151
|
Targeting of phage particles towards endothelial cells by antibodies selected through a multi-parameter selection strategy. Sci Rep 2017; 7:42230. [PMID: 28186116 PMCID: PMC5301479 DOI: 10.1038/srep42230] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 01/06/2017] [Indexed: 12/18/2022] Open
Abstract
One of the hallmarks of cancer is sustained angiogenesis. Here, normal endothelial cells are activated, and their formation of new blood vessels leads to continued tumour growth. An improved patient condition is often observed when angiogenesis is prevented or normalized through targeting of these genomically stable endothelial cells. However, intracellular targets constitute a challenge in therapy, as the agents modulating these targets have to be delivered and internalized specifically to the endothelial cells. Selection of antibodies binding specifically to certain cell types is well established. It is nonetheless a challenge to ensure that the binding of antibodies to the target cell will mediate internalization. Previously selection of such antibodies has been performed targeting cancer cell lines; most often using either monovalent display or polyvalent display. In this article, we describe selections that isolate internalizing antibodies by sequential combining monovalent and polyvalent display using two types of helper phages, one which increases display valence and one which reduces background. One of the selected antibodies was found to mediate internalization into human endothelial cells, although our results confirms that the single stranded nature of the DNA packaged into phage particles may limit applications aimed at targeting nucleic acids in mammalian cells.
Collapse
|
152
|
Mozhi A, Ahmad I, Okeke CI, Li C, Liang XJ. pH-sensitive polymeric micelles for the Co-delivery of proapoptotic peptide and anticancer drug for synergistic cancer therapy. RSC Adv 2017. [DOI: 10.1039/c6ra27054a] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
pH-sensitive polymeric micelles for targeted co-delivery of mitochondria-damaged proapoptotic peptide and DTX for synergistic cancer therapy.
Collapse
Affiliation(s)
- Anbu Mozhi
- Laboratory of Controllable Nanopharmaceuticals
- Chinese Academy of Sciences (CAS)
- Beijing 100190
- China
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
| | - Israr Ahmad
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
- National Center for Nanoscience and Technology
- Beijing 100190
- China
- University of Chinese Academy of Sciences
| | - Chukwunweike Ikechukwu Okeke
- Laboratory of Controllable Nanopharmaceuticals
- Chinese Academy of Sciences (CAS)
- Beijing 100190
- China
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
| | - Chan Li
- Laboratory of Controllable Nanopharmaceuticals
- Chinese Academy of Sciences (CAS)
- Beijing 100190
- China
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
| | - Xing-Jie Liang
- Laboratory of Controllable Nanopharmaceuticals
- Chinese Academy of Sciences (CAS)
- Beijing 100190
- China
- Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety
| |
Collapse
|
153
|
Abramczyk H, Brozek-Pluska B, Surmacki J, Tondusson M, Freysz E. Photostability of biological systems—Femtosecond dynamics of zinc tetrasulfonated phthalocyanine at cancerous and noncancerous human Breast tissues. J Photochem Photobiol A Chem 2017. [DOI: 10.1016/j.jphotochem.2016.08.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
154
|
Design of novel chemotherapeutic delivery systems for colon cancer therapy based on oral polymeric nanoparticles. Ther Deliv 2017; 8:29-47. [DOI: 10.4155/tde-2016-0058] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Oral delivery of cancer chemotherapeutic drugs (CCDs) is subject matter in the 21st century, which changes the dosage regimens of oncotherapy with enhancement in patient’s life and deducts the cost of therapy. The present report explored on the nano-oncology such as polymeric nanoparticles (PNPs) as an oral CCDs delivery vehicle, showing great potential for colon cancer treatment. Proof-of-concept in vitro and in vivo results for delivery of CCDs using various oral PNPs are included in this review from the literatures. Subsequently, the gastrointestinal barriers for oral chemotherapy have been highlighted. Furthermore, PNPs achieving better accumulation in the cancer region by desirable quality of their passive- and active-targeting phenomena have also been highlighted.
Collapse
|
155
|
Fabian KL, Storkus WJ. Immunotherapeutic Targeting of Tumor-Associated Blood Vessels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1036:191-211. [PMID: 29275473 DOI: 10.1007/978-3-319-67577-0_13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Pathological angiogenesis occurs during tumor progression and leads in the formation of an abnormal vasculature in the tumor microenvironment (TME). The tumor vasculature is disorganized, tortuous and leaky, resulting in high interstitial pressure and hypoxia in the TME, all of which are events that support tumor growth and survival. Given the sustaining role of the tumor vasculature, it has become an increasingly attractive target for the development of anti-cancer therapies. Antibodies, tyrosine kinase inhibitors and cancer vaccines that target pro-angiogenic factors, angiogenesis-associated receptors or tumor blood vessel-associated antigens continue to be developed and tested for therapeutic efficacy. Preferred anti-angiogenic protocols include those that "normalize" the tumor-associated vasculature which reduce hypoxia and improve tumor blood perfusion, resulting in tumor cell apoptosis, decreased immunosuppression, and enhanced effector immune cell infiltration/tumoricidal action within the TME.
