1
|
Yu J, Xu X, Griffin JI, Mu Q, Ho RJY. Drug Combination Nanoparticles Containing Gemcitabine and Paclitaxel Enable Orthotopic 4T1 Breast Tumor Regression. Cancers (Basel) 2024; 16:2792. [PMID: 39199565 PMCID: PMC11352501 DOI: 10.3390/cancers16162792] [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: 05/24/2024] [Revised: 07/25/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024] Open
Abstract
Early diagnosis, intervention, and therapeutic advancements have extended the lives of breast cancer patients; however, even with molecularly targeted therapies, many patients eventually progress to metastatic cancer. Recent data suggest that residual breast cancer cells often reside in the lymphatic system before rapidly spreading through the bloodstream. To address this challenge, an effective drug combination composed of gemcitabine (G) and paclitaxel (T) is administered intravenously in sequence at the metastatic stage, but intravenous GT infusion may limit lymphatic GT drug accessibility and asynchronous drug exposure in cancer cells within the lymph. To determine whether co-localization of intracellular gemcitabine and paclitaxel (referred to as GT) could overcome these limitations and enhance the efficacy of GT, we have evaluated a previously reported GT drug-combination formulated in nanoparticle (referred to as GT-in-DcNP) evaluated in an orthotopic breast tumor model. Previously, with indocyanine green-labeled nanoparticles, we reported that GT-in-DcNP particles after subcutaneous dosing were taken up rapidly and preferentially into the lymph instead of blood vessels. The pharmacokinetic study showed enhanced co-localization of GT within the tumors and likely through lymphatic access, before drug apparency in the plasma leading to apparent long-acting plasma time-course. The mechanisms may be related to significantly greater inhibitions of tumor growth-by 100 to 140 times-in both sub-iliac and axillary regions compared to the equivalent dosing with free-and-soluble GT formulation. Furthermore, GT-in-DcNP exhibited dose-dependent effects with significant tumor regression. In contrast, even at the highest dose of free GT combination, only a modest tumor growth reduction was notable. Preliminary studies with MDA-231-HM human breast cancer in an orthotopic xenograft model indicated that GT-in-DcNP may be effective in suppressing human breast tumor growth. Taken together, the synchronized delivery of GT-in-DcNP to mammary tumors through the lymphatic system offers enhanced cellular retention and greater efficacy.
Collapse
Affiliation(s)
- Jesse Yu
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA; (J.Y.); (X.X.)
| | - Xiaolin Xu
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA; (J.Y.); (X.X.)
| | - James Ian Griffin
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA; (J.Y.); (X.X.)
| | - Qingxin Mu
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA; (J.Y.); (X.X.)
| | - Rodney J. Y. Ho
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA; (J.Y.); (X.X.)
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
2
|
Laxane N, Yadav KS. QbD-based co-loading of paclitaxel and imatinib mesylate by protamine-coated PLGA nanoparticles effective on breast cancer cells. Nanomedicine (Lond) 2024; 19:2211-2227. [PMID: 38934510 PMCID: PMC11486237 DOI: 10.1080/17435889.2024.2353557] [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: 01/22/2024] [Accepted: 05/07/2024] [Indexed: 06/28/2024] Open
Abstract
Aim: Paclitaxel and imatinib mesylate are drugs used in the treatment of breast cancer. Conventional drug-delivery systems have limitations in the effective treatment of breast cancer using the drugs.Materials & methods: Combination index studies were used to identify the optimum ratio of both drugs showing maximum synergistic effect. Using a systematic quality-by-design approach, protamine-coated PLGA nanoparticles co-loaded with paclitaxel and imatinib mesylate were formulated. Further characterization and cell line evaluations were performed.Results: Encapsulation efficiency obtained was 92.54% for paclitaxel and 75.12% for imatinib mesylate. A sustained (24 h) and controlled zero-order drug release was obtained.Conclusion: Formulated nanoparticles had a low IC50 value and enhanced cellular uptake.
