the Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Uri Raviv
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 91904, Israel; The Center for Nanoscience and Nanotechnology of the Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
33
|
Alawak M, Abu Dayyih A, Mahmoud G, Tariq I, Duse L, Goergen N, Engelhardt K, Reddy Pinnapireddy S, Jedelská J, Awak M, König AM, Brüßler J, Bartsch JW, Bakowsky U. ADAM 8 as a novel target for doxorubicin delivery to TNBC cells using magnetic thermosensitive liposomes. Eur J Pharm Biopharm 2020; 158:390-400. [PMID: 33338603 DOI: 10.1016/j.ejpb.2020.12.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 11/21/2020] [Accepted: 12/13/2020] [Indexed: 01/04/2023]
Abstract
Metastatic breast cancer is one of the most common causes of cancer-related death in women worldwide. The transmembrane metalloprotease-disintegrin (ADAM8) protein is highly overexpressed in triple-negative breast cancer (TNBC) cells and potentiates tumor cell invasion and extracellular matrix remodeling. Exploiting the high expression levels of ADAM8 in TNBC cells by delivering anti-ADAM8 antibodies efficiently to the targeted site can be a promising strategy for therapy of TNBC. For instance, a targeted approach with the aid of ultra-high field magnetic resonance imaging (UHF-MRI) activatable thermosensitive liposomes (LipTS-GD) could specifically increase the intracellular accumulation of cytotoxic drugs. The surface of doxorubicin-loaded LipTS-GD was modified by covalent coupling of MAB1031 antibody (LipTS-GD-MAB) in order to target the overexpressed ADAM8 in ADAM8 positive MDA-MB-231 cells. Physicochemical characterization of these liposomes was performed using size, surface morphology and UHF-MRI imaging analysis. In vitro cell targeting was investigated by the washing and circulation method. Intracellular trafficking and lysosomal colocalization were assessed by fluorescence microscopy. Cell viability, biocompatibility and in-ovo CAM assays were performed to determine the effectiveness and safety profiles of liposome formulations. Our results show specific binding and induction of doxorubicin release after LipTS-GD-MAB treatment caused a higher cytotoxic effect at the cellular target site.
Collapse
Affiliation(s)
- Mohamad Alawak
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, 35037 Marburg, Germany
| | - Alice Abu Dayyih
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, 35037 Marburg, Germany
| | - Gihan Mahmoud
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, 35037 Marburg, Germany; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Helwan University, Ain Helwan, 11795 Cairo, Egypt
| | - Imran Tariq
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, 35037 Marburg, Germany; Punjab University College of Pharmacy, University of the Punjab, 54000 Lahore, Pakistan
| | - Lili Duse
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, 35037 Marburg, Germany
| | - Nathalie Goergen
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, 35037 Marburg, Germany
| | - Konrad Engelhardt
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, 35037 Marburg, Germany
| | | | - Jarmila Jedelská
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, 35037 Marburg, Germany
| | - Muhannad Awak
- Department of Neurosurgery, Wolfsburg Hospital, 38440 Wolfsburg, Germany
| | - Alexander M König
- Department of Diagnostic and Interventional Radiology, University of Marburg, 35032 Marburg, Germany
| | - Jana Brüßler
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, 35037 Marburg, Germany
| | - Jörg W Bartsch
- Department of Neurosurgery, University of Marburg, University Hospital Marburg, 35032 Marburg, Germany
| | - Udo Bakowsky
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, 35037 Marburg, Germany.
| |
Collapse
|
34
|
Chemotherapy Options beyond the First Line in HER-Negative Metastatic Breast Cancer. JOURNAL OF ONCOLOGY 2020; 2020:9645294. [PMID: 33312203 PMCID: PMC7719522 DOI: 10.1155/2020/9645294] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 10/05/2020] [Accepted: 11/11/2020] [Indexed: 12/24/2022]
Abstract
Despite the recent advances in the biological understanding of breast cancer (BC), chemotherapy still represents a key component in the armamentarium for this disease. Different agents are available as mono-chemotherapy options in patients with locally advanced or metastatic BC (MBC) who progress after a first- and second-line treatment with anthracyclines and taxanes. However, no clear indication exists on what the best option is in some populations, such as heavily pretreated, elderly patients, triple-negative BC (TNBC), and those who do not respond to the first-line therapy. In this article, we summarize available literature evidence on different chemotherapy agents used beyond the first-line, in locally advanced or MBC patients, including rechallenge with anthracyclines and taxanes, antimetabolite and antimicrotubule agents, such as vinorelbine, capecitabine, eribulin, ixabepilone, and the newest developed agents, such as vinflunine, irinotecan, and etirinotecan.
