1
|
Fang S, Li Y, Wu W, He K, Patil N, Sharma S, A K, Thatoi DN, Mubarakali A. Combining computational and experimental approaches: a novel pH-responsive PVA-stabilized MXene nanocarriers/doxorubicin delivery system with enhanced efficacy for targeted lung cancer therapeutics. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03994-3. [PMID: 40299025 DOI: 10.1007/s00210-025-03994-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/27/2025] [Indexed: 04/30/2025]
Abstract
While advancements have been made in cancer treatment, achieving effective localized therapy remains a significant challenge. Major obstacles include the inefficiency of drug delivery methods and the side effects linked to traditional chemotherapeutics. In this study, we present an innovative delivery system designed to transport doxorubicin (DOX) directly to the lungs. This system employs PVA-stabilized DOX-loaded MXene, aiming to improve targeted delivery and drug efficacy while minimizing toxicity. Our approach represents a promising advancement in the optimization of cancer therapeutics. Using in silico and computational methods, we evaluated the interactions between PVA, DOX, and MXene. Characterization techniques demonstrated that the synthesized PVA@Mxene/DOX exhibited favorable physicochemical properties. We assessed the anticancer potential of PVA@Mxene/DOX through the MTT assay, in vitro migration assay, and apoptosis assay. The findings revealed that the developed anticancer PVA@Mxene/DOX displayed a layered structure with controlled release kinetics. Notably, it significantly reduced cancer cell growth (P < 0.05), induced apoptosis in cancer cells, and inhibited their migration. These results suggest that PVA@Mxene/DOX holds promise as an effective anticancer agent to enhance lung cancer treatment and improve patient care.
Collapse
Affiliation(s)
- Shan Fang
- Department of Respiratory and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277, Jiefang Road, Wuhan City, Hubei Province, 430022, China
| | - Yuan Li
- Department of Respiratory Medicine, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, 030013, China
| | - Wenjuan Wu
- Department of Medical Oncology, The First Affiliated Hospital of Hebei North University, No.36, Changqing District, Zhangjiakou, 075000, China
| | - Kun He
- Department of Emergency, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
| | - Nagaraj Patil
- Department of Mechanical Engineering, School of Engineering and Technology, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Shubham Sharma
- Department of Technical Sciences, Western Caspian University, Baku, Azerbaijan
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
- Jadara University Research Center, Jadara University, Irbid, Jordan
| | - Karthikeyan A
- Department of Mechanical Engineering, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Dhirendra Nath Thatoi
- Department of Mechanical Engineering, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751030, India
| | - Azath Mubarakali
- Department of Informatics and Computer Systems, College of Computer Science, King Khalid University, Abha, Kingdom of Saudi Arabia
| |
Collapse
|
2
|
Abbaspour S, Mohamadzadeh M, Shojaosadati SA. Protein-based nanocarriers for paclitaxel (PTX) delivery in cancer treatment: A review. Int J Biol Macromol 2025; 310:143068. [PMID: 40220831 DOI: 10.1016/j.ijbiomac.2025.143068] [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: 03/01/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
Paclitaxel (PTX) is recognized as one of the most potent chemotherapy agents and is widely used to treat various cancers, including ovarian, lung, breast, head, and neck cancer. Due to the limited solubility and high toxicity of PTX, its use in cancer treatment is challenging and limited. Hence, strategies have been devised to improve the solubility and bioavailability of paclitaxel. In recent years, biocompatible nanocarriers have garnered attention due to their desirable properties, including increased permeability, targeted delivery, extended circulatory half-life, and biological drug delivery for the delivery of chemotherapeutic drugs. Protein nanostructures have been widely studied for the delivery of paclitaxel due to their significant advantages, such as safety, low toxicity, availability, and relatively easy preparation. This review article reviews recent advances in the development of protein-based drug delivery systems for loading and releasing paclitaxel. These nanocarriers have great potential to improve paclitaxel's antitumor properties and efficacy. Therefore, in the future, the integration of the pharmaceutical industry and artificial intelligence techniques will provide more opportunities for research and development in the pharmaceutical field.
