1
|
Zhu C, Liu Y, Ji X, Si Y, Tao X, Zhang X, Yin L. Enhanced Antitumor Efficacy of Cytarabine and Idarubicin in Acute Myeloid Leukemia Using Liposomal Formulation: In Vitro and In Vivo Studies. Pharmaceutics 2024; 16:1220. [PMID: 39339256 PMCID: PMC11434936 DOI: 10.3390/pharmaceutics16091220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/05/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Background: Acute myeloid leukemia (AML) is the most common type of acute leukemia among adults with the recommend therapy of combination of cytarabine and idarubicin in the induction phase. The uncoordinated pharmacokinetics prevent adequate control of drug ratio following systemic administration. Therefore, the dual-loaded liposomes containing cytarabine and idarubicin for synergistic effects were proposed and investigated. Methods: The molar ratio of cytarabine and idarubicin for synergistic effects was investigated. The dual-loaded liposomes were prepared and characterized by particle size, zeta potential, encapsulation efficiency, cryo-Transmission electron microscopy (cryo-TEM), and in vitro stability. The in vitro cytotoxicity and cell uptake of liposomes were determined within CCRF-CEM cells. The PK experiments was carried out in male SD rats. The in vivo antitumor effect was carried out within CD-1 nude female mice. The antitumor mechanism of liposomes was investigated. Results: The synergistic molar ratios were found to be in the range of 20:1~40:1. The size distribution of the dual-loaded liposomes was approximately 100 nm with PDI ≤ 0.1, a zeta potential of approximately -30 mV, an entrapment efficiency of cytarabine and idarubicin of >95% with spherical structure and uniform distribution, and in vitro stability for 21 d. The drugs in the liposomes can be quickly uptaken by the leukemia cells. The PK experiments showed that the molar ratio of cytarabine to idarubicin in plasma was maintained at 30:1 within 4 h. The efficacy of liposomes was significantly enhanced. Conclusions: The dual-loaded liposomes containing cytarabine and idarubicin showed enhanced antitumor efficacy.
Collapse
Affiliation(s)
- Chunxia Zhu
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmaceutics, Nanjing Chia-Tai Tianqing Pharmaceutical Co., Ltd., Nanjing 210046, China
| | - Yang Liu
- Department of Pharmaceutics, Nanjing Chia-Tai Tianqing Pharmaceutical Co., Ltd., Nanjing 210046, China
| | - Xiaojun Ji
- Department of Pharmaceutics, Nanjing Chia-Tai Tianqing Pharmaceutical Co., Ltd., Nanjing 210046, China
| | - Yaxuan Si
- Department of Pharmaceutics, Nanjing Chia-Tai Tianqing Pharmaceutical Co., Ltd., Nanjing 210046, China
| | - Xianhao Tao
- Department of Pharmaceutics, Nanjing Chia-Tai Tianqing Pharmaceutical Co., Ltd., Nanjing 210046, China
| | - Xiaohua Zhang
- Department of Pharmaceutics, Nanjing Chia-Tai Tianqing Pharmaceutical Co., Ltd., Nanjing 210046, China
| | - Lifang Yin
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Nanjing 210009, China
- State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing 210009, China
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
2
|
Yang S, Aggarwal K, Jurczyszak J, Brown N, Sridhar S. Nanomedicine Therapies for Pediatric Diseases. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1996. [PMID: 39420230 PMCID: PMC11493394 DOI: 10.1002/wnan.1996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 07/18/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024]
Abstract
In 2020, the top 10 causes of death among children and adolescents between the ages of 1 and 19 years old included cancer, congenital anomalies, heart disease, and chronic respiratory disease; all these conditions are potentially treatable with medical intervention. However, children exhibit specific physiological and developmental characteristics that can significantly impact drug pharmacokinetics, pharmacodynamics, and safety profile. These factors illustrate the importance of a heightened focus on pediatric drug development. Traditional drugs lack proper circulation, permeability, targeting, accumulation, and release, and they often require dose adjustments or modifications, which can result in suboptimal therapeutic outcomes and increased risks of adverse effects in pediatric patients. Nanomedicines have emerged as efficient drug delivery systems because of their unique properties, which can improve the solubility and stability of drugs by encapsulating them in different forms of nanoparticles. This review discusses the challenges of pediatric therapy, and the current state of nanomedicines for pediatric diseases in terms of Food and Drug Administration-approved nanomedicines, the types of diseases treated or diagnosed, and preclinical studies that have the potential to be translated to the clinic. In summary, nanomedicine holds significant potential for addressing the unique and pressing challenges associated with diagnosing and treating pediatric diseases.
Collapse
Affiliation(s)
- Shicheng Yang
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
| | - Kushi Aggarwal
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, Massachusetts, USA
| | - Jillian Jurczyszak
- Cancer Nanomedicine Co-Ops for Undergraduate Research Experience (CaNCURE), Northeastern University, Boston, Massachusetts, USA
| | - Needa Brown
- Department of Physics, Northeastern University, Boston, Massachusetts, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Srinivas Sridhar
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts, USA
- Department of Physics, Northeastern University, Boston, Massachusetts, USA
- Department of Radiation Oncology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Konstantinidis I, Tsokkou S, Grigoriadis S, Chrysavgi L, Gavriilaki E. Cardiotoxicity in Acute Myeloid Leukemia in Adults: A Scoping Study. Cancers (Basel) 2024; 16:2474. [PMID: 39001536 PMCID: PMC11240574 DOI: 10.3390/cancers16132474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 06/29/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Introduction: According to the National Cancer Institute of the NIH, acute myeloid leukemia (AML) is a rapidly growing cancer with a large quantity of myeloblasts. AML is most often observed in adults over the age of 35, accounting for 1% of all cancer types. In 2023, the number of new cases being reported was estimated to reach around 20,380 in total and the rate of mortality in the same year was 1.9%, or 11,310 cases, in the US. Purpose: This scoping study aims to extensively assess and explore the degree of cardiotoxicity in patients with AML that can be caused due to pharmaceutical treatments prescribed by hematologists. This is achieved by performing extensive searches of different scientific databases including PubMed, Scopus, and ScienceDirect. Results: A variety of options are available that are summarized in tables included herein, with each having their advantages and risks of adverse effects, among these being cardiotoxicity. Important medications found to play a significant role include gemtuzumab ozogamicin, venetoclax, and vyxeos. Conclusions: It is understandable that being familiar with all the treatment options available and every potential adverse effect is impossible. However, hematologists and, in general, physicians must try to be updated with the most recent information released to improve the quality of life of their patients and minimize the risk of additional complications.
Collapse
Affiliation(s)
- Ioannis Konstantinidis
- 2nd Propaedeutic Department of Internal Medicine, Ippokratio University Hospital, Department of Medicine, Faculty of Health and Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Research Team "Histologistas", Interinstitutional Postgraduate Program "Health and Environmental Factors", Department of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Sophia Tsokkou
- 2nd Propaedeutic Department of Internal Medicine, Ippokratio University Hospital, Department of Medicine, Faculty of Health and Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Research Team "Histologistas", Interinstitutional Postgraduate Program "Health and Environmental Factors", Department of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | | | - Lalayianni Chrysavgi
- Hematology Department, BMT Unit, G Papanicolaou Hospital, 57010 Thessaloniki, Greece
| | - Eleni Gavriilaki
- 2nd Propaedeutic Department of Internal Medicine, Ippokratio University Hospital, Department of Medicine, Faculty of Health and Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
4
|
Kalra J, Baker J, Sun X, Kyle A, Minchinton A, Bally MB. Accumulation of liposomes in metastatic tumor sites is not necessary for anti-cancer drug efficacy. J Transl Med 2024; 22:621. [PMID: 38961395 PMCID: PMC11223361 DOI: 10.1186/s12967-024-05428-9] [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: 02/27/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND The tumor microenvironment is profoundly heterogeneous particularly when comparing sites of metastases. Establishing the extent of this heterogeneity may provide guidance on how best to design lipid-based drug delivery systems to treat metastatic disease. Building on our previous research, the current study employs a murine model of metastatic cancer to explore the distribution of ~ 100 nm liposomes. METHODS Female NCr nude mice were inoculated with a fluorescently labeled, Her2/neu-positive, trastuzumab-resistant breast cancer cell line, JIMT-1mkate, either in the mammary fat pad to create an orthotopic tumor (OT), or via intracardiac injection (IC) to establish tumors throughout the body. Animals were dosed with fluorescent and radio-labeled liposomes. In vivo and ex vivo fluorescent imaging was used to track liposome distribution over a period of 48 h. Liposome distribution in orthotopic tumors was compared to sites of tumor growth that arose following IC injection. RESULTS A significant amount of inter-vessel heterogeneity for DiR distribution was observed, with most tumor blood vessels showing little to no presence of the DiR-labelled liposomes. Further, there was limited extravascular distribution of DiR liposomes in the perivascular regions around DiR-positive vessels. While all OT tumors contained at least some DiR-positive vessels, many metastases had very little or none. Despite the apparent limited distribution of liposomes within metastases, two liposomal drug formulations, Irinophore C and Doxil, showed similar efficacy for both the OT and IC JIMT-1mkate models. CONCLUSION These findings suggest that liposomal formulations achieve therapeutic benefits through mechanisms that extend beyond the enhanced permeability and retention effect.
Collapse
Affiliation(s)
- Jessica Kalra
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC, Canada.
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada.
| | - Jennifer Baker
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - XuXin Sun
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Alastair Kyle
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Andrew Minchinton
- Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Marcel B Bally
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
- NanoMedicine Innovation Network, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
5
|
Sadat SMA, Vakili MR, Abd-El Hafeez SI, Paladino M, Hall DG, Weinfeld M, Lavasanifar A. Synergistic Nanomedicine Delivering Topoisomerase I Toxin (SN-38) and Inhibitors of Polynucleotide Kinase 3'-Phosphatase (PNKP) for Enhanced Treatment of Colorectal Cancer. Mol Pharm 2024; 21:3240-3255. [PMID: 38785196 DOI: 10.1021/acs.molpharmaceut.4c00007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Inhibitors of a DNA repair enzyme known as polynucleotide kinase 3'-phosphatase (PNKP) are expected to show synergistic cytotoxicity in combination with topoisomerase I (TOP1) inhibitors in cancer. In this study, the synergistic cytotoxicity of a novel inhibitor of PNKP, i.e., A83B4C63, with a potent TOP1 inhibitor, i.e., SN-38, against colorectal cancer cells was investigated. Polymeric micelles (PMs) for preferred tumor delivery of A83B4C63, developed through physical encapsulation of this compound in methoxy poly(ethylene oxide)-poly(α-benzyl carboxylate-ε-caprolactone) (mPEO-b-PBCL) micelles, were combined with SN-38 in free or PM form. The PM form of SN-38 was prepared through chemical conjugation of SN-38 to the functional end group of mPEO-b-PBCL and further assembly of mPEO-b-PBCL-SN-38 in water. Moreover, mixed micelles composed of mPEO-b-PBCL and mPEO-b-PBCL-SN-38 were used to co-load A83B4C63 and SN-38 in the same nanoformulation. The loading content (% w/w) of the SN-38 and A83B4C63 to mPEO-b-PBCL in the co-loaded formulation was 7.91 ± 0.66 and 16.13 ± 0.11% (w/w), respectively, compared to 15.67 ± 0.34 (% w/w) and 23.06 ± 0.63 (% w/w) for mPEO-b-PBCL micelles loading individual drugs. Notably, the average diameter of PMs co-encapsulating both SN-38 and A83B4C63 was larger than that of PMs encapsulating either of these compounds alone but still lower than 60 nm. The release of A83B4C63 from PMs co-encapsulating both drugs was 76.36 ± 1.41% within 24 h, which was significantly higher than that of A83B4C63-encapsulated micelles (42.70 ± 0.72%). In contrast, the release of SN-38 from PMs co-encapsulating both drugs was 44.15 ± 2.61% at 24 h, which was significantly lower than that of SN-38-conjugated PMs (74.16 ± 3.65%). Cytotoxicity evaluations by the MTS assay as analyzed by the Combenefit software suggested a clear synergy between PM/A83B4C63 (at a concentration range of 10-40 μM) and free SN-38 (at a concentration range of 0.001-1 μM). The synergistic cytotoxic concentration range for SN-38 was narrowed down to 0.1-1 or 0.01-1 μM when combined with PM/A83B4C63 at 10 or 20-40 μM, respectively. In general, PMs co-encapsulating A83B4C63 and SN-38 at drug concentrations within the synergistic range (10 μM for A83B4C63 and 0.05-1 μM for SN-38) showed slightly less enhancement of SN-38 anticancer activity than a combination of individual micelles, i.e., A83B4C63 PMs + SN-38 PMs at the same molar concentrations. This was attributed to the slower release of SN-38 from the SN-38 and A83B4C63 co-encapsulated PMs compared to PMs only encapsulating SN-38. Cotreatment of cells with TOP1 inhibitors and A83B4C63 formulation enhanced the expression level of γ-HA2X, cleaved PARP, caspase-3, and caspase-7 in most cases. This trend was more consistent and notable for PMs co-encapsulating both A83B4C63 and SN-38. The overall result from the study shows a synergy between PMs of SN-38 and A83B4C63 as a mixture of two PMs for individual drugs or PMs co-encapsulating both drugs.