Collapse
|
35
|
Ghasemi K, Vaseghi G, Mansourian M. Pharmacological interventions for preventing anthracycline-induced clinical and subclinical cardiotoxicity: A network meta-analysis of metastatic breast cancer. J Oncol Pharm Pract 2020; 27:414-427. [PMID: 33081570 DOI: 10.1177/1078155220965674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Doxorubicin- and epirubicin-induced cardiotoxicities are life threatening for those suffering from breast cancer. Comparing the effects of different strategies on the prevention of these agent-induced cardiotoxicities remains unexplored.Data sources: A comprehensive review of clinical trials was performed on the prevention of epirubicin- and/or doxorubicin-induced cardiotoxicity in HER2-positive metastatic breast cancer patients. The reduction in ejection fraction was directed at evaluating cardiac toxicity.Data summary: Fourteen articles evaluated cardiotoxicity as a condition among 2945 individuals, evaluating doxorubicin, epirubicin, Liposomal Doxorubicin (LD), Pegylated Liposomal Doxorubicin (PLD), dexrazoxane plus doxorubicin or epirubicin, and Angiotensin-Converting Enzyme Inhibitors (ACEIs) plus doxorubicin. Pooled Odds Ratio (OR) of 0.043 with a 95% credible interval (CrI) between 0.005 and 0.22 indicated that the dexrazoxane plus epirubicin reduced the number of cardiac events compared with doxorubicin. Furthermore, doxorubicin and epirubicin represented the most effective interventions with a 52% probability of success. Also, the best treatment for reducing Congestive Heart Failure (CHF) was dexrazoxane plus epirubicin with a probability of 43%. For the Left Ventricular Ejection Fraction (LVEF) reduction outcome, ACEIs plus doxorubicin was ranked first with a success probability of 61.2% and they could significantly prevent the reduction in LVEF compared with LD, epirubicin, or doxorubicin. CONCLUSION Our data suggested that angiotensin-converting enzyme inhibitors and dexrazoxane plus epirubicin were the most effective interventions for preventing cardiotoxicity and CHF. However, ACEIs plus doxorubicin was the best treatment for preventing LVEF reduction.
Collapse
Affiliation(s)
- Khojasteh Ghasemi
- Department of Epidemiology and Biostatistics, Health School, Isfahan University of Medical Sciences, Isfahan, Iran.,Student Research Committee, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Golnaz Vaseghi
- Applied Physiology Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medicine Sciences, Isfahan, Iran
| | - Marjan Mansourian
- Department of Epidemiology and Biostatistics, Health School, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
36
|
Steinborn B, Lächelt U. Metal-organic Nanopharmaceuticals. Pharm Nanotechnol 2020; 8:163-190. [PMID: 32316907 DOI: 10.2174/2211738508666200421113215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/31/2020] [Accepted: 04/07/2020] [Indexed: 11/22/2022]
Abstract
Coordinative interactions between multivalent metal ions and drug derivatives with Lewis base functions give rise to nanoscale coordination polymers (NCPs) as delivery systems. As the pharmacologically active agent constitutes a main building block of the nanomaterial, the resulting drug loadings are typically very high. By additionally selecting metal ions with favorable pharmacological or physicochemical properties, the obtained NCPs are predominantly composed of active components which serve individual purposes, such as pharmacotherapy, photosensitization, multimodal imaging, chemodynamic therapy or radiosensitization. By this approach, the assembly of drug molecules into NCPs modulates pharmacokinetics, combines pharmacological drug action with specific characteristics of metal components and provides a strategy to generate tailorable multifunctional nanoparticles. This article reviews different applications and recent examples of such highly functional nanopharmaceuticals with a high 'material economy'. Lay Summary: Nanoparticles, that are small enough to circulate in the bloodstream and can carry cargo molecules, such as drugs, imaging or contrast agents, are attractive materials for pharmaceutical applications. A high loading capacity is a generally aspired parameter of nanopharmaceuticals to minimize patient exposure to unnecessary nanomaterial. Pharmaceutical agents containing Lewis base functions in their molecular structure can directly be assembled into metal-organic nanopharmaceuticals by coordinative interaction with metal ions. Such coordination polymers generally feature extraordinarily high loading capacities and the flexibility to encapsulate different agents for a simultaneous delivery in combination therapy or 'theranostic' applications.
