1
|
Habashy KJ, Dmello C, Chen L, Arrieta VA, Kim KS, Gould A, Youngblood MW, Bouchoux G, Burdett KB, Zhang H, Canney M, Stupp R, Sonabend AM. Paclitaxel and Carboplatin in Combination with Low-intensity Pulsed Ultrasound for Glioblastoma. Clin Cancer Res 2024; 30:1619-1629. [PMID: 38295144 DOI: 10.1158/1078-0432.ccr-23-2367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 11/22/2023] [Accepted: 01/29/2024] [Indexed: 02/02/2024]
Abstract
PURPOSE We recently reported on clinical trials for patients with recurrent glioblastoma where low-intensity pulsed ultrasound and microbubbles (LIPU/MB) improved paclitaxel or carboplatin delivery into the brain. Here, we report variable local tumor control with paclitaxel at the maximal/target dose in our phase I trial (NCT04528680). To address this, we investigated the combination of paclitaxel with carboplatin in preclinical glioma models. EXPERIMENTAL DESIGN We performed MRI-based analysis to evaluate disease control in patients from our trial. We studied the cytotoxicity of paclitaxel and carboplatin against 11 human glioma lines as monotherapy and in combination at concentrations derived from human intraoperative studies. Synergy was assessed with the Loewe model and the survival benefit evaluated in two xenografts. We examined the effects on cell cycle progression, DNA damage, and apoptosis. RESULTS Patients treated with paclitaxel and LIPU/MB exhibited variable local tumor control, which correlated with overall survival. We observed limited cross-resistance to paclitaxel and carboplatin in glioma lines, with almost a third of them being exclusively susceptible to one drug. This combination led to susceptibility of 81% of lines and synergy in 55% of them. The combination proved more efficacious in two intracranial xenografts when administered with LIPU/MB, leading to complementary effects on cell cycle arrest. CONCLUSIONS Combining paclitaxel and carboplatin in gliomas may be more efficacious than monotherapy, as in other cancers, due to synergy and independent susceptibility to each drug. These results form the basis for an ongoing phase II trial (NCT04528680) where we investigate this combination with LIPU/MB.
Collapse
Affiliation(s)
- Karl J Habashy
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Crismita Dmello
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Li Chen
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Victor A Arrieta
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Kwang-Soo Kim
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Andrew Gould
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Mark W Youngblood
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | | - Kirsten B Burdett
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Hui Zhang
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | | - Roger Stupp
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Adam M Sonabend
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Northwestern Medicine Lou and Jean Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
2
|
Dey T, Ghosh A, Sanyal A, Charles CJ, Pokharel S, Nair L, Singh M, Kaity S, Ravichandiran V, Kaur K, Roy S. Surface engineered nanodiamonds: mechanistic intervention in biomedical applications for diagnosis and treatment of cancer. Biomed Mater 2024; 19:032003. [PMID: 38574581 DOI: 10.1088/1748-605x/ad3abb] [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: 10/01/2023] [Accepted: 04/04/2024] [Indexed: 04/06/2024]
Abstract
In terms of biomedical tools, nanodiamonds (ND) are a more recent innovation. Their size typically ranges between 4 to 100 nm. ND are produced via a variety of methods and are known for their physical toughness, durability, and chemical stability. Studies have revealed that surface modifications and functionalization have a significant influence on the optical and electrical properties of the nanomaterial. Consequently, surface functional groups of NDs have applications in a variety of domains, including drug administration, gene delivery, immunotherapy for cancer treatment, and bio-imaging to diagnose cancer. Additionally, their biocompatibility is a critical requisite for theirin vivoandin vitrointerventions. This review delves into these aspects and focuses on the recent advances in surface modification strategies of NDs for various biomedical applications surrounding cancer diagnosis and treatment. Furthermore, the prognosis of its clinical translation has also been discussed.
Collapse
Affiliation(s)
- Tanima Dey
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneshwar 751024, Odisha, India
| | - Anushikha Ghosh
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneshwar 751024, Odisha, India
| | - Arka Sanyal
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneshwar 751024, Odisha, India
| | | | - Sahas Pokharel
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneshwar 751024, Odisha, India
| | - Lakshmi Nair
- Department of Pharmaceutical Sciences, Assam Central University, Silchar 788011, Assam, India
| | - Manjari Singh
- Department of Pharmaceutical Sciences, Assam Central University, Silchar 788011, Assam, India
| | - Santanu Kaity
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical, Education and Research, Kolkata, West Bengal 700054, India
| | - Velayutham Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical, Education and Research, Kolkata, West Bengal 700054, India
| | - Kulwinder Kaur
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons, Dublin 2 D02YN77, Ireland
- Department of Pharmacy & Biomolecular Science, Royal College of Surgeons, Dublin 2 D02YN77, Ireland
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical, Education and Research, Kolkata, West Bengal 700054, India
| |
Collapse
|
3
|
Dobrovolskaia MA. Lessons learned from immunological characterization of nanomaterials at the Nanotechnology Characterization Laboratory. Front Immunol 2022; 13:984252. [PMID: 36304452 PMCID: PMC9592561 DOI: 10.3389/fimmu.2022.984252] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Nanotechnology carriers have become common in pharmaceutical products because of their benefits to drug delivery, including reduced toxicities and improved efficacy of active pharmaceutical ingredients due to targeted delivery, prolonged circulation time, and controlled payload release. While available examples of reduced drug toxicity through formulation using a nanocarrier are encouraging, current data also demonstrate that nanoparticles may change a drug’s biodistribution and alter its toxicity profile. Moreover, individual components of nanoparticles and excipients commonly used in formulations are often not immunologically inert and contribute to the overall immune responses to nanotechnology-formulated products. Said immune responses may be beneficial or adverse depending on the indication, dose, dose regimen, and route of administration. Therefore, comprehensive toxicology studies are of paramount importance even when previously known drugs, components, and excipients are used in nanoformulations. Recent data also suggest that, despite decades of research directed at hiding nanocarriers from the immune recognition, the immune system’s inherent property of clearing particulate materials can be leveraged to improve the therapeutic efficacy of drugs formulated using nanoparticles. Herein, I review current knowledge about nanoparticles’ interaction with the immune system and how these interactions contribute to nanotechnology-formulated drug products’ safety and efficacy through the lens of over a decade of nanoparticle characterization at the Nanotechnology Characterization Laboratory.
Collapse
|
4
|
Vène E, Jarnouen K, Ribault C, Vlach M, Verres Y, Bourgeois M, Lepareur N, Cammas-Marion S, Loyer P. Circumsporozoite Protein of Plasmodium berghei- and George Baker Virus A-Derived Peptides Trigger Efficient Cell Internalization of Bioconjugates and Functionalized Poly(ethylene glycol)- b-poly(benzyl malate)-Based Nanoparticles in Human Hepatoma Cells. Pharmaceutics 2022; 14:804. [PMID: 35456637 PMCID: PMC9028075 DOI: 10.3390/pharmaceutics14040804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/31/2022] [Accepted: 04/03/2022] [Indexed: 02/04/2023] Open
Abstract
In order to identify the peptides, selected from the literature, that exhibit the strongest tropism towards human hepatoma cells, cell uptake assays were performed using biotinylated synthetic peptides bound to fluorescent streptavidin or engrafted onto nanoparticles (NPs), prepared from biotin-poly(ethylene glycol)-block-poly(benzyl malate) (Biot-PEG-b-PMLABe) via streptavidin bridging. Two peptides, derived from the circumsporozoite protein of Plasmodium berghei- (CPB) and George Baker (GB) Virus A (GBVA10-9), strongly enhanced the endocytosis of both streptavidin conjugates and NPs in hepatoma cells, compared to primary human hepatocytes and non-hepatic cells. Unexpectedly, the uptake of CPB- and GBVA10-9 functionalized PEG-b-PMLABe-based NPs by hepatoma cells involved, at least in part, the peptide binding to apolipoproteins, which would promote NP's interactions with cell membrane receptors of HDL particles. In addition, CPB and GBVA10-9 peptide-streptavidin conjugates favored the uptake by hepatoma cells over that of the human macrophages, known to strongly internalize nanoparticles by phagocytosis. These two peptides are promising candidate ligands for targeting hepatocellular carcinomas.
Collapse
Affiliation(s)
- Elise Vène
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- Pôle Pharmacie, Service Hospitalo-Universitaire de Pharmacie, CHU Rennes, F-35033 Rennes, France
| | - Kathleen Jarnouen
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
| | - Catherine Ribault
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
| | - Manuel Vlach
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- INRAE, Institut AGRO, PEGASE UMR 1348, F-35590 Saint-Gilles, France
| | - Yann Verres
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
| | - Mickaël Bourgeois
- CRCINA, Inserm, CNRS, Université de Nantes, F-44000 Nantes, France;
- ARRONAX Cyclotron, F-44817 Saint Herblain, France
| | - Nicolas Lepareur
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- Comprehensive Cancer Center Eugène Marquis, F-35000 Rennes, France
| | - Sandrine Cammas-Marion
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
- Institut des Sciences Chimiques de Rennes (ISCR), Ecole Nationale Supérieure de Chimie de Rennes, CNRS UMR 6226, University of Rennes, F-35042 Rennes, France
| | - Pascal Loyer
- Institut NUMECAN (Nutrition Metabolisms and Cancer), Inserm, UMR-S 1241, INRAE UMR-A 1341, Univ Rennes, F-35000 Rennes, France; (E.V.); (K.J.); (C.R.); (M.V.); (Y.V.)