Collapse
Affiliation(s)
- Neha Laxane
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's, NMIMS Deemed to be University, Mumbai, 400056, India
| | - Khushwant S Yadav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's, NMIMS Deemed to be University, Mumbai, 400056, India
| |
Collapse
|
3
|
Boggio E, Gigliotti CL, Stoppa I, Pantham D, Sacchetti S, Rolla R, Grattarola M, Monge C, Pizzimenti S, Dianzani U, Dianzani C, Battaglia L. Exploiting Nanomedicine for Cancer Polychemotherapy: Recent Advances and Clinical Applications. Pharmaceutics 2023; 15:937. [PMID: 36986798 PMCID: PMC10057931 DOI: 10.3390/pharmaceutics15030937] [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: 01/20/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023] Open
Abstract
The most important limitations of chemotherapeutic agents are severe side effects and the development of multi-drug resistance. Recently, the clinical successes achieved with immunotherapy have revolutionized the treatment of several advanced-stage malignancies, but most patients do not respond and many of them develop immune-related adverse events. Loading synergistic combinations of different anti-tumor drugs in nanocarriers may enhance their efficacy and reduce life-threatening toxicities. Thereafter, nanomedicines may synergize with pharmacological, immunological, and physical combined treatments, and should be increasingly integrated in multimodal combination therapy regimens. The goal of this manuscript is to provide better understanding and key considerations for developing new combined nanomedicines and nanotheranostics. We will clarify the potential of combined nanomedicine strategies that are designed to target different steps of the cancer growth as well as its microenvironment and immunity interactions. Moreover, we will describe relevant experiments in animal models and discuss issues raised by translation in the human setting.
Collapse
Affiliation(s)
- Elena Boggio
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Casimiro Luca Gigliotti
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Ian Stoppa
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Deepika Pantham
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Sara Sacchetti
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, 28100 Novara, Italy
- Ospedale Universitario Maggiore della Carità, 28100 Novara, Italy
| | - Roberta Rolla
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, 28100 Novara, Italy
- Ospedale Universitario Maggiore della Carità, 28100 Novara, Italy
| | - Margherita Grattarola
- Dipartimento di Scienze Cliniche e Biologiche, Università degli Studi di Torino, Corso Raffaello 30, 10125 Torino, Italy
| | - Chiara Monge
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, 10125 Torino, Italy
| | - Stefania Pizzimenti
- Dipartimento di Scienze Cliniche e Biologiche, Università degli Studi di Torino, Corso Raffaello 30, 10125 Torino, Italy
| | - Umberto Dianzani
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, 28100 Novara, Italy
- Ospedale Universitario Maggiore della Carità, 28100 Novara, Italy
| | - Chiara Dianzani
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, 10125 Torino, Italy
| | - Luigi Battaglia
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, 10125 Torino, Italy
- Centro Interdipartimentale Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, Università degli Studi di Torino, 10124 Torino, Italy
| |
Collapse
|
4
|
Yu J, Mu Q, Fung M, Xu X, Zhu L, Ho RJY. Challenges and opportunities in metastatic breast cancer treatments: Nano-drug combinations delivered preferentially to metastatic cells may enhance therapeutic response. Pharmacol Ther 2022; 236:108108. [PMID: 34999182 PMCID: PMC9256851 DOI: 10.1016/j.pharmthera.2022.108108] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/12/2021] [Accepted: 01/03/2022] [Indexed: 02/07/2023]
Abstract
Despite advances in breast cancer treatments and related 5-year survival outcomes, metastatic breast cancer cures remain elusive. The current standard of care includes a combination of surgery, radiation therapy and drug therapy. However, even the most advanced procedures and treatments do not prevent breast cancer recurrence and metastasis. Once metastasis occurs, patient prognosis is poor. Recent elucidation of the spatiotemporal transit of metastatic cancer cells from primary tumor sites to distant sites provide an opportunity to integrate knowledge of drug disposition in our effort to enhance drug localization and exposure in cancer laden tissues . Novel technologies have been developed, but could be further refined to facilitate the distribution of drugs to target cancer cells and tissues. The purpose of this review is to highlight the challenges in metastatic breast cancer treatment and focus on novel drug combination and nanotechnology approaches to overcome the challenges. With improved definition of metastatic tissue target, directed localization and retention of multiple, pharmacologically active drugs to tissues and cells of interest may overcome the limitations in breast cancer treatment that may lead to a cure for breast cancer.