Collapse
Affiliation(s)
- Benjamin Steinborn
- Department of Pharmacy and Center for NanoScience (CeNS), LMU Munich, 81377 Munich, Germany
| | - Ulrich Lächelt
- Department of Pharmacy and Center for NanoScience (CeNS), LMU Munich, 81377 Munich, Germany
| |
Collapse
|
37
|
Doerr V, Montalvo RN, Kwon OS, Talbert EE, Hain BA, Houston FE, Smuder AJ. Prevention of Doxorubicin-Induced Autophagy Attenuates Oxidative Stress and Skeletal Muscle Dysfunction. Antioxidants (Basel) 2020; 9:antiox9030263. [PMID: 32210013 PMCID: PMC7139604 DOI: 10.3390/antiox9030263] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/16/2020] [Accepted: 03/20/2020] [Indexed: 12/31/2022] Open
Abstract
Clinical use of the chemotherapeutic doxorubicin (DOX) promotes skeletal muscle atrophy and weakness, adversely affecting patient mobility and strength. Although the mechanisms responsible for DOX-induced skeletal muscle dysfunction remain unclear, studies implicate the significant production of reactive oxygen species (ROS) in this pathology. Supraphysiological ROS levels can enhance protein degradation via autophagy, and it is established that DOX upregulates autophagic signaling in skeletal muscle. To determine the precise contribution of accelerated autophagy to DOX-induced skeletal muscle dysfunction, we inhibited autophagy in the soleus via transduction of a dominant negative mutation of the autophagy related 5 (ATG5) protein. Targeted inhibition of autophagy prevented soleus muscle atrophy and contractile dysfunction acutely following DOX administration, which was associated with a reduction in mitochondrial ROS and maintenance of mitochondrial respiratory capacity. These beneficial modifications were potentially the result of enhanced transcription of antioxidant response element-related genes and increased antioxidant capacity. Specifically, our results showed significant upregulation of peroxisome proliferator-activated receptor gamma co-activator 1-alpha, nuclear respiratory factor-1, nuclear factor erythroid-2-related factor-2, nicotinamide-adenine dinucleotide phosphate quinone dehydrogenase-1, and catalase in the soleus with DOX treatment when autophagy was inhibited. These findings establish a significant role of autophagy in the development of oxidative stress and skeletal muscle weakness following DOX administration.
Collapse
Affiliation(s)
- Vivian Doerr
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA; (V.D.); (R.N.M.)
| | - Ryan N. Montalvo
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA; (V.D.); (R.N.M.)
| | - Oh Sung Kwon
- Department of Kinesiology, University of Connecticut, Storrs, CT 06269, USA;
| | - Erin E. Talbert
- Department of Health and Human Physiology, University of Iowa, Iowa City, IA 52242, USA;
| | - Brian A. Hain
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA;
| | - Fraser E. Houston
- Department of Health Sciences and Human Performance, University of Tampa, Tampa, FL 33606, USA;
| | - Ashley J. Smuder
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA; (V.D.); (R.N.M.)