| |
Collapse
|
5
|
Allemailem KS, Almatroudi A, Alrumaihi F, Almatroodi SA, Alkurbi MO, Basfar GT, Rahmani AH, Khan AA. Novel Approaches of Dysregulating Lysosome Functions in Cancer Cells by Specific Drugs and Its Nanoformulations: A Smart Approach of Modern Therapeutics. Int J Nanomedicine 2021; 16:5065-5098. [PMID: 34345172 PMCID: PMC8324981 DOI: 10.2147/ijn.s321343] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/08/2021] [Indexed: 01/18/2023] Open
Abstract
The smart strategy of cancer cells to bypass the caspase-dependent apoptotic pathway has led to the discovery of novel anti-cancer approaches including the targeting of lysosomes. Recent discoveries observed that lysosomes perform far beyond just recycling of cellular waste, as these organelles are metabolically very active and mediate several signalling pathways to sense the cellular metabolic status. These organelles also play a significant role in mediating the immune system functions. Thus, direct or indirect lysosome-targeting with different drugs can be considered a novel therapeutic approach in different disease including cancer. Recently, some anticancer lysosomotropic drugs (eg, nortriptyline, siramesine, desipramine) and their nanoformulations have been engineered to specifically accumulate within these organelles. These drugs can enhance lysosome membrane permeabilization (LMP) or disrupt the activity of resident enzymes and protein complexes, like v-ATPase and mTORC1. Other anticancer drugs like doxorubicin, quinacrine, chloroquine and DQ661 have also been used which act through multi-target points. In addition, autophagy inhibitors, ferroptosis inducers and fluorescent probes have also been used as novel theranostic agents. Several lysosome-specific drug nanoformulations like mixed charge and peptide conjugated gold nanoparticles (AuNPs), Au-ZnO hybrid NPs, TPP-PEG-biotin NPs, octadecyl-rhodamine-B and cationic liposomes, etc. have been synthesized by diverse methods. These nanoformulations can target cathepsins, glucose-regulated protein 78, or other lysosome specific proteins in different cancers. The specific targeting of cancer cell lysosomes with drug nanoformulations is quite recent and faces tremendous challenges like toxicity concerns to normal tissues, which may be resolved in future research. The anticancer applications of these nanoformulations have led them up to various stages of clinical trials. Here in this review article, we present the recent updates about the lysosome ultrastructure, its cross-talk with other organelles, and the novel strategies of targeting this organelle in tumor cells as a recent innovative approach of cancer management.
Collapse
Affiliation(s)
- Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Saleh A Almatroodi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Mohammad O Alkurbi
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ghaiyda Talal Basfar
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Amjad Ali Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
6
|
Mthimkhulu NP, Mosiane KS, Nweke EE, Balogun M, Fru P. Prospects of Delivering Natural Compounds by Polymer-Drug Conjugates in Cancer Therapeutics. Anticancer Agents Med Chem 2021; 22:1699-1713. [PMID: 33874874 DOI: 10.2174/1871520621666210419094623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/01/2021] [Accepted: 03/08/2021] [Indexed: 11/22/2022]
Abstract
Synthetic chemotherapeutics have played a crucial role in minimizing mostly palliative symptoms associated with cancer; however, they have also created other problems such as system toxicity due to a lack of specificity. This has led to the development of polymer-drug conjugates amongst other novel drug delivery systems. Most of the formulations designed using delivery systems consist of synthetic drugs and face issues such as drug resistance, which has already rendered drugs such as antibiotics ineffective. This is further exacerbated by toxicity due to long term use. Given these problems and the fact that conjugation of synthetic compounds to polymers has been relatively slow with no formulation on the market after a decade of extensive studies, the focus has shifted to using this platform with medicinal plant extracts to improve solubility, specificity and increase drug release of medicinal and herbal bioactives. In recent years, various plant extracts such as flavonoids, tannins and terpenoids have been studied extensively using this approach. The success of formulations developed using novel drug-delivery systems is highly dependent on the tumour microenvironment especially on the enhanced permeability and retention effect. As a result, the compromised lymphatic network and 'leaky' vasculature exhibited by tumour cells act as a guiding principle in the delivering of these formulations. This review focuses on the state of the polymer-drug conjugates and their exploration with natural compounds, the progress and difficulties thus far, and future directions concerning cancer treatment.
Collapse
Affiliation(s)
- Nompumelelo P Mthimkhulu
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193. South Africa
| | - Karabo S Mosiane
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193. South Africa
| | - Ekene E Nweke
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193. South Africa
| | - Mohammed Balogun
- Biopolymer Modification and Therapeutics Lab, Materials Science & Manufacturing, Council for Scientific and Industrial Research, Meiring Naude Road, Brummeria, Pretoria 0001. South Africa
| | - Pascaline Fru
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193. South Africa
| |
Collapse
|
7
|
Long Q, Zhu W, Guo L, Pu L. RGD-Conjugated Resveratrol HSA Nanoparticles as a Novel Delivery System in Ovarian Cancer Therapy. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:5747-5756. [PMID: 33408463 PMCID: PMC7779302 DOI: 10.2147/dddt.s248950] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 08/12/2020] [Indexed: 11/23/2022]
Abstract
Background To establish a novel delivery system of RGD-conjugated resveratrol human serum albumin (HAS) nanoparticles in ovarian cancer therapy. Methods The nanoparticles system was characterized for physicochemical properties, the stability in the serum and in vitro release. The comparison between RVT injection, HSA-RVT NPs and RGD-HSA-RVT NPs regarding tissue distributions and pharmacokinetics was also carried out using mice as the animal models. Results The results showed that RGD-HSA-RVT NPs were characterized of small particle size about 128.2 nm and negative zeta potential about -21.42 mV, and drug controlled to release slowly on a biphasic pattern. Compared with control groups, RGD-HSA-RVT NPs showed the higher cellular uptake and cell inhibition rates. In vivo data showed that RGD-HSA-RVT NPs have good tumor enrichment characteristics and a significant difference in tumor inhibition, compared with the control group. Conclusion RGD-conjugated resveratrol HSA nanoparticles are an ideal drug delivery system, which can play a role in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Qifang Long
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Weipei Zhu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Liangsheng Guo
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Li Pu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
8
|
Confeld MI, Mamnoon B, Feng L, Jensen-Smith H, Ray P, Froberg J, Kim J, Hollingsworth MA, Quadir M, Choi Y, Mallik S. Targeting the Tumor Core: Hypoxia-Responsive Nanoparticles for the Delivery of Chemotherapy to Pancreatic Tumors. Mol Pharm 2020; 17:2849-2863. [PMID: 32521162 DOI: 10.1021/acs.molpharmaceut.0c00247] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In pancreatic ductal adenocarcinoma (PDAC), early onset of hypoxia triggers remodeling of the extracellular matrix, epithelial-to-mesenchymal transition, increased cell survival, the formation of cancer stem cells, and drug resistance. Hypoxia in PDAC is also associated with the development of collagen-rich, fibrous extracellular stroma (desmoplasia), resulting in severely impaired drug penetration. To overcome these daunting challenges, we created polymer nanoparticles (polymersomes) that target and penetrate pancreatic tumors, reach the hypoxic niches, undergo rapid structural destabilization, and release the encapsulated drugs. In vitro studies indicated a high cellular uptake of the polymersomes and increased cytotoxicity of the drugs under hypoxia compared to unencapsulated drugs. The polymersomes decreased tumor growth by nearly 250% and significantly increased necrosis within the tumors by 60% in mice compared to untreated controls. We anticipate that these polymer nanoparticles possess a considerable translational potential for delivering drugs to solid hypoxic tumors.
Collapse
Affiliation(s)
- Matthew I Confeld
- Pharmaceutical Sciences Department, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Babak Mamnoon
- Pharmaceutical Sciences Department, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Li Feng
- Pharmaceutical Sciences Department, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Heather Jensen-Smith
- Fred & Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Priyanka Ray
- Coatings and Polymeric Materials Department, North Dakota State University, Fargo, North Dakota 58108, United States
| | - James Froberg
- Physics Department, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Jiha Kim
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58102, United States
| | - Michael A Hollingsworth
- Fred & Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Mohiuddin Quadir
- Coatings and Polymeric Materials Department, North Dakota State University, Fargo, North Dakota 58108, United States
| | - Yongki Choi
- Physics Department, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Sanku Mallik
- Pharmaceutical Sciences Department, North Dakota State University, Fargo, North Dakota 58105, United States
| |
Collapse
|
9
|
Parisi A, Palluzzi E, Cortellini A, Sidoni T, Cocciolone V, Lanfiuti Baldi P, Porzio G, Ficorella C, Cannita K. First-line carboplatin/nab-paclitaxel in advanced ovarian cancer patients, after hypersensitivity reaction to solvent-based taxanes: a single-institution experience. Clin Transl Oncol 2020; 22:158-162. [PMID: 31041717 DOI: 10.1007/s12094-019-02122-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 04/19/2019] [Indexed: 10/26/2022]
Abstract
One of the major challenges related to solvent-based taxanes administration in clinical practice is the high rate of hypersensitivity reactions (HSRs). Nab-paclitaxel is a solvent-free, albumin-bound, paclitaxel, which minimize the risk of HSR occurrence. In this single-institution, retrospective analysis, we evaluated stage IIIc-IV epithelial ovarian cancer (EOC) patients, treated with first-line carboplatin/nab-paclitaxel (± bevacizumab), after the occurrence of an HSR with solvent-based paclitaxel (and/or docetaxel). Between April 2012 and December 2018, ten patients (20.8%) received carboplatin/nab-paclitaxel (± bevacizumab) after the occurrence of an HSR to solvent-based taxanes. Among the evaluable patients, ORR was 100%. At median follow-up of 28.5 months, median PFS was 16.7 months, and median OS was 65.4 months, respectively. Median received dose intensity (DI) was 86% and 80% of the projected DI for nab-paclitaxel and carboplatin, respectively. There were no treatment-related grade 4 adverse events. Most relevant treatment-related grade 3 adverse events were: asthenia (10%), hypertransaminasemia (10%), neutropenia (20%), thrombocytopenia (20%), and anemia (10%). No HSR recurrence was observed. The high rate of HSR occurrence could limit first-line treatment options in clinical practice. Carboplatin/nab-paclitaxel association could represent a valid treatment option in this setting.