Collapse
Affiliation(s)
- Jesse Yu
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA
| | - Qingxin Mu
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA
| | - Millie Fung
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA
| | - Xiaolin Xu
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA
| | - Linxi Zhu
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA
| | - Rodney J Y Ho
- Department of Pharmaceutics, University of Washington, Seattle, WA 98195, USA; Department of Bioengineering, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
5
|
Behl A, Sarwalia P, Kumar S, Behera C, Mintoo MJ, Datta TK, Gupta PN, Chhillar AK. Codelivery of Gemcitabine and MUC1 Inhibitor Using PEG-PCL Nanoparticles for Breast Cancer Therapy. Mol Pharm 2022; 19:2429-2440. [PMID: 35639628 DOI: 10.1021/acs.molpharmaceut.2c00175] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In breast cancer therapy, Gemcitabine (Gem) is an antineoplastic antimetabolite with greater anticancer efficacy and tolerability. However, effectiveness of Gem is limited by its off-target effects. The synergistic potential of MUC1 (mucin 1) inhibitors and Gem-loaded polymeric nanoparticles (NPs) was discussed in this work in order to reduce dose-related toxicities and enhance the therapeutic efficacy. The double emulsion solvent evaporation method was used to prepare poly(ethylene glycol) methyl ether-block-poly-caprolactone (PEG-PCL)-loaded Gem and MUC 1 inhibitor NPs. The average size of Gem and MUC 1 inhibitor-loaded NPs was 128 nm, with a spherical shape. Twin-loaded NPs containing Gem and the MUC1 inhibitor decreased IC50 and behaved synergistically. Furthermore, in vitro mechanistic studies, that is, loss of MMP, clonogenic assay, Annexin V FITC assay, and Western blotting to confirm apoptosis with simultaneous induction of autophagy using acridine orange (AO) staining were performed in this study. Furthermore, the investigated NPs upon combination exhibited greater loss of MMP and decreased clonogenic potential with simultaneous induction of autophagy in MCF-7 cells. Annexin V FITC clearly showed the percentage of apoptosis while Western blotting protein expression analysis revealed an increase in caspase-3 activity with simultaneous decrease in Bcl-2 protein expression, a hallmark of apoptosis. The effectiveness of the Ehrlich ascites solid (EAT) mice treated with Gem-MUC1 inhibitor NPs was higher than that of the animals treated alone. Overall, the combined administration of Gem and MUC1 inhibitor-loaded NPs was found to be more efficacious than Gem and MUC1 inhibitor delivered separately.