Collapse
Affiliation(s)
- Sams M A Sadat
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Mohammad Reza Vakili
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Sara I Abd-El Hafeez
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
- Department of Pharmaceutics, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
| | - Marco Paladino
- Department of Chemistry, Faculty of Science, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Dennis G Hall
- Department of Chemistry, Faculty of Science, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Michael Weinfeld
- Department of Oncology, Cross Cancer Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
- Department of Chemical and Material Engineering, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| |
Collapse
|
6
|
Yuan R, Xu ZJ, Zhang SK, Cao XY, Dai AG, Song L. New evidence for a role of DANCR in cancers: a comprehensive review. J Transl Med 2024; 22:569. [PMID: 38877534 PMCID: PMC11177382 DOI: 10.1186/s12967-024-05246-z] [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: 03/22/2024] [Accepted: 04/28/2024] [Indexed: 06/16/2024] Open
Abstract
Cancer remains a leading cause of mortality and poses a substantial threat to public health. Studies have revealed that Long noncoding RNA DANCR is a cytoplasmic lncRNA whose aberrant expression plays a pivotal role in various cancer types. Within tumour biology, DANCR exerts regulatory control over crucial processes such as proliferation, invasion, metastasis, angiogenesis, inflammatory responses, cellular energy metabolism reprogramming, and apoptosis. By acting as a competitive endogenous RNA for miRNAs and by interacting with proteins and mRNAs at the molecular level, DANCR contributes significantly to cancer progression. Elevated DANCR levels have also been linked to heightened resistance to anticancer drugs. Moreover, the detection of circulating DANCR holds promise as a valuable biomarker for aiding in the clinical differentiation of different cancer types. This article offers a comprehensive review and elucidation of the primary functions and molecular mechanisms through which DANCR influences tumours.
Collapse
Affiliation(s)
- Rong Yuan
- School of Medicine, Hunan University of Chinese Medicine, 300 Xueshi Road, Hanpu Science and Teaching Park, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, 300 Xueshi Road, Hanpu Science and Teaching Park, Changsha, 410208, Hunan, China
| | - Zhao-Jun Xu
- Department of Cardiothoracic Surgery, the First Affiliated Hospital, Hunan University of Chinese Medicine, 97 Shaoshan Road, Changsha, 410007, Hunan, China
| | - Sheng-Kang Zhang
- Department of Cardiothoracic Surgery, the First Affiliated Hospital, Hunan University of Chinese Medicine, 97 Shaoshan Road, Changsha, 410007, Hunan, China
| | - Xian-Ya Cao
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, 300 Xueshi Road, Hanpu Science and Teaching Park, Changsha, 410208, Hunan, China
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China
| | - Ai-Guo Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, 300 Xueshi Road, Hanpu Science and Teaching Park, Changsha, 410208, Hunan, China.
- Department of Respiratory Diseases, School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410021, Hunan, China.
| | - Lan Song
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, 300 Xueshi Road, Hanpu Science and Teaching Park, Changsha, 410208, Hunan, China.
- Department of Biochemistry and Molecular Biology, School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
7
|
Pinheiro M, Mu Q. Editorial: Editor's challenge: Dr. Qingxin Mu - how can nanomedicine approaches advance multi-targeting strategy in combination cancer therapy? Front Oncol 2024; 14:1437497. [PMID: 38919540 PMCID: PMC11196825 DOI: 10.3389/fonc.2024.1437497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 06/04/2024] [Indexed: 06/27/2024] Open
Affiliation(s)
- Marina Pinheiro
- Department of Chemistry, Faculdade de Farmácia, Universidade do Porto, Porto, Portugal
- Public Health Unit, Local Health Unit Barcelos/Esposende, Barcelos, Portugal
| | - Qingxin Mu
- Department of Pharmaceutics, University of Washington, Seattle, WA, United States
| |
Collapse
|
8
|
Lammers T. Nanomedicine Tumor Targeting. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312169. [PMID: 38361435 DOI: 10.1002/adma.202312169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/24/2024] [Indexed: 02/17/2024]
Abstract
Nanomedicines are extensively explored for cancer therapy. By delivering drug molecules more efficiently to pathological sites and by attenuating their accumulation in healthy organs and tissues, nanomedicine formulations aim to improve the balance between drug efficacy and toxicity. More than 20 cancer nanomedicines are approved for clinical use, and hundreds of formulations are in (pre)clinical development. Over the years, several key pitfalls have been identified as bottlenecks in nanomedicine tumor targeting and translation. These go beyond materials- and production-related issues, and particularly also encompass biological barriers and pathophysiological heterogeneity. In this manuscript, the author describes the most important principles, progress, and products in nanomedicine tumor targeting, delineates key current problems and challenges, and discusses the most promising future prospects to create clinical impact.
Collapse
Affiliation(s)
- Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Center for Biohyhrid Medical Systems, University Hospital RWTH Aachen, Forckenbeckstrasse 55, 52074, Aachen, Germany
| |
Collapse
|
9
|
Forsberg M, Konopleva M. AML treatment: conventional chemotherapy and emerging novel agents. Trends Pharmacol Sci 2024; 45:430-448. [PMID: 38643058 DOI: 10.1016/j.tips.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/25/2024] [Accepted: 03/25/2024] [Indexed: 04/22/2024]
Abstract
Acute myeloid leukemia (AML) is driven by complex mutations and cytogenetic abnormalities with profound tumoral heterogeneity, making it challenging to treat. Ten years ago, the 5-year survival rate of patients with AML was only 29% with conventional chemotherapy and stem cell transplantation. All attempts to improve conventional therapy over the previous 40 years had failed. Now, new genomic, immunological, and molecular insights have led to a renaissance in AML therapy. Improvements to standard chemotherapy and a wave of new targeted therapies have been developed. However, how best to incorporate these advances into frontline therapy and sequence them in relapse is not firmly established. In this review, we highlight current treatments of AML, targeted agents, and pioneering attempts to synthesize these developments into a rational standard of care (SoC).
Collapse
Affiliation(s)
- Mark Forsberg
- Montefiore Einstein Cancer Center, Department of Oncology, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Marina Konopleva
- Montefiore Einstein Cancer Center, Department of Oncology, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| |
Collapse
|
10
|
Li K, Chen W, Ma L, Yan L, Wang B. Approaches for reducing chemo/radiation-induced cardiotoxicity by nanoparticles. ENVIRONMENTAL RESEARCH 2024; 244:117264. [PMID: 37776941 DOI: 10.1016/j.envres.2023.117264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
Nanoparticles are fascinating and encouraging carriers for cancer treatment due to their extraordinary properties and potential applications in targeted drug delivery, treatment, and diagnosis. Experimental studies including in vitro and in vivo examinations show that nanoparticles can cause a revolution in different aspects of cancer therapy. Normal tissue toxicity and early and late consequences are the major limitations of cancer therapy by radiotherapy and chemotherapy. However, the delivery of drugs into tumors or reducing the accumulation of drugs in normal tissues can permit a more satisfactory response of malignancies to therapy with more inferior side effects. Cardiac toxicity is one of the major problems for chemotherapy and radiotherapy. Therefore, several experimental studies have been performed to minimize the degenerative impacts of cancer treatment on the heart and also enhance the influences of radiotherapy and chemotherapy agents in cancers. This review article emphasizes the benefits of nanoparticle-based drug delivery techniques, including minimizing the exposure of the heart to anticancer drugs, enhancing the accumulation of drugs in cancers, and expanding the effectiveness of radiotherapy. The article also discusses the challenges and problems accompanied with nanoparticle-based drug delivery techniques such as toxicity, which need to be addressed through further research. Moreover, the article emphasizes the importance of developing safe and effective nanoparticle-based therapies that can be translated into clinical practice.
Collapse
Affiliation(s)
- Ketao Li
- Department of Cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang, 310022, China
| | - Wan Chen
- Department of Cardiology, Jiulongpo First People's Hospital, Chongqing, 400051, China
| | - Liping Ma
- Department of Cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang, 310022, China
| | - Laixing Yan
- Department of Cardiology, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, Zhejiang, 310022, China
| | - Bing Wang
- Department of Cardiology, Zouping People's Hospital, Zouping, shandong, 256299, China.
| |
Collapse
|
11
|
Fernandes S, Cassani M, Cavalieri F, Forte G, Caruso F. Emerging Strategies for Immunotherapy of Solid Tumors Using Lipid-Based Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305769. [PMID: 38054651 PMCID: PMC10885677 DOI: 10.1002/advs.202305769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/09/2023] [Indexed: 12/07/2023]
Abstract
The application of lipid-based nanoparticles for COVID-19 vaccines and transthyretin-mediated amyloidosis treatment have highlighted their potential for translation to cancer therapy. However, their use in delivering drugs to solid tumors is limited by ineffective targeting, heterogeneous organ distribution, systemic inflammatory responses, and insufficient drug accumulation at the tumor. Instead, the use of lipid-based nanoparticles to remotely activate immune system responses is an emerging effective strategy. Despite this approach showing potential for treating hematological cancers, its application to treat solid tumors is hampered by the selection of eligible targets, tumor heterogeneity, and ineffective penetration of activated T cells within the tumor. Notwithstanding, the use of lipid-based nanoparticles for immunotherapy is projected to revolutionize cancer therapy, with the ultimate goal of rendering cancer a chronic disease. However, the translational success is likely to depend on the use of predictive tumor models in preclinical studies, simulating the complexity of the tumor microenvironment (e.g., the fibrotic extracellular matrix that impairs therapeutic outcomes) and stimulating tumor progression. This review compiles recent advances in the field of antitumor lipid-based nanoparticles and highlights emerging therapeutic approaches (e.g., mechanotherapy) to modulate tumor stiffness and improve T cell infiltration, and the use of organoids to better guide therapeutic outcomes.
Collapse
Affiliation(s)
- Soraia Fernandes
- Center for Translational Medicine (CTM)International Clinical Research Centre (ICRC)St. Anne HospitalBrno656 91Czech Republic
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Marco Cassani
- Center for Translational Medicine (CTM)International Clinical Research Centre (ICRC)St. Anne HospitalBrno656 91Czech Republic
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| | - Francesca Cavalieri
- School of ScienceRMIT UniversityMelbourneVictoria3000Australia
- Dipartimento di Scienze e Tecnologie ChimicheUniversita di Roma “Tor Vergata”Via della Ricerca Scientifica 1Rome00133Italy
| | - Giancarlo Forte
- Center for Translational Medicine (CTM)International Clinical Research Centre (ICRC)St. Anne HospitalBrno656 91Czech Republic
- School of Cardiovascular and Metabolic Medicine & SciencesKing's College LondonLondonSE5 9NUUK
| | - Frank Caruso
- Department of Chemical EngineeringThe University of MelbourneParkvilleVictoria3010Australia
| |
Collapse
|
12
|
Nguyen TM, Joyce P, Ross DM, Bremmell K, Jambhrunkar M, Wong SS, Prestidge CA. Combating Acute Myeloid Leukemia via Sphingosine Kinase 1 Inhibitor-Nanomedicine Combination Therapy with Cytarabine or Venetoclax. Pharmaceutics 2024; 16:209. [PMID: 38399263 PMCID: PMC10893145 DOI: 10.3390/pharmaceutics16020209] [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: 12/25/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
MP-A08 is a novel sphingosine kinase 1 (SPHK1) inhibitor with activity against acute myeloid leukemia (AML). A rationally designed liposome-based encapsulation and delivery system has been shown to overcome the physicochemical challenges of MP-A08 and enable its effective delivery for improved efficacy and survival of mice engrafted with human AML in preclinical models. To establish therapies that overcome AML's heterogeneous nature, here we explored the combination of MP-A08-loaded liposomes with both the standard chemotherapy, cytarabine, and the targeted therapy, venetoclax, against human AML cell lines. Cytarabine (over the dose range of 0.1-0.5 µM) in combination with MP-A08 liposomes showed significant synergistic effects (as confirmed by the Chou-Talalay Combination Index) against the chemosensitised human AML cell lines MV4-11 and OCI-AML3. Venetoclax (over the dose range of 0.5-250 nM) in combination with MP-A08 liposomes showed significant synergistic effects against the chemosensitised human AML cell lines, particularly in venetoclax-resistant human AML cells. This strong synergistic effect is due to multiple mechanisms of action, i.e., inhibiting MCL-1 through SPHK1 inhibition, leading to ceramide accumulation, activation of protein kinase R, ATF4 upregulation, and NOXA activation, ultimately resulting in MCL-1 degradation. These combination therapies warrant further consideration and investigation in the search for a more comprehensive treatment strategy for AML.
Collapse
Affiliation(s)
- Thao M. Nguyen
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (T.M.N.); (P.J.); (K.B.); (M.J.); (S.S.W.)
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5001, Australia;
| | - Paul Joyce
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (T.M.N.); (P.J.); (K.B.); (M.J.); (S.S.W.)
| | - David M. Ross
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5001, Australia;
- Department of Haematology, Flinders University and Medical Centre, Adelaide, SA 5001, Australia
- Department of Haematology and Bone Marrow Transplantation, Royal Adelaide Hospital, Adelaide, SA 5001, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia
| | - Kristen Bremmell
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (T.M.N.); (P.J.); (K.B.); (M.J.); (S.S.W.)
| | - Manasi Jambhrunkar
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (T.M.N.); (P.J.); (K.B.); (M.J.); (S.S.W.)
| | - Sook S. Wong
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (T.M.N.); (P.J.); (K.B.); (M.J.); (S.S.W.)
| | - Clive A. Prestidge
- Centre for Pharmaceutical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; (T.M.N.); (P.J.); (K.B.); (M.J.); (S.S.W.)
| |
Collapse
|
13
|
Becker M, Farina KA, Mascarenhas J. Acute myeloid leukemia: Current understanding and management. JAAPA 2024; 37:34-39. [PMID: 38128137 DOI: 10.1097/01.jaa.0000995680.52352.b5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
ABSTRACT Although relatively rare, acute myeloid leukemia (AML) is the most common type of acute leukemia in adults. AML is associated with poor 5-year overall survival and prompt treatment is critical. Classifying AML based on World Health Organization criteria is important for determining prognosis and applying a risk-adapted treatment approach. Throughout therapy, patients require comprehensive supportive care measures with blood product transfusions, antimicrobial treatment, and frequent monitoring for chemotherapy-related complications. This article provides an overview of AML and its treatments. Clinicians in all specialties must be able to recognize the early signs of AML and ensure their patients seek appropriate expert medical care with a hematologist/oncologist.