Collapse
Affiliation(s)
- Sakineh Abbaspour
- Biotechnology Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | | | - Seyed Abbas Shojaosadati
- Biotechnology Department, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
3
|
Qiao JX, Guo DY, Tian H, Wang ZP, Fan QQ, Tian Y, Sun J, Zhang XF, Zou JB, Cheng JX, Luan F, Zhai BT. Research progress of paclitaxel nanodrug delivery system in the treatment of triple-negative breast cancer. Mater Today Bio 2024; 29:101358. [PMID: 39677523 PMCID: PMC11638641 DOI: 10.1016/j.mtbio.2024.101358] [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: 07/16/2024] [Revised: 10/27/2024] [Accepted: 11/21/2024] [Indexed: 12/17/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, characterized by the loss or low expression of estrogen receptor (ER), human epidermal growth factor receptor 2 (HER2) and progesterone receptor (PR). Due to the lack of clear therapeutic targets, paclitaxel (PTX) is often used as a first-line standard chemotherapy drug for the treatment of high-risk and locally advanced TNBC. PTX is a diterpenoid alkaloid extracted and purified from Taxus plants, functioning as an anticancer agent by inducing and promoting tubulin polymerization, inhibiting spindle formation in cancer cells, and preventing mitosis. However, its clinical application is limited by low solubility and high toxicity. Nanodrug delivery system (NDDS) is one of the feasible methods to improve the water solubility of PTX and reduce side effects. In this review, we summarize the latest advancements in PTX-targeted NDDS, as well as its combination with other codelivery therapies for TNBC treatment. NDDS includes passive targeting, active targeting, stimuli-responsive, codelivery, and multimode strategies. These systems have good prospects in improving the bioavailability of PTX, enhancing tumor targeting, reducing toxicity, controlling drug release, and reverse tumor multidrug resistance (MDR). This review provides valuable insights into the clinical development and application of PTX-targeted NDDS in the treatment of TNBC.
Collapse
Affiliation(s)
- Jia-xin Qiao
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Dong-yan Guo
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Huan Tian
- Department of Pharmacy, National Old Pharmacist Inheritance Studio, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, China
| | - Zhan-peng Wang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Qiang-qiang Fan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Yuan Tian
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jing Sun
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Xiao-fei Zhang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jun-bo Zou
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jiang-xue Cheng
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Fei Luan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Bing-tao Zhai
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| |
Collapse
|
4
|
Wenger V, Zeiser R. Editorial: Current concepts of cellular and biological drugs to modulate regulatory T cell activity in the clinic, volume II. Front Immunol 2023; 14:1221904. [PMID: 37383231 PMCID: PMC10294709 DOI: 10.3389/fimmu.2023.1221904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 06/01/2023] [Indexed: 06/30/2023] Open
Affiliation(s)
- Valentin Wenger
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signaling Research Centre for Biological Signalling Studies (BIOSS) Freiburg and CIBSS – Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
5
|
Wenhao Zhou, Hu H, Wang T. Study on Modification of Paclitaxel and Its Antitumor Preparation. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2023. [DOI: 10.1134/s1068162023020255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
|
6
|
Gong L, Zhou H, Zhang S, Wang C, Fu K, Ma C, Zhang Y, Peng C, Li Y. CD44-Targeting Drug Delivery System of Exosomes Loading Forsythiaside A Combats Liver Fibrosis via Regulating NLRP3-Mediated Pyroptosis. Adv Healthc Mater 2023; 12:e2202228. [PMID: 36603210 DOI: 10.1002/adhm.202202228] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/26/2022] [Indexed: 01/06/2023]
Abstract
Liver fibrosis is a progressive pathological process induced by various stimuli and may progress to liver cirrhosis and cancer. Forsythiaside A (FA) is an active ingredient extracted from traditional Chinese medicine Forsythiae Fructus and has prominent hepatoprotective activities. However, the unsatisfactory pharmacokinetic properties restrict its clinical application. In this study, the nanocarrier of CD44-specific ligand Hyaluronic acid (HA)-modified milk-derived exosomes (mExo) encapsulated with FA (HA-mExo-FA) is developed. As a result, HA modification could deliver drug-loaded exosomes to the target cells and form a specific ligand-receptor interaction with CD44, thus improving the anti-liver fibrosis effect of FA. In vitro findings indicate that HA-mExo-FA could inhibit TGF-β1-induced LX2 cell proliferation, reduce α-SMA and collagen gene and protein levels, and promote the apoptosis of activated LX2 cells. In vivo results demonstrate that HA-mExo-FA could improve liver morphology and function changes in zebrafish larvae. The anti-liver fibrosis mechanism of HA-mExo-FA may be attributed to the inhibition of NLRP3-mediated pyroptosis. In addition, the effect of HA-mExo-FA on TAA-induced increase in NLRP3 production is attenuated by NLRP3 inhibitor MCC950. Collectively, this study demonstrates the promising application of HA-mExo-FA in drug delivery with high specificity and provides a powerful and novel delivery platform for liver fibrosis therapy.