Collapse
Affiliation(s)
- Akanksha Behl
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak, Haryana 124 001, India
| | - Parul Sarwalia
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, Haryana 132001, India
| | - Sushil Kumar
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, Haryana 132001, India
| | - Chittaranjan Behera
- PK-PD Tox and Formulation Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Mubashir Javed Mintoo
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Tirtha Kumar Datta
- Animal Biotechnology Centre, ICAR-National Dairy Research Institute, Karnal, Haryana 132001, India
| | - Prem N Gupta
- PK-PD Tox and Formulation Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Anil K Chhillar
- Centre for Biotechnology, Maharshi Dayanand University, Rohtak, Haryana 124 001, India
| |
Collapse
|
6
|
Liu Y, Ran Y, Ge Y, Raza F, Li S, Zafar H, Wu Y, Paiva-Santos AC, Yu C, Sun M, Zhu Y, Li F. pH-Sensitive Peptide Hydrogels as a Combination Drug Delivery System for Cancer Treatment. Pharmaceutics 2022; 14:pharmaceutics14030652. [PMID: 35336026 PMCID: PMC8948763 DOI: 10.3390/pharmaceutics14030652] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/01/2022] [Accepted: 03/11/2022] [Indexed: 01/09/2023] Open
Abstract
Conventional antitumor chemotherapeutics generally have shortcomings in terms of dissolubility, selectivity and drug action time, and it has been difficult to achieve high antitumor efficacy with single-drug therapy. At present, combination therapy with two or more drugs is widely used in the treatment of cancer, but a shortcoming is that the drugs do not reach the target at the same time, resulting in a reduction in efficacy. Therefore, it is necessary to design a carrier that can release two drugs at the same site. We designed an injectable pH-responsive OE peptide hydrogel as a carrier material for the antitumor drugs gemcitabine (GEM) and paclitaxel (PTX) that can release drugs at the tumor site simultaneously to achieve the antitumor effect. After determining the optimal gelation concentration of the OE polypeptide, we conducted an in vitro release study to prove its pH sensitivity. The release of PTX from the OE hydrogel in the medium at pH 5.8 and pH 7.4 was 96.90% and 38.98% in 7 days. The release of GEM from the OE hydrogel in media with pH of 5.8 and 7.4 was 99.99% and 99.63% in 3 days. Transmission electron microscopy (TEM) and circular dichroism (CD) experiments were used to observe the microstructure of the peptides. The circular dichroism of OE showed a single negative peak shape when under neutral conditions, indicating a β-folded structure, while under acidic conditions, it presented characteristics of a random coil. Rheological experiments were used to investigate the mechanical strength of this peptide hydrogel. Furthermore, the treatment effect of the drug-loaded peptide hydrogel was demonstrated through in vitro and in vivo experiments. The results show that the peptide hydrogels have different structures at different pH values and are highly sensitive to pH. They can reach the tumor site by injection and are induced by the tumor microenvironment to release antitumor drugs slowly and continuously. This biologically functional material has a promising future in drug delivery for combination drugs.
Collapse
Affiliation(s)
- Yuanfen Liu
- Department of Pharmacy, Jiangsu Health Vocational College, Nanjing 211800, China;
| | - Yingchun Ran
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Department of Emergency, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China;
| | - Yu Ge
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (Y.G.); (Y.W.); (C.Y.); (M.S.)
| | - Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China;
- Correspondence: (F.R.); (Y.Z.); (F.L.)
| | - Shasha Li
- College of Pharmacy, Xinjiang Medical University, Ürümqi 830000, China;
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China;
| | - Yiqun Wu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (Y.G.); (Y.W.); (C.Y.); (M.S.)
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal;
| | - Chenyang Yu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (Y.G.); (Y.W.); (C.Y.); (M.S.)
| | - Meng Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (Y.G.); (Y.W.); (C.Y.); (M.S.)
| | - Ying Zhu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (Y.G.); (Y.W.); (C.Y.); (M.S.)
- Correspondence: (F.R.); (Y.Z.); (F.L.)
| | - Fei Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China; (Y.G.); (Y.W.); (C.Y.); (M.S.)
- Correspondence: (F.R.); (Y.Z.); (F.L.)