Collapse
Affiliation(s)
- A Parisi
- Medical Oncology, St. Salvatore Hospital, Via Vetoio, 67100, L'Aquila, Italy.
- Departement of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy.
| | - E Palluzzi
- Gynecologic Oncology Unit, Department of Women and Children Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - A Cortellini
- Medical Oncology, St. Salvatore Hospital, Via Vetoio, 67100, L'Aquila, Italy
- Departement of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - T Sidoni
- Departement of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - V Cocciolone
- Departement of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - P Lanfiuti Baldi
- Departement of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - G Porzio
- Medical Oncology, St. Salvatore Hospital, Via Vetoio, 67100, L'Aquila, Italy
- Departement of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - C Ficorella
- Medical Oncology, St. Salvatore Hospital, Via Vetoio, 67100, L'Aquila, Italy
- Departement of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - K Cannita
- Departement of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
10
|
Formulation optimization of an ephrin A2 targeted immunoliposome encapsulating reversibly modified taxane prodrugs. J Control Release 2019; 310:47-57. [PMID: 31400383 DOI: 10.1016/j.jconrel.2019.08.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 08/01/2019] [Accepted: 08/07/2019] [Indexed: 12/19/2022]
Abstract
Ephrin A2 targeted immunoliposomes incorporating pH-sensitive taxane prodrugs were developed for sustained delivery of active drug to solid tumors. Here we describe the systematic formulation development and characterization of these immunoliposomes. We synthesized both paclitaxel and docetaxel prodrugs to formulate as ephrin A2-targeted liposomes stabilized in the aqueous core with sucroseoctasulfate (SOS). The optimized lipid formulation was comprised of egg-sphingomyelin, cholesterol, and polyethylene glycol distearoyl glycerol (PEG-DSG). The formulations examined had a high efficiency of prodrug encapsulation (as high as 114 mol% taxane per mole phospholipid) and subsequent stability (>3 years at 2-8 °C). The taxane prodrug was stabilized with extraliposomal citric acid and subsequently loaded into liposomes containing a gradient of SOS, resulting in highly stable SOS-drug complexes being formed inside the liposome. The internal prodrug and SOS concentrations were optimized for their impact on in vivo drug release and drug degradation. Cryo-electron microscope images revealed dense prodrug-SOS complex in the aqueous core of the immunoliposomes. Ephrin A2-targeted taxane liposomes exhibited sub-nanomolar (0.69 nM) apparent equilibrium dissociation constant toward the extracellular domain of the ephrin A2 receptor, long circulation half-life (8-12 h) in mouse plasma, a release rate dependent on intraliposomal drug concentration and stable long-term storage. At an equitoxic dose of 50 mg taxane/kg, ephrin A2-targeted liposomal prodrug showed greater antitumor activity than 10 mg/kg of docetaxel in A549 non-small cell lung, as well as MDA-MB-436 and SUM149 triple negative breast cancer xenograft models. The lead molecule entered a Phase I clinical trial in patients with solid tumors (NCT03076372).
Collapse
|
11
|
Meng L, Gan S, Zhou Y, Cheng Y, Ding Y, Tong X, Wu J, Hu Y, Yuan A. Oxygen-rich chemotherapyviamodified Abraxane to inhibit the growth and metastasis of triple-negative breast cancer. Biomater Sci 2019; 7:168-177. [DOI: 10.1039/c8bm00753e] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Oxygen-rich chemotherapyviaMnO2modified Abraxane® for inhibiting growth and metastasis of triple negative breast cancer.
Collapse
Affiliation(s)
- Lingtong Meng
- State Key Laboratory of Pharmaceutical Biotechnology
- Medical School and School of life science
- Nanjing University
- Nanjing 210093
- China
| | - Shaoju Gan
- State Key Laboratory of Pharmaceutical Biotechnology
- Medical School and School of life science
- Nanjing University
- Nanjing 210093
- China
| | - Ya Zhou
- State Key Laboratory of Pharmaceutical Biotechnology
- Medical School and School of life science
- Nanjing University
- Nanjing 210093
- China
| | - Yali Cheng
- State Key Laboratory of Pharmaceutical Biotechnology
- Medical School and School of life science
- Nanjing University
- Nanjing 210093
- China
| | - Yawen Ding
- State Key Laboratory of Pharmaceutical Biotechnology
- Medical School and School of life science
- Nanjing University
- Nanjing 210093
- China
| | - Xiaoning Tong
- State Key Laboratory of Pharmaceutical Biotechnology
- Medical School and School of life science
- Nanjing University
- Nanjing 210093
- China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology
- Medical School and School of life science
- Nanjing University
- Nanjing 210093
- China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology
- Medical School and School of life science
- Nanjing University
- Nanjing 210093
- China
| | - Ahu Yuan
- State Key Laboratory of Pharmaceutical Biotechnology
- Medical School and School of life science
- Nanjing University
- Nanjing 210093
- China
| |
Collapse
|
12
|
Kavoosi F, Modaresi F, Sanaei M, Rezaei Z. Medical and dental applications of nanomedicines. APMIS 2018; 126:795-803. [PMID: 30264432 DOI: 10.1111/apm.12890] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 09/03/2018] [Indexed: 12/25/2022]
Abstract
Nanoparticles are tiny materials with nanosized components less than 100 nm in at least one dimension with physicochemical properties, which make them very attractive for medical application. These compounds have been evaluated as potential medicines for several decades. Nanotechnology has provided advances in the various fields of health sciences such as diagnosis, prevention and treatment by application of the agents named nanomedicines, including proteins, polymers, micelles, dendrimers, liposomes, emulsions, nanocapsules and nanoparticles. These materials can act as a scaffold, gene/drug delivery, tumor suppressor, conjugated with surgical implant, etc. They can also use as a nanocomposite, artificial tooth and dental caries preventing agent in the dentistry science. This current review tries to summarize recent applications of nanomedicine in the medical and dental fields.
Collapse
Affiliation(s)
- Fraidoon Kavoosi
- Research Center for Non-communicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Farzan Modaresi
- Department of Microbiology, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran.,Department of Advanced Medical Sciences and Technology, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Masumeh Sanaei
- Research Center for Non-communicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Zahra Rezaei
- Student Research Committee, Jahrom University of Medical Sciences, Jahrom, Iran
| |
Collapse
|
13
|
Hyaluronic Acid Layer-By-Layer (LbL) Nanoparticles for Synergistic Chemo-Phototherapy. Pharm Res 2018; 35:196. [PMID: 30143878 DOI: 10.1007/s11095-018-2480-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/11/2018] [Indexed: 02/05/2023]
Abstract
PURPOSE The aim of this study was to design hyaluronic acid (HA) layer-by-layer (LbL) nanoparticles, which carried paclitaxel (PTX) and Indocyanine green (ICG) to both tumor cells and tumor associated cells to achieve synergistic chemo-photothermal therapeutic effect. METHODS The LbL-engineered nanoparticles (PDIH) were prepared by dopamine self-polymerization on PTX nanocrystal to form thin, surface-adherent polydopamine (PDA) films, which subsequently absorbed ICG and HA. The tumor cell and tumor associated cell targeting and antitumor efficacy of PDIH were investigated both in vitro an in vivo using 4 T1 murine mammary cancer cell lines and mice bearing orthotopic 4 T1 breast tumor. RESULTS PDIH presented a long-rod shape in TEM and showed enhanced photothermal effect and cytotoxicity upon NIR laser irradiation both in vitro and in vivo. PDIH also displayed high target ability to CD44 overexpressed tumor cells and tumor associated cells mediated by HA. In vivo antitumor study indicated that PDIH therapeutic strategy could achieve remarkable antitumor efficacy. CONCLUSION PDIH showed excellent tumor-targeting property and chemo-photothermal therapeutic efficacy.
Collapse
|
14
|
Li R, Zheng K, Yuan C, Chen Z, Huang M. Be Active or Not: the Relative Contribution of Active and Passive Tumor Targeting of Nanomaterials. Nanotheranostics 2017; 1:346-357. [PMID: 29071198 PMCID: PMC5646738 DOI: 10.7150/ntno.19380] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 06/21/2017] [Indexed: 12/23/2022] Open
Abstract
Malignant tumor (cancer) remains as one of the deadliest diseases throughout the world, despite its overall mortality drops. Nanomaterials (NMs) have been widely studied as diagnostic and/or therapeutic agents for tumors. A feature of NMs, compared to small molecules, is that NMs can be concentrated passively in tumors through enhanced permeability and retention (EPR) effect. In the meantime, NMs can be engineered to target toward tumor specific markers in an active manner, e.g., receptor-mediated targeting. The relative contribution of the EPR effect and the receptor-mediated targeting to NM accumulation in tumor tissues has not been clearly defined yet. Here, we tackle this fundamental issue by reviewing previous studies. First, we summarize the current knowledge on these two tumor targeting strategies of NMs, and on how NMs arrive to tumors from blood circulation. We then demonstrate that contribution of the active and passive effects to total accumulation of NMs in tumors varies with time. Over time, the receptor-mediated targeting contributes more than the EPR effect with a ratio of 3 in the case of urokinase-type plasminogen activator receptor (uPAR)-mediated targeting and human serum albumin (HSA)-mediated EPR effect. Therefore, this review highlights the dynamics of active and passive targeting of NMs on their accumulation at tumor sites, and is valuable for future design of NMs in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Rui Li
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, Henan, 450002, China
| | - Ke Zheng
- Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Cai Yuan
- Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Zhuo Chen
- Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, 350002, China
| | - Mingdong Huang
- Fuzhou University, Fuzhou, Fujian, 350116, China.,Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, 350002, China
| |
Collapse
|
15
|
Ren L, Liu X, Wang Q, Zhang L, Deng G, Zhou F, Lu J. Facile fabrication of a magnetically smart PTX-loaded Cys–Fe3O4/CuS@BSA nano-drug for imaging-guided chemo-photothermal therapy. Dalton Trans 2017; 46:2204-2213. [DOI: 10.1039/c6dt04308a] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A PTX-loaded Cys-Fe3O4/CuS@BSA nano-drug was synthesized for MR and NIR imaging-guided chemo-photothermal combination therapy of cancer via a facile fabrication method.