Collapse
Affiliation(s)
- Lihong Gong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Honglin Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Shenglin Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Cheng Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ke Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Cheng Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yafang Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| |
Collapse
|
7
|
Kwon SH, Faruque HA, Kee H, Kim E, Park S. Exosome-based hybrid nanostructures for enhanced tumor targeting and hyperthermia therapy. Colloids Surf B Biointerfaces 2021; 205:111915. [PMID: 34130212 DOI: 10.1016/j.colsurfb.2021.111915] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/29/2021] [Accepted: 06/06/2021] [Indexed: 02/06/2023]
Abstract
Recently, natural exosomes have attracted attention as an ideal drug carrier to overcome the limitations of existing drug delivery systems which are toxicity induction and low cancer-targeting performance. In this study, we propose an exosome-based hybrid nanostructure (EHN) with improved targeting ability and therapeutic efficacy against colorectal cancer by using exosomes isolated from the tumor cell line as a drug carrier. The proposed EHN can have high biocompatibility by using exosomes, a biologically derived material, and show improved targeting performance by adding a tumor-targeting ligand (folic acid). In addition, the proposed EHN is capable of chemotherapy because doxorubicin, an anticancer drug, is encapsulated by the exosome with high efficiency, and it can induce hyperthermia therapy because of the magnetic nanoparticles (MNPs) attached to the surface of exosomes. Through in vitro and in vivo experiments using a xenograft tumor mouse model, it was confirmed that the proposed EHN could exhibit increased apoptosis and excellent tumor growth inhibition ability. Therefore, the proposed EHN is expected to overcome the limitations of existing drug delivery systems and be utilized as an effective drug delivery system in cancer treatment.
Collapse
Affiliation(s)
- Su-Hyun Kwon
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea; Department of Chemistry, College of Science, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, Republic of Korea
| | - Hasan Al Faruque
- Companion Diagnostics and Medical Technology Research Group, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Hyeonwoo Kee
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea
| | - Eunjoo Kim
- Companion Diagnostics and Medical Technology Research Group, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea.
| | - Sukho Park
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, 42988, Republic of Korea.
| |
Collapse
|
8
|
Extracellular Vesicles as an Efficient and Versatile System for Drug Delivery. Cells 2020; 9:cells9102191. [PMID: 33003285 PMCID: PMC7600121 DOI: 10.3390/cells9102191] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/24/2020] [Accepted: 05/30/2020] [Indexed: 12/12/2022] Open
Abstract
Despite the recent advances in drug development, the majority of novel therapeutics have not been successfully translated into clinical applications. One of the major factors hindering their clinical translation is the lack of a safe, non-immunogenic delivery system with high target specificity upon systemic administration. In this respect, extracellular vesicles (EVs), as natural carriers of bioactive cargo, have emerged as a promising solution and can be further modified to improve their therapeutic efficacy. In this review, we provide an overview of the biogenesis pathways, biochemical features, and isolation methods of EVs with an emphasis on their many intrinsic properties that make them desirable as drug carriers. We then describe in detail the current advances in EV therapeutics, focusing on how EVs can be engineered to achieve improved target specificity, better circulation kinetics, and efficient encapsulation of therapeutic payloads. We also identify the challenges and obstacles ahead for clinical translation and provide an outlook on the future perspective of EV-based therapeutics.