- Correspondence:
| |
Collapse
|
38
|
Alanazi AM, Fadda L, Alhusaini A, Ahmad R, Hasan IH, Mahmoud AM. Liposomal Resveratrol and/or Carvedilol Attenuate Doxorubicin-Induced Cardiotoxicity by Modulating Inflammation, Oxidative Stress and S100A1 in Rats. Antioxidants (Basel) 2020; 9:antiox9020159. [PMID: 32079097 PMCID: PMC7070570 DOI: 10.3390/antiox9020159] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/12/2020] [Accepted: 02/14/2020] [Indexed: 12/23/2022] Open
Abstract
Doxorubicin (DOX) is a cytotoxic anthracycline antibiotic and one of the important chemotherapeutic agents for different types of cancers. DOX treatment is associated with adverse effects, particularly cardiac dysfunction. This study examined the cardioprotective effects of carvedilol (CAR) and/or resveratrol (RES) and liposomal RES (LIPO-RES) against DOX-induced cardiomyopathy, pointing to their modulatory effect on oxidative stress, inflammation, S100A1 and sarco/endoplasmic reticulum calcium ATPase2a (SERCA2a). Rats received CAR (30 mg/kg) and/or RES (20 mg/kg) or LIPO-RES (20 mg/kg) for 6 weeks and were challenged with DOX (2 mg/kg) twice per week from week 2 to week 6. DOX-administered rats exhibited a significant increase in serum creatine kinase-MB (CK-MB), troponin-I and lactate dehydrogenase (LDH) along with histological alterations, reflecting cardiac cell injury. Cardiac toll-like receptor 4 (TLR-4), inducible nitric oxide synthase (iNOS), tumor necrosis factor (TNF)-α and interleukin (IL)-6 protein expression were up-regulated, and lipid peroxidation was increased in DOX-administered rats. Treatment with CAR, RES or LIPO-RES as well as their alternative combinations ameliorated all observed biochemical and histological alterations with the most potent effect exerted by CAR/LIPO-RES. All treatments increased cardiac antioxidants, and the expression of S100A1 and SERCA2a. In conclusion, the present study conferred new evidence on the protective effects of CAR and its combination with either RES or LIPO-RES on DOX-induced inflammation, oxidative stress and calcium dysregulation.
Collapse
Affiliation(s)
- Abeer M. Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.M.A.); (L.F.); (R.A.); (I.H.H.)
| | - Laila Fadda
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.M.A.); (L.F.); (R.A.); (I.H.H.)
| | - Ahlam Alhusaini
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.M.A.); (L.F.); (R.A.); (I.H.H.)
- Correspondence: (A.A.); (A.M.M.)
| | - Rehab Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.M.A.); (L.F.); (R.A.); (I.H.H.)
| | - Iman H. Hasan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (A.M.A.); (L.F.); (R.A.); (I.H.H.)
| | - Ayman M. Mahmoud
- Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62514, Egypt
- Correspondence: (A.A.); (A.M.M.)
| |
Collapse
|
39
|
Yazdani H, Kaul E, Bazgir A, Maysinger D, Kakkar A. Telodendrimer-Based Macromolecular Drug Design using 1,3-Dipolar Cycloaddition for Applications in Biology. Molecules 2020; 25:E857. [PMID: 32075239 PMCID: PMC7071137 DOI: 10.3390/molecules25040857] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 12/20/2022] Open
Abstract
An architectural polymer containing hydrophobic isoxazole-based dendron and hydrophilic polyethylene glycol linear tail is prepared by a combination of the robust ZnCl2 catalyzed alkyne-nitrile oxide 1,3-dipolar cycloaddition and esterification chemistry. This water soluble amphiphilic telodendrimer acts as a macromolecular biologically active agent and shows concentration dependent reduction of glioblastoma (U251) cell survival.