| |
Collapse
|
7
|
Polymeric Nanoparticles: Exploring the Current Drug Development and Therapeutic Insight of Breast Cancer Treatment and Recommendations. Polymers (Basel) 2021; 13:polym13244400. [PMID: 34960948 PMCID: PMC8703470 DOI: 10.3390/polym13244400] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 02/06/2023] Open
Abstract
This manuscript aims to provide the latest update on polymeric nanoparticle drug delivery system for breast cancer treatment after 2015 and how research-oriented it is based on the available research data. Therefore, the authors have chosen breast cancer which is the most frequent and common reason for mortality in women worldwide. The first-line treatment for breast cancer treatment is chemotherapy, apart from surgery, radiation and hormonal therapy. Chemotherapy is associated with lesser therapeutics and undesirable side effects and hence. In addition, drug resistance affects the therapeutic dose to the target site. Although various nano-based formulations have been developed for effective treatment, the polymeric nanoparticles effectively avoid the lacunae of conventional chemotherapy. There has been an effort made to understand the chemotherapy drugs and their conventional formulation-related problems for better targeting and effective drug delivery for breast cancer treatment. Thus, the polymeric nanoparticles as a strategy overcome the associated problems with resulting dose reduction, enhanced bioavailability, reduced side effects, etc. This present review has compiled the research reports published from 2015 to 2021 from different databases, such as PubMed, Google Scholar, ScienceDirect, which are related to breast cancer treatment in which the drug delivery of numerous chemotherapeutic agents alone or in combination, including phytoconstituents formulated into various polymer-based nanoparticles.
Collapse
|
8
|
Drug Resistance in Metastatic Breast Cancer: Tumor Targeted Nanomedicine to the Rescue. Int J Mol Sci 2021; 22:ijms22094673. [PMID: 33925129 PMCID: PMC8125767 DOI: 10.3390/ijms22094673] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/25/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
Breast cancer, specifically metastatic breast, is a leading cause of morbidity and mortality in women. This is mainly due to relapse and reoccurrence of tumor. The primary reason for cancer relapse is the development of multidrug resistance (MDR) hampering the treatment and prognosis. MDR can occur due to a multitude of molecular events, including increased expression of efflux transporters such as P-gp, BCRP, or MRP1; epithelial to mesenchymal transition; and resistance development in breast cancer stem cells. Excessive dose dumping in chemotherapy can cause intrinsic anti-cancer MDR to appear prior to chemotherapy and after the treatment. Hence, novel targeted nanomedicines encapsulating chemotherapeutics and gene therapy products may assist to overcome cancer drug resistance. Targeted nanomedicines offer innovative strategies to overcome the limitations of conventional chemotherapy while permitting enhanced selectivity to cancer cells. Targeted nanotheranostics permit targeted drug release, precise breast cancer diagnosis, and importantly, the ability to overcome MDR. The article discusses various nanomedicines designed to selectively target breast cancer, triple negative breast cancer, and breast cancer stem cells. In addition, the review discusses recent approaches, including combination nanoparticles (NPs), theranostic NPs, and stimuli sensitive or “smart” NPs. Recent innovations in microRNA NPs and personalized medicine NPs are also discussed. Future perspective research for complex targeted and multi-stage responsive nanomedicines for metastatic breast cancer is discussed.
Collapse
|
9
|
Paroha S, Verma J, Dubey RD, Dewangan RP, Molugulu N, Bapat RA, Sahoo PK, Kesharwani P. Recent advances and prospects in gemcitabine drug delivery systems. Int J Pharm 2021; 592:120043. [DOI: 10.1016/j.ijpharm.2020.120043] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 10/17/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022]
|
10
|
Yu J, Mu Q, Perazzolo S, Griffin JI, Zhu L, McConnachie LA, Shen DD, Ho RJ. Novel Long-Acting Drug Combination Nanoparticles Composed of Gemcitabine and Paclitaxel Enhance Localization of Both Drugs in Metastatic Breast Cancer Nodules. Pharm Res 2020; 37:197. [PMID: 32968837 PMCID: PMC8686529 DOI: 10.1007/s11095-020-02888-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/21/2020] [Indexed: 12/25/2022]
Abstract
PURPOSE To develop drug-combination nanoparticles (DcNPs) composed of hydrophilic gemcitabine (G) and hydrophobic paclitaxel (T) and deliver both drugs to metastatic cancer cells. METHODS GT DcNPs were evaluated based on particle size and drug association efficiency (AE%). The effect of DcNP on GT plasma time-course and tissue distribution was characterized in mice and a pharmacokinetic model was developed. A GT distribution study into cancer nodules (derived from 4 T1 cells) was performed. RESULTS An optimized GT DcNP composition (d = 59.2 nm ±9.2 nm) was found to be suitable for IV formulation. Plasma exposure of G and T were enhanced 61-fold and 3.8-fold when given in DcNP form compared to the conventional formulation, respectively. Mechanism based pharmacokinetic modeling and simulation show that both G and T remain highly associated to DcNPs in vivo (G: 98%, T:75%). GT DcNPs have minimal distribution to healthy organs with selective distribution and retention in tumor burdened tissue. Tumor bearing lungs had a 5-fold higher tissue-to-plasma ratio of gemcitabine in GT DcNPs compared to healthy lungs. CONCLUSIONS DcNPs can deliver hydrophilic G and hydrophobic T together to cancer nodules and produce long acting exposure, likely due to stable GT association to DcNPs in vivo.