Collapse
Affiliation(s)
- Kellsye L Fabian
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Dermatology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
156
|
Fan Z, Ley K. Leukocyte arrest: Biomechanics and molecular mechanisms of β2 integrin activation. Biorheology 2016; 52:353-77. [PMID: 26684674 DOI: 10.3233/bir-15085] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Integrins are a group of heterodimeric transmembrane receptors that play essential roles in cell-cell and cell-matrix interaction. Integrins are important in many physiological processes and diseases. Integrins acquire affinity to their ligand by undergoing molecular conformational changes called activation. Here we review the molecular biomechanics during conformational changes of integrins, integrin functions in leukocyte biorheology (adhesive functions during rolling and arrest) and molecules involved in integrin activation.
Collapse
Affiliation(s)
- Zhichao Fan
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA
| | - Klaus Ley
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037, USA.,Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
157
|
Nemudraya AA, Kuligina EV, Ilyichev AA, Fomin AS, Stepanov GA, Savelyeva AV, Koval OA, Richter VA. Selection of antitumor displayed peptides for the specific delivery of the anticancer drug lactaptin. Oncol Lett 2016; 12:4547-4555. [PMID: 28105163 PMCID: PMC5228560 DOI: 10.3892/ol.2016.5266] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 09/09/2016] [Indexed: 11/24/2022] Open
Abstract
It has been previously demonstrated that lactaptin, the proteolytic fragment of human milk protein κ-casein, induces the death of various cultured cancer cells. The recombinant analog of lactaptin, RL2, effectively induces the apoptosis of mouse hepatocarcinoma-1 (HA-1) tumor cells in vitro and suppress the growth of HA-1 tumors and metastases in vivo. The antitumor drug Lactaptin developed on the basis of RL2 has been successful in preclinical trials. Lactaptin shows its efficiency in relation to mouse and human cancer cells and tumors. However, Lactaptin, as with the majority of protein-based therapeutic drugs, is distributed evenly throughout the organism, which reduces its antitumor efficacy. To develop the targeted delivery of lactaptin, the present study selected tumor-specific peptides by screening a phage display peptide library in vivo on A/Sn strain mice with subcutaneously transplanted HA-1 cells. Two genetic constructs were made for the production of recombinant fusion proteins composed of RL2 and the selected tumor-targeting peptide. In vitro experiments involving HA-1, MDA-MB-231 and MCF-7 cells cultures demonstrated that the fusion proteins induce apoptotic death in mouse and human tumor cells, as with RL2. The in vivo experiments involving the mouse HA-1 tumor model demonstrated that the tumor fluorescence intensity of the Cy5-fusion protein conjugates is higher than that of RL2-Cy5. As conjugation of the tumor-specific peptides to RL2 provided retention of RL2 in the tumor tissues, fusion proteins composed of lactaptin and peptides specific for human tumors are deemed promising to improve the antitumor efficiency of lactaptin.
Collapse
Affiliation(s)
- Anna Andreevna Nemudraya
- Laboratory of Biotechnology, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of The Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Elena Vladimirovna Kuligina
- Laboratory of Biotechnology, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of The Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Alexandr Alexeevich Ilyichev
- Department of Bioengineering, State Research Center of Virology and Biotechnology VECTOR, 630559 Koltsovo, Russia; Department of Biology, Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Alexandr Sergeevich Fomin
- Laboratory of Biotechnology, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of The Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Grigory Alexandrovich Stepanov
- Laboratory of Biotechnology, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of The Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Anna Valentinovna Savelyeva
- Laboratory of Biotechnology, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of The Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Olga Alexandrovna Koval
- Laboratory of Biotechnology, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of The Russian Academy of Sciences, 630090 Novosibirsk, Russia; Department of Biology, Faculty of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Vladimir Alexandrovich Richter
- Laboratory of Biotechnology, Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of The Russian Academy of Sciences, 630090 Novosibirsk, Russia
| |
Collapse
|
158
|
Liu C, Yao S, Li X, Wang F, Jiang Y. iRGD-mediated core-shell nanoparticles loading carmustine and O 6-benzylguanine for glioma therapy. J Drug Target 2016; 25:235-246. [PMID: 27646474 DOI: 10.1080/1061186x.2016.1238091] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
iRGD (internalizing RGD) with high affinity to αν integrins was reported to enhance tumor penetrability by binding to neuropilin-1 (NRP-1). Based on our previous study, chitosan surface-modified poly (lactide-co-glycolides) nanoparticles (PLGA/CS NPs), loaded with carmustine (BCNU) and its sensitizer (O6-benzylguanine, BG) showed stronger anti-tumor effect than free drugs. In present study, PLGA/CS NPs (NPs) with core-shell structure were prepared and modified with iRGD or mPEG. F98, C6 or U87 cell lines with different receptors levels were selected for in vitro and in vivo studies. After administration of iRGD-mediated NPs, including iRGD-modified NPs (iRGD-NPs) and co-administration of iRGD and NPs (iRGD + NPs), their effects on glioma were compared with NPs. iRGD-NPs showed stronger cytotoxicity and cellular uptake than other groups. iRGD-NPs and iRGD + NPs displayed deeper tumor penetration and stronger anti-invasion effect on three dimensional (3D) glioma spheroids than NPs. On F98 glioma-bearing mice model, iRGD-mediated NPs showed enhanced crossing BBB ability and brain tumor accumulation levels. Correspondingly, the median survival time of iRGD + NPs, iRGD-NPs and NPs groups were 58, 49 and 34.5 days, respectively. Present studies supported the iRGD-mediated strategy to improve the efficacy of antitumor drug delivery system. Importantly, co-administration of iRGD may be a greater way over the conjugation of iRGD.