Collapse
Affiliation(s)
- Hossein Yazdani
- Department of Chemistry, McGill University, 801 Sherbrooke St. West, Montréal, QC H3A 0B8, Canada;
- Department of Chemistry, Shahid Beheshti University G.C., Tehran 1983963113, Iran;
| | - Esha Kaul
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montréal, QC H3G 1Y6, Canada;
| | - Ayoob Bazgir
- Department of Chemistry, Shahid Beheshti University G.C., Tehran 1983963113, Iran;
| | - Dusica Maysinger
- Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Montréal, QC H3G 1Y6, Canada;
| | - Ashok Kakkar
- Department of Chemistry, McGill University, 801 Sherbrooke St. West, Montréal, QC H3A 0B8, Canada;
| |
Collapse
|
40
|
Huang X, Lin H, Huang F, Xie Y, Wong KH, Chen X, Wu D, Lu A, Yang Z. Targeting Approaches of Nanomedicines in Acute Myeloid Leukemia. Dose Response 2019; 17:1559325819887048. [PMID: 31853234 PMCID: PMC6906351 DOI: 10.1177/1559325819887048] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 09/10/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy, which is commonly
associated with high incidence and mortality among adult patients. The standard
induction regimen for AML has been substantially unchanged over the past 40
years, for which novel nanomedicines have represented a promising strategy in
AML therapies. Despite developments of multiple nanoparticles formulated with
drugs or genes, less there is not much information available about approaches in
AML is available. This review presents an overview of nanomedicines currently
being evaluated in AML. First, it briefly summarized conventional chemotherapies
in use. Second, nanomedicines presently ongoing in clinical trials or
preclinical researches were classified and described, with illustrative examples
from recent literatures. Finally, limitations and potential safety issues
concerns in clinical translation of AML treatment were discussed as well.
Collapse
Affiliation(s)
- Xiao Huang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Hai Lin
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Feng Huang
- Institute of Acupuncture & Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuning Xie
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Ka Hong Wong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Xiaoyu Chen
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Dongyue Wu
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Aiping Lu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Zhijun Yang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
41
|
de Oliveira Silva J, Fernandes RS, Ramos Oda CM, Ferreira TH, Machado Botelho AF, Martins Melo M, de Miranda MC, Assis Gomes D, Dantas Cassali G, Townsend DM, Rubello D, Oliveira MC, de Barros ALB. Folate-coated, long-circulating and pH-sensitive liposomes enhance doxorubicin antitumor effect in a breast cancer animal model. Biomed Pharmacother 2019; 118:109323. [PMID: 31400669 PMCID: PMC7104811 DOI: 10.1016/j.biopha.2019.109323] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/23/2019] [Accepted: 07/26/2019] [Indexed: 11/08/2022] Open
Abstract
Long circulating pH-sensitive liposomes have been shown to effectively deliver doxorubicin (DOX) to tumors and reduce its toxic effects. Folic acid receptors are upregulated in a wide variety of solid, epithelial tumors, including breast cancer. In order to improve liposomal endocytosis and antitumor activity, folic acid has been added to nanoparticles surfaces to exploit overexpression of folate receptors in tumor cells. The purpose of this study was to evaluate the antitumor activity in vitro and in vivo of long circulating pH-sensitive folate-coated DOX-loaded liposomes (SpHL-DOX-Fol) in a 4T1 breast cancer model system in vitro and in vivo. Biodistribution studies were performed and in vivo electrocardiographic parameters were evaluated. A higher tumor uptake for radiolabeled SpHL-Fol (99mTc-SpHL-Fol) 4 h after intravenous administration was observed in comparision with non-folate-coated liposomes (99mTc-SpHL). Antitumor activity showed that SpHL-DOX-Fol treatment led to a 68% growth arrest and drastically reduce pulmonary metastasis foci. Additionally, eletrocardiographic parameters analysis revealed no dispersion in the QT and QTc interval was observed in liposomal treated mice. In summary, this novel multifunctional nanoplatform deomonstrated higher tumor uptake and antitumor activity. SpHL-DOX-Fol represents a drug delivery platform to improve DOX tumor delivery and reduce dose-limiting toxicity.