Collapse
Affiliation(s)
- Lanfang Ren
- School of Chemistry and Chemical Engineering
- Shanghai University of Engineering Science
- Shanghai 201620
- P. R. China
| | - Xijian Liu
- School of Chemistry and Chemical Engineering
- Shanghai University of Engineering Science
- Shanghai 201620
- P. R. China
| | - Qian Wang
- Orthopedic Traumatology
- Trauma Center
- Shanghai First People's Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200080
| | - Lijuan Zhang
- School of Chemistry and Chemical Engineering
- Shanghai University of Engineering Science
- Shanghai 201620
- P. R. China
| | - Guoying Deng
- Orthopedic Traumatology
- Trauma Center
- Shanghai First People's Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200080
| | - Feng Zhou
- Orthopedic Traumatology
- Trauma Center
- Shanghai First People's Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai 200080
| | - Jie Lu
- School of Chemistry and Chemical Engineering
- Shanghai University of Engineering Science
- Shanghai 201620
- P. R. China
| |
Collapse
|
16
|
Tai X, Wang Y, Zhang L, Yang Y, Shi K, Ruan S, Liu Y, Gao H, Zhang Z, He Q. Cabazitaxel and indocyanine green co-delivery tumor-targeting nanoparticle for improved antitumor efficacy and minimized drug toxicity. J Drug Target 2016; 25:179-187. [PMID: 27608834 DOI: 10.1080/1061186x.2016.1233975] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cabazitaxel (CBX) is an effective antineoplastic agent for the treatment of many kinds of cancers. However, the poor water solubility remains a serious deterrent to the utilization of CBX as a commercial drug. In this study, we designed a strategy that integrated CBX into albumin nanoparticles (ANs) formed with human serum albumin (HSA) to improve the water solubility and targeting ability. Meanwhile, we utilized a photothermal agent-indocyanine green (ICG), which could cooperate with CBX to enhance the antitumor effect. The obtained ANs containing ICG and CBX (AN-ICG-CBX) exhibited good mono-dispersity. In vitro cytotoxicity study showed the effectiveness of CBX and ICG, respectively, whereas AN-ICG-CBX with irradiation exhibited the most efficient antiproliferative ability (83.7%). In vivo safety evaluation studies demonstrated the safety of AN-ICG-CBX. Furthermore, the in vivo antitumor study indicated that the AN-ICG-CBX with irradiation achieved higher tumor inhibition rate (91.3%) compared with CBX-encapsulated AN (AN-CBX) (83.3%) or ICG-encapsulated AN (AN-ICG) plus irradiation (60.1%) in 4T1 tumor-bearing mice. To sum up, a safety and effective formulation AN-ICG-CBX was developed in this study and successfully reduced the drug toxicity, improved the targeting efficiency and enhanced the therapeutic effects, becoming a promising candidate for clinical application.
Collapse
Affiliation(s)
- Xiaowei Tai
- a Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy , Sichuan University , Chengdu , China
| | - Yang Wang
- a Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy , Sichuan University , Chengdu , China
| | - Li Zhang
- a Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy , Sichuan University , Chengdu , China
| | - Yuting Yang
- a Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy , Sichuan University , Chengdu , China
| | - Kairong Shi
- a Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy , Sichuan University , Chengdu , China
| | - Shaobo Ruan
- a Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy , Sichuan University , Chengdu , China
| | - Yayuan Liu
- a Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy , Sichuan University , Chengdu , China
| | - Huile Gao
- a Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy , Sichuan University , Chengdu , China
| | - Zhirong Zhang
- a Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy , Sichuan University , Chengdu , China
| | - Qin He
- a Key Laboratory of Drug Targeting and Drug Delivery Systems, West China School of Pharmacy , Sichuan University , Chengdu , China
| |
Collapse
|
17
|
Dobrovolskaia MA, Shurin M, Shvedova AA. Current understanding of interactions between nanoparticles and the immune system. Toxicol Appl Pharmacol 2016; 299:78-89. [PMID: 26739622 PMCID: PMC4811709 DOI: 10.1016/j.taap.2015.12.022] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 12/24/2015] [Accepted: 12/26/2015] [Indexed: 10/22/2022]
Abstract
The delivery of drugs, antigens, and imaging agents benefits from using nanotechnology-based carriers. The successful translation of nanoformulations to the clinic involves thorough assessment of their safety profiles, which, among other end-points, includes evaluation of immunotoxicity. The past decade of research focusing on nanoparticle interaction with the immune system has been fruitful in terms of understanding the basics of nanoparticle immunocompatibility, developing a bioanalytical infrastructure to screen for nanoparticle-mediated immune reactions, beginning to uncover the mechanisms of nanoparticle immunotoxicity, and utilizing current knowledge about the structure-activity relationship between nanoparticles' physicochemical properties and their effects on the immune system to guide safe drug delivery. In the present review, we focus on the most prominent pieces of the nanoparticle-immune system puzzle and discuss the achievements, disappointments, and lessons learned over the past 15years of research on the immunotoxicity of engineered nanomaterials.
Collapse
Affiliation(s)
- Marina A Dobrovolskaia
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NCI at Frederick, Frederick, MD 21702, USA.
| | - Michael Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Anna A Shvedova
- Health Effects Laboratory Division, National Institute of Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV 26505, USA; Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Recent advances in nanotechnology have addressed some of the issues related to lack of selectivity and nonspecific toxicities associated with conventional chemotherapy. Nanoparticles are therapeutic carriers that can be fine tuned for specific application and for passive or active tumor targeting. RECENT FINDINGS Although the nanoparticle field is rapidly expanding, there are to date only six nanoparticle-based drug delivery platforms and two antibody-drug conjugates that are clinically approved for cancer therapy. Here, we review the clinical data of liposomal anthracyclines, nanoparticle formulations of paclitaxel and trastuzumab emtansine. We then briefly comment on efficacy and safety issues of nanoparticles, as well as on the next-generation nanoparticles for cancer therapy. SUMMARY The emerging development of cancer nanotechnology offers the opportunity of reinvestigating the potential of cytotoxic agents, improving tumor targeting and drug delivery, leading to better safety profile and antitumor activity. Adding specificity to nanoparticles may allow personalization of cancer therapy using chemotherapy.
Collapse
|
19
|
Carraher CE, Roner MR, Moric-Johnson A, Miller L, Barot G, Sookdeo N. Ability of simple organotin polyethers to inhibit pancreatic cancer. JOURNAL OF MACROMOLECULAR SCIENCE PART A-PURE AND APPLIED CHEMISTRY 2016. [DOI: 10.1080/10601325.2016.1120172] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
20
|
Siqing F, Culotta KS, Falchook GS, Hong DS, Myers AL, Zhang YP, Naing A, Janku F, Hou MM, Kurzrock R. Pharmacokinetic evaluation of nanoparticle albumin-bound paclitaxel delivered via hepatic arterial infusion in patients with predominantly hepatic metastases. Cancer Chemother Pharmacol 2015; 77:357-64. [DOI: 10.1007/s00280-015-2946-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 12/11/2015] [Indexed: 11/29/2022]
|
21
|
|
22
|
Leon-Ferre RA, Markovic SN. Nab-paclitaxel in patients with metastatic melanoma. Expert Rev Anticancer Ther 2015; 15:1371-7. [DOI: 10.1586/14737140.2015.1110024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
23
|
Affiliation(s)
- Yuanzeng Min
- Laboratory of Nano- and Translational Medicine, Carolina Institute of Nanomedicine, Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Joseph M Caster
- Laboratory of Nano- and Translational Medicine, Carolina Institute of Nanomedicine, Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Michael J Eblan
- Laboratory of Nano- and Translational Medicine, Carolina Institute of Nanomedicine, Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill , Chapel Hill, North Carolina 27599, United States
| | - Andrew Z Wang
- Laboratory of Nano- and Translational Medicine, Carolina Institute of Nanomedicine, Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill , Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
24
|
Phase I trial of biochemotherapy with cisplatin, temozolomide, and dose escalation of nab-paclitaxel combined with interleukin-2 and interferon-α in patients with metastatic melanoma. Melanoma Res 2015; 24:342-8. [PMID: 24743052 DOI: 10.1097/cmr.0000000000000062] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The primary objective of this study was to determine the safety, toxicity, and maximum tolerated dose of nanoparticle albumin-bound (nab)-paclitaxel as part of biochemotherapy for metastatic melanoma and to determine whether substituting nab-paclitaxel for less potent agents could increase response rates and duration. Treatment consisted of intravenous cisplatin (20 mg/m) on days 1-4, oral temozolomide (250 mg/m) on days 1-3, subcutaneous interferon-α (5×10 IU/m) on days 1-5, and continuous intravenous interleukin-2 (9×10 IU/m) for 96 h on days 1-4. A standard 3+3 dose escalation method was used; the nab-paclitaxel starting dose was 100 mg/m on day 1 and 70 mg/m on day 5. The treatment cycle was repeated every 3 weeks and toxicity was assessed weekly. Ten patients were enrolled. Dose-limiting toxicities included diarrhea, transaminasemia, and neutropenia. The maximum tolerated dose was not identified because the nab-paclitaxel dose on day 1 at the lowest planned dose (80 mg/m) caused dose-limiting toxicity in two of five patients. Of the nine patients who were evaluable for response, five had a partial response. The median time to disease progression was 5.30 months and the median overall survival was 8.73 months. Six patients developed central nervous system metastasis at a median of 5.33 months after treatment initiation. Biochemotherapy including nab-paclitaxel according to the doses and schedule regimen used in the present study has significant toxicity. Substituting dacarbazine with temozolomide did not prevent central nervous system metastasis in patients with metastatic melanoma.