Collapse
Affiliation(s)
- Michelle Becker
- Michelle Becker practices in the adult leukemia program at the Icahn School of Medicine at Mount Sinai in New York, N.Y. Kyle A. Farina is a clinical pharmacy manager, working on the leukemia service at Mount Sinai Hospital in New York, N.Y. John Mascarenhas is director of the adult leukemia program, leader of the Myeloproliferative Disorders Clinical Research Program in the Division of Hematology/Oncology at the Tisch Cancer Institute at Mount Sinai Hospital, and a professor of medicine at the Icahn School of Medicine at Mount Sinai. Dr. Farina discloses that he is a consultant and speaker for Bristol Myers Squibb. Dr. Mascarenhas discloses that he is a consultant for Celgene Corp., Bristol Myers Squibb Co., Incyte Inc., F. Hoffmann-La Roche AG, PharmaEssentia Corp., Geron Corp., CTI Biopharma Corp., MorphoSys AG, Abbvie Inc., Kartos Therapeutics, Novartis AG, Sierra Oncology Inc., GSK plc, Karyopharm Therapeutics Inc., Galecto Inc., Imago BioSciences Inc., and Pfizer Inc., and receives research funding from Bristol Myers Squibb, Abbvie, CTI Biopharma, Incyte, Merck & Co., Novartis, Roche, Kartos, PharmaEssentia, and Geron. The authors have disclosed no other potential conflicts of interest, financial or otherwise
| | | | | |
Collapse
|
14
|
Rey G, Daguenet E, Bonjean P, Devillier R, Fegueux N, Forcade E, Srour M, Chevallier P, Robin M, Suarez F, Micol JB, Labussière-Wallet H, Bilger K, Daguindau E, Bay JO, Fayard A, Bulabois CE, Nguyen-Quoc S, Genthon A, Orvain C, Turlure P, Loschi M, Poiré X, Guillerm G, Beguin Y, Maillard N, Mear JB, Chalayer E, Cornillon J, Tavernier E. Allogeneic hematopoietic stem cell transplantation for adults with therapy-related acute myeloid leukaemia: a retrospective multicentre study on behalf of the SFGM-TC. Bone Marrow Transplant 2023; 58:1331-1338. [PMID: 37653054 DOI: 10.1038/s41409-023-02082-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 07/20/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023]
Abstract
We report the results from a multicentre retrospective study of 220 adult patients who underwent allogeneic hematopoietic stem cell transplantation (alloHSCT) for therapy-related acute myeloid leukaemia (t-AML). Median age at t-AML diagnosis was 56 years, with a prior history of haematological (45%) or breast (34%). Median time from cytotoxic exposure to t-AML diagnosis was 54.7 months. At transplant, around 20% of patients had measurable residual disease and 3% of patients were not in complete remission. The median follow-up was 21.4 months (Q1-Q3, 5.9-52.8). At 12 months, overall survival (OS), event-free survival (EFS), and graft-versus-host-disease (GVHD)-free-relapse-free survival (GRFS) were 60.7% (95% CI 54.6-67.5), 52.8% (95% CI 46.5-68.4), and 44.1% (95% CI 37.6-51.8), respectively. At 5 years, OS, EFS, and GRFS were 44.1% (95% CI 37.4-52.1), 40.4% (95% CI 33.9-48.1), and 35.3% (95% CI 28.8-43.3), respectively. At last follow-up, 44% of patients were in complete remission (n = 96) and transplant-related mortality accounted for 21% of all deaths (n = 119). Multivariable analysis revealed that uncontrolled t-AML at transplant was associated with lower EFS (HR 1.94, 95% CI 1.0-3.7, p = 0.041). In conclusion, alloHSCT for t-AML shows encouraging results and offers additional opportunity with the emergence of novel pre-graft therapies.
Collapse
Affiliation(s)
- Gaëlle Rey
- Département d'hématologie clinique, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Priest-en-Jarez, France
| | - Elisabeth Daguenet
- Département Universitaire de Recherche et d'Enseignement, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Priest-en-Jarez, France
| | - Paul Bonjean
- Unité de Recherche Clinique Innovation Pharmacologique, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Priest-en-Jarez, France
| | | | - Nathalie Fegueux
- Hôpital Saint Eloi, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Edouard Forcade
- Centre Hospitalier Universitaire Haut-Lévêque Magellan, Bordeaux, France
| | - Micha Srour
- Hôpital Claude Hurriez, Centre Hospitalier Universitaire de Lille, Lille, France
| | | | - Marie Robin
- Hôpital Saint-Louis, APHP, Université de Paris Cité, Paris, France
| | | | | | | | - Karin Bilger
- Centre Hospitalier Universitaire Hautepierre, Strasbourg, France
| | | | - Jacques-Olivier Bay
- Hôpital Estaing, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Amandine Fayard
- Hôpital Estaing, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | | | | | | | | | - Pascal Turlure
- Centre Hospitalier Universitaire Dupuytren, Limoges, France
| | - Michael Loschi
- Hôpital de l'Archet, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Xavier Poiré
- Cliniques Universitaires St. Luc, Brussels, Belgium
| | - Gaëlle Guillerm
- Centre Hospitalier Universitaire Augustin Morvan, Brest, France
| | - Yves Beguin
- CHU of Liège and University of Liège, Liège, Belgium
| | | | | | - Emilie Chalayer
- Département d'hématologie clinique, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Priest-en-Jarez, France
| | - Jérôme Cornillon
- Département d'hématologie clinique, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Priest-en-Jarez, France
| | - Emmanuelle Tavernier
- Département d'hématologie clinique, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Priest-en-Jarez, France.
| |
Collapse
|
15
|
Gabizon A, Shmeeda H, Draper B, Parente-Pereira A, Maher J, Carrascal-Miniño A, de Rosales RTM, La-Beck NM. Harnessing Nanomedicine to Potentiate the Chemo-Immunotherapeutic Effects of Doxorubicin and Alendronate Co-Encapsulated in Pegylated Liposomes. Pharmaceutics 2023; 15:2606. [PMID: 38004584 PMCID: PMC10675201 DOI: 10.3390/pharmaceutics15112606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Encapsulation of Doxorubicin (Dox), a potent cytotoxic agent and immunogenic cell death inducer, in pegylated (Stealth) liposomes, is well known to have major pharmacologic advantages over treatment with free Dox. Reformulation of alendronate (Ald), a potent amino-bisphosphonate, by encapsulation in pegylated liposomes, results in significant immune modulatory effects through interaction with tumor-associated macrophages and activation of a subset of gamma-delta T lymphocytes. We present here recent findings of our research work with a formulation of Dox and Ald co-encapsulated in pegylated liposomes (PLAD) and discuss its pharmacological properties vis-à-vis free Dox and the current clinical formulation of pegylated liposomal Dox. PLAD is a robust formulation with high and reproducible remote loading of Dox and high stability in plasma. Results of biodistribution studies, imaging with radionuclide-labeled liposomes, and therapeutic studies as a single agent and in combination with immune checkpoint inhibitors or gamma-delta T lymphocytes suggest that PLAD is a unique product with distinct tumor microenvironmental interactions and distinct pharmacologic properties when compared with free Dox and the clinical formulation of pegylated liposomal Dox. These results underscore the potential added value of PLAD for chemo-immunotherapy of cancer and the relevance of the co-encapsulation approach in nanomedicine.
Collapse
Affiliation(s)
- Alberto Gabizon
- Nano-Oncology Research Center, Oncology Institute, Shaare Zedek Medical Center, Jerusalem 9103102, Israel;
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112102, Israel
| | - Hilary Shmeeda
- Nano-Oncology Research Center, Oncology Institute, Shaare Zedek Medical Center, Jerusalem 9103102, Israel;
| | - Benjamin Draper
- King’s College London, School of Cancer and Pharmaceutical Sciences, Guy’s Cancer Centre, Great Maze Pond, London SE1 9RT, UK; (B.D.); (A.P.-P.); (J.M.)
| | - Ana Parente-Pereira
- King’s College London, School of Cancer and Pharmaceutical Sciences, Guy’s Cancer Centre, Great Maze Pond, London SE1 9RT, UK; (B.D.); (A.P.-P.); (J.M.)
| | - John Maher
- King’s College London, School of Cancer and Pharmaceutical Sciences, Guy’s Cancer Centre, Great Maze Pond, London SE1 9RT, UK; (B.D.); (A.P.-P.); (J.M.)
| | - Amaia Carrascal-Miniño
- King’s College London, School of Biomedical Engineering & Imaging Sciences, St. Thomas’ Hospital, London SE1 7EH, UK; (A.C.-M.); (R.T.M.d.R.)
| | - Rafael T. M. de Rosales
- King’s College London, School of Biomedical Engineering & Imaging Sciences, St. Thomas’ Hospital, London SE1 7EH, UK; (A.C.-M.); (R.T.M.d.R.)
| | - Ninh M. La-Beck
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA;
| |
Collapse
|
16
|
Zhao DK, Liang J, Huang XY, Shen S, Wang J. Organoids technology for advancing the clinical translation of cancer nanomedicine. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1892. [PMID: 37088100 DOI: 10.1002/wnan.1892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 04/25/2023]
Abstract
The past decades have witnessed the rapid development and widespread application of nanomedicines in cancer treatment; however, the clinical translation of experimental findings has been low, as evidenced by the low percentage of commercialized nanomedicines. Incomplete understanding of nanomedicine-tumor interactions and inappropriate evaluation models are two important challenges limiting the clinical translation of cancer nanomedicines. Currently, nanomedicine-tumor interaction and therapeutic effects are mainly investigated using cell lines or mouse models, which do not recapitulate the complex tumor microenvironment in human patients. Thus, information obtained from cell lines and mouse models cannot provide adequate guidance for the rational redesign of nanomedicine. Compared with other preclinical models, tumor organoids constructed from patient-derived tumor tissues are superior in retaining the key histopathological, genetic, and phenotypic features of the parent tumor. We speculate that organoid technology would help elucidate nanomedicine-tumor interaction in the tumor microenvironment and guide the design of nanomedicine, making it a reliable tool to accurately predict drug responses in patients with cancer. This review highlighted the advantages of drug delivery systems in cancer treatment, challenges limiting the clinical translation of antitumor nanomedicines, and potential application of patient-derived organoids (PDO) in nanomedicine. We propose that combining organoids and nanotechnology would facilitate the development of safe and effective cancer nanomedicines and accelerate their clinical application. This review discussed the potential translational value of integrative research using organoids and cancer nanomedicine. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Dong-Kun Zhao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
| | - Jie Liang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Xiao-Yi Huang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| | - Song Shen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, China
| |
Collapse
|
17
|
Martins C, Sarmento B. Multi-ligand functionalized blood-to-tumor sequential targeting strategies in the field of glioblastoma nanomedicine. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1893. [PMID: 37186374 DOI: 10.1002/wnan.1893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 05/17/2023]
Abstract
Glioblastoma (GBM) is an unmet clinical need characterized by a standard of care (SOC) 5-year survival rate of only 5%, and a treatment mostly palliative. Significant hurdles in GBM therapies include an effective penetration of therapeutics through the brain protective barrier, namely the blood-brain barrier (BBB), and a successful therapeutic delivery to brain-invading tumor cells post-BBB crossing. These hurdles, along with the poor prognosis and critical heterogeneity of the disease, have shifted attention to treatment modalities with capacity to precisely and sequentially target (i) BBB cells, inducing blood-to-brain transport, and (ii) GBM cells, leading to a higher therapeutic accumulation at the tumor site. This sequential targeting allows therapeutic molecules to reach the brain parenchyma and compromise molecular processes that support tumor cell invasion. Besides improving formulation and pharmacokinetics constraints of drugs, nanomedicines offer the possibility of being surface functionalized with multiple possibilities of targeting ligands, while delivering the desired therapeutic cargos to the biological sites of interest. Targeting ligands exploit the site-specific expression or overexpression of specific molecules on BBB and GBM cells, triggering brain plus tumor transport. Since the efficacy of single-ligand functionalized nanomedicines is limited due to the GBM anatomical site (brain) and disease complexity, this review presents an overview of multi-ligand functionalized, BBB and GBM sequentially- and dual-targeted nanomedicines reported in literature over the last 10 years. The role of the BBB in GBM progression, treatment options, and the multiple possibilities of currently available targeting ligands will be summarized. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Cláudia Martins
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Bruno Sarmento
- I3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- IUCS-CESPU, Gandra, Portugal
| |
Collapse
|
18
|
Kelvin JM, Chimenti ML, Zhang DY, Williams EK, Moore SG, Humber GM, Baxter TA, Birnbaum LA, Qui M, Zecca H, Thapa A, Jain J, Jui NT, Wang X, Fu H, Du Y, Kemp ML, Lam WA, Graham DK, DeRyckere D, Dreaden EC. Development of constitutively synergistic nanoformulations to enhance chemosensitivity in T-cell leukemia. J Control Release 2023; 361:470-482. [PMID: 37543290 PMCID: PMC10544718 DOI: 10.1016/j.jconrel.2023.07.045] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/27/2023] [Accepted: 07/28/2023] [Indexed: 08/07/2023]
Abstract
Advances in multiagent chemotherapy have led to recent improvements in survival for patients with acute lymphoblastic leukemia (ALL); however, a significant fraction do not respond to frontline chemotherapy or later relapse with recurrent disease, after which long-term survival rates remain low. To develop new, effective treatment options for these patients, we conducted a series of high-throughput combination drug screens to identify chemotherapies that synergize in a lineage-specific manner with MRX-2843, a small molecule dual MERTK and FLT3 kinase inhibitor currently in clinical testing for treatment of relapsed/refractory leukemias and solid tumors. Using experimental and computational approaches, we found that MRX-2843 synergized strongly-and in a ratio-dependent manner-with vincristine to inhibit both B-ALL and T-ALL cell line expansion. Based on these findings, we developed multiagent lipid nanoparticle formulations of these drugs that not only delivered defined drug ratios intracellularly in T-ALL, but also improved anti-leukemia activity following drug encapsulation. Synergistic and additive interactions were recapitulated in primary T-ALL patient samples treated with MRX-2843 and vincristine nanoparticle formulations, suggesting their clinical relevance. Moreover, the nanoparticle formulations reduced disease burden and prolonged survival in an orthotopic murine xenograft model of early thymic precursor T-ALL (ETP-ALL), with both agents contributing to therapeutic activity in a dose-dependent manner. In contrast, nanoparticles containing MRX-2843 alone were ineffective in this model. Thus, MRX-2843 increased the sensitivity of ETP-ALL cells to vincristine in vivo. In this context, the additive particles, containing a higher dose of MRX-2843, provided more effective disease control than the synergistic particles. In contrast, particles containing an even higher, antagonistic ratio of MRX-2843 and vincristine were less effective. Thus, both the drug dose and the ratio-dependent interaction between MRX-2843 and vincristine significantly impacted therapeutic activity in vivo. Together, these findings present a systematic approach to high-throughput combination drug screening and multiagent drug delivery that maximizes the therapeutic potential of combined MRX-2843 and vincristine in T-ALL and describe a novel translational agent that could be used to enhance therapeutic responses to vincristine in patients with T-ALL. This broadly generalizable approach could also be applied to develop other constitutively synergistic combination products for the treatment of cancer and other diseases.