Collapse
Affiliation(s)
- Juliana de Oliveira Silva
- Department Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Renata Salgado Fernandes
- Department Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Caroline Mari Ramos Oda
- Department Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Tiago Hilário Ferreira
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ana Flávia Machado Botelho
- Department of Veterinary Medicine, School of Veterinary and Zootechny, Universidade Federal de Goiás, Goiânia, Goiás, Brazil
| | - Marília Martins Melo
- Department of Veterinary Clinical and Surgery, School of Veterinary, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marcelo Coutinho de Miranda
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Dawidson Assis Gomes
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Geovanni Dantas Cassali
- Department of General Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Danyelle M Townsend
- Department of Drug Discovery and Pharmaceutical Sciences, Medical University of South Carolina, USA
| | - Domenico Rubello
- Department of Radiology, Molecular Imaging, Interventional Radiology, NeuroRadiology, Medical Physics, Pathology, Biomarkers Unit, Clinical Laboratory, Microbiology Unit, Rovigo & Adria Hospital, Rovigo, Italy
| | - Mônica Cristina Oliveira
- Department Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - André Luís Branco de Barros
- Department of Clinical and Toxicological Analyses, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
42
|
Hassan T, Jinho P, Hytham H. G, Masters AR, Abdel-Aleem JA, Abdelrahman SI, Abdelrahman AA, Lyle LT, Yeo Y. Development of Liposomal Gemcitabine with High Drug Loading Capacity. Mol Pharm 2019; 16:2858-2871. [PMID: 31136710 PMCID: PMC6662591 DOI: 10.1021/acs.molpharmaceut.8b01284] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Liposomes are widely used for systemic delivery of chemotherapeutic agents to reduce their nonspecific side effects. Gemcitabine (Gem) makes a great candidate for liposomal encapsulation due to the short half-life and nonspecific side effects; however, it has been difficult to achieve liposomal Gem with high drug loading capacity. Remote loading, which uses a transmembrane pH gradient to induce an influx of drug and locks the drug in the core as a sulfate complex, does not serve Gem as efficiently as doxorubicin (Dox) due to the low p Ka value of Gem. Existing studies have attempted to improve Gem loading capacity in liposomes by employing lipophilic Gem derivatives or creating a high-concentration gradient for active loading into the hydrophilic cores (small volume loading). In this study, we combine the remote loading approach and small volume loading or hypertonic loading, a new approach to induce the influx of Gem into the preformed liposomes by high osmotic pressure, to achieve a Gem loading capacity of 9.4-10.3 wt % in contrast to 0.14-3.8 wt % of the conventional methods. Liposomal Gem showed a good stability during storage, sustained-release over 120 h in vitro, enhanced cellular uptake, and improved cytotoxicity as compared to free Gem. Liposomal Gem showed a synergistic effect with liposomal Dox on Huh7 hepatocellular carcinoma cells. A mixture of liposomal Gem and liposomal Dox delivered both drugs to the tumor more efficiently than a free drug mixture and showed a relatively good anti-tumor effect in a xenograft model of hepatocellular carcinoma. This study shows that bioactive liposomal Gem with high drug loading capacity can be produced by remote loading combined with additional approaches to increase drug influx into the liposomes.
Collapse
Affiliation(s)
- Tamam Hassan
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Park Jinho
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
| | - Gadalla Hytham H.
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Andrea R. Masters
- Clinical Pharmacology Analytical Core, Indiana University Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jelan A. Abdel-Aleem
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Sayed I. Abdelrahman
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - Aly A. Abdelrahman
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut 71526, Egypt
| | - L. Tiffany Lyle
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA
| | - Yoon Yeo
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, IN 47907, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
43
|
Alberto M, Brandl A, Garg PK, Gül-Klein S, Dahlmann M, Stein U, Rau B. Pressurized intraperitoneal aerosol chemotherapy and its effect on gastric-cancer-derived peritoneal metastases: an overview. Clin Exp Metastasis 2019; 36:1-14. [PMID: 30715654 DOI: 10.1007/s10585-019-09955-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 01/17/2019] [Indexed: 12/19/2022]
Abstract
This manuscript aspires to portray a review of the current literature focusing on manifest peritoneal metastasis (PM) derived from gastric cancer and its treatment options. Despite the development of chemotherapy and multimodal treatment options during the last decades, mortality remains high worldwide. After refreshing important epidemiological considerations, the molecular mechanisms currently accepted through which PM occurs are revised. Palliative chemotherapy is the only recommended treatment option for patients with PM of gastric cancer according to the National Comprehensive Cancer Network guidelines, although cytoreductive surgery in combination with hyperthermic intraperitoneal chemotherapy demonstrated promising results in selected patients with regional PM and localized intraabdominal tumor spread. A novel treatment named pressurized intraperitoneal aerosol chemotherapy may have a promising future in improving overall survival with an acceptable postoperative complication rate and stabilizing quality of life during treatment. Additionally, the procedure has been proved to be safe for the patient and medical personnel and a feasible, repeatable method to deter metastatic proliferation. This overview comprehensively addresses this novel and promising treatment in the context of a scientifically and clinically challenging disease.