Collapse
Affiliation(s)
- Chang Liu
- a Key Laboratory of Smart Drug Delivery, Ministry of Education and PLA, Department of pharmaceutics , School of Pharmacy, Fudan University , Shanghai , China
| | - Sen Yao
- a Key Laboratory of Smart Drug Delivery, Ministry of Education and PLA, Department of pharmaceutics , School of Pharmacy, Fudan University , Shanghai , China
| | - Xuqian Li
- a Key Laboratory of Smart Drug Delivery, Ministry of Education and PLA, Department of pharmaceutics , School of Pharmacy, Fudan University , Shanghai , China
| | - Feng Wang
- a Key Laboratory of Smart Drug Delivery, Ministry of Education and PLA, Department of pharmaceutics , School of Pharmacy, Fudan University , Shanghai , China
| | - Yanyan Jiang
- a Key Laboratory of Smart Drug Delivery, Ministry of Education and PLA, Department of pharmaceutics , School of Pharmacy, Fudan University , Shanghai , China
| |
Collapse
|
159
|
Nemudraya AA, Makartsova AA, Fomin AS, Nushtaeva AA, Koval OA, Richter VA, Kuligina EV. Tumor-Specific Peptide, Selected from a Phage Peptide Library, Enhances Antitumor Activity of Lactaptin. PLoS One 2016; 11:e0160980. [PMID: 27513518 PMCID: PMC4981335 DOI: 10.1371/journal.pone.0160980] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 07/27/2016] [Indexed: 01/04/2023] Open
Abstract
A recombinant analogue of lactaptin (RL2), a new potential anticancer molecule, induces apoptosis in cultured tumor cells. The tumor suppression efficacy of RL2 was shown against mouse hepatoma-1 cells and MDA-MB-231 human breast adenocarcinoma cells. The RL2-based therapeutic drug lactaptin is distributed evenly throughout the organism, which reduces its antitumor efficacy. In the current study, we obtained a genetic construct that allows production of the recombinant fusion protein T3-RL2, consisting of RL2 and T3 peptide (YTYDPWLIFPAN), in E. coli cells. T3 peptide was selected from a phage peptide library as a result of two screenings: in vitro using MDA-MB-231 cell culture and in vivo using a mouse xenograft model of breast cancer MDA-MB-231. It was shown that the displayed peptide T3 provides binding and internalization of phage particles by MDA-MB-231 cells and their specific accumulation in MDA-MB-231 tumor tissue. In addition, based on the nucleotide sequences coding RL2 and the known tumor-targeting peptide iRGD, we obtained genetic constructs that provide synthesis of fusion proteins RL2-iRGD and RL-iRGD-His. We studied the cytotoxic activity of fusion proteins T3-RL2, RL2-iRGD and RL-iRGD-His in vitro using MDA-MB-231 and MCF-7 human adenocarcinoma cells. The in vitro results showed that the fusion proteins inhibit proliferation of both cell cultures, and their cytotoxic activity is higher than that of RL2. In vivo experiments on the study of the antitumor efficacy of the obtained fusion proteins demonstrated that T3-RL2 protein significantly inhibits MDA-MB-231 tumor growth in a xenograft model compared with RL2, while the antitumor effect of RL2-iRGD and RL-iRGD-His proteins is comparable to the effect of RL2.
Collapse
Affiliation(s)
- Anna A. Nemudraya
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
- * E-mail:
| | - Anna A. Makartsova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Alexandr S. Fomin
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Anna A. Nushtaeva
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Olga A. Koval
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Vladimir A. Richter
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| | - Elena V. Kuligina
- Institute of Chemical Biology and Fundamental Medicine SB RAS, Novosibirsk, Russia
| |
Collapse
|
160
|
Herranz-Blanco B, Shahbazi MA, Correia AR, Balasubramanian V, Kohout T, Hirvonen J, Santos HA. pH-Switch Nanoprecipitation of Polymeric Nanoparticles for Multimodal Cancer Targeting and Intracellular Triggered Delivery of Doxorubicin. Adv Healthc Mater 2016; 5:1904-16. [PMID: 27245691 DOI: 10.1002/adhm.201600160] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 04/15/2016] [Indexed: 11/10/2022]
Abstract
Theranostic nanoparticles are emerging as potent tools for noninvasive diagnosis, treatment, and monitoring of solid tumors. Herein, an advanced targeted and multistimuli responsive theranostic platform is presented for the intracellular triggered delivery of doxorubicin. The system consists of a polymeric-drug conjugate solid nanoparticle containing encapsulated superparamagnetic iron oxide nanoparticles (IO@PNP) and decorated with a tumor homing peptide, iRGD. The production of this nanosystem is based on a pH-switch nanoprecipitation method in organic-free solvents, making it ideal for biomedical applications. The nanosystem shows sufficient magnetization saturation for magnetically guided therapy along with reduced cytotoxicity and hemolytic effects. IO@PNP are largely internalized by endothelial and metastatic cancer cells and iRGD decorated IO@PNP moderately enhance their internalization into endothelial cells, while no enhancement is found for the metastatic cancer cells. Poly(ethylene glycol)-block-poly(histidine) with pH-responsive and proton-sponge properties promotes prompt lysosomal escape once the nanoparticles are endocyted. In addition, the polymer-doxorubicin conjugate solid nanoparticles show both intracellular lysosomal escape and efficient translocation of doxorubicin to the nuclei of the cells via cleavage of the amide bond. Overall, IO@PNP-doxorubicin and the iRGD decorated counterpart demonstrate to enhance the toxicity of doxorubicin in cancer cells by improving the intracellular delivery of the drug carried in the IO@PNP.