Collapse
|
25
|
Chen Q, Liang C, Wang C, Liu Z. An imagable and photothermal "Abraxane-like" nanodrug for combination cancer therapy to treat subcutaneous and metastatic breast tumors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:903-10. [PMID: 25504416 DOI: 10.1002/adma.201404308] [Citation(s) in RCA: 353] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 10/28/2014] [Indexed: 05/28/2023]
Abstract
A new "Abraxane-like" nanotheranostic formulation self-assembled from three clinically approved agents, human serum albumin, paclitaxel, and indocyanine green, is developed. Utilizing such a newly formulated nanodrug, combined photothermal and chemotherapy is conducted under the guidance of imaging, not only effectively eliminating subcutaneous tumors, but also significantly inhibiting the development of metastatic tumors in this proof-of-concept study.
Collapse
Affiliation(s)
- Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou, Jiangsu, 215123, China
| | | | | | | |
Collapse
|
26
|
Townley HE, Zheng Y, Goldsmith J, Zheng YY, Stratford MRL, Dobson PJ, Ahmed AA. A novel biosensor for quantitative monitoring of on-target activity of paclitaxel. NANOSCALE 2015; 7:1127-1135. [PMID: 25483994 DOI: 10.1039/c4nr01105h] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
This study describes a system for quantifying paclitaxel activity using the C-terminus of α-tubulin as a biomarker. Following stabilization of microtubules with paclitaxel, a specific detyrosination reaction occurs at the C-terminus of α-tubulin which could be used to assess efficacy. A fluorescence resonance energy transfer (FRET) based biosensor was synthesized comprising a short peptide that corresponded to the C-terminus of α-tubulin, a fluorophore (Abz), and a quencher (Dnp). The fluorophore added to the end of the peptide can be released upon enzymatic detyrosination. In addition, a single fluorophore-tagged peptide was also conjugated to mesoporous silica nanoparticles to examine the feasibility of combining the drug with the peptide biomarker. As a proof of concept, we found that the degree of peptide cleavage, and therefore enzymatic activity, was directly correlated with exogenous bovine carboxypeptidase (CPA) an enzyme that mimics endogenous detyrosination. In addition, we show that cell lysates obtained from paclitaxel-treated cancer cells competed with exogenous CPA for biosensor cleavage in a paclitaxel dose-dependent manner. Our work provides strong evidence for the feasibility of combining paclitaxel with a novel biosensor in a multi-load nanoparticle.
Collapse
Affiliation(s)
- H E Townley
- Department of Engineering Sciences, Parks Road, Oxford, OX1 3PJ, UK.
| | | | | | | | | | | | | |
Collapse
|
27
|
Viúdez A, Ramírez N, Hernández-García I, Carvalho F, Vera R, Hidalgo M. Nab-paclitaxel: A flattering facelift. Crit Rev Oncol Hematol 2014; 92:166-80. [DOI: 10.1016/j.critrevonc.2014.06.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 06/03/2014] [Accepted: 06/20/2014] [Indexed: 12/21/2022] Open
|
28
|
Dawidczyk CM, Kim C, Park JH, Russell LM, Lee KH, Pomper MG, Searson PC. State-of-the-art in design rules for drug delivery platforms: lessons learned from FDA-approved nanomedicines. J Control Release 2014; 187:133-44. [PMID: 24874289 DOI: 10.1016/j.jconrel.2014.05.036] [Citation(s) in RCA: 366] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 05/14/2014] [Accepted: 05/17/2014] [Indexed: 12/14/2022]
Abstract
The ability to efficiently deliver a drug to a tumor site is dependent on a wide range of physiologically imposed design constraints. Nanotechnology provides the possibility of creating delivery vehicles where these design constraints can be decoupled, allowing new approaches for reducing the unwanted side effects of systemic delivery, increasing targeting efficiency and efficacy. Here we review the design strategies of the two FDA-approved antibody-drug conjugates (Brentuximab vedotin and Trastuzumab emtansine) and the four FDA-approved nanoparticle-based drug delivery platforms (Doxil, DaunoXome, Marqibo, and Abraxane) in the context of the challenges associated with systemic targeted delivery of a drug to a solid tumor. The lessons learned from these nanomedicines provide an important insight into the key challenges associated with the development of new platforms for systemic delivery of anti-cancer drugs.
Collapse
Affiliation(s)
- Charlene M Dawidczyk
- Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Johns Hopkins Center of Cancer Nanotechnology Excellence, 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA.; Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Chloe Kim
- Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Johns Hopkins Center of Cancer Nanotechnology Excellence, 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA.; Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Jea Ho Park
- Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Johns Hopkins Center of Cancer Nanotechnology Excellence, 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA.; Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Luisa M Russell
- Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Johns Hopkins Center of Cancer Nanotechnology Excellence, 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA.; Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Kwan Hyi Lee
- KIST Biomedical Research Institute, 5 Hwarangno 14-gil, Seongbuk-gu, Seoul 136-791, Republic of Korea
| | - Martin G Pomper
- Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Johns Hopkins Center of Cancer Nanotechnology Excellence, 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA.; Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA.
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA; Johns Hopkins Center of Cancer Nanotechnology Excellence, 100 Croft Hall, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA.; Department of Materials Science and Engineering, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA.
| |
Collapse
|
29
|
Stirland DL, Nichols JW, Miura S, Bae YH. Mind the gap: a survey of how cancer drug carriers are susceptible to the gap between research and practice. J Control Release 2013; 172:1045-64. [PMID: 24096014 PMCID: PMC3889175 DOI: 10.1016/j.jconrel.2013.09.026] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Revised: 09/07/2013] [Accepted: 09/25/2013] [Indexed: 11/23/2022]
Abstract
With countless research papers using preclinical models and showing the superiority of nanoparticle design over current drug therapies used to treat cancers, it is surprising how deficient the translation of these nano-sized drug carriers into the clinical setting is. This review article seeks to compare the preclinical and clinical results for Doxil®, PK1, Abraxane®, Genexol-PM®, Xyotax™, NC-6004, Mylotarg®, PK2, and CALAA-01. While not comprehensive, it covers nano-sized drug carriers designed to improve the efficacy of common drugs used in chemotherapy. While not always available or comparable, effort was made to compare the pharmacokinetics, toxicity, and efficacy between the animal and human studies. Discussion is provided to suggest what might be causing the gap. Finally, suggestions and encouragement are dispensed for the potential that nano-sized drug carriers hold.
Collapse
Affiliation(s)
- Darren Lars Stirland
- University of Utah; Department of Bioengineering; College of Engineering; Salt Lake City; UT 84112; United States
| | - Joseph W. Nichols
- University of Utah; Department of Bioengineering; College of Engineering; Salt Lake City; UT 84112; United States
| | - Seiji Miura
- Fuji Research Laboratories, Pharmaceutical Division, Kowa Co. Ltd., 332–1 Ohnoshinden, Fuji, Shizuoka, Japan
- University of Utah, Department of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, Salt Lake City, UT 84112, United States
| | - You Han Bae
- University of Utah, Department of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, Salt Lake City, UT 84112, United States
| |
Collapse
|
30
|
Socinski MA, Okamoto I, Hon JK, Hirsh V, Dakhil SR, Page RD, Orsini J, Yamamoto N, Zhang H, Renschler MF. Safety and efficacy analysis by histology of weekly nab-paclitaxel in combination with carboplatin as first-line therapy in patients with advanced non-small-cell lung cancer. Ann Oncol 2013; 24:2390-6. [PMID: 23842283 DOI: 10.1093/annonc/mdt235] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND This analysis compared the efficacy and safety outcomes by histology of nab-paclitaxel (nab-P) plus carboplatin (C) versus solvent-based paclitaxel (sb-P) plus C in patients with advanced non-small-cell lung cancer (NSCLC) based on preplanned stratification factors specified in the phase III trial protocol. PATIENTS AND METHODS Patients with untreated stage III/IV NSCLC received 100 mg/m(2) nab-P weekly and C (area under the curve, AUC = 6) every 3 weeks (q3w) or 200 mg/m(2) sb-P plus C (AUC = 6) q3w. Primary end point was objective overall response rate (ORR). RESULTS nab-P/C versus sb-P/C produced a significantly higher ORR (41% versus 24%; response rate ratio [RRR] 1.680; P < 0.001) in patients with squamous cell (SCC) NSCLC. For nab-P/C versus sb-P/C, ORRs were 26% versus 27% (RRR 0.966; P = 0.814) in patients with adenocarcinoma, 33% versus 15% (RRR 2.167; P = 0.323) in patients with large cell carcinoma (LC), and 24% versus 15% (RRR 1.593; P = 0.372) in patients with not otherwise specified histology. Median overall survival for nab-P/C versus sb-P/C in patients with SCC was 10.7 versus 9.5 months (HR 0.890; P = 0.310), and 12.4 versus 10.6 months (HR 1.208; P = 0.721) for patients with LC. nab-P/C produced significantly (P < 0.05) less grade 3/4 neuropathy and arthralgia, whereas sb-P/C produced less thrombocytopenia and anemia. CONCLUSION(S) First-line nab-P/C demonstrated a favorable risk-benefit profile in patients with NSCLC regardless of histology.