Collapse
Affiliation(s)
- James M Kelvin
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA
| | - Madison L Chimenti
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Dan Y Zhang
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Evelyn K Williams
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA
| | - Samuel G Moore
- Systems Mass Spectrometry Core Facility, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Gabrielle M Humber
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Travon A Baxter
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Lacey A Birnbaum
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA
| | - Min Qui
- Department of Pharmacology and Chemical Biology, Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Henry Zecca
- Department of Chemistry, Emory University, Atlanta, GA 30322, USA
| | - Aashis Thapa
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA
| | - Juhi Jain
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Nathan T Jui
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA; Department of Chemistry, Emory University, Atlanta, GA 30322, USA
| | - Xiaodong Wang
- Center for Integrative Chemical Biology and Drug Discovery, Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA; Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology, Emory Chemical Biology Discovery Center, Emory University School of Medicine, Atlanta, GA 30322, USA; Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Melissa L Kemp
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA; Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Wilbur A Lam
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA; Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA; Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Douglas K Graham
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Deborah DeRyckere
- Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA.
| | - Erik C Dreaden
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30322, USA; Department of Pediatrics, Emory School of Medicine, Atlanta, GA 30322, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA; Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA 30322, USA; Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
19
|
Nguyen TM, Jambhrunkar M, Wong SS, Ross DM, Joyce P, Finnie JW, Manavis J, Bremmell K, Pitman MR, Prestidge CA. Targeting Acute Myeloid Leukemia Using Sphingosine Kinase 1 Inhibitor-Loaded Liposomes. Mol Pharm 2023; 20:3937-3946. [PMID: 37463151 DOI: 10.1021/acs.molpharmaceut.3c00078] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Acute myeloid leukemia (AML) kills 75% of patients and represents a major clinical challenge with a need to improve on current treatment approaches. Targeting sphingosine kinase 1 with a novel ATP-competitive-inhibitor, MP-A08, induces cell death in AML. However, limitations in MP-A08's "drug-like properties" (solubility, biodistribution, and potency) hinder its pathway to the clinic. This study demonstrates a liposome-based delivery system of MP-A08 that exhibits enhanced MP-A08 potency against AML cells. MP-A08-liposomes increased MP-A08 efficacy against patient AML cells (>140-fold) and significantly prolonged overall survival of mice with human AML disease (P = 0.03). The significant antileukemic property of MP-A08-liposomes could be attributed to its enhanced specificity, bioaccessibility, and delivery to the bone marrow, as demonstrated in the pharmacokinetic and biodistribution studies. Our findings indicate that MP-A08-liposomes have potential as a novel treatment for AML.
Collapse
Affiliation(s)
- Thao M Nguyen
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia5001, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5001, Australia
| | - Manasi Jambhrunkar
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia5001, Australia
| | - Sook S Wong
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia5001, Australia
| | - David M Ross
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, South Australia 5001, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5001, Australia
- Department of Haematology, Flinders University and Medical Centre, Adelaide, South Australia 5001, Australia
- Department of Haematology and Bone Marrow Transplantation, Royal Adelaide Hospital, Adelaide, South Australia 5001, Australia
| | - Paul Joyce
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia5001, Australia
| | - John W Finnie
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5001, Australia
| | - Jim Manavis
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5001, Australia
| | - Kristen Bremmell
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia5001, Australia
| | - Melissa R Pitman
- School of Biological Sciences, University of Adelaide, Adelaide, South Australia 5001, Australia
| | - Clive A Prestidge
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia5001, Australia
| |
Collapse
|
20
|
Al-Antary ET, Gupte A, Carter J, Kaafarani M, Howard M, Edwards H, Ge Y, Taub JW. Curing childhood cancer the "Natural" Way: Nature as the source of chemotherapy agents. Biochem Pharmacol 2023; 213:115630. [PMID: 37263301 DOI: 10.1016/j.bcp.2023.115630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/24/2023] [Accepted: 05/24/2023] [Indexed: 06/03/2023]
Abstract
For many centuries, products of natural origin from plants, marine, microbes and soil micro-organisms have been studied by numerous researchers across the world to yield many of the chemotherapeutic agents we use in this modern era. There has been a tremendous gain in knowledge from various screening and separating techniques which led to the discovery of biologically active small molecules from natural products. Preclinical studies testing the antitumor activities of these agents against tumor cell lines and xenograft animal models were the gateway to the clinical trials in humans leading to the approval of these agents that are in clinical use today. This review summarizes how various chemotherapeutic agents were discovered from products of natural origin, their preclinical development, and their indications in both pediatric and adult oncology. Many of these natural products have contributed to the very high cure rates of both pediatric leukemias and solid tumors.
Collapse
Affiliation(s)
- Eman T Al-Antary
- Division of Pediatric Hematology/Oncology, Children's Hospital of Michigan, Detroit, MI, USA; Discipline of Pediatrics, Central Michigan University, Mt. Pleasant, MI, USA
| | - Avanti Gupte
- Division of Pediatric Hematology/Oncology, Children's Hospital of Michigan, Detroit, MI, USA; Discipline of Pediatrics, Central Michigan University, Mt. Pleasant, MI, USA
| | - Jenna Carter
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA; MD/PhD Program, Wayne State University School of Medicine, Detroit, MI, USA
| | | | | | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yubin Ge
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA; Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jeffrey W Taub
- Division of Pediatric Hematology/Oncology, Children's Hospital of Michigan, Detroit, MI, USA; Discipline of Pediatrics, Central Michigan University, Mt. Pleasant, MI, USA; Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA; Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
21
|
Huber S, Baer C, Hutter S, Dicker F, Meggendorfer M, Pohlkamp C, Kern W, Haferlach T, Haferlach C, Hoermann G. AML classification in the year 2023: How to avoid a Babylonian confusion of languages. Leukemia 2023; 37:1413-1420. [PMID: 37120689 PMCID: PMC10317829 DOI: 10.1038/s41375-023-01909-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/01/2023]
Abstract
In parallel to the 5th edition of the World Health Organization Classification of Haematolymphoid Tumours (WHO 2022), an alternative International Consensus Classification (ICC) has been proposed. To evaluate the impact of the new classifications on AML diagnoses and ELN-based risk classification, we analyzed 717 MDS and 734 AML non-therapy-related patients diagnosed according to the revised 4th WHO edition (WHO 2017) by whole genome and transcriptome sequencing. In both new classifications, the purely morphologically defined AML entities decreased from 13% to 5%. Myelodysplasia-related (MR) AML increased from 22% to 28% (WHO 2022) and 26% (ICC). Other genetically-defined AML remained the largest group, and the abandoned AML-RUNX1 was mainly reclassified as AML-MR (WHO 2022: 77%; ICC: 96%). Different inclusion criteria of AML-CEBPA and AML-MR (i.a. exclusion of TP53 mutated cases according to ICC) were associated with differences in overall survival. In conclusion, both classifications focus on more genetics-based definitions with similar basic concepts and a large degree of agreement. The remaining non-comparability (e.g., TP53 mutated AML) needs additional studies to definitely answer open questions on disease categorization in an unbiased way.
Collapse
Affiliation(s)
- Sandra Huber
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Constance Baer
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Stephan Hutter
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Frank Dicker
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Manja Meggendorfer
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Christian Pohlkamp
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Wolfgang Kern
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Torsten Haferlach
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Claudia Haferlach
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Gregor Hoermann
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany.
| |
Collapse
|
22
|
Yusuf A, Almotairy ARZ, Henidi H, Alshehri OY, Aldughaim MS. Nanoparticles as Drug Delivery Systems: A Review of the Implication of Nanoparticles' Physicochemical Properties on Responses in Biological Systems. Polymers (Basel) 2023; 15:polym15071596. [PMID: 37050210 PMCID: PMC10096782 DOI: 10.3390/polym15071596] [Citation(s) in RCA: 113] [Impact Index Per Article: 113.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/11/2023] [Accepted: 03/14/2023] [Indexed: 04/14/2023] Open
Abstract
In the last four decades, nanotechnology has gained momentum with no sign of slowing down. The application of inventions or products from nanotechnology has revolutionised all aspects of everyday life ranging from medical applications to its impact on the food industry. Nanoparticles have made it possible to significantly extend the shelf lives of food product, improve intracellular delivery of hydrophobic drugs and improve the efficacy of specific therapeutics such as anticancer agents. As a consequence, nanotechnology has not only impacted the global standard of living but has also impacted the global economy. In this review, the characteristics of nanoparticles that confers them with suitable and potentially toxic biological effects, as well as their applications in different biological fields and nanoparticle-based drugs and delivery systems in biomedicine including nano-based drugs currently approved by the U.S. Food and Drug Administration (FDA) are discussed. The possible consequence of continuous exposure to nanoparticles due to the increased use of nanotechnology and possible solution is also highlighted.
Collapse
Affiliation(s)
- Azeez Yusuf
- Irish Centre for Genetic Lung Disease, Department of Medicine, RCSI University of Medicine and Health Sciences, Beaumont Hospital, D02 YN77 Dublin, Ireland
| | | | - Hanan Henidi
- Research Department, Health Sciences Research Center, Princess Nourah bint Abdulrahman University, Riyadh 84428, Saudi Arabia
| | - Ohoud Y Alshehri
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11564, Saudi Arabia
| | - Mohammed S Aldughaim
- Research Center, King Fahad Medical City, Riyadh Second Health Cluster, Riyadh 11451, Saudi Arabia
| |
Collapse
|
23
|
Kotsiafti A, Giannakas K, Christoforou P, Liapis K. Progress toward Better Treatment of Therapy-Related AML. Cancers (Basel) 2023; 15:cancers15061658. [PMID: 36980546 PMCID: PMC10046015 DOI: 10.3390/cancers15061658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Therapy-related acute myeloid leukemia (t-AML) comprises 10-20% of all newly diagnosed cases of AML and is related to previous use of chemotherapy or ionizing radiotherapy for an unrelated malignant non-myeloid disorder or autoimmune disease. Classic examples include alkylating agents and topoisomerase II inhibitors, whereas newer targeted therapies such as poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitors have emerged as causative agents. Typically, t-AML is characterized by adverse karyotypic abnormalities and molecular lesions that confer a poor prognosis. Nevertheless, there are also cases of t-AML without poor-risk features. The management of these patients remains controversial. We describe the causes and pathophysiology of t-AML, putting emphasis on its mutational heterogeneity, and present recent advances in its treatment including CPX-351, hypomethylating agent plus venetoclax combination, and novel, molecularly targeted agents that promise to improve the cure rates. Evidence supporting personalized medicine for patients with t-AML is presented, as well as the authors' clinical recommendations.