Collapse
Affiliation(s)
- Miguel Alberto
- Department of Surgery, Campus Virchow Klinikum - Campus Mitte, Charité - University Hospital Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Andreas Brandl
- Department of Surgery, Campus Virchow Klinikum - Campus Mitte, Charité - University Hospital Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Pankaj Kumar Garg
- Department of Surgery, Guru Teg Bahadur Hospital, University College of Medical Sciences, University of Delhi, Delhi, India
| | - Safak Gül-Klein
- Department of Surgery, Campus Virchow Klinikum - Campus Mitte, Charité - University Hospital Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Mathias Dahlmann
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité University Hospital Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
- German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Ulrike Stein
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité University Hospital Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Robert-Rössle-Straße 10, 13125, Berlin, Germany
- German Cancer Consortium (DKTK), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Beate Rau
- Department of Surgery, Campus Virchow Klinikum - Campus Mitte, Charité - University Hospital Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
44
|
Yoon CS, Kim HK, Mishchenko NP, Vasileva EA, Fedoreyev SA, Stonik VA, Han J. Spinochrome D Attenuates Doxorubicin-Induced Cardiomyocyte Death via Improving Glutathione Metabolism and Attenuating Oxidative Stress. Mar Drugs 2018; 17:md17010002. [PMID: 30577438 PMCID: PMC6356724 DOI: 10.3390/md17010002] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/13/2018] [Accepted: 12/15/2018] [Indexed: 12/15/2022] Open
Abstract
Doxorubicin, an anthracycline from Streptomyces peucetius, exhibits antitumor activity against various cancers. However, doxorubicin is cardiotoxic at cumulative doses, causing increases in intracellular reactive oxygen species in the heart. Spinochrome D (SpD) has a structure of 2,3,5,6,8-pentahydroxy-1,4-naphthoquinone and is a structural analogue of well-known sea urchin pigment echinochrome A. We previously reported that echinochrome A is cardioprotective against doxorubicin toxicity. In the present study, we assessed the cardioprotective effects of SpD against doxorubicin and determined the underlying mechanism. ¹H-NMR-based metabolomics and mass spectrometry-based proteomics were utilized to characterize the metabolites and proteins induced by SpD in a human cardiomyocyte cell line (AC16) and human breast cancer cell line (MCF-7). Multivariate analyses identified 12 discriminating metabolites (variable importance in projection > 1.0) and 1814 proteins from SpD-treated AC16 cells. Proteomics and metabolomics analyses showed that glutathione metabolism was significantly influenced by SpD treatment in AC16 cells. SpD treatment increased ATP production and the oxygen consumption rate in D-galactose-treated AC16 cells. SpD protected AC16 cells from doxorubicin cytotoxicity, but it did not affect the anticancer properties. With SpD treatment, the mitochondrial membrane potential and mitochondrial calcium localization were significantly different between cardiomyocytes and cancer cell lines. Our findings suggest that SpD could be cardioprotective against the cytotoxicity of doxorubicin.
Collapse
Affiliation(s)
- Chang Shin Yoon
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center (CMDC), Inje University, Busan 614-735, Korea.
| | - Hyoung Kyu Kim
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center (CMDC), Inje University, Busan 614-735, Korea.
| | - Natalia P Mishchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Vladivostok 690022, Russia.
| | - Elena A Vasileva
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Vladivostok 690022, Russia.
| | - Sergey A Fedoreyev
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Vladivostok 690022, Russia.
| | - Valentin A Stonik
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Vladivostok 690022, Russia.
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center (CMDC), Inje University, Busan 614-735, Korea.
| |
Collapse
|