Collapse
Affiliation(s)
- Bárbara Herranz-Blanco
- Division of Pharmaceutical Chemistry and Technology; Faculty of Pharmacy; University of Helsinki; Viikinkaari 5 E (P. O. Box 56) 00014 Helsinki Finland
| | - Mohammad-Ali Shahbazi
- Division of Pharmaceutical Chemistry and Technology; Faculty of Pharmacy; University of Helsinki; Viikinkaari 5 E (P. O. Box 56) 00014 Helsinki Finland
| | - Alexandra R. Correia
- Division of Pharmaceutical Chemistry and Technology; Faculty of Pharmacy; University of Helsinki; Viikinkaari 5 E (P. O. Box 56) 00014 Helsinki Finland
| | - Vimalkumar Balasubramanian
- Division of Pharmaceutical Chemistry and Technology; Faculty of Pharmacy; University of Helsinki; Viikinkaari 5 E (P. O. Box 56) 00014 Helsinki Finland
| | - Tomáš Kohout
- Department of Physics; University of Helsinki; Gustaf Hällströmin katu 2a (P. O. Box 64) 00560 Helsinki Finland
| | - Jouni Hirvonen
- Division of Pharmaceutical Chemistry and Technology; Faculty of Pharmacy; University of Helsinki; Viikinkaari 5 E (P. O. Box 56) 00014 Helsinki Finland
| | - Hélder A. Santos
- Division of Pharmaceutical Chemistry and Technology; Faculty of Pharmacy; University of Helsinki; Viikinkaari 5 E (P. O. Box 56) 00014 Helsinki Finland
| |
Collapse
|
161
|
Thejass P, Kuttan G. Inhibition of Endothelial Cell Differentiation and Proinflammatory Cytokine Production During Angiogenesis by Allyl Isothiocyanate and Phenyl Isothiocyanate. Integr Cancer Ther 2016; 6:389-99. [DOI: 10.1177/1534735407309084] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Angiogenesis is a crucial step in the growth and metastasis of cancers. The activation of endothelial cells and their further behavior are very critical during angiogenesis. The authors analyze the effect of allyl isothiocyanate (AITC) and phenyl isothiocyanate (PITC) on angiogenesis in an in vitro model using human umbilical vein endothelial cells (HUVECs). AITC and PITC significantly inhibited endothelial cell migration, invasion, and tube formation. 3H-thymidine proliferation assay showed that AITC and PITC significantly inhibited the proliferation of HUVECs in vitro. The authors also studied the effect of AITC and PITC on the serum cytokine profiles of angiogenesis-induced animals and found that these compounds are highly potent in the downregulation of vascular endothelial growth factor (VEGF) and proinflammatory cytokines such as interleukin (IL)—1β , IL-6, granulocyte macrophage colony-stimulating factor (GM-CSF), and tumor necrosis factor α (TNF-α). Treatment with these compounds showed an elevation in the levels of IL-2 and tissue inhibitor of metalloproteinases (TIMP)—1, which are antiangiogenic factors. Moreover, studies using B16F-10 melanoma cells showed that both AITC and PITC significantly reduced VEGF mRNA expression. These findings suggest that AITC and PITC act as angiogenesis inhibitors through the downregulation of VEGF and proinflammatory cytokines such as IL-1β, IL-6, GM-CSF, and TNF-α and upregulation of IL-2 and TIMP.
Collapse
Affiliation(s)
- P. Thejass
- Amala Cancer Research Centre, Amala Nagar, Thrissur, Kerala State, India
| | - Girija Kuttan
- Amala Cancer Research Centre, Amala Nagar, Thrissur, Kerala State, India,
| |
Collapse
|
162
|
A peptide for targeted, systemic delivery of imaging and therapeutic compounds into acute brain injuries. Nat Commun 2016; 7:11980. [PMID: 27351915 PMCID: PMC4931241 DOI: 10.1038/ncomms11980] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 05/18/2016] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is a major health and socio-economic problem, but no pharmacological agent is currently approved for the treatment of acute TBI. Thus, there is a great need for advances in this field. Here, we describe a short peptide (sequence CAQK) identified by in vivo phage display screening in mice with acute brain injury. The CAQK peptide selectively binds to injured mouse and human brain, and systemically injected CAQK specifically homes to sites of brain injury in mouse models. The CAQK target is a proteoglycan complex upregulated in brain injuries. Coupling to CAQK increased injury site accumulation of systemically administered molecules ranging from a drug-sized molecule to nanoparticles. CAQK-coated nanoparticles containing silencing oligonucleotides provided the first evidence of gene silencing in injured brain parenchyma by systemically administered siRNA. These findings present an effective targeting strategy for the delivery of therapeutics in clinical management of acute brain injuries.