Collapse
Affiliation(s)
- Jesse Yu
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Qingxin Mu
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Simone Perazzolo
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - James I Griffin
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Linxi Zhu
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Lisa A McConnachie
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Danny D Shen
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA
| | - Rodney Jy Ho
- Departments of Pharmaceutics, University of Washington, Seattle, Washington, 98195, USA.
- Departments of Bioengineering, University of Washington, Seattle, Washington, 98195, USA.
| |
Collapse
|
11
|
Madamsetty VS, Pal K, Dutta SK, Wang E, Mukhopadhyay D. Targeted Dual Intervention-Oriented Drug-Encapsulated (DIODE) Nanoformulations for Improved Treatment of Pancreatic Cancer. Cancers (Basel) 2020; 12:cancers12051189. [PMID: 32397114 PMCID: PMC7281578 DOI: 10.3390/cancers12051189] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/24/2020] [Accepted: 05/06/2020] [Indexed: 12/14/2022] Open
Abstract
Despite recent advancements, effective treatment for pancreatic ductal adenocarcinoma (PDAC) has remained elusive. The overall survival rate in PDAC patients has been dismally low due to resistance to standard therapies. In fact, the failure of monotherapies to provide long-term survival benefits in patients led to ascension of several combination therapies for PDAC treatment. However, these combination therapies provided modest survival improvements while increasing treatment-related adverse side effects. Hence, recent developments in drug delivery methods hold the potential for enhancing therapeutic benefits by offering cocktail drug loading and minimizing chemotherapy-associated side effects. Nanoformulations-aided deliveries of anticancer agents have been a success in recent years. Yet, improving the tumor-targeted delivery of drugs to PDAC remains a major hurdle. In the present paper, we developed several new tumor-targeted dual intervention-oriented drug-encapsulated (DIODE) liposomes. We successfully formulated liposomes loaded with gemcitabine (G), paclitaxel (P), erlotinib (E), XL-184 (c-Met inhibitor, X), and their combinations (GP, GE, and GX) and evaluated their in vitro and in vivo efficacies. Our novel DIODE liposomal formulations improved median survival in comparison with gemcitabine-loaded liposomes or vehicle. Our findings are suggestive of the importance of the targeted delivery for combination therapies in improving pancreatic cancer treatment.