Collapse
Affiliation(s)
| | | | - Panagiotis Christoforou
- Pathophysiology Department, National and Kapodistrian University of Athens, 157 72 Athens, Greece
| | - Konstantinos Liapis
- Dragana Campus, Democritus University of Thrace Medical School, 681 00 Alexandroupolis, Greece
| |
Collapse
|
24
|
Sarkar M, Wang Y, Ekpenyong O, Liang D, Xie H. Pharmacokinetic behaviors of soft nanoparticulate formulations of chemotherapeutics. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1846. [PMID: 35979879 PMCID: PMC9938089 DOI: 10.1002/wnan.1846] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/17/2022] [Accepted: 07/12/2022] [Indexed: 11/10/2022]
Abstract
Chemotherapeutic treatment with conventional drug formulations pose numerous challenges, such as poor solubility, high cytotoxicity and serious off-target side effects, low bioavailability, and ultimately subtherapeutic tumoral concentration leading to poor therapeutic outcomes. In the field of Nanomedicine, advances in nanotechnology have been applied with great success to design and develop novel nanoparticle-based formulations for the treatment of various types of cancer. The approval of the first nanomedicine, Doxil® (liposomal doxorubicin) in 1995, paved the path for further development for various types of novel delivery platforms. Several different types of nanoparticles, especially organic (soft) nanoparticles (liposomes, polymeric micelles, and albumin-bound nanoparticles), have been developed and approved for several anticancer drugs. Nanoparticulate drug delivery platform have facilitated to overcome of these challenges and offered key advantages of improved bioavailability, higher intra-tumoral concentration of the drug, reduced toxicity, and improved efficacy. This review introduces various commonly used nanoparticulate systems in biomedical research and their pharmacokinetic (PK) attributes, then focuses on the various physicochemical and physiological factors affecting the in vivo disposition of chemotherapeutic agents encapsulated in nanoparticles in recent years. Further, it provides a review of the current landscape of soft nanoparticulate formulations for the two most widely investigated anticancer drugs, paclitaxel, and doxorubicin, that are either approved or under investigation. Formulation details, PK profiles, and therapeutic outcomes of these novel strategies have been discussed individually and in comparison, to traditional formulations. This article is categorized under: Nanotechnology Approaches to Biology > Cells at the Nanoscale Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Mahua Sarkar
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas, USA
| | - Yang Wang
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas, USA
| | | | - Dong Liang
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas, USA
| | - Huan Xie
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas, USA
| |
Collapse
|
25
|
Jia Y, Jiang Y, He Y, Zhang W, Zou J, Magar KT, Boucetta H, Teng C, He W. Approved Nanomedicine against Diseases. Pharmaceutics 2023; 15:774. [PMID: 36986635 PMCID: PMC10059816 DOI: 10.3390/pharmaceutics15030774] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/08/2023] [Accepted: 02/18/2023] [Indexed: 03/03/2023] Open
Abstract
Nanomedicine is a branch of medicine using nanotechnology to prevent and treat diseases. Nanotechnology represents one of the most effective approaches in elevating a drug's treatment efficacy and reducing toxicity by improving drug solubility, altering biodistribution, and controlling the release. The development of nanotechnology and materials has brought a profound revolution to medicine, significantly affecting the treatment of various major diseases such as cancer, injection, and cardiovascular diseases. Nanomedicine has experienced explosive growth in the past few years. Although the clinical transition of nanomedicine is not very satisfactory, traditional drugs still occupy a dominant position in formulation development, but increasingly active drugs have adopted nanoscale forms to limit side effects and improve efficacy. The review summarized the approved nanomedicine, its indications, and the properties of commonly used nanocarriers and nanotechnology.
Collapse
Affiliation(s)
- Yuanchao Jia
- Nanjing Vtrying Pharmatech Co., Ltd., Nanjing 211122, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yuxin Jiang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yonglong He
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Wanting Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jiahui Zou
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | | | - Hamza Boucetta
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Chao Teng
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| |
Collapse
|
26
|
Swetha KL, Maravajjala KS, Li SD, Singh MS, Roy A. Breaking the niche: multidimensional nanotherapeutics for tumor microenvironment modulation. Drug Deliv Transl Res 2023; 13:105-134. [PMID: 35697894 DOI: 10.1007/s13346-022-01194-7] [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: 06/01/2022] [Indexed: 12/13/2022]
Abstract
Most of the current antitumor therapeutics were developed targeting the cancer cells only. Unfortunately, in the majority of tumors, this single-dimensional therapy is found to be ineffective. Advanced research has shown that cancer is a multicellular disorder. The tumor microenvironment (TME), which is made by a complex network of the bulk tumor cells and other supporting cells, plays a crucial role in tumor progression. Understanding the importance of the TME in tumor growth, different treatment modalities have been developed targeting these supporting cells. Recent clinical results suggest that simultaneously targeting multiple components of the tumor ecosystem with drug combinations can be highly effective. This type of "multidimensional" therapy has a high potential for cancer treatment. However, tumor-specific delivery of such multi-drug combinations remains a challenge. Nanomedicine could be utilized for the tumor-targeted delivery of such multidimensional therapeutics. In this review, we first give a brief overview of the major components of TME. We then highlight the latest developments in nanoparticle-based combination therapies, where one drug targets cancer cells and other drug targets tumor-supporting components in the TME for a synergistic effect. We include the latest preclinical and clinical studies and discuss innovative nanoparticle-mediated targeting strategies.
Collapse
Affiliation(s)
- K Laxmi Swetha
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India
| | - Kavya Sree Maravajjala
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, The University of British Columbia, 2405 Westbrook Mall, Vancouver, BC, Canada
| | - Manu Smriti Singh
- Department of Biotechnology, Bennett University, Greater Noida, Uttar Pradesh, 201310, India. .,Center of Excellence for Nanosensors and Nanomedicine, Bennett University, Greater Noida, Uttar Pradesh, 201310, India.
| | - Aniruddha Roy
- Department of Pharmacy, Birla Institute of Technology & Science, Vidya Vihar, Pilani, Rajasthan, 333031, India.
| |
Collapse
|
27
|
Lipid-Nanoparticle-Mediated Delivery of Docetaxel Prodrug for Exploiting Full Potential of Gold Nanoparticles in the Treatment of Pancreatic Cancer. Cancers (Basel) 2022; 14:cancers14246137. [PMID: 36551622 PMCID: PMC9776798 DOI: 10.3390/cancers14246137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/07/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Current chemoradiation therapy suffers from normal tissue toxicity. Thus, we are proposing incorporating gold nanoparticles (GNPs) and docetaxel (DTX), as they have shown very promising synergetic radiosensitization effects. Here, we explored the effect of a DTX prodrug encapsulated in lipid nanoparticles (LNPDTX-P) on GNP uptake in pancreatic cancer models in vitro and in vivo. For the in vitro experiment, a pancreatic cancer cell line, MIA PaCa-2, was cultured and dosed with 1 nM GNPs and 45 nM free DTX or an equivalent dose of LNPDTX-P. For the in vivo experiment, MIA PaCa-2 cells were implanted subcutaneously in NRG mice, and the mice were dosed with 2 mg/kg of GNPs and 6 mg/kg of DTX or an equivalent dose of LNPDTX-P. The results show that LNPDTX-P-treated tumour samples had double the amount GNPs compared to control samples, both in vitro and in vivo. The results are very promising, as LNPDTX-P have superior targeting of tumour tissues compared to free DTX due to their nanosize and their ability to be functionalized. Because of their minimal toxicity to normal tissues, both GNPs and LNPDTX-P could be ideal radiosensitization candidates in radiotherapy and would produce very promising synergistic therapeutic outcomes.
Collapse
|
28
|
Parodi A, Kolesova EP, Voronina MV, Frolova AS, Kostyushev D, Trushina DB, Akasov R, Pallaeva T, Zamyatnin AA. Anticancer Nanotherapeutics in Clinical Trials: The Work behind Clinical Translation of Nanomedicine. Int J Mol Sci 2022; 23:13368. [PMID: 36362156 PMCID: PMC9656556 DOI: 10.3390/ijms232113368] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 10/04/2023] Open
Abstract
The ultimate goal of nanomedicine has always been the generation of translational technologies that can ameliorate current therapies. Cancer disease represented the primary target of nanotechnology applied to medicine, since its clinical management is characterized by very toxic therapeutics. In this effort, nanomedicine showed the potential to improve the targeting of different drugs by improving their pharmacokinetics properties and to provide the means to generate new concept of treatments based on physical treatments and biologics. In this review, we considered different platforms that reached the clinical trial investigation, providing an objective analysis about their physical and chemical properties and the working mechanism at the basis of their tumoritr opic properties. With this review, we aim to help other scientists in the field in conceiving their delivering platforms for clinical translation by providing solid examples of technologies that eventually were tested and sometimes approved for human therapy.
Collapse
Affiliation(s)
- Alessandro Parodi
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Ekaterina P. Kolesova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Maya V. Voronina
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Anastasia S. Frolova
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Dmitry Kostyushev
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Daria B. Trushina
- Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Federal Scientific Research Center «Crystallography and Photonics», Russian Academy of Sciences, 119333 Moscow, Russia
| | - Roman Akasov
- Institute of Molecular Theranostics, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Federal Scientific Research Center «Crystallography and Photonics», Russian Academy of Sciences, 119333 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Tatiana Pallaeva
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia
- Federal Scientific Research Center «Crystallography and Photonics», Russian Academy of Sciences, 119333 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Andrey A. Zamyatnin
- Scientific Center for Translation Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| |
Collapse
|
29
|
He Y, Zhang W, Xiao Q, Fan L, Huang D, Chen W, He W. Liposomes and liposome-like nanoparticles: From anti-fungal infection to the COVID-19 pandemic treatment. Asian J Pharm Sci 2022; 17:817-837. [PMID: 36415834 PMCID: PMC9671608 DOI: 10.1016/j.ajps.2022.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/18/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022] Open
Abstract
The liposome is the first nanomedicine transformed into the market and applied to human patients. Since then, such phospholipid bilayer vesicles have undergone technological advancements in delivering small molecular-weight compounds and biological drugs. Numerous investigations about liposome uses were conducted in different treatment fields, including anti-tumor, anti-fungal, anti-bacterial, and clinical analgesia, owing to liposome's ability to reduce drug cytotoxicity and improve the therapeutic efficacy and combinatorial delivery. In particular, two liposomal vaccines were approved in 2021 to combat COVID-19. Herein, the clinically used liposomes are reviewed by introducing various liposomal preparations in detail that are currently proceeding in the clinic or on the market. Finally, we discuss the challenges of developing liposomes and cutting-edge liposomal delivery for biological drugs and combination therapy.
Collapse
Affiliation(s)
- Yonglong He
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Wanting Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Qingqing Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Lifang Fan
- Jiangsu Aosaikang Pharmaceutical Co., Ltd., Nanjing 211112, China
| | - Dechun Huang
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Wei Chen
- School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| |
Collapse
|
30
|
Renault-Mahieux M, Mignet N, Seguin J, Alhareth K, Paul M, Andrieux K. Co-encapsulation of flavonoids with anti-cancer drugs: a challenge ahead. Int J Pharm 2022; 623:121942. [PMID: 35728717 DOI: 10.1016/j.ijpharm.2022.121942] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/24/2022] [Accepted: 06/15/2022] [Indexed: 11/29/2022]
Abstract
Flavonoids have been considered as promising molecules for cancer treatment due to their pleiotropic properties such as anti-carcinogenic, anti-angiogenic or efflux proteins inhibition. However, due to their lipophilic properties and their chemical instability, vectorization seems compulsory to administer flavonoids. Flavonoids have been co-encapsulated with other anti-cancer agents in a broad range of nanocarriers aiming to i) achieve a synergistic/additive effect at the tumor site, ii) delay drug resistance apparition by combining agents with different action mechanisms or iii) administer a lower dose of the anti-cancer drug, reducing its toxicity. However, co-encapsulation could lead to a change in the nanoparticles' diameter and drug-loading, as well as a decrease in their stability during storage. The preparation process should also take into accounts the physico-chemical properties of both the flavonoid and the anti-cancer agent. Moreover, the co-encapsulation could affect the release and activity of each drug. This review aims to study the formulation, preparation and characterization strategies of these co-loaded nanomedicines, as well as their stability. The in vitro assays to predict the nanomedicines' behavior in biological fluids, as well as their in vivo efficacy, are also discussed. A special focus concerns the evaluation of their synergistic effect on tumor treatment.
Collapse
Affiliation(s)
- Morgane Renault-Mahieux
- Université Paris Cité, CNRS, Inserm, UTCBS, F-75006 Paris, France; Pharmacy Department, AP-HP, Henri Mondor Hospital Group, F-94010, France.
| | - Nathalie Mignet
- Université Paris Cité, CNRS, Inserm, UTCBS, F-75006 Paris, France.
| | - Johanne Seguin
- Université Paris Cité, CNRS, Inserm, UTCBS, F-75006 Paris, France.
| | - Khair Alhareth
- Université Paris Cité, CNRS, Inserm, UTCBS, F-75006 Paris, France.
| | - Muriel Paul
- Pharmacy Department, AP-HP, Henri Mondor Hospital Group, F-94010, France.
| | - Karine Andrieux
- Université Paris Cité, CNRS, Inserm, UTCBS, F-75006 Paris, France.
| |
Collapse
|
31
|
Nsairat H, Khater D, Sayed U, Odeh F, Al Bawab A, Alshaer W. Liposomes: structure, composition, types, and clinical applications. Heliyon 2022; 8:e09394. [PMID: 35600452 PMCID: PMC9118483 DOI: 10.1016/j.heliyon.2022.e09394] [Citation(s) in RCA: 284] [Impact Index Per Article: 142.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/19/2022] [Accepted: 05/06/2022] [Indexed: 12/18/2022] Open
Abstract
Liposomes are now considered the most commonly used nanocarriers for various potentially active hydrophobic and hydrophilic molecules due to their high biocompatibility, biodegradability, and low immunogenicity. Liposomes also proved to enhance drug solubility and controlled distribution, as well as their capacity for surface modifications for targeted, prolonged, and sustained release. Based on the composition, liposomes can be considered to have evolved from conventional, long-circulating, targeted, and immune-liposomes to stimuli-responsive and actively targeted liposomes. Many liposomal-based drug delivery systems are currently clinically approved to treat several diseases, such as cancer, fungal and viral infections; more liposomes have reached advanced phases in clinical trials. This review describes liposomes structure, composition, preparation methods, and clinical applications.