Collapse
Affiliation(s)
- M A Socinski
- Division of Hematology/Oncology, University of Pittsburgh Cancer Institute, PA 15232, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Batus M, Waheed S, Ruby C, Petersen L, Bines SD, Kaufman HL. Optimal management of metastatic melanoma: current strategies and future directions. Am J Clin Dermatol 2013; 14:179-94. [PMID: 23677693 PMCID: PMC3913474 DOI: 10.1007/s40257-013-0025-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Melanoma is increasing in incidence and remains a major public health threat. Although the disease may be curable when identified early, advanced melanoma is characterized by widespread metastatic disease and a median survival of less than 10 months. In recent years, however, major advances in our understanding of the molecular nature of melanoma and the interaction of melanoma cells with the immune system have resulted in several new therapeutic strategies that are showing significant clinical benefit. Current therapeutic approaches include surgical resection of metastatic disease, chemotherapy, immunotherapy, and targeted therapy. Dacarbazine, interleukin-2, ipilimumab, and vemurafenib are now approved for the treatment of advanced melanoma. In addition, new combination chemotherapy regimens, monoclonal antibodies blocking the programmed death-1 (PD-1)/PD-ligand 1 pathway, and targeted therapy against CKIT, mitogen-activated protein/extracellular signal-regulated kinase (MEK), and other putative signaling pathways in melanoma are beginning to show promise in early-phase clinical trials. Further research on these modalities alone and in combination will likely be the focus of future clinical investigation and may impact the outcomes for patients with advanced melanoma.
Collapse
Affiliation(s)
- Marta Batus
- Rush University Melanoma Program and Departments of Medicine, General Surgery and Immunology and Microbiology, Rush University Medical Center, 1725 W. Harrison Street, Room 845, Chicago, IL 60612, USA
| | | | | | | | | | | |
Collapse
|
32
|
Phase I/II study of albumin-bound nab-paclitaxel plus gemcitabine administered to Chinese patients with advanced pancreatic cancer. Cancer Chemother Pharmacol 2013; 71:1065-72. [PMID: 23483298 DOI: 10.1007/s00280-013-2102-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 01/22/2013] [Indexed: 12/13/2022]
Abstract
PURPOSE The primary objective of this study was to evaluate the dose-limiting toxicities (DLTs) and identify the maximum-tolerated dose (MTD) and recommended dose of nab-paclitaxel plus gemcitabine as a first-line treatment in Chinese patients with advanced pancreatic ductal adenocarcinoma (PDA). METHODS Patients with previously untreated advanced PDA were treated with nab-paclitaxel followed by gemcitabine (1,000 mg/m(2)) administered intravenously for 30 min on days 1 and 8 and repeated every 21 days. RESULTS Patients received nab-paclitaxel at the following dose levels: 80 mg/m(2) (n = 3), 100 mg/m(2) (n = 6), and 120 mg/m(2) (n = 12). The DLTs evaluated were elevated alanine aminotransferase and febrile neutropenia. However, there had no two out of three to six patients experienced DLTs, the MTD was not met. A total of 93 cycles were administered. The most common grade 3/4 toxicities were neutropenia (9.52 %), thrombocytopenia (4.76 %), and sensory neuropathy (4.76 %). For 12 patients receiving 120 mg/m(2), the overall response rate and disease control rate were 41.67 and 83.33 %, respectively, and the median progression-free survival and overall survival were 5.23 and 12.17 months, respectively. CONCLUSIONS Treatment with albumin-bound nab-paclitaxel (120 mg/m(2)) plus gemcitabine has a favorable safety profile with an encouraging antitumor effect in Chinese patients.
Collapse
|
33
|
Abstract
The treatment of metastasized urothelial cancer has been evolving in recent years. In particular, in the second-line setting after the failure of platinum-containing therapy, options are few and besides vinflunine, the recently approved standard in Europe, well-designed highly selective clinical trials may be possible alternatives for patients in this palliative situation. However, targeted therapy approaches have not achieved the same results in urothelial cancer as for instance in renal cell carcinoma. Many of the new targeted drugs have been investigated as single agents in phase II clinical trials without convincing oncologic outcome. This review aims to highlight the most relevant clinical studies examining targeted agents in the second-line setting of metastasized transitional carcinoma of the urothelium.
Collapse
|
34
|
Hertz DL, Walko CM, Bridges AS, Hull JH, Herendeen J, Rollins K, Watkins PB, Dees EC. Pilot study of rosiglitazone as an in vivo probe of paclitaxel exposure. Br J Clin Pharmacol 2012; 74:197-200. [PMID: 22680343 DOI: 10.1111/j.1365-2125.2012.04165.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
WHAT IS ALREADY KNOWN ABOUT THIS SUBJECT • Paclitaxel and rosiglitazone are primarily metabolized by CYP2C8 and their in vitro metabolism by human liver microsomes is correlated. Probe assays that quantify the in vivo activity of CYP enzymes which are important in drug metabolism have been developed for use in clinical pharmacology research. A probe of CYP2C8 that is easy to administer and interpret may be valuable for individualized dosing of paclitaxel. WHAT THIS STUDY ADDS • This pilot study demonstrates for the first time that there is an in vivo correlation between paclitaxel and rosiglitazone exposure. The finding, that a single rosiglitazone plasma concentration after oral dosing may explain significant variance in paclitaxel exposure, suggests that rosiglitazone may satisfy the requirements of a clinically useful in vivo probe. However, it is acknowledged that there is a need for further studies evaluating the use of rosiglitazone as a CYP2C8 probe and quantifying the relationship, in order to guide dosing of narrow therapeutic index drugs metabolized primarily by CYP2C8, such as paclitaxel. AIMS To evaluate the use of rosiglitazone and the erythromycin breath test (ERMBT), as probes of CYP2C8 and CYP3A4, respectively, to explain inter-individual variability in paclitaxel exposure. METHODS The concentration of rosiglitazone at 3 h and ERMBT results were included in a regression model to explain the variability in paclitaxel exposure in 14 subjects. RESULTS Rosiglitazone concentration was significantly correlated with paclitaxel exposure (P= 0.018) while ERMBT had no predictive value (P= 0.47). CONCLUSIONS The correlation between the exposure of rosiglitazone and paclitaxel likely reflects mutual dependence on the activity of CYP2C8. Rosiglitazone or similar agents may have value as in vivo probes of CYP2C8 activity.
Collapse
Affiliation(s)
- Daniel L Hertz
- UNC Eshelman School of Pharmacy, Division of Pharmacotherapy and Experimental Therapeutics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Bellmunt J. What are the expected developments in the medical treatment of bladder cancer. Eur J Cancer 2011; 47 Suppl 3:S294-7. [DOI: 10.1016/s0959-8049(11)70177-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
36
|
Chirgwin J, Chua SL. Management of breast cancer with nanoparticle albumin-bound (nab)-paclitaxel combination regimens: a clinical review. Breast 2011; 20:394-406. [PMID: 21839635 DOI: 10.1016/j.breast.2011.06.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Revised: 05/12/2011] [Accepted: 06/22/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Monotherapy with nanoparticle albumin-bound (nab)-paclitaxel has demonstrated improved efficacy and safety compared with solvent-based paclitaxel and docetaxel. DESIGN A comprehensive review of all reported studies of nab-paclitaxel combinations with other agents in all breast cancer settings was undertaken. RESULTS Most studies reviewed are small, phase II and non-comparative. Combinations studied included nab-paclitaxel plus trastuzumab and/or bevacizumab (with or without additional cytotoxic agents), gemcitabine, capecitabine, carboplatin, or lapatinib. The majority of metastatic and neoadjuvant studies demonstrated satisfactory efficacy and safety for nab-paclitaxel combinations, although conclusions regarding comparison with solvent-based taxane (SBT) regimens are not possible. The two adjuvant studies confirmed the safety of nab-paclitaxel combinations in this setting. CONCLUSIONS Although results of this review indicate that nab-paclitaxel may be an appropriate substitute for SBTs in combination regimens, additional research is required to confirm the place and cost effectiveness of these combinations before nab-paclitaxel could be recommended routinely in all settings.
Collapse
Affiliation(s)
- J Chirgwin
- Department of Medical Oncology, Box Hill and Maroondah Hospitals, Maroondah Breast Clinic, 20 Grey St, Ringwood East, Melbourne, VIC 3135, Australia.
| | | |
Collapse
|
37
|
Vishnu P, Roy V. nab-paclitaxel: a novel formulation of taxane for treatment of breast cancer. ACTA ACUST UNITED AC 2011; 6:495-506. [PMID: 20597612 DOI: 10.2217/whe.10.42] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Breast cancer is the most common type of cancer diagnosed in women. Anthracyclines and taxanes are the most active and widely used chemotherapeutic agents in the treatment of both early-stage and advanced breast cancer. In the past decade, novel formulations of these cytotoxic agents have been developed to improve efficacy and decrease toxicity. nab-paclitaxel is a solvent-free, albumin-bound 130-nm particle form of paclitaxel (Abraxane, Abraxis Bioscience, CA, USA), which was developed to avoid toxicities associated with the Cremophor vehicle used in solvent-based paclitaxel. In a Phase III study, nab-paclitaxel demonstrated higher response rates, a better safety profile compared with conventional paclitaxel, and improved survival in patients receiving it as second-line therapy. Based on this pivotal trial, nab-paclitaxel is now approved in the USA for treatment of breast cancer after failure of combination chemotherapy for metastatic disease or relapse within 6 months of adjuvant therapy where prior therapy included an anthracycline unless clinically contraindicated. Recently, several Phase II studies have suggested a role for nab-paclitaxel as a single agent and in combination with other agents for first-line treatment of metastatic breast cancer. Studies are ongoing to explore the use of nab-paclitaxel in other solid tumors such as non-small-cell lung cancer, ovarian cancer and malignant melanoma.