Collapse
|
163
|
Tsiambas E, Lefas AY, Georgiannos SN, Ragos V, Fotiades PP, Grapsa D, Stamatelopoulos A, Kavantzas N, Patsouris E, Syrigos K. EGFR gene deregulation mechanisms in lung adenocarcinoma: A molecular review. Pathol Res Pract 2016; 212:672-7. [PMID: 27461822 DOI: 10.1016/j.prp.2016.06.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 05/22/2016] [Accepted: 06/15/2016] [Indexed: 12/26/2022]
Abstract
For the last two decades, evolution in molecular biology has expanded our knowledge in decoding a broad spectrum of genomic imbalances that progressively lead normal cells to a neoplastic state and finally to complete malignant transformation. Concerning oncogenes and signaling transduction pathways mediated by them, identification of specific gene alterations remains a critical process for handling patients by applying targeted therapeutic regimens. The epidermal growth factor receptor (EGFR) signaling pathway plays a crucial role in regulating cell proliferation, differentiation and apoptosis in normal cells. EGFR mutations and amplification represent the gene's main deregulation mechanisms in cancers of different histo-genetic origin. Furthermore, intra-cancer molecular heterogeneity due to clonal rise and expansion mainly explains the variable resistance to novel anti-EGFR monoclonal antibody (mAb), and also tyrosine kinase inhibitors (TKIs). According to recently published 2015 WHO new classification, lung cancer is the leading cause of death related to cancer and its incidence is still on the increase worldwide. The majority of patients suffering from lung cancer are diagnosed with epithelial tumors (adenocarcinoma predominantly and squamous cell carcinoma represent ∼85% of all pathologically defined lung cancer cases). In those patients, EGFR-activating somatic mutations in exons 18/19/20/21 modify patients' sensitivity (i.e. exon 21 L858R, exon 19 LREA deletion) or resistance (ie exon 20 T790M and/or insertion) to TKI mediated targeted therapeutic strategies. Additionally, the role of specific micro-RNAs that affect EGFR regulation is under investigation. In the current review, we focused on EGFR gene/protein structural and functional aspects and the corresponding alterations that occur mainly in lung adenocarcinoma to critically modify its molecular landscape.
Collapse
Affiliation(s)
- Evangelos Tsiambas
- Dept of IHC & Mol Biology, 401 GAH, Athens, Greece; Dept of Pathology, Medical School, University of Athens, Greece.
| | | | | | - Vasileios Ragos
- Dept of Maxillofacial, School of Medicine, University of Ioannina, Greece
| | | | - Dimitra Grapsa
- 3rd Dept of Medicine, Athens School of Medicine, "Sotiria" General Hospital, Athens, Greece
| | | | | | | | - Konstantinos Syrigos
- 3rd Dept of Medicine, Athens School of Medicine, "Sotiria" General Hospital, Athens, Greece
| |
Collapse
|
164
|
Lao X, Li B, Liu M, Shen C, Yu T, Gao X, Zheng H. A modified thymosin alpha 1 inhibits the growth of breast cancer both in vitro and in vivo: suppressment of cell proliferation, inducible cell apoptosis and enhancement of targeted anticancer effects. Apoptosis 2016; 20:1307-20. [PMID: 26283169 DOI: 10.1007/s10495-015-1151-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Thymosin alpha 1 (Tα1) is commonly used for treating several diseases; however its usage has been limited because of poor penetration of the target tissue, such as tumor cells. In the present study, Tα1-iRGD, a peptide by conjugating Tα1 with the iRGD fragment, was evaluated its performance in MCF-7 and MDA-MB-231 human breast cancer cells. Compared with the wild-type peptide, Tα1-iRGD was more selective in binding tumor cells in the cell attachment assay. Furthermore, the MTT assay confirmed that Tα1-iRGD proved more effective in significantly inhibiting the growth of MCF-7 cells in contrast to the general inhibition displayed by Tα1. Further, conjugation of Tα1 with iRGD preserved the immunomodulatory activity of the drug by increasing the proliferation of mouse spleen lymphocytes. Further, compared with Tα1 treatment, Tα1-iRGD treatment of MCF-7 cells considerably increased the number of cells undergoing apoptosis, resulting in a dose-dependent inhibition of cancer cell growth, which was associated with a much better effect on up-regulation of the expression of BCL2-associated X protein (Bax), caspase 9, etc. More importantly, treatment with Ta1-iRGD was more efficacious than treatment with Ta1 in vivo. This study highlights the importance of iRGD on enhancement of cell penetration and tumor accumulation. In summary, our findings demonstrate that the novel modified Tα1 developed in this study has the potential to be used for treating breast cancer.
Collapse
Affiliation(s)
- Xingzhen Lao
- School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China,
| | | | | | | | | | | | | |
Collapse
|
165
|
Pombero A, Garcia-Lopez R, Martinez S. Brain mesenchymal stem cells: physiology and pathological implications. Dev Growth Differ 2016; 58:469-80. [PMID: 27273235 DOI: 10.1111/dgd.12296] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 05/03/2016] [Accepted: 05/03/2016] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are defined as progenitor cells that give rise to a number of unique, differentiated mesenchymal cell types. This concept has progressively evolved towards an all-encompassing concept including multipotent perivascular cells of almost any tissue. In central nervous system, pericytes are involved in blood-brain barrier, and angiogenesis and vascular tone regulation. They form the neurovascular unit (NVU) together with endothelial cells, astrocytes and neurons. This functional structure provides an optimal microenvironment for neural proliferation in the adult brain. Neurovascular niche include both diffusible signals and direct contact with endothelial and pericytes, which are a source of diffusible neurotrophic signals that affect neural precursors. Therefore, MSCs/pericyte properties such as differentiation capability, as well as immunoregulatory and paracrine effects make them a potential resource in regenerative medicine.