Collapse
Affiliation(s)
- Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Dima Khater
- Department of Chemistry, Faculty of Arts and Science, Applied Science Private University, Amman, Jordan
| | - Usama Sayed
- Department of Biology, The University of Jordan, Amman, 11942, Jordan
| | - Fadwa Odeh
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan
| | - Abeer Al Bawab
- Department of Chemistry, The University of Jordan, Amman, 11942, Jordan.,Hamdi Mango Center for Scientific Research, The University of Jordan, Amman, 11942, Jordan
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, 11942, Jordan
| |
Collapse
|
32
|
Bhattacharjee S. Craft of Co-encapsulation in Nanomedicine: A Struggle To Achieve Synergy through Reciprocity. ACS Pharmacol Transl Sci 2022; 5:278-298. [PMID: 35592431 PMCID: PMC9112416 DOI: 10.1021/acsptsci.2c00033] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Indexed: 12/19/2022]
Abstract
Achieving synergism, often by combination therapy via codelivery of chemotherapeutic agents, remains the mainstay of treating multidrug-resistance cases in cancer and microbial strains. With a typical core-shell architecture and surface functionalization to ensure facilitated targeting of tissues, nanocarriers are emerging as a promising platform toward gaining such synergism. Co-encapsulation of disparate theranostic agents in nanocarriers-from chemotherapeutic molecules to imaging or photothermal modalities-can not only address the issue of protecting the labile drug payload from a hostile biochemical environment but may also ensure optimized drug release as a mainstay of synergistic effect. However, the fate of co-encapsulated molecules, influenced by temporospatial proximity, remains unpredictable and marred with events with deleterious impact on therapeutic efficacy, including molecular rearrangement, aggregation, and denaturation. Thus, more than just an art of confining multiple therapeutics into a 3D nanoscale space, a co-encapsulated nanocarrier, while aiming for synergism, should strive toward achieving a harmonious cohabitation of the encapsulated molecules that, despite proximity and opportunities for interaction, remain innocuous toward each other and ensure molecular integrity. This account will inspect the current progress in co-encapsulation in nanocarriers and distill out the key points toward accomplishing such synergism through reciprocity.
Collapse
Affiliation(s)
- Sourav Bhattacharjee
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
33
|
Wang T, Zhang X, Jia M, Yang A, Liu J, Wen T, Meng J, Xu H. Hydrophilic Realgar Nanocrystals Prolong the Survival of Refractory Acute Myeloid Leukemia Mice Through Inducing Multi-Lineage Differentiation and Apoptosis. Int J Nanomedicine 2022; 17:2191-2202. [PMID: 35599749 PMCID: PMC9122054 DOI: 10.2147/ijn.s358469] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/11/2022] [Indexed: 12/25/2022] Open
Affiliation(s)
- Tao Wang
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China
| | - Xue Zhang
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China
| | - Mengfan Jia
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China
| | - Aiyun Yang
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China
| | - Jian Liu
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China
| | - Tao Wen
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China
| | - Jie Meng
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China
| | - Haiyan Xu
- Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China
- Correspondence: Haiyan Xu; Jie Meng, Department of Biomedical Engineering, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, People’s Republic of China, Tel +8610 69156437; +8610 65135502, Email ;
| |
Collapse
|
34
|
Cheng S, Xiao P, Wang J, Li Z, Gao L, Zheng J, Hu Y, Ding X, Ling J, Lu Q, Pan J, Li B, Lu J, Wang Y, Ribeiro RC, Hu S. Decitabine combined with minimally myelosuppressive therapy for induction of remission in pediatric high-risk acute myeloid leukemia with chromosome 5q deletion: a report of three cases. Int J Hematol 2022; 116:146-151. [PMID: 35181851 DOI: 10.1007/s12185-022-03309-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 10/19/2022]
Abstract
Cases of pediatric acute myeloid leukemia (AML) with complex karyotypes including chromosome 5 abnormalities are rare and have a very poor prognosis. Management of AML with monosomy 5/del(5q) has been inconsistent. We treated three adolescents with this AML subtype using combined low-dose cytarabine and mitoxantrone, concurrently with decitabine and G-CSF, for remission induction. Decitabine was also included in the conditioning regimen before hematopoietic cell transplantation (HCT). All three patients achieved complete remission after treatment with this combination therapy. The treatment was well tolerated, and the patients are alive and free of disease at 3.6, 3.2, and 3.0 years after HCT, respectively.
Collapse
Affiliation(s)
- Shengqin Cheng
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Peifang Xiao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Juxiang Wang
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China.,Department of Hematology and Oncology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhiheng Li
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Li Gao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Jiajia Zheng
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Yixin Hu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Xin Ding
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Jing Ling
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Qin Lu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Jian Pan
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Bohan Li
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Jun Lu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Yi Wang
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China
| | - Raul C Ribeiro
- Department of Oncology, Division of Leukemia/Lymphoma, and Department of Global Pediatric Medicine, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Shaoyan Hu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou Industrial Park, No. 92 Zhongnan Street, Suzhou, 215025, People's Republic of China.
| |
Collapse
|
35
|
Souri M, Soltani M, Moradi Kashkooli F, Kiani Shahvandi M, Chiani M, Shariati FS, Mehrabi MR, Munn LL. Towards principled design of cancer nanomedicine to accelerate clinical translation. Mater Today Bio 2022; 13:100208. [PMID: 35198957 PMCID: PMC8841842 DOI: 10.1016/j.mtbio.2022.100208] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 02/08/2023] Open
Abstract
Nanotechnology in medical applications, especially in oncology as drug delivery systems, has recently shown promising results. However, although these advances have been promising in the pre-clinical stages, the clinical translation of this technology is challenging. To create drug delivery systems with increased treatment efficacy for clinical translation, the physicochemical characteristics of nanoparticles such as size, shape, elasticity (flexibility/rigidity), surface chemistry, and surface charge can be specified to optimize efficiency for a given application. Consequently, interdisciplinary researchers have focused on producing biocompatible materials, production technologies, or new formulations for efficient loading, and high stability. The effects of design parameters can be studied in vitro, in vivo, or using computational models, with the goal of understanding how they affect nanoparticle biophysics and their interactions with cells. The present review summarizes the advances and technologies in the production and design of cancer nanomedicines to achieve clinical translation and commercialization. We also highlight existing challenges and opportunities in the field.
Collapse
Key Words
- CFL, Cell-free layer
- CGMD, Coarse-grained molecular dynamic
- Clinical translation
- DPD, Dissipative particle dynamic
- Drug delivery
- Drug loading
- ECM, Extracellular matrix
- EPR, Permeability and retention
- IFP, Interstitial fluid pressure
- MD, Molecular dynamic
- MDR, Multidrug resistance
- MEC, Minimum effective concentration
- MMPs, Matrix metalloproteinases
- MPS, Mononuclear phagocyte system
- MTA, Multi-tadpole assemblies
- MTC, Minimum toxic concentration
- Nanomedicine
- Nanoparticle design
- RBC, Red blood cell
- TAF, Tumor-associated fibroblast
- TAM, Tumor-associated macrophage
- TIMPs, Tissue inhibitor of metalloproteinases
- TME, Tumor microenvironment
- Tumor microenvironment
Collapse
Affiliation(s)
- Mohammad Souri
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
- Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - M. Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
- Department of Electrical and Computer Engineering, University of Waterloo, ON, Canada
- Centre for Biotechnology and Bioengineering (CBB), University of Waterloo, Waterloo, ON, Canada
- Advanced Bioengineering Initiative Center, Computational Medicine Center, K. N. Toosi University of Technology, Tehran, Iran
| | | | | | - Mohsen Chiani
- Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Lance L. Munn
- Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| |
Collapse
|
36
|
Biomembrane-based nanostructures for cancer targeting and therapy: From synthetic liposomes to natural biomembranes and membrane-vesicles. Adv Drug Deliv Rev 2021; 178:113974. [PMID: 34530015 DOI: 10.1016/j.addr.2021.113974] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/29/2021] [Accepted: 09/08/2021] [Indexed: 12/14/2022]
Abstract
The translational success of liposomes in chemotherapeutics has already demonstrated the great potential of biomembrane-based nanostructure in effective drug delivery. Meanwhile, increasing efforts are being dedicated to the application of naturally derived lipid membranes, including cellular membranes and extracellular vesicles in anti-cancer therapies. While synthetic liposomes support superior multifunctional flexibility, natural biomembrane materials possess interesting biomimetic properties and can also be further engineered for intelligent design. Despite being remarkably different from each other in production and composition, the phospholipid bilayer structure in common allows liposomes, cell membrane-derived nanomaterials, and extracellular vesicles to be modified, functionalized, and exploited in many similar manners against challenges posed by tumor-targeted drug delivery. This review will summarize the recent advancements in engineering the membrane-derived nanostructures with "intelligent" modules to respond, regulate, and target tumor cells and the microenvironment to fight against malignancy. We will also discuss perspectives of combining engineered functionalities with naturally occurring activity for enhanced cancer therapy.
Collapse
|
37
|
Garcia-Manero G, Döhner H, Wei AH, La Torre I, Skikne B, Beach CL, Santini V. Oral Azacitidine (CC-486) for the Treatment of Myeloid Malignancies. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 22:236-250. [PMID: 34758945 DOI: 10.1016/j.clml.2021.09.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/22/2021] [Accepted: 09/27/2021] [Indexed: 02/07/2023]
Abstract
Epigenetic dysregulation leads to aberrant DNA hypermethylation and is common in acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS). A large number of clinical trials in AML, MDS, and other hematologic malignancies have assessed hypomethylating agents (HMAs), used alone or in combination with other drugs, in the frontline, maintenance, relapsed/refractory, and peritransplant settings. Effective maintenance therapy has long been a goal for patients with AML in remission. Previous large, randomized clinical trials of maintenance with HMAs or other agents had not shown meaningful improvement in overall survival. Oral azacitidine (Oral-AZA [CC-486]) is approved in the United States, Canada, and European Union for treatment of adult patients with AML in first complete remission (CR) or CR with incomplete blood count recovery (CRi) following intensive induction chemotherapy who are ineligible for hematopoietic cell transplant. Regulatory approvals of Oral-AZA were based on outcomes from the randomized, phase III QUAZAR AML-001 trial, which showed a median overall survival advantage of 9.9 months with Oral-AZA versus placebo. Oral-AZA allows convenient extended AZA dosing for 14 days per 28-day treatment cycle, which is not feasible with injectable AZA. Focusing on AML and MDS, this report reviews the rationale for the use of orally bioavailable AZA and its potential use in all-oral combination therapy regimens; the unique pharmacokinetic and pharmacodynamic profile of Oral-AZA compared with injectable AZA; the clinical safety and efficacy of Oral-AZA maintenance therapy in patients with AML in first remission and for treatment of patients with active MDS; and ongoing Oral-AZA clinical trials.
Collapse
Affiliation(s)
| | - Hartmut Döhner
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
| | - Andrew H Wei
- Department of Clinical Haematology, The Alfred Hospital, Melbourne, Australia; Monash University, Australian Centre for Blood Diseases, Melbourne, Australia
| | | | - Barry Skikne
- Bristol-Myers Squibb Company, Princeton, NJ; Department of Hematology, University of Kansas Medical Center, Kansas City, KS
| | - C L Beach
- Bristol-Myers Squibb Company, Princeton, NJ
| | - Valeria Santini
- MDS Unit, Hematology, AOU Careggi, University of Florence, Florence, Italy
| |
Collapse
|
38
|
Xu MQ, Zhong T, Yao X, Li ZY, Li H, Wang JR, Feng ZH, Zhang X. Effect of XlogP and hansen solubility parameters on the prediction of small molecule modified docetaxel, doxorubicin and irinotecan conjugates forming stable nanoparticles. Drug Deliv 2021; 28:1603-1615. [PMID: 34319209 PMCID: PMC8330778 DOI: 10.1080/10717544.2021.1958107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Small molecule-chemotherapeutic drug conjugate nanoparticles (SMCDC NPs) has a great advantage in improving drug loading. However, the factors which influence these conjugates forming stable nanoparticles (NPs) are currently unclear. In our previous studies, we synthesized a series of fatty acid-paclitaxel conjugates and suggested that the changes in the hydrophobic parameters (XlogP), solubility parameters and crystallinity of these fatty acid-paclitaxel conjugates were the key factors for affecting these small molecule-chemotherapeutic drug conjugates (SMCDCs) forming stable NPs in water. Here, we selected clinically widely used chemotherapeutic drug (docetaxel (DTX), doxorubicin (DOX) and irinotecan (Ir)) as model drug, and chose three straight-chain fatty acids (acetic acid (Ac), hexanoic acid (HA) and stearic acid (SA)) and one branched small molecule (N-(tert-butoxycarbonyl) glycine (B-G)) to synthesize 12 SMCDCs. Our results indicated that our prediction criterions obtained from paclitaxel conjugates were also appropriated for these synthesized SMCDCs. We suggested that the present studies expanded the scope of application of the above-mentioned influencing factors, provided research ideas for the rational design of SMCDC forming NPs and a basis for screening NPs with good anticancer activity.