Collapse
Affiliation(s)
- Prakash Vishnu
- Mayo Clinic, Division of Hematology Oncology, Jacksonville, FL, USA
| | | |
Collapse
|
38
|
Fu S, Naing A, Moulder SL, Culotta KS, Madoff DC, Ng CS, Madden TL, Falchook GS, Hong DS, Kurzrock R. Phase I trial of hepatic arterial infusion of nanoparticle albumin-bound paclitaxel: toxicity, pharmacokinetics, and activity. Mol Cancer Ther 2011; 10:1300-7. [PMID: 21571911 DOI: 10.1158/1535-7163.mct-11-0259] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Because liver involvement in patients with metastatic cancer has limited options and poor outcomes, we conducted a phase I study to determine the safety, activity, and pharmacokinetic characteristics of hepatic arterial infusion of nanoparticle albumin-bound paclitaxel (HAI nab-paclitaxel). Cohorts of three patients having predominant hepatic metastases received HAI nab-paclitaxel at three dose levels (180, 220, and 260 mg/m(2), respectively) infused for more than 1 hour every 3 weeks (3 + 3 design). Some patients participated in comparative pharmacokinetic studies (i.v. vs. HAI), receiving their first course i.v., to determine peak concentrations and effect of first-pass hepatic extraction compared with subsequent courses administered by HAI. The highest dose level was expanded to determine the safety and activity of HAI nab-paclitaxel. Thirty-eight patients were treated. There were no dose-limiting toxicities at doses up to 260 mg/m(2). Common adverse events included alopecia, fatigue, myelosuppresion, nausea, and vomiting. Three patients had stable disease for 4 or more months and 2 patients (1 of 12 with breast cancer and 1 of 1 with cervical cancer) achieved a partial response lasting for 5 and 15 months, respectively. Peak concentrations were lower (∼50%) with greater hepatic extraction of drug (∼42%) following HAI than i.v. infusion based on area under the curve comparison of drug exposure. HAI nab-paclitaxel showed partial hepatic extraction. At doses 260 mg/m(2) or less given for 1 hour every 3 weeks, the treatment was well-tolerated and showed activity in advanced cancer patients with predominant liver metastases.
Collapse
Affiliation(s)
- Siqing Fu
- Department of Investigational Cancer Therapeutics, Unit 0455, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Safety and pharmacokinetic study of nab-paclitaxel plus carboplatin in chemotherapy-naïve patients with advanced non–small cell lung cancer. Invest New Drugs 2011; 30:1132-7. [DOI: 10.1007/s10637-011-9674-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 04/18/2011] [Indexed: 11/26/2022]
|
40
|
Vishnu P, Roy V. Safety and Efficacy of nab-Paclitaxel in the Treatment of Patients with Breast Cancer. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2011; 5:53-65. [PMID: 21603258 PMCID: PMC3091407 DOI: 10.4137/bcbcr.s5857] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Taxanes are highly active chemotherapeutic agents in the treatment of early-stage and metastatic breast cancer. Novel formulations have been developed to improve efficacy and decrease toxicity associated with these cytotoxic agents. nab-paclitaxel is a solvent free, albumin-bound 130-nanometer particle formulation of paclitaxel (Abraxane(®), Abraxis Bioscience), which was developed to avoid toxicities of the Cremophor vehicle used in solvent-based paclitaxel. In a phase III clinical trial, nab-paclitaxel demonstrated higher response rates, better safety and side-effect profile compared to conventional paclitaxel, and improved survival in patients receiving it as second line therapy. Higher doses can be administered over a shorter infusion time without the need for special infusion sets or pre-medications. It is now approved in the US for treatment of breast cancer after failure of combination chemotherapy for metastatic disease or relapse within 6 months of adjuvant therapy, where prior therapy included an anthracycline. Recently, several phase II studies have suggested a role for nab-paclitaxel as a single agent and in combination with other agents for first-line treatment of metastatic breast cancer.
Collapse
Affiliation(s)
- Prakash Vishnu
- Division of Hematology Oncology, Mayo Clinic, Jacksonville, FL, USA
| | | |
Collapse
|
41
|
Conlin AK, Seidman AD, Bach A, Lake D, Dickler M, D'Andrea G, Traina T, Danso M, Brufsky AM, Saleh M, Clawson A, Hudis CA. Phase II trial of weekly nanoparticle albumin-bound paclitaxel with carboplatin and trastuzumab as first-line therapy for women with HER2-overexpressing metastatic breast cancer. Clin Breast Cancer 2010; 10:281-7. [PMID: 20705560 DOI: 10.3816/cbc.2010.n.036] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE This multicenter phase II trial evaluated the efficacy and safety of weekly nanoparticle albumin-bound paclitaxel with carboplatin and weekly trastuzumab as first-line therapy for women with HER2-overexpressing metastatic breast cancer (MBC). PATIENTS AND METHODS We treated 32 patients who had measurable MBC that was HER2-positive defined by an immunohistochemical staining score of 3+ or gene amplification by fluorescence in situ hybridization, required for those with an IHC of 2+. Patients were treated with albumin-bound paclitaxel 100 mg/m2 and carboplatin at area under the curve (AUC) = 2 on days 1, 8, and 15 of a 28-day cycle. Trastuzumab was administered at 2 mg/kg weekly after a loading dose of 4 mg/kg. Because of hypersensitivity reactions occurring during carboplatin infusion numbers 6-8 in 4 of the first 13 patients with this premedication-free regimen, the protocol was amended for carboplatin and dosed at AUC = 6 day 1 each 28-day cycle, in lieu of introducing steroid prophylaxis. Patients were treated with 6 cycles and allowed to continue with all 3 drugs or trastuzumab alone if free of progression and unacceptable toxicity after 6 cycles. RESULTS The overall response rate (ORR) was 62.5% (95% CI, 45.7%-79.3%) with 3 confirmed complete responders (CRs; 9%) and 17 confirmed partial responses (PRs; 53%). An additional 6 patients (19%) had stable disease (SD) for greater than 16 weeks for a clinical benefit rate (ORR + SD > 16 weeks) of 81%. As of April 16, 2009, 20 patients (63%) had progressed with a median progression-free survival (PFS) of 16.6 months (95% CI, 7.5-26.5 months). Antitumor activity was similar for patients treated with weekly carboplatin and every-4-week carboplatin (ORR, 65% vs. 67%, respectively). Hematologic toxicities were the only grade 4 toxicities noted and were infrequent with grade 4 neutropenia in 3 patients (9%) and 1 febrile neutropenia. Grade 2/3 peripheral neuropathy was uncommon (13%/3%). CONCLUSION Weekly albumin-bound paclitaxel with carboplatin and trastuzumab is highly active in HER2-overexpressing MBC. In the absence of corticosteroid premedication, which we avoided with albumin-bound paclitaxel, carboplatin seems best dosed every 4 weeks rather than weekly because of carboplatin-associated hypersensitivity reactions. The regimen was very well tolerated with few grade 3 and 4 nonhematologic toxicities experienced, and severe hematologic toxicity and peripheral neuropathy were infrequent.
Collapse
Affiliation(s)
- Alison K Conlin
- Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kottschade LA, Suman VJ, Amatruda T, McWilliams RR, Mattar BI, Nikcevich DA, Behrens R, Fitch TR, Jaslowski AJ, Markovic SN. A phase II trial of nab-paclitaxel (ABI-007) and carboplatin in patients with unresectable stage IV melanoma : a North Central Cancer Treatment Group Study, N057E(1). Cancer 2010; 117:1704-10. [PMID: 21472717 DOI: 10.1002/cncr.25659] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Revised: 08/17/2010] [Accepted: 08/18/2010] [Indexed: 11/06/2022]
Abstract
BACKGROUND There is increasing evidence that paclitaxel and carboplatin are clinically active in the treatment of metastatic melanoma (MM). ABI-007 is an albumin-bound formulation of paclitaxel that has demonstrated single-agent activity against metastatic melanoma. METHODS A parallel phase II trial was conducted in patients with unresectable stage IV melanoma who were either chemotherapy naive (CN) or previously treated (PT). The treatment regimen consisted of ABI-007 (100 mg/m(2) ) and carboplatin area under the curve (AUC2) administered on days 1, 8, and 15 every 28 days. The primary aim of this study was objective response rate (RECIST). RESULTS Seventy-six patients (41 CN and 35 PT) were enrolled between November 2006 and July 2007. Three patients withdrew consent prior to starting treatment. The median number of treatment cycles was 4. There were 10 (25.6%) responses (1 complete response [CR] and 9 partial responses [PRs]) in the CN cohort (90% CI, 16.7%-42.3%) and 3 (8.8%) responses (3 PRs) in the PT cohort (90% CI, 2.5%-21.3%). Median progression-free survival was 4.5 months in the CN cohort and 4.1 months in the PT cohort. Median overall survival (OS) was 11.1 months in the CN group and 10.9 months in the PT group. Severe toxicities in both groups (Common Terminology Criteria for Adverse Effects v.3.0 ≥grade 3) included neutropenia, thrombocytopenia, neurosensory problems, fatigue, nausea, and vomiting. CONCLUSIONS The weekly combination of ABI-007 and carboplatin appears to be moderately well tolerated, with promising clinical activity as therapy in patients who are chemotherapy naive and with modest antitumor activity in those previously treated.