Collapse
|
12
|
Samanta K, Setua S, Kumari S, Jaggi M, Yallapu MM, Chauhan SC. Gemcitabine Combination Nano Therapies for Pancreatic Cancer. Pharmaceutics 2019; 11:E574. [PMID: 31689930 PMCID: PMC6920852 DOI: 10.3390/pharmaceutics11110574] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 10/28/2019] [Accepted: 10/28/2019] [Indexed: 12/15/2022] Open
Abstract
Pancreatic cancer is one of the deadliest causes of cancer-related death in the United States, with a 5-year overall survival rate of 6 to 8%. These statistics suggest that immediate medical attention is needed. Gemcitabine (GEM) is the gold standard first-line single chemotherapy agent for pancreatic cancer but, after a few months, cells develop chemoresistance. Multiple clinical and experimental investigations have demonstrated that a combination or co-administration of other drugs as chemotherapies with GEM lead to superior therapeutic benefits. However, such combination therapies often induce severe systemic toxicities. Thus, developing strategies to deliver a combination of chemotherapeutic agents more securely to patients is needed. Nanoparticle-mediated delivery can offer to load a cocktail of drugs, increase stability and availability, on-demand and tumor-specific delivery while minimizing chemotherapy-associated adverse effects. This review discusses the available drugs being co-administered with GEM and the limitations associated during the process of co-administration. This review also helps in providing knowledge of the significant number of delivery platforms being used to overcome problems related to gemcitabine-based co-delivery of other chemotherapeutic drugs, thereby focusing on how nanocarriers have been fabricated, considering the modes of action, targeting receptors, pharmacology of chemo drugs incorporated with GEM, and the differences in the physiological environment where the targeting is to be done. This review also documents the focus on novel mucin-targeted nanotechnology which is under development for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Kamalika Samanta
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Saini Setua
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Sonam Kumari
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Meena Jaggi
- Department of Immunology and Microbiology, Institute for Cancer Immunotherapy, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA.
| | - Murali M Yallapu
- Department of Immunology and Microbiology, Institute for Cancer Immunotherapy, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA.
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, Institute for Cancer Immunotherapy, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78503, USA.
| |
Collapse
|
13
|
Malliappan SP, Kandasamy P, Chidambaram S, Venkatasubbu D, Perumal SK, Sugumaran A. Breast Cancer Targeted Treatment Strategies: Promising Nanocarrier Approaches. Anticancer Agents Med Chem 2019; 20:1300-1310. [PMID: 31642415 DOI: 10.2174/1871520619666191022175003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 08/20/2019] [Accepted: 08/20/2019] [Indexed: 12/18/2022]
Abstract
Breast cancer is the second most common cancer that causes death among women worldwide. Incidence of breast cancer is increasing worldwide, and the age at which breast cancer develops has shifted from 50- 70 years to 30-40 years. Chemotherapy is the most commonly used effective treatment strategy to combat breast cancer. However, one of the major drawbacks is low selective site-specificity and the consequent toxic insult to normal healthy cells. The nanocarrier system is consistently utilised to minimise the various limitations involved in the conventional treatment of breast cancer. The nanocarrier based targeted drug delivery system provides better bioavailability, prolonged circulation with an effective accumulation of drugs at the tumour site either by active or passive drug targeting. Active targeting has been achieved by receptor/protein anchoring and externally guided magnetic nanocarriers, whereas passive targeting accomplished by employing the access to the tunnel via leaky tumour vasculature, utilising the tumour microenvironment, because the nanocarrier systems can reduce the toxicity to normal cells. As of now a few nanocarrier systems have been approved by FDA, and various nanoformulations are in the pipeline at the preclinical and clinical development for targeting breast cancer; among them, polymeric micelles, microemulsions, magnetic microemulsions, liposomes, dendrimers, carbon nanotubes, and magnetic Nanoparticles (NPs) are the most common. The current review highlights the active and passive targeting potential of nanocarriers in breast cancer and discusses their role in targeting breast cancer without affecting normal healthy cells.