Collapse
Affiliation(s)
- Mei-Qi Xu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Ting Zhong
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xin Yao
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zhuo-Yue Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Hui Li
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jing-Ru Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Zhen-Han Feng
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xuan Zhang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing, China.,Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing, China
| |
Collapse
|
39
|
Renault-Mahieux M, Vieillard V, Seguin J, Espeau P, Le DT, Lai-Kuen R, Mignet N, Paul M, Andrieux K. Co-Encapsulation of Fisetin and Cisplatin into Liposomes for Glioma Therapy: From Formulation to Cell Evaluation. Pharmaceutics 2021; 13:pharmaceutics13070970. [PMID: 34206986 PMCID: PMC8309049 DOI: 10.3390/pharmaceutics13070970] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/14/2021] [Accepted: 06/23/2021] [Indexed: 12/13/2022] Open
Abstract
(1) Background: Glioblastoma (GBM) is the most frequent cerebral tumor. It almost always relapses and there is no validated treatment for second-line GBM. We proposed the coencapsulation of fisetin and cisplatin into liposomes, aiming to (i) obtain a synergistic effect by combining the anti-angiogenic effect of fisetin with the cytotoxic effect of cisplatin, and (ii) administrate fisetin, highly insoluble in water. The design of a liposomal formulation able to encapsulate, retain and deliver both drugs appeared a challenge. (2) Methods: Liposomes with increasing ratios of cholesterol/DOPC were prepared and characterized in term of size, PDI and stability. The incorporation of fisetin was explored using DSC. The antiangiogneic and cytotoxic activities of the selected formulation were assayed in vitro. (3) Results: We successfully developed an optimized liposomal formulation incorporating both drugs, composed by DOPC/cholesterol/DODA-GLY-PEG2000 at a molar ratio of 75.3/20.8/3.9, with a diameter of 173 ± 8 nm (PDI = 0.12 ± 0.01) and a fisetin and cisplatin drug loading of 1.7 ± 0.3% and 0.8 ± 0.1%, respectively, with a relative stability over time. The maximum incorporation of fisetin into the bilayer was determined at 3.2% w/w. Then, the antiangiogenic activity of fisetin was maintained after encapsulation. The formulation showed an additive effect of cisplatin and fisetin on GBM cells; (4) Conclusions: The developed co-loaded formulation was able to retain the activity of fisetin, was effective against GBM cells and is promising for further in vivo experimentations.
Collapse
Affiliation(s)
- Morgane Renault-Mahieux
- Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Paris, 4 Avenue de l’Observatoire, 75006 Paris, France; (M.R.-M.); (J.S.); (P.E.); (D.T.L.); (N.M.)
- Henri Mondor Hospital Group, Pharmacy Department, Assistance Publique—Hôpitaux de Paris (AP-HP), 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France; (V.V.); (M.P.)
| | - Victoire Vieillard
- Henri Mondor Hospital Group, Pharmacy Department, Assistance Publique—Hôpitaux de Paris (AP-HP), 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France; (V.V.); (M.P.)
| | - Johanne Seguin
- Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Paris, 4 Avenue de l’Observatoire, 75006 Paris, France; (M.R.-M.); (J.S.); (P.E.); (D.T.L.); (N.M.)
| | - Philippe Espeau
- Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Paris, 4 Avenue de l’Observatoire, 75006 Paris, France; (M.R.-M.); (J.S.); (P.E.); (D.T.L.); (N.M.)
| | - Dang Tri Le
- Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Paris, 4 Avenue de l’Observatoire, 75006 Paris, France; (M.R.-M.); (J.S.); (P.E.); (D.T.L.); (N.M.)
| | - René Lai-Kuen
- UMS3612 Centre National de la Recherche Scientifique (CNRS), US25 Institut NATIONAL de la Santé et de la Recherche Médicale (INSERM), Plateforme Mutualisée de l’Institut du Médicament (P-MIM), Plateau Technique Imagerie Cellulaire et Moléculaire, Université de Paris, 75006 Paris, France;
| | - Nathalie Mignet
- Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Paris, 4 Avenue de l’Observatoire, 75006 Paris, France; (M.R.-M.); (J.S.); (P.E.); (D.T.L.); (N.M.)
| | - Muriel Paul
- Henri Mondor Hospital Group, Pharmacy Department, Assistance Publique—Hôpitaux de Paris (AP-HP), 51 Avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France; (V.V.); (M.P.)
| | - Karine Andrieux
- Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Paris, 4 Avenue de l’Observatoire, 75006 Paris, France; (M.R.-M.); (J.S.); (P.E.); (D.T.L.); (N.M.)
- Correspondence: ; Tel.: +33-(0)1-53-73-97-63
| |
Collapse
|
40
|
Kumar S, Nagpal R, Kumar A, Ashraf MU, Bae YS. Immunotherapeutic Potential of m6A-Modifiers and MicroRNAs in Controlling Acute Myeloid Leukaemia. Biomedicines 2021; 9:690. [PMID: 34207299 PMCID: PMC8234128 DOI: 10.3390/biomedicines9060690] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/30/2021] [Accepted: 06/09/2021] [Indexed: 02/06/2023] Open
Abstract
Epigenetic alterations have contributed greatly to human carcinogenesis. Conventional epigenetic studies have been predominantly focused on DNA methylation, histone modifications, and chromatin remodelling. Epitranscriptomics is an emerging field that encompasses the study of RNA modifications that do not affect the RNA sequence but affect functionality via a series of RNA binding proteins called writer, reader and eraser. Several kinds of epi-RNA modifications are known, such as 6-methyladenosine (m6A), 5-methylcytidine (m5C), and 1-methyladenosine. M6A modification is the most studied and has large therapeutic implications. In this review, we have summarised the therapeutic potential of m6A-modifiers in controlling haematological disorders, especially acute myeloid leukaemia (AML). AML is a type of blood cancer affecting specific subsets of blood-forming hematopoietic stem/progenitor cells (HSPCs), which proliferate rapidly and acquire self-renewal capacities with impaired terminal cell-differentiation and apoptosis leading to abnormal accumulation of white blood cells, and thus, an alternative therapeutic approach is required urgently. Here, we have described how RNA m6A-modification machineries EEE (Editor/writer: Mettl3, Mettl14; Eraser/remover: FTO, ALKBH5, and Effector/reader: YTHDF-1/2) could be reformed into potential druggable candidates or as RNA-modifying drugs (RMD) to treat leukaemia. Moreover, we have shed light on the role of microRNAs and suppressors of cytokine signalling (SOCS/CISH) in increasing anti-tumour immunity towards leukaemia. We anticipate, our investigation will provide fundamental knowledge in nurturing the potential of RNA modifiers in discovering novel therapeutics or immunotherapeutic procedures.
Collapse
Affiliation(s)
- Sunil Kumar
- Department of Biological Sciences, Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea;
- Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea
| | - Ravinder Nagpal
- Department of Nutrition & Integrative Physiology, Florida State University, Tallahassee, FL 32306, USA;
| | - Amit Kumar
- Medical Writer, Quebec City, QC G1X 3E1, Canada;
| | - Muhammad Umer Ashraf
- Department of Biological Sciences, Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea;
- Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea
| | - Yong-Soo Bae
- Department of Biological Sciences, Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea;
- Science Research Center (SRC) for Immune Research on Non-lymphoid Organ (CIRNO), Sungkyunkwan University, Jangan-gu, Suwon 16419, Gyeonggi-do, Korea
| |
Collapse
|
41
|
Szumilak M, Wiktorowska-Owczarek A, Stanczak A. Hybrid Drugs-A Strategy for Overcoming Anticancer Drug Resistance? Molecules 2021; 26:2601. [PMID: 33946916 PMCID: PMC8124695 DOI: 10.3390/molecules26092601] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023] Open
Abstract
Despite enormous progress in the treatment of many malignancies, the development of cancer resistance is still an important reason for cancer chemotherapy failure. Increasing knowledge of cancers' molecular complexity and mechanisms of their resistance to anticancer drugs, as well as extensive clinical experience, indicate that an effective fight against cancer requires a multidimensional approach. Multi-target chemotherapy may be achieved using drugs combination, co-delivery of medicines, or designing hybrid drugs. Hybrid drugs simultaneously targeting many points of signaling networks and various structures within a cancer cell have been extensively explored in recent years. The single hybrid agent can modulate multiple targets involved in cancer cell proliferation, possesses a simpler pharmacokinetic profile to reduce the possibility of drug interactions occurrence, and facilitates the process of drug development. Moreover, a single medication is expected to enhance patient compliance due to a less complicated treatment regimen, as well as a diminished number of adverse reactions and toxicity in comparison to a combination of drugs. As a consequence, many efforts have been made to design hybrid molecules of different chemical structures and functions as a means to circumvent drug resistance. The enormous number of studies in this field encouraged us to review the available literature and present selected research results highlighting the possible role of hybrid drugs in overcoming cancer drug resistance.
Collapse
Affiliation(s)
- Marta Szumilak
- Department of Hospital Pharmacy, Faculty of Pharmacy, Medical University of Lodz, 1 Muszynskiego Street, 90-151 Lodz, Poland
| | - Anna Wiktorowska-Owczarek
- Department of Pharmacology and Toxicology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland;
| | - Andrzej Stanczak
- Department of Community Pharmacy, Faculty of Pharmacy, Medical University of Lodz, 1 Muszynskiego Street, 90-151 Lodz, Poland;
| |
Collapse
|
42
|
Panina SB, Pei J, Kirienko NV. Mitochondrial metabolism as a target for acute myeloid leukemia treatment. Cancer Metab 2021; 9:17. [PMID: 33883040 PMCID: PMC8058979 DOI: 10.1186/s40170-021-00253-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemias (AML) are a group of aggressive hematologic malignancies resulting from acquired genetic mutations in hematopoietic stem cells that affect patients of all ages. Despite decades of research, standard chemotherapy still remains ineffective for some AML subtypes and is often inappropriate for older patients or those with comorbidities. Recently, a number of studies have identified unique mitochondrial alterations that lead to metabolic vulnerabilities in AML cells that may present viable treatment targets. These include mtDNA, dependency on oxidative phosphorylation, mitochondrial metabolism, and pro-survival signaling, as well as reactive oxygen species generation and mitochondrial dynamics. Moreover, some mitochondria-targeting chemotherapeutics and their combinations with other compounds have been FDA-approved for AML treatment. Here, we review recent studies that illuminate the effects of drugs and synergistic drug combinations that target diverse biomolecules and metabolic pathways related to mitochondria and their promise in experimental studies, clinical trials, and existing chemotherapeutic regimens.
Collapse
Affiliation(s)
| | - Jingqi Pei
- Department of BioSciences, Rice University, Houston, TX, USA
| | | |
Collapse
|
43
|
Richardson DR, Swoboda DM, Moore DT, Johnson SM, Chan O, Galeotti J, Esparza S, Hussaini MO, Van Deventer H, Foster MC, Coombs CC, Montgomery ND, Sallman DA, Zeidner JF. Genomic characteristics and prognostic significance of co-mutated ASXL1/SRSF2 acute myeloid leukemia. Am J Hematol 2021; 96:462-470. [PMID: 33502020 DOI: 10.1002/ajh.26110] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/12/2021] [Accepted: 01/17/2021] [Indexed: 12/24/2022]
Abstract
The ASXL1 and SRSF2 mutations in AML are frequently found in patients with preexisting myeloid malignancies and are individually associated with poor outcomes. In this multi-institutional retrospective analysis, we assessed the genetic features and clinical outcomes of 43 patients with ASXL1mut SRSF2mut AML and compared outcomes to patients with either ASXL1 (n = 57) or SRSF2 (n = 70) mutations. Twenty-six (60%) had secondary-AML (s-AML). Variant allele fractions suggested that SRSF2 mutations preceded ASXL1 mutational events. Median overall survival (OS) was 7.0 months (95% CI:3.8,15.3) and was significantly longer in patients with de novo vs s-AML (15.3 vs 6.4 months, respectively; P = .04 on adjusted analysis). Compared to ASXL1mut SRSF2wt and ASXL1wt SRSF2mut , co-mutated patients had a 1.4 and 1.6 times increase in the probability of death, respectively (P = .049), with a trend towards inferior OS (median OS = 7.0 vs 11.5 vs 10.9 months, respectively; P = .10). Multivariable analysis suggests this difference in OS is attributable to the high proportion of s-AML patients in the co-mutated cohort (60% vs 32% and 23%, respectively). Although this study is limited by the retrospective data collection and the relatively small sample size, these data suggest that ASXL1mut SRSF2mut AML is a distinct subgroup of AML frequently associated with s-AML and differs from ASXL1mut SRSF2wt /ASXL1wt SRSF2mut with respect to etiology and leukemogenesis.