Collapse
|
43
|
A dose finding study of weekly and every-3-week nab-Paclitaxel followed by carboplatin as first-line therapy in patients with advanced non-small cell lung cancer. J Thorac Oncol 2010; 5:852-61. [PMID: 20521351 DOI: 10.1097/jto.0b013e3181d5e39e] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION This nonrandomized study aimed to identify the optimal dose of every-3-week (q3w) and weekly nab-paclitaxel plus q3w carboplatin as first-line therapy in patients with advanced non-small cell lung cancer (NSCLC) for a phase 3 trial. METHODS Previously untreated patients with advanced NSCLC enrolled sequentially into seven cohorts (25 patients/cohort, N = 175). Cohorts 1 to 4 and 5 to 7 received nab-paclitaxel q3w and weekly, respectively. Patients were evaluated for efficacy and safety. RESULTS The most common treatment-related > or = grade 3 adverse events were neutropenia (60%), neuropathy (19%), fatigue (9%), and thrombocytopenia (29%) (no grade 4 neuropathy or fatigue). A 100 mg/m(2) weekly nab-paclitaxel produced less serious adverse events than other doses/schedules. Response rate (RR) was greater in the weekly versus q3w cohorts (47% vs. 30%). Median progression-free survival (PFS) ranged from 4.8 to 6.9 months, and overall survival (OS) ranged from 8.3 to 15.0 months (all cohorts). Patients receiving 100 mg/m(2) weekly nab-paclitaxel achieved 48% RR with 6.2 and 11.3 months of PFS and OS, respectively. In a retrospective analysis, patients with nonsquamous cell carcinoma receiving weekly nab-paclitaxel had significantly improved RR (59.4% vs. 23.5%, respectively, p = 0.003), and >2 months longer PFS and OS compared with q3w schedule. In patients with squamous cell carcinoma, the q3w schedule significantly increased PFS by 3 months (p = 0.014) and OS by >2 months (no difference in RR) compared with the weekly schedule. CONCLUSION nab-Paclitaxel plus carboplatin is an effective therapy for advanced NSCLC. Based on favorable efficacy and safety profiles, a phase 3, randomized, multicenter study comparing 100 mg/m(2) weekly nab-paclitaxel plus q3w carboplatin to solvent-based paclitaxel plus carboplatin has enrolled patients.
Collapse
|
44
|
Abstract
Microtubules are dynamic filamentous cytoskeletal proteins composed of tubulin and are an important therapeutic target in tumour cells. Agents that bind to microtubules have been part of the pharmacopoeia of anticancer therapy for decades and until the advent of targeted therapy, microtubules were the only alternative to DNA as a therapeutic target in cancer. The screening of a range of botanical species and marine organisms has yielded promising new antitubulin agents with novel properties. In the current search for novel microtubule-binding agents, enhanced tumour specificity, reduced neurotoxicity and insensitivity to chemoresistance mechanisms are the three main objectives.
Collapse
Affiliation(s)
- Charles Dumontet
- INSERM 590, Faculté Rockefeller, 8 Avenue Rockefeller, 69008 Lyon, France and Université Lyon 1, ISPB, Lyon, F-69003, France.
| | | |
Collapse
|
45
|
Salazar R, Plummer R, Oaknin A, Robinson A, Pardo B, Soto-Matos A, Yovine A, Szyldergemajn S, Calvert AH. Phase I study of weekly plitidepsin as 1-hour infusion combined with carboplatin in patients with advanced solid tumors or lymphomas. Invest New Drugs 2010; 29:1406-13. [PMID: 20623160 DOI: 10.1007/s10637-010-9488-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 06/24/2010] [Indexed: 11/28/2022]
Abstract
This dose-escalating phase I clinical trial was designed to determine the recommended dose (RD) and to assess the safety and feasibility of weekly plitidepsin (1-hour i.v. infusion, Days 1, 8 and 15) combined with carboplatin (1-hour i.v. infusion, Day 1, after plitidepsin) in 4-week (q4wk) cycles given to patients with advanced solid tumors or lymphomas. Twenty patients were enrolled and evaluable for both safety and efficacy. The starting dose was plitidepsin 1.8 mg/m(2) and carboplatin area under the curve (AUC) = 5 min*mg/ml; dose escalation proceeded based on worst toxicity in the previous cohort. The maximum tolerated dose (MTD) was plitidepsin 3.0 mg/m(2) and carboplatin AUC = 5 min*mg/ml, with grade 3 transaminase increases as the most common dose-limiting toxicities (DLTs). The RD for phase II studies was plitidepsin 2.4 mg/m(2) and carboplatin AUC = 5 min*mg/ml, with fatigue, myalgia and nausea as the most common drug-related adverse events (AEs). No unexpected toxicity was seen. Twelve patients (60%), ten of whom were heavily pretreated (≥2 previous chemotherapy lines) showed stable disease (SD), with a median time to progression (TTP) of 4.4 months. In conclusion, plitidepsin 2.4 mg/m(2) and carboplatin AUC = 5 min*mg/ml is a safe dose for future phase II studies evaluating the use of this combination in cancer patients potentially sensitive to platinum-based therapy.
Collapse
Affiliation(s)
- Ramón Salazar
- Instituto Catalán de Oncología, Ctra. Gran Vía, s/n, L'Hospitalet de Llobregat, 08907, Barcelona, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Zolnik BS, González-Fernández A, Sadrieh N, Dobrovolskaia MA. Nanoparticles and the immune system. Endocrinology 2010; 151:458-65. [PMID: 20016026 PMCID: PMC2817614 DOI: 10.1210/en.2009-1082] [Citation(s) in RCA: 599] [Impact Index Per Article: 39.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Accepted: 11/19/2009] [Indexed: 11/19/2022]
Abstract
Today nanotechnology is finding growing applications in industry, biology, and medicine. The clear benefits of using nanosized products in various biological and medical applications are often challenged by concerns about the lack of adequate data regarding their toxicity. One area of interest involves the interactions between nanoparticles and the components of the immune system. Nanoparticles can be engineered to either avoid immune system recognition or specifically inhibit or enhance the immune responses. We review herein reported observations on nanoparticle-mediated immunostimulation and immunosuppression, focusing on possible theories regarding how manipulation of particle physicochemical properties can influence their interaction with immune cells to attain desirable immunomodulation and avoid undesirable immunotoxicity.
Collapse
Affiliation(s)
- Banu S Zolnik
- Center for Drug Evaluation and Research, Office of Pharmaceutical Science, Food and Drug Administration, Silver Spring, Maryland 20993, USA
| | | | | | | |
Collapse
|
47
|
Masellis AM, Sielaff TD, Bender GP. Successful treatment of metastatic pancreatic adenocarcinoma with combination chemotherapy regimens. Int J Clin Oncol 2009; 14:478-81. [DOI: 10.1007/s10147-008-0873-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Accepted: 12/23/2008] [Indexed: 01/17/2023]
|
48
|
Steinhauser I, Langer K, Strebhardt K, Spänkuch B. Uptake of plasmid-loaded nanoparticles in breast cancer cells and effect on Plk1 expression. J Drug Target 2009; 17:627-37. [PMID: 19591537 DOI: 10.1080/10611860903118823] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The development of nucleic acid-based drugs for cancer therapeutic application has shown promising results in the past. However the delivery of these drugs to target cells is one problem which remains to be resolved. Nanoparticles have been described as promising strategies to deliver drugs into target cells. Human serum albumin (HSA) nanoparticles conjugated to trastuzumab for a cell type-specific targeting of human epidermal growth factor receptor 2 (HER2)-overexpressing cells were developed with incorporated expression plasmids for small hairpin RNAs (shRNAs) targeting polo-like kinase 1 (Plk1). Plk1 is a promising target for such an approach because it is overexpressed in all known cancer types and is a negative prognostic factor. Receptor-mediated uptake of the trastuzumab-modified nanoparticles into HER2-positive cells could be observed leading to reduced Plk1 expression. Taken together, HSA nanoparticles represent promising tools to deliver expression plasmids for shRNAs into target cells and should be further evaluated with regard to a therapeutic application of RNA interference in cancer therapy.
Collapse
Affiliation(s)
- Isabel Steinhauser
- Institute of Pharmaceutical Technology, Biocenter of Goethe-University, Frankfurt, Germany
| | | | | | | |
Collapse
|
49
|
Yang AS, Chapman PB. The history and future of chemotherapy for melanoma. Hematol Oncol Clin North Am 2009; 23:583-97, x. [PMID: 19464604 DOI: 10.1016/j.hoc.2009.03.006] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Melanoma is considered a chemotherapy-resistant cancer, but in reality there are several chemotherapy drugs with significant single-agent activity. Response rates to combination regimens are reproducibly higher than with standard dacarbazine, but of the randomized trials comparing combination regimens with dacarbazine, none were of sufficient size to detect a realistic effect on survival. Similarly, adjuvant chemotherapy has not had a realistic test in melanoma. Response to chemotherapy is associated reproducibly with better survival rates suggesting that regimens with higher response rates are needed. Recent observations suggest that combining antiangiogenic agents with either dacarbazine or temozolomide can double response rates. These combinations are worthy of further investigation and might serve as a foundation on which to build a combination regimen that improves overall survival in metastatic melanoma patients.
Collapse
Affiliation(s)
- Arvin S Yang
- Melanoma/Sarcoma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | | |
Collapse
|
50
|
Mansfield AS, Markovic SN. Novel therapeutics for the treatment of metastatic melanoma. Future Oncol 2009; 5:543-57. [PMID: 19450181 DOI: 10.2217/fon.09.15] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Metastatic malignant melanoma is an incurable disease with a median survival of 8.5 months and a probability of surviving 5 years after the diagnosis of less than 5%. To date, no systemic therapy has meaningfully changed these survival end points. Currently, in the USA the FDA has approved three agents for the treatment of metastatic melanoma: hydroxyurea, dacarbazine and interleukin-2. None of these have demonstrated a meaningfully prolonged survival of patients with metastatic melanoma. Therefore, a number of innovative therapeutic strategies have been pursued to improve outcomes, including immune therapy, tyrosine kinase inhibitors and angiogenesis inhibitors. Herein, we review some of the recent advances in novel therapeutic developments for the treatment of metastatic melanoma.
Collapse
Affiliation(s)
- Aaron S Mansfield
- Department of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| | | |
Collapse
|