Collapse
|
9
|
Ciruelos E, Apellániz-Ruiz M, Cantos B, Martinez-Jáñez N, Bueno-Muiño C, Echarri MJ, Enrech S, Guerra JA, Manso L, Pascual T, Dominguez C, Gonzalo JF, Sanz JL, Rodriguez-Antona C, Sepúlveda JM. A Pilot, Phase II, Randomized, Open-Label Clinical Trial Comparing the Neurotoxicity of Three Dose Regimens of Nab-Paclitaxel to That of Solvent-Based Paclitaxel as the First-Line Treatment for Patients with Human Epidermal Growth Factor Receptor Type 2-Negative Metastatic Breast Cancer. Oncologist 2019; 24:e1024-e1033. [PMID: 31023863 DOI: 10.1634/theoncologist.2017-0664] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/28/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND This study aimed to characterize the neurotoxicity of three different regimens of nab-paclitaxel compared with a standard regimen of solvent-based (sb) paclitaxel for the first-line treatment of HER2-negative metastatic breast cancer based on the Total Neurotoxicity Score (TNS), a tool specifically developed to assess chemotherapy-induced neurotoxicity. MATERIALS AND METHODS This was a randomized, open-label study testing 4-week cycles of 80 mg/m2 sb-paclitaxel (PACL80/w) on days 1, 8, and 15; 100 mg/m2 nab-paclitaxel on days 1, 8, and 15 (NAB100/w); 150 mg/m2 nab-paclitaxel on days 1, 8, and 15 (NAB150/w); and 150 mg/m2 nab-paclitaxel on days 1 and 15 (NAB150/2w). In addition to the TNS, neuropathy was assessed using the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE). Tumor response and quality of life were also evaluated. RESULTS Neurotoxicity, as evaluated by the TNS, did not significantly differ between the sb-paclitaxel group and any of the nab-paclitaxel groups. The frequency of (any grade) polyneuropathy, as measured by the NCI-CTCAE, was lower in the PACL80/w (n = 7, 50%) and NAB150/2w (n = 10, 62.5%) groups than in the NAB100/w (n = 13, 81.3%) or NAB150/w (n = 11, 78.6%) group. Although the differences were not statistically significant, compared with the other groups, in the NAB150/w group, the time to occurrence of grade ≥2 polyneuropathy was shorter, and the median time to recovery from grade ≥2 polyneuropathy was longer. Dose delays and reductions due to neurotoxicity and impact of neurotoxicity on the patients' experience of symptoms and functional limitations was greater with NAB150/w. Among the seven polymorphisms selected for genotyping, the variant alleles of EPHA5-rs7349683, EPHA6-rs301927, and EPHA8-rs209709 were associated with an increased risk of paclitaxel-induced neuropathy. CONCLUSION The results of this exploratory study showed that, regardless of the dose, nab-paclitaxel did not differ from sb-paclitaxel in terms of neurotoxicity as evaluated with the TNS. However, results from NCI-CTCAE, dose delays and reductions, and functional tools consistently indicate that NAB150/w regimen is associated with a greater risk of chemotherapy-induced neuropathy. Thus, our results question the superiority of the TNS over NCI-CTCAE for evaluating chemotherapy-induced neuropathy and guiding treatment decisions in this context. The selection of the nab-paclitaxel regimen should be individualized based on the clinical context and potentially supported by pharmacogenetic analysis. Registry: EudraCT, 2012-002361-36; NCT01763710 IMPLICATIONS FOR PRACTICE: The results of this study call into question the superiority of the Total Neurotoxicity Score over the National Cancer Institute Common Terminology Criteria for Adverse Events for evaluating chemotherapy-induced neuropathy and guiding treatment decisions in this context and suggest that a regimen of 150 mg/m2 nab-paclitaxel administered on days 1, 8, and 15 is associated with a greater risk of chemotherapy-induced neuropathy and hematological toxicity compared with other lower-dose nab-paclitaxel regimens or a standard regimen of solvent-based paclitaxel. The selection of the nab-paclitaxel regimen should be individualized based on the clinical context and could benefit from pharmacogenetics analysis.