Collapse
Affiliation(s)
- Sivakumar P Malliappan
- Center for Molecular Biology, Institute of Research and Development, Duy Tan University, 03 Quang Trung, Da Nang, Vietnam
| | - Palanivel Kandasamy
- Institute of Biochemistry and Molecular Medicine (IBMM), University of Bern, CH-3012 Bern, Switzerland
| | - Siva Chidambaram
- Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur-603203, India
| | - Devanand Venkatasubbu
- Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur-603203, India
| | - Sathish K Perumal
- Department of Plant Science, Bharathidasan University, Tiruchirappalli, India
| | - Abimanyu Sugumaran
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur-603203, India
| |
Collapse
|
14
|
Chang R, Zou Q, Xing R, Yan X. Peptide‐Based Supramolecular Nanodrugs as a New Generation of Therapeutic Toolboxes against Cancer. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900048] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Rui Chang
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences Beijing 100190 China
- School of Chemical EngineeringUniversity of Chinese Academy of Sciences Beijing 100049 China
| | - Qianli Zou
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences Beijing 100190 China
| | - Ruirui Xing
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences Beijing 100190 China
| | - Xuehai Yan
- State Key Laboratory of Biochemical EngineeringInstitute of Process EngineeringChinese Academy of Sciences Beijing 100190 China
- School of Chemical EngineeringUniversity of Chinese Academy of Sciences Beijing 100049 China
- Center for MesoscienceInstitute of Process EngineeringChinese Academy of Sciences Beijing 100190 China
| |
Collapse
|
15
|
Lei M, Sha S, Wang X, Wang J, Du X, Miao H, Zhou H, Bai E, Shi J, Zhu Y. Co-delivery of paclitaxel and gemcitabine via a self-assembling nanoparticle for targeted treatment of breast cancer. RSC Adv 2019; 9:5512-5520. [PMID: 35515924 PMCID: PMC9060788 DOI: 10.1039/c9ra00276f] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 02/07/2019] [Indexed: 11/27/2022] Open
Abstract
Multi-functional nanoparticles can be used to improve the treatment index and reduce side effects of anti-tumor drugs. Herein, we developed a kind of multi-functional and highly biocompatible nanoparticle (NP) loaded with folic acid (FA), paclitaxel (PTX) and gemcitabine (GEM) via self-assembly to target cancer cells. The transmission electron microscopy (TEM) results showed that multi-functional FA targeting nanoparticles (MF-FA NPs) exhibited spherical morphology and favorable structural stability in aqueous solution. In addition, NPs (MF-FA NPs and MF NPs) exhibited comparable proliferation inhibition to breast cancer cell 4T1 compared with the pure drug. In in vivo antitumor studies, NPs showed an obviously enhanced anti-tumor efficacy compared with the pure drug. Furthermore, MF-FA NPs displayed higher tumor growth inhibition than MF NPs due to the specific targeting of FA to cancer cells. Consequently, the novel MF-FA NPs could be used as a potential chemotherapeutic formulation for breast cancer therapy. Preparation of MF-FA nanoparticles and the release behavior of drugs in tumor cells.![]()
Collapse
Affiliation(s)
- Meng Lei
- College of Science
- Nanjing Forestry University
- Nanjing 210037
- PR China
| | - Sijia Sha
- College of Science
- Nanjing Forestry University
- Nanjing 210037
- PR China
| | - Xueyuan Wang
- College of Life Science
- Nanjing Normal University
- Nanjing 210046
- PR China
| | - Jia Wang
- Jiangsu Chia Tai Fenghai Pharmaceutical Co. Ltd
- Nanjing 210046
- PR China
| | - Xiao Du
- Department of Pharmaceutics
- School of Pharmacy
- China Pharmaceutical University
- Nanjing 210009
- PR China
| | - Hang Miao
- College of Science
- Nanjing Forestry University
- Nanjing 210037
- PR China
| | - Hui Zhou
- College of Life Science
- Nanjing Normal University
- Nanjing 210046
- PR China
| | - Enhe Bai
- College of Life Science
- Nanjing Normal University
- Nanjing 210046
- PR China
| | - Jingmiao Shi
- Jiangsu Chia Tai Fenghai Pharmaceutical Co. Ltd
- Nanjing 210046
- PR China
| | - Yongqiang Zhu
- College of Life Science
- Nanjing Normal University
- Nanjing 210046
- PR China
| |
Collapse
|