Collapse
Affiliation(s)
- Ana Pombero
- Intituto Murciano de Investigación Biosanitaria (IMIB)-Arrixaca, University of Murcia, Murcia, Spain
| | - Raquel Garcia-Lopez
- Instituto de Neurociencias, Universidad Miguel Hernandez-Consejo Superior de Investigaciones, Av Ramon y Cajal s/n, San Juan de Alicante, 03550, Spain
| | - Salvador Martinez
- Instituto de Neurociencias, Universidad Miguel Hernandez-Consejo Superior de Investigaciones, Av Ramon y Cajal s/n, San Juan de Alicante, 03550, Spain
| |
Collapse
|
166
|
Molecular Imaging of Angiogenesis and Vascular Remodeling in Cardiovascular Pathology. J Clin Med 2016; 5:jcm5060057. [PMID: 27275836 PMCID: PMC4929412 DOI: 10.3390/jcm5060057] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 05/19/2016] [Accepted: 05/31/2016] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis and vascular remodeling are involved in a wide array of cardiovascular diseases, from myocardial ischemia and peripheral arterial disease, to atherosclerosis and aortic aneurysm. Molecular imaging techniques to detect and quantify key molecular and cellular players in angiogenesis and vascular remodeling (e.g., vascular endothelial growth factor and its receptors, αvβ3 integrin, and matrix metalloproteinases) can advance vascular biology research and serve as clinical tools for early diagnosis, risk stratification, and selection of patients who would benefit most from therapeutic interventions. To target these key mediators, a number of molecular imaging techniques have been developed and evaluated in animal models of angiogenesis and vascular remodeling. This review of the state of the art molecular imaging of angiogenesis and vascular (and valvular) remodeling, will focus mostly on nuclear imaging techniques (positron emission tomography and single photon emission tomography) that offer high potential for clinical translation.
Collapse
|
167
|
Lunasin suppresses the migration and invasion of breast cancer cells by inhibiting matrix metalloproteinase-2/-9 via the FAK/Akt/ERK and NF-κB signaling pathways. Oncol Rep 2016; 36:253-62. [PMID: 27175819 DOI: 10.3892/or.2016.4798] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/01/2016] [Indexed: 11/05/2022] Open
Abstract
Lunasin is a naturally existing bioactive peptide with an Arg-Gly-Asp (RGD) motif, which competes with integrins to bind with the extracellular matrix (ECM) consequently suppressing the integrin-mediated signaling pathway. Owing to the RGD motif, lunasin has been proven as an effective anti-inflammatory, antitumor and antimetastatic agent in many types of cancer. However, knowledge of its inhibitory effect on metastasis and the related mechanism of action in breast cancer cells is limited. In this study, the inhibitory effect of lunasin on the proliferation, migration and invasion of two typical breast cancer cell lines, ER-negative MDA-MB-231 with αVβ3 expression and ER-positive MCF-7 with αVβ5/α5β1 expression, were examined in vitro as well the related mechanisms. The results demonstrated that lunasin (10-20 µM) effectively inhibited the migration and invasion activity and expression of matrix metalloproteinase (MMP)‑2/-9 in both breast cancer cell lines. Meanwhile, we also found that lunasin inhibited the phosphorylation of focal adhesion kinase (FAK), Src, Akt, ERK and nucleus translocation of NF-κB, which indicates that, possibly via competing with αVβ3 or αVβ5/α5β1 integrin, lunasin suppresses the metastasis of breast cancer cells through integrin-mediated FAK/Akt/ERK and NF-κB signaling pathways followed by downregulation of the activity and expression of MMP-2/-9.
Collapse
|
168
|
King A, Ndifon C, Lui S, Widdows K, Kotamraju VR, Agemy L, Teesalu T, Glazier JD, Cellesi F, Tirelli N, Aplin JD, Ruoslahti E, Harris LK. Tumor-homing peptides as tools for targeted delivery of payloads to the placenta. SCIENCE ADVANCES 2016; 2:e1600349. [PMID: 27386551 PMCID: PMC4928982 DOI: 10.1126/sciadv.1600349] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 04/08/2016] [Indexed: 05/08/2023]
Abstract
The availability of therapeutics to treat pregnancy complications is severely lacking mainly because of the risk of causing harm to the fetus. As enhancement of placental growth and function can alleviate maternal symptoms and improve fetal growth in animal models, we have developed a method for targeted delivery of payloads to the placenta. We show that the tumor-homing peptide sequences CGKRK and iRGD bind selectively to the placental surface of humans and mice and do not interfere with normal development. Peptide-coated nanoparticles intravenously injected into pregnant mice accumulated within the mouse placenta, whereas control nanoparticles exhibited reduced binding and/or fetal transfer. We used targeted liposomes to efficiently deliver cargoes of carboxyfluorescein and insulin-like growth factor 2 to the mouse placenta; the latter significantly increased mean placental weight when administered to healthy animals and significantly improved fetal weight distribution in a well-characterized model of fetal growth restriction. These data provide proof of principle for targeted delivery of drugs to the placenta and provide a novel platform for the development of placenta-specific therapeutics.