Collapse
Affiliation(s)
- Daniel R. Richardson
- Lineberger Comprehensive Cancer Center The University of North Carolina School of Medicine Chapel Hill North Carolina USA
- Division of Hematology, Department of Medicine The University of North Carolina School of Medicine Chapel Hill North Carolina USA
- The Cecil G. Sheps Center for Health Services Research University of North Carolina at Chapel Hill Chapel Hill North Carolina USA
| | - David M. Swoboda
- Department of Malignant Hematology H. Lee Moffitt Cancer Center and Research Institute Tampa Florida USA
| | - Dominic T. Moore
- Lineberger Comprehensive Cancer Center The University of North Carolina School of Medicine Chapel Hill North Carolina USA
| | - Steven M. Johnson
- Department of Pathology and Laboratory Medicine The University of North Carolina School of Medicine Chapel Hill North Carolina USA
| | - Onyee Chan
- Department of Malignant Hematology H. Lee Moffitt Cancer Center and Research Institute Tampa Florida USA
| | - Jonathan Galeotti
- Department of Pathology and Laboratory Medicine The University of North Carolina School of Medicine Chapel Hill North Carolina USA
| | - Sonia Esparza
- Lineberger Comprehensive Cancer Center The University of North Carolina School of Medicine Chapel Hill North Carolina USA
- Division of Hematology, Department of Medicine The University of North Carolina School of Medicine Chapel Hill North Carolina USA
| | - Mohammad O. Hussaini
- Department of Malignant Hematology H. Lee Moffitt Cancer Center and Research Institute Tampa Florida USA
| | - Hendrick Van Deventer
- Lineberger Comprehensive Cancer Center The University of North Carolina School of Medicine Chapel Hill North Carolina USA
- Division of Hematology, Department of Medicine The University of North Carolina School of Medicine Chapel Hill North Carolina USA
| | - Matthew C. Foster
- Lineberger Comprehensive Cancer Center The University of North Carolina School of Medicine Chapel Hill North Carolina USA
- Division of Hematology, Department of Medicine The University of North Carolina School of Medicine Chapel Hill North Carolina USA
| | - Catherine C. Coombs
- Lineberger Comprehensive Cancer Center The University of North Carolina School of Medicine Chapel Hill North Carolina USA
- Division of Hematology, Department of Medicine The University of North Carolina School of Medicine Chapel Hill North Carolina USA
| | - Nathan D. Montgomery
- Lineberger Comprehensive Cancer Center The University of North Carolina School of Medicine Chapel Hill North Carolina USA
- Department of Pathology and Laboratory Medicine The University of North Carolina School of Medicine Chapel Hill North Carolina USA
| | - David A. Sallman
- Department of Malignant Hematology H. Lee Moffitt Cancer Center and Research Institute Tampa Florida USA
| | - Joshua F. Zeidner
- Lineberger Comprehensive Cancer Center The University of North Carolina School of Medicine Chapel Hill North Carolina USA
- Division of Hematology, Department of Medicine The University of North Carolina School of Medicine Chapel Hill North Carolina USA
| |
Collapse
|
44
|
Ismailov RM, Saenz DT, Gascon P, Nucci M, Khasanova ZD. Improving awareness of several combination therapies for acute myeloid leukemia among oncology and hematology team members in Colorado, USA. Hematol Transfus Cell Ther 2021; 44:358-364. [PMID: 33622645 PMCID: PMC9477762 DOI: 10.1016/j.htct.2021.01.001] [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: 08/30/2020] [Revised: 10/23/2020] [Accepted: 01/07/2021] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Although several combination therapies for acute myeloid leukemia (AML) have emerged recently, there has been a lack of published surveys and educational projects focused on these important treatment options. We aimed to improve the oncology team members' knowledge and awareness of several FDA approved combination therapies for AML, including glasdegib (DAURISMO®), venetoclax (VENCLEXTA®), GO (MYOLOTARG®),CPX-351 (VYXEOS®), and midostaurin (RYDAPT®). Additionally, we aimed to examine these teams' perspectives, views, and attitudes towards these topics and finally identify barriers to the implementationof such therapies in clinical practice. METHOD Initially, we developed booklets and then distributed them to each participating oncology and hematology office. Subsequently, all participating oncology and hematology team members were asked to complete an anonymous online survey to test their knowledge of and attitudes toward the subjects. MAIN RESULTS There was a total of 52 survey respondents. The correct answer regarding various combination therapies for AML was identified by nearly 70% or more of survey takers. The level of awareness of project subjects significantly improved after reading our printing materials. Many survey respondents were motivated to learn more about combination therapies for AML as well as discuss these topics with others. CONCLUSIONS Our booklets effectively improved understanding and awareness of combination therapies for AML. Future studies should explore awareness, knowledge, and perception of other new and emerging combination therapies for AML among oncology and hematology team members in other areas.
Collapse
Affiliation(s)
- Rovshan M Ismailov
- Complex Mechanisms of Disease, Aging and Trauma (CMDAT) Research Foundation, Denver, CO, United States.
| | - Dyana T Saenz
- The University of Texas M.D. Anderson Cancer Center, Houston TX, United States
| | - Pere Gascon
- Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain
| | - Marcio Nucci
- Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Zaytuna D Khasanova
- Complex Mechanisms of Disease, Aging and Trauma (CMDAT) Research Foundation, Denver, CO, United States
| |
Collapse
|
45
|
Controversies in the recent (2016) World Health Organization classification of acute myeloid leukemia. Best Pract Res Clin Haematol 2021; 34:101249. [PMID: 33762104 DOI: 10.1016/j.beha.2021.101249] [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: 12/20/2022]
Abstract
The current World Health Organization (WHO) Classification of acute myeloid leukemia (AML), developed in 2016 and published in 2017, codifies the defining features of AML and recognizes several subtypes based on clinical, morphologic, and genetic features. This classification is widely used for the purposes of assigning patients to specific therapeutic approaches and entry into clinical trials. Although the WHO Classification ultimately has its origins in the original 1976 French-American-British Classification, it has been periodically updated by the incorporation of a large body of evidence and input from both diagnosticians and clinicians who study and treat AML. Nevertheless, the recent accumulation of genetic data on the molecular underpinnings of myeloid neoplasms as well as numerous recently approved novel therapies have highlighted areas of controversy in how we currently define and classify AML; the 2016 WHO Classification will continually be revised and updated in future versions based on these advances. The purpose of this review is to explore areas of potential refinement in the current WHO Classification of AML, both in terms of its criteria defining the disease as well as the specific disease subtypes.
Collapse
|
46
|
Carter JL, Hege K, Yang J, Kalpage HA, Su Y, Edwards H, Hüttemann M, Taub JW, Ge Y. Targeting multiple signaling pathways: the new approach to acute myeloid leukemia therapy. Signal Transduct Target Ther 2020; 5:288. [PMID: 33335095 PMCID: PMC7746731 DOI: 10.1038/s41392-020-00361-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is the most common form of acute leukemia in adults and the second most common form of acute leukemia in children. Despite this, very little improvement in survival rates has been achieved over the past few decades. This is partially due to the heterogeneity of AML and the need for more targeted therapeutics than the traditional cytotoxic chemotherapies that have been a mainstay in therapy for the past 50 years. In the past 20 years, research has been diversifying the approach to treating AML by investigating molecular pathways uniquely relevant to AML cell proliferation and survival. Here we review the development of novel therapeutics in targeting apoptosis, receptor tyrosine kinase (RTK) signaling, hedgehog (HH) pathway, mitochondrial function, DNA repair, and c-Myc signaling. There has been an impressive effort into better understanding the diversity of AML cell characteristics and here we highlight important preclinical studies that have supported therapeutic development and continue to promote new ways to target AML cells. In addition, we describe clinical investigations that have led to FDA approval of new targeted AML therapies and ongoing clinical trials of novel therapies targeting AML survival pathways. We also describe the complexity of targeting leukemia stem cells (LSCs) as an approach to addressing relapse and remission in AML and targetable pathways that are unique to LSC survival. This comprehensive review details what we currently understand about the signaling pathways that support AML cell survival and the exceptional ways in which we disrupt them.
Collapse
Affiliation(s)
- Jenna L Carter
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA.,MD/PhD Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Katie Hege
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jay Yang
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Hasini A Kalpage
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yongwei Su
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.,National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, China
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jeffrey W Taub
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA. .,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA. .,Division of Pediatric Hematology/Oncology, Children's Hospital of Michigan, Detroit, MI, USA. .,Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Yubin Ge
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI, USA. .,Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA. .,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
47
|
Zhang J, Zhou L, Zhang Y, He H, Yin T, Gou J, Wang Y, Tang X. Contrastive Studies of Cytarabine/Daunorubicin Dual-Loaded Liposomes Prepared by pH Gradient and Cu 2+ Gradient Method. AAPS PharmSciTech 2020; 21:325. [PMID: 33206247 DOI: 10.1208/s12249-020-01867-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/28/2020] [Indexed: 12/16/2022] Open
Abstract
Conventional combination chemotherapy often leads to unsatisfactory clinical outcomes due to the different distribution characteristics in vivo and the superimposed systemic toxicity of the drug cocktail. Co-encapsulated nano preparations have been gradually developed in recent years. In this work, cytarabine (Ara-C)/daunorubicin (DNR) liposomes were prepared by the pH gradient (ADL-pH) and Cu2+ gradient (ADL-Cu) methods. Ara-C did not show significant release from either ADL-Cu or ADL-pH in vitro during 168 h, which related to its logPoct. Different drug-loading patterns showed different release characteristics of DNR due to the different existence forms, ADL-pH contains the citrate form, while in ADL-Cu, there is the Cu2+ complex. To evaluate the release behavior, daunorubicin liposome (DL) and daunorubicin-Cu2+ complex (DNR-Cu) were prepared. The addition of EDTA in the release medium significantly increased the release rate of DNR from DL-Cu, while lower pH accelerated DNR release from both DL-pH and DL-Cu. The PK confirmed that ADL-Cu and ADL-pH could prolong the drug circulation time, and ADL-Cu had a mean retention time 1.5 times that of ADL-pH. Furthermore, both liposomes allowed the two drugs to maintain a relatively constant plasma concentration ratio for a prolonged time. Cytotoxicity assays showed that Ara-C/DNR with a molar ratio of 5:1 and 3:1 exhibited an excellent synergistic effect, which was more obvious at 5:1. In vitro antitumor results revealed that ADL-Cu exhibited more cytotoxicity than ADL-pH. All factors tested in this work suggest the considerable potential of ADL-Cu and ADL-pH for anticancer treatment.
Collapse
|
48
|
Gurnari C, Pagliuca S, Visconte V. Deciphering the Therapeutic Resistance in Acute Myeloid Leukemia. Int J Mol Sci 2020; 21:ijms21228505. [PMID: 33198085 PMCID: PMC7697160 DOI: 10.3390/ijms21228505] [Citation(s) in RCA: 8] [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: 10/20/2020] [Revised: 11/05/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is a clonal hematopoietic disorder characterized by abnormal proliferation, lack of cellular differentiation, and infiltration of bone marrow, peripheral blood, or other organs. Induction failure and in general resistance to chemotherapeutic agents represent a hindrance for improving survival outcomes in AML. Here, we review the latest insights in AML biology concerning refractoriness to therapies with a specific focus on cytarabine and daunorubicin which still represent milestones agents for inducing therapeutic response and disease eradication. However, failure to achieve complete remission in AML is still high especially in elderly patients (40-60% in patients >65 years old). Several lines of basic and clinical research have been employed to improve the achievement of complete remission. These lines of research include molecular targeted therapy and more recently immunotherapy. In terms of molecular targeted therapies, specific attention is given to DNMT3A and TP53 mutant AML by reviewing the mechanisms underlying epigenetic therapies' (e.g., hypomethylating agents) resistance and providing critical points and hints for possible future therapies overcoming AML refractoriness.
Collapse
Affiliation(s)
- Carmelo Gurnari
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.G.); (S.P.)
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Simona Pagliuca
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.G.); (S.P.)
| | - Valeria Visconte
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.G.); (S.P.)
- Correspondence: ; Tel.: +1-216-445-6895
| |
Collapse
|
49
|
Pérez-López A, Martín-Sabroso C, Torres-Suárez AI, Aparicio-Blanco J. Timeline of Translational Formulation Technologies for Cancer Therapy: Successes, Failures, and Lessons Learned Therefrom. Pharmaceutics 2020; 12:E1028. [PMID: 33126622 PMCID: PMC7692572 DOI: 10.3390/pharmaceutics12111028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
Over the past few decades, the field of cancer therapy has seen a significant change in the way in which formulations are designed and developed, resulting in more efficient products that allow us to ultimately achieve improved drug bioavailability, efficacy, and safety. However, although many formulations have entered the market, many others have fallen by the wayside leaving the scientific community with several lessons to learn. The successes (and failures) achieved with formulations that have been approved in Europe and/or by the FDA for the three major types of cancer therapy (peptide-based therapy, chemotherapy, and radiotherapy) are reviewed herein, covering the period from the approval of the first prolonged-release system for hormonal therapy to the appearance of the first biodegradable microspheres intended for chemoembolization in 2020. In addition, those products that have entered phase III clinical trials that have been active over the last five years are summarized in order to outline future research trends and possibilities that lie ahead to develop clinically translatable formulations for cancer treatment.
Collapse
Affiliation(s)
- Alexandre Pérez-López
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (A.P.-L.); (C.M.-S.); (J.A.-B.)
| | - Cristina Martín-Sabroso
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (A.P.-L.); (C.M.-S.); (J.A.-B.)
- Institute of Industrial Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
| | - Ana Isabel Torres-Suárez
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (A.P.-L.); (C.M.-S.); (J.A.-B.)
- Institute of Industrial Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
| | - Juan Aparicio-Blanco
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain; (A.P.-L.); (C.M.-S.); (J.A.-B.)
- Institute of Industrial Pharmacy, Complutense University of Madrid, 28040 Madrid, Spain
| |
Collapse
|
50
|
A compound combination screening approach with potential to identify new treatment options for paediatric acute myeloid leukaemia. Sci Rep 2020; 10:18514. [PMID: 33116257 PMCID: PMC7595190 DOI: 10.1038/s41598-020-75453-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023] Open
Abstract
Paediatric acute myeloid leukaemia (AML) is a heterogeneous disease characterised by genetics and morphology. The introduction of intensive chemotherapy treatments together with patient stratification and supportive therapy has resulted in a moderate improvement in patient prognosis. However, overall survival rates remain unacceptably poor, with only 65% of patients surviving longer than 5 years. Recently age-specific differences in AML have been identified, highlighting the need for tailored treatments for paediatric patients. Combination therapies have the potential to improve patient prognosis, while minimising harmful side-effects. In the laboratory setting, identifying key combinations from large drug libraries can be resource-intensive, prohibiting discovery and translation into the clinic. To minimise redundancy and maximise discovery, we undertook a multiplex screen of 80 apoptotic-inducing agents in paediatric AML pre-clinical models. The screen was designed using an all-pairs testing algorithm, which ensured that all pairs of compounds could be tested, while minimising the number of wells used. We identified a combination of ABT-737, a Bcl-2 family inhibitor and Purvalanol A, a CDK inhibitor, as a potential targeted therapy for AML patients with an MLL rearrangement and an FLT3-ITD. Our approach has the potential to reduce resource-intensity and time associated with the identification of novel combination therapies.
Collapse
|