Collapse
Affiliation(s)
- Eva Ciruelos
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
- Oncosur Study Group, Madrid, Spain
| | - María Apellániz-Ruiz
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Blanca Cantos
- Oncosur Study Group, Madrid, Spain
- Medical Oncology Department, Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| | - Noelia Martinez-Jáñez
- Oncosur Study Group, Madrid, Spain
- Medical Oncology Department, Hospital Ramón y Cajal, Madrid, Spain
| | - Coralia Bueno-Muiño
- Oncosur Study Group, Madrid, Spain
- Medical Oncology Department, Hospital Infanta Cristina, Parla, Spain
| | - Maria-Jose Echarri
- Oncosur Study Group, Madrid, Spain
- Medical Oncology Department, Hospital Severo Ochoa, Leganes, Spain
| | - Santos Enrech
- Oncosur Study Group, Madrid, Spain
- Medical Oncology Department, Hospital Universitario de Getafe, Getafe, Spain
| | - Juan-Antonio Guerra
- Oncosur Study Group, Madrid, Spain
- Medical Oncology Department, Hospital de Fuenlabrada, Fuenlabrada, Spain
| | - Luis Manso
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
- Oncosur Study Group, Madrid, Spain
| | - Tomas Pascual
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
- Oncosur Study Group, Madrid, Spain
| | - Cristina Dominguez
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
- Oncosur Study Group, Madrid, Spain
| | - Juan-Francisco Gonzalo
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
- Oncosur Study Group, Madrid, Spain
| | - Juan-Luis Sanz
- Clinical Research Department, Apoyo a la Investigación Clínica en España (APICES), Madrid, Spain
| | - Cristina Rodriguez-Antona
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- Neurology Division, Neuromuscular Unit, ISCIII Center for Biomedical Research on Rare Diseases (CIBERER), Madrid, Spain
| | - Juan-Manuel Sepúlveda
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
- Oncosur Study Group, Madrid, Spain
| |
Collapse
|
10
|
Nanomedicines: The magic bullets reaching their target? Eur J Pharm Sci 2018; 128:73-80. [PMID: 30465818 DOI: 10.1016/j.ejps.2018.11.019] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/29/2018] [Accepted: 11/18/2018] [Indexed: 12/11/2022]
Abstract
Nanomedicines, since the approval of the first one in the 1950s, have been accompanied by expectations of higher efficiency and efficacy, compared to less complex drugs. The fulfilment of those expectations has been slower than anticipated, due to the high complexity of nanomedicine drugs combined with a lack of scientific understanding of nanomedicine interactions with biological systems. The unique properties of their size and their surface composition create difficulties in their physicochemical characterization, and as a consequence, difficulty in assessing the similarity of follow-on products (nanosimilars) to originator nanomedicines. During the 2018 European Federation for Pharmaceutical Sciences (EUFEPS) annual meeting "Crossing the barrier for future medicines" in Athens, there were several sessions on nanomedicines organised by the EUFEPS Nanomedicine Network. This review focuses on the session "Nanomedicines and nanosimilars: how to assess similar?", discussing the nature of nanomedicines, the regulatory aspects of the topic and the impact of practical use and handling of such medicinal products. Emphasis is put on the consequences their nanosize-related properties have on the establishment of their critical quality attributes and how this affects the demonstration of bioequivalence of nanosimilars to their originator products. The lack of an appropriate and harmonized regulatory evaluation procedure and the absence of corresponding education are also discussed, especially the uncertainty surrounding the practical use of nanosimilars, including the higher healthcare cost due to less than satisfactory number of safe and efficacious nanosimilars in the market.
Collapse
|
11
|
Schettini F, Giuliano M, De Placido S, Arpino G. Nab-paclitaxel for the treatment of triple-negative breast cancer: Rationale, clinical data and future perspectives. Cancer Treat Rev 2016; 50:129-141. [PMID: 27665540 DOI: 10.1016/j.ctrv.2016.09.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/14/2016] [Accepted: 09/01/2016] [Indexed: 01/05/2023]
Abstract
Triple-negative breast cancer (TNBC) accounts for ∼10-20% of breast cancers and is associated with relatively poor prognosis, earlier disease recurrence and higher number of visceral metastases. Despite an increasing understanding of the molecular heterogeneity of TNBC, clinical trials of targeted agents have thus far been disappointing; chemotherapy, in particular with anthracycline and taxanes, remains the backbone medical management for both early and metastatic TNBC. Nab-paclitaxel is a solvent-free, albumin-bound, nanoparticle formulation of paclitaxel and represents a novel formulation of an established, effective chemotherapeutic agent. Nab-paclitaxel has been specifically designed to overcome the limitations of conventional taxane formulations, including the barriers to effective drug delivery of highly lipophilic agents. It has shown significant efficacy and better tolerability than conventional taxanes in metastatic breast cancer and is approved for use in this setting. Increasing evidence suggests that nab-paclitaxel is effective in patients with more aggressive tumours, as seen in TNBC. Indeed, results of Phase II/III studies indicate that nab-paclitaxel may be effective as neoadjuvant treatment of TNBC. This article reviews the rationale and evidence supporting a role for nab-paclitaxel in the treatment of TNBC, including ongoing studies such as ADAPT-TN and tnAcity. In addition, the article reviews ongoing research into targeted therapies and immuno-oncology for the treatment of TNBC, and explores the potential role, current evidence and ongoing studies of nab-paclitaxel as the chemotherapy partner in combination with immunotherapy, where the unique properties of this taxane, including the lack of requirement for steroid pre-medication, may present an advantage.