Collapse
Affiliation(s)
- Anna King
- Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, Manchester M13 9WL, UK
- Academic Health Science Centre, St Mary’s Hospital, Oxford Road, Manchester M13 9WL, UK
| | - Cornelia Ndifon
- Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, Manchester M13 9WL, UK
- Academic Health Science Centre, St Mary’s Hospital, Oxford Road, Manchester M13 9WL, UK
| | - Sylvia Lui
- Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, Manchester M13 9WL, UK
- Academic Health Science Centre, St Mary’s Hospital, Oxford Road, Manchester M13 9WL, UK
| | - Kate Widdows
- Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, Manchester M13 9WL, UK
- Academic Health Science Centre, St Mary’s Hospital, Oxford Road, Manchester M13 9WL, UK
| | - Venkata R. Kotamraju
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
- Center for Nanomedicine and Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106–9610, USA
| | - Lilach Agemy
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
- Center for Nanomedicine and Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106–9610, USA
| | - Tambet Teesalu
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
- Center for Nanomedicine and Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106–9610, USA
| | - Jocelyn D. Glazier
- Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, Manchester M13 9WL, UK
- Academic Health Science Centre, St Mary’s Hospital, Oxford Road, Manchester M13 9WL, UK
| | - Francesco Cellesi
- School of Pharmacy, University of Manchester, Stopford Building, Oxford Road, Manchester M13 9PT, UK
| | - Nicola Tirelli
- School of Pharmacy, University of Manchester, Stopford Building, Oxford Road, Manchester M13 9PT, UK
- Institute of Inflammation and Repair, Faculty of Medical and Human Sciences, University of Manchester, Manchester M13 9PT, UK
| | - John D. Aplin
- Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, Manchester M13 9WL, UK
- Academic Health Science Centre, St Mary’s Hospital, Oxford Road, Manchester M13 9WL, UK
| | - Erkki Ruoslahti
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
- Center for Nanomedicine and Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA 93106–9610, USA
| | - Lynda K. Harris
- Maternal and Fetal Health Research Centre, Institute of Human Development, University of Manchester, Manchester M13 9WL, UK
- Academic Health Science Centre, St Mary’s Hospital, Oxford Road, Manchester M13 9WL, UK
- School of Pharmacy, University of Manchester, Stopford Building, Oxford Road, Manchester M13 9PT, UK
- Corresponding author.
| |
Collapse
|
169
|
iRGD decorated lipid-polymer hybrid nanoparticles for targeted co-delivery of doxorubicin and sorafenib to enhance anti-hepatocellular carcinoma efficacy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1303-11. [PMID: 26964482 DOI: 10.1016/j.nano.2016.01.017] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 01/26/2016] [Accepted: 01/29/2016] [Indexed: 11/21/2022]
Abstract
The combination of doxorubicin (DOX) with sorafenib (SOR) has proven an effective strategy to enhance anti-hepatocellular carcinoma (HCC) efficacy. However, respective in vivo pharmacokinetic profiles and different endocytosis capacities of these two drugs greatly hinder their current application. Herein, the tumor-targeting peptide iRGD decorated lipid-polymer hybrid nanoparticles (NPs) with a shell-core structure were developed for co-delivery of DOX and SOR (DOX+SOR/iRGD NPs). After the drug ratio was optimized, the stabilized DOX+SOR/iRGD NPs were prepared. Through the iRGD-integrin recognition, DOX+SOR/iRGD NPs showed synergistic cytotoxicity, pro-apoptotic ability and enhanced internalization rate in human liver cancer HepG2 cells. In vivo pharmacokinetic result demonstrated that an extended circulation and bioavailability of DOX+SOR/iRGD NPs than free drugs. More importantly, DOX+SOR/iRGD NPs significantly enhanced antitumor efficiency in HCC xenograft mouse models. Overall, this study describes a promising nanoparticulate drug co-delivery strategy to combine clinical anticancer drugs and enhance anti-HCC efficacy.
Collapse
|
170
|
Synthesis of Novel Protected Nα(ω-Drug) Amino Acid Building Units for Facile Preparation of Anticancer Drug-Conjugates. Int J Pept Res Ther 2016. [DOI: 10.1007/s10989-015-9509-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
171
|
Scodeller P. Extracellular Matrix Degrading Enzymes for Nanocarrier-Based Anticancer Therapy. INTRACELLULAR DELIVERY III 2016. [DOI: 10.1007/978-3-319-43525-1_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
172
|
Yu X, Trase I, Ren M, Duval K, Guo X, Chen Z. Design of Nanoparticle-Based Carriers for Targeted Drug Delivery. JOURNAL OF NANOMATERIALS 2016; 2016:1087250. [PMID: 27398083 PMCID: PMC4936496 DOI: 10.1155/2016/1087250] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Nanoparticles have shown promise as both drug delivery vehicles and direct antitumor systems, but they must be properly designed in order to maximize efficacy. Computational modeling is often used both to design new nanoparticles and to better understand existing ones. Modeled processes include the release of drugs at the tumor site and the physical interaction between the nanoparticle and cancer cells. In this article, we provide an overview of three different targeted drug delivery methods (passive targeting, active targeting and physical targeting), compare methods of action, advantages, limitations, and the current stage of research. For the most commonly used nanoparticle carriers, fabrication methods are also reviewed. This is followed by a review of computational simulations and models on nanoparticle-based drug delivery.