Collapse
Affiliation(s)
- Francesco Schettini
- Medical Oncology, Department of Clinical and Surgical Medicine, University of Naples Federico II, Pansini 5, 80131 Naples, Italy.
| | - Mario Giuliano
- Medical Oncology, Department of Clinical and Surgical Medicine, University of Naples Federico II, Pansini 5, 80131 Naples, Italy; Lester and Sue Smith Breast Center, Baylor College of Medicine, 1 Baylor Plaza, 77030 Houston, TX, USA.
| | - Sabino De Placido
- Medical Oncology, Department of Clinical and Surgical Medicine, University of Naples Federico II, Pansini 5, 80131 Naples, Italy.
| | - Grazia Arpino
- Medical Oncology, Department of Clinical and Surgical Medicine, University of Naples Federico II, Pansini 5, 80131 Naples, Italy.
| |
Collapse
|
12
|
Palumbo R, Sottotetti F, Bernardo A. Targeted chemotherapy with nanoparticle albumin-bound paclitaxel (nab-paclitaxel) in metastatic breast cancer: which benefit for which patients? Ther Adv Med Oncol 2016; 8:209-29. [PMID: 27239239 DOI: 10.1177/1758834016639873] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The therapeutic goals in metastatic breast cancer (MBC) remain palliative in nature, aimed at controlling symptoms, improving or maintaining quality of life and prolonging survival. The advent of new drugs and new formulations of standard agents has led to better outcomes in patients with advanced or metastatic disease. These developments have also allowed a tailored therapeutic approach, in which the molecular biology of the tumour, the treatment history, and patient attitudes are taken into account in the decision-making process. Targeting drug delivery to the tumour is a promising mean of increasing the therapeutic index of highly active agents such as the taxanes, and nanoparticle albumin-bound paclitaxel (nab-paclitaxel), the first nanotechnology-based drug developed in cancer treatment, is one such advance. Data from randomized trials support the efficacy of single-agent nab-paclitaxel as first-line and further treatment lines in MBC at the registered 3-weekly schedule of 260 mg/m(2), but emerging evidence suggests its activity as a weekly regimen or combined with other agents in various clinical scenarios. Thus, nab-paclitaxel seems to offer flexibility in terms of dosing schedules, allowing physicians to tailor the dose according to different clinical situations. This paper reviews the clinical trial background for nab-paclitaxel in MBC, focusing on specific 'difficult-to-treat' patient populations, such as taxane-pretreated or elderly women, as well as those with triple-negative, HER2-positive and poor-prognostic-factors disease. Moving beyond evidence-based information, 'real life' available experiences are also discussed with the aim of providing an update for daily clinical practice.
Collapse
Affiliation(s)
- Raffaella Palumbo
- Departmental Unit of Oncology, Fondazione Salvatore Maugeri, Via Maugeri 10, 27100 Pavia, Italy
| | - Federico Sottotetti
- Departmental Unit of Oncology, IRCCS-Fondazione Salvatore Maugeri, Pavia, Italy
| | - Antonio Bernardo
- Departmental Unit of Oncology, IRCCS-Fondazione Salvatore Maugeri, Pavia, Italy
| |
Collapse
|
13
|
Abstract
The optimal sequence of systemic chemotherapy in metastatic breast cancer (MBC) is unknown. We report the case of a woman who was successfully treated with nanoparticle albumin-bound (nab)-paclitaxel for triple negative MBC in our institution. In November 2008, a 48-year-old woman underwent surgical treatment for a triple negative invasive ductal breast cancer and subsequently received adjuvant chemotherapy with fluorouracil/epirubicin/cyclophosphamide and radiotherapy. Sixteen months after surgery, she presented with a left chest wall metastatasis. The patient received combination therapy with conventional paclitaxel (90 mg/m² weekly for 3 out of 4 weeks [QW 3/4]) and bevacizumab (10 mg/kg every 2 weeks [Q2W]) as first-line treatment for MBC (six cycles; March to September 2010) and achieved a partial response at the metastatic site. Bevacizumab monotherapy was continued until disease progression (April 2011) with the development of a single infraclavicular lymph node metastasis and an increase in the dimensions of the left chest wall lesion. From May to December 2011, the patient received nab-paclitaxel 260 mg/m² every 3 weeks (Q3W) as second-line treatment (11 cycles). After three cycles, the left chest wall lesion and the infraclavicular lymph node metastasis were undetectable and the patient was considered to have achieved a complete response. Treatment was well tolerated with no significant toxicity or need for dose reduction. Given our case, here we review the clinical evidence and discuss the potential role of nab-paclitaxel for the treatment of triple negative MBC, a subgroup typically characterized as having aggressive disease and limited treatment options.
Collapse
|
14
|
Munagala R, Aqil F, Jeyabalan J, Gupta RC. Bovine milk-derived exosomes for drug delivery. Cancer Lett 2015; 371:48-61. [PMID: 26604130 DOI: 10.1016/j.canlet.2015.10.020] [Citation(s) in RCA: 668] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/21/2015] [Accepted: 10/21/2015] [Indexed: 12/20/2022]
Abstract
Exosomes are biological nanovesicles that are involved in cell-cell communication via the functionally-active cargo (such as miRNA, mRNA, DNA and proteins). Because of their nanosize, exosomes are explored as nanodevices for the development of new therapeutic applications. However, bulk, safe and cost-effective production of exosomes is not available. Here, we show that bovine milk can serve as a scalable source of exosomes that can act as a carrier for chemotherapeutic/chemopreventive agents. Drug-loaded exosomes showed significantly higher efficacy compared to free drug in cell culture studies and against lung tumor xenografts in vivo. Moreover, tumor targeting ligands such as folate increased cancer-cell targeting of the exosomes resulting in enhanced tumor reduction. Milk exosomes exhibited cross-species tolerance with no adverse immune and inflammatory response. Thus, we show the versatility of milk exosomes with respect to the cargo it can carry and ability to achieve tumor targetability. This is the first report to identify a biocompatible and cost-effective means of exosomes to enhance oral bioavailability, improve efficacy and safety of drugs.
Collapse
Affiliation(s)
- Radha Munagala
- Department of Medicine, University of Louisville, Louisville, KY 40202.,James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202
| | - Farrukh Aqil
- Department of Medicine, University of Louisville, Louisville, KY 40202.,James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202
| | | | - Ramesh C Gupta
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202.,Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY 40202
| |
Collapse
|
15
|
Poon W, Heinmiller A, Zhang X, Nadeau JL. Determination of biodistribution of ultrasmall, near-infrared emitting gold nanoparticles by photoacoustic and fluorescence imaging. JOURNAL OF BIOMEDICAL OPTICS 2015; 20:066007. [PMID: 26102572 DOI: 10.1117/1.jbo.20.6.066007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/28/2015] [Indexed: 06/04/2023]
Abstract
This study compares fluorescence and photoacoustic (PA) imaging of ex vivo tumors and organs from tumor-bearing mice injected intravenously with ultrasmall (<3 nm ) tiopronin-capped Au nanoparticles and compares the data with inductively coupled plasma mass spectrometry (ICP-MS). Good agreement is seen in particle distributions and concentrations at the organ level. The spatial resolution from the imaging techniques allows for localization of the particles within organ structures. Although the particles do not have a plasmon peak, their absorbance in the near-infrared (NIR) is sufficient for PA excitation. PA imaging shows an increase of signal as particle concentrations increase, with changes in spectrum if particles aggregate. Fluorescence imaging using the particles’ native NIR emission shows agreement in general intensity in each organ, though quenching of emission can be seen at very high concentrations. Both of these imaging techniques are noninvasive and labor-saving alternatives to organ digestion and ICP-MS and may provide insight into cellular distribution of particles. The simple construct avoids the use of toxic semiconductor materials or dyes, relying upon the gold itself for both the fluorescence and PA signal. This provides a useful alternative to more complex approaches to multimodal imaging and one that is readily translatable to the clinic.
Collapse
Affiliation(s)
- Wilson Poon
- McGill University, Department of Biomedical Engineering, 3775 University Street, Montréal, Quebec H3A 2B4, CanadabUniversity of Toronto, Institute of Biomaterials and Biomedical Engineering, 160 College Street, Toronto, Ontario M5S 3E1, Canada
| | - Andrew Heinmiller
- VisualSonics Inc., 6100-3080 Yonge Street, Toronto, Ontario M4N 3N1, Canada
| | - Xuan Zhang
- McGill University, Department of Biomedical Engineering, 3775 University Street, Montréal, Quebec H3A 2B4, Canada
| | - Jay L Nadeau
- McGill University, Department of Biomedical Engineering, 3775 University Street, Montréal, Quebec H3A 2B4, Canada
| |
Collapse
|
16
|
Palumbo R, Sottotetti F, Trifirò G, Piazza E, Ferzi A, Gambaro A, Spinapolice EG, Pozzi E, Tagliaferri B, Teragni C, Bernardo A. Nanoparticle albumin-bound paclitaxel (nab-paclitaxel) as second-line chemotherapy in HER2-negative, taxane-pretreated metastatic breast cancer patients: prospective evaluation of activity, safety, and quality of life. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:2189-99. [PMID: 25931813 PMCID: PMC4404936 DOI: 10.2147/dddt.s79563] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND A prospective, multicenter trial was undertaken to assess the activity, safety, and quality of life of nanoparticle albumin-bound paclitaxel (nab-paclitaxel) as second-line chemotherapy in HER2-negative, taxane-pretreated metastatic breast cancer (MBC). PATIENTS AND METHODS Fifty-two women with HER2-negative MBC who were candidates for second-line chemotherapy for the metastatic disease were enrolled and treated at three centers in Northern Italy. All patients had previously received taxane-based chemotherapy in the adjuvant or first-line metastatic setting. Single-agent nab-paclitaxel was given at the dose of 260 mg/m(2) as a 30-minute intravenous infusion on day 1 each treatment cycle, which lasted 3 weeks, in the outpatient setting. No steroid or antihistamine premedication was provided. Treatment was stopped for documented disease progression, unacceptable toxicity, or patient refusal. RESULTS All of the enrolled patients were evaluable for the study endpoints. The objective response rate was 48% (95% CI, 31.5%-61.3%) and included complete responses from 13.5%. Disease stabilization was obtained in 19 patients and lasted >6 months in 15 of them; the overall clinical benefit rate was 77%. The median time to response was 70 days (range 52-86 days). The median progression-free survival time was 8.9 months (95% CI, 8.0-11.6 months, range 5-21+ months). The median overall survival point has not yet been reached. Toxicities were expected and manageable with good patient compliance and preserved quality of life in patients given long-term treatment. CONCLUSION Our results showed that single-agent nab-paclitaxel 260 mg/m(2) every 3 weeks is an effective and well tolerated regimen as second-line chemotherapy in HER2-negative, taxane-pretreated MBC patients, and that it produced interesting values of objective response rate and progression-free survival without the concern of significant toxicity. Specifically, the present study shows that such a regimen is a valid therapeutic option for that 'difficult to treat' patient population represented by women who at the time of disease relapse have already received the most active agents in the adjuvant and/or metastatic setting (ie, conventional taxanes).
Collapse
Affiliation(s)
- Raffaella Palumbo
- Departmental Unit of Oncology, IRCCS Fondazione Salvatore Maugeri, Pavia, Italy
| | - Federico Sottotetti
- Departmental Unit of Oncology, IRCCS Fondazione Salvatore Maugeri, Pavia, Italy
| | - Giuseppe Trifirò
- Unit of Nuclear Medicine, IRCCS Fondazione Salvatore Maugeri, Pavia, Italy
| | - Elena Piazza
- Medical Oncology Luigi Sacco Hospital, Milano, Italy
| | | | - Anna Gambaro
- Medical Oncology Luigi Sacco Hospital, Milano, Italy
| | | | - Emma Pozzi
- Departmental Unit of Oncology, IRCCS Fondazione Salvatore Maugeri, Pavia, Italy
| | - Barbara Tagliaferri
- Departmental Unit of Oncology, IRCCS Fondazione Salvatore Maugeri, Pavia, Italy
| | - Cristina Teragni
- Departmental Unit of Oncology, IRCCS Fondazione Salvatore Maugeri, Pavia, Italy
| | - Antonio Bernardo
- Departmental Unit of Oncology, IRCCS Fondazione Salvatore Maugeri, Pavia, Italy
| |
Collapse
|