Collapse
Affiliation(s)
- Xiaojiao Yu
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Ian Trase
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Muqing Ren
- Department of Chemical Physics, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Kayla Duval
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Xing Guo
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | - Zi Chen
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
173
|
Stojanovic V, Cunin F, Durand JO, Garcia M, Gary-Bobo M. Potential of porous silicon nanoparticles as an emerging platform for cancer theranostics. J Mater Chem B 2016; 4:7050-7059. [DOI: 10.1039/c6tb01829g] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Currently, nanoscience is a major part of biomedical research, due to material advances that aid the development of new tools and techniques to replace traditional methods. Here we describe the theranostic potential of multifunctional porous silicon nanoparticles to target, image and treat cancer.
Collapse
Affiliation(s)
- V. Stojanovic
- Institut des Biomolécules Max Mousseron
- UMR5247CNRS-UM
- 34093 Montpellier Cedex 05
- France
| | - F. Cunin
- Institut Charles Gerhardt Montpellier
- UMR5253CNRS-ENSCM-UM
- Ecole Nationale Supérieure de Chimie Montpellier
- 8 rue de l'Ecole Normale
- 34296 Montpellier
| | - J. O. Durand
- Institut Charles Gerhardt Montpellier
- UMR5253CNRS-ENSCM-UM
- Ecole Nationale Supérieure de Chimie Montpellier
- 8 rue de l'Ecole Normale
- 34296 Montpellier
| | - M. Garcia
- Institut des Biomolécules Max Mousseron
- UMR5247CNRS-UM
- 34093 Montpellier Cedex 05
- France
| | - M. Gary-Bobo
- Institut des Biomolécules Max Mousseron
- UMR5247CNRS-UM
- 34093 Montpellier Cedex 05
- France
| |
Collapse
|
174
|
Abstract
Bispecific antibodies (BsAbs) recognize two different epitopes. This dual specificity opens up a wide range of applications, including redirecting T cells to tumor cells, blocking two different signaling pathways simultaneously, dual targeting of different disease mediators, and delivering payloads to targeted sites. The approval of catumaxomab (anti-EpCAM and anti-CD3) and blinatumomab (anti-CD19 and anti-CD3) has become a major milestone in the development of bsAbs. Currently, more than 60 different bsAb formats exist, some of them making their way into the clinical pipeline. This review summarizes diverse formats of bsAbs and their clinical applications and sheds light on strategies to optimize the design of bsAbs.
Collapse
Affiliation(s)
- Gaowei Fan
- National Center for Clinical Laboratories, Beijing Hospital, No 1 Dahua Road, Dongdan, Beijing, 100730, China.
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Zujian Wang
- Shunyi District Maternal and Child Health Hospital of Beijing City, Beijing, 101300, China.
| | - Mingju Hao
- National Center for Clinical Laboratories, Beijing Hospital, No 1 Dahua Road, Dongdan, Beijing, 100730, China.
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Jinming Li
- National Center for Clinical Laboratories, Beijing Hospital, No 1 Dahua Road, Dongdan, Beijing, 100730, China.
- Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
175
|
Wolfram J, Shen H, Ferrari M. Multistage vector (MSV) therapeutics. J Control Release 2015; 219:406-415. [PMID: 26264836 PMCID: PMC4656100 DOI: 10.1016/j.jconrel.2015.08.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/01/2015] [Accepted: 08/04/2015] [Indexed: 12/21/2022]
Abstract
One of the greatest challenges in the field of medicine is obtaining controlled distribution of systemically administered therapeutic agents within the body. Indeed, biological barriers such as physical compartmentalization, pressure gradients, and excretion pathways adversely affect localized delivery of drugs to pathological tissue. The diverse nature of these barriers requires the use of multifunctional drug delivery vehicles that can overcome a wide range of sequential obstacles. In this review, we explore the role of multifunctionality in nanomedicine by primarily focusing on multistage vectors (MSVs). The MSV is an example of a promising therapeutic platform that incorporates several components, including a microparticle, nanoparticles, and small molecules. In particular, these components are activated in a sequential manner in order to successively address transport barriers.
Collapse
Affiliation(s)
- Joy Wolfram
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, National Center for Nanoscience & Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Haifa Shen
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
176
|
Feitelson MA, Arzumanyan A, Kulathinal RJ, Blain SW, Holcombe RF, Mahajna J, Marino M, Martinez-Chantar ML, Nawroth R, Sanchez-Garcia I, Sharma D, Saxena NK, Singh N, Vlachostergios PJ, Guo S, Honoki K, Fujii H, Georgakilas AG, Bilsland A, Amedei A, Niccolai E, Amin A, Ashraf SS, Boosani CS, Guha G, Ciriolo MR, Aquilano K, Chen S, Mohammed SI, Azmi AS, Bhakta D, Halicka D, Keith WN, Nowsheen S. Sustained proliferation in cancer: Mechanisms and novel therapeutic targets. Semin Cancer Biol 2015; 35 Suppl:S25-S54. [PMID: 25892662 PMCID: PMC4898971 DOI: 10.1016/j.semcancer.2015.02.006] [Citation(s) in RCA: 444] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 02/20/2015] [Accepted: 02/23/2015] [Indexed: 02/08/2023]
Abstract
Proliferation is an important part of cancer development and progression. This is manifest by altered expression and/or activity of cell cycle related proteins. Constitutive activation of many signal transduction pathways also stimulates cell growth. Early steps in tumor development are associated with a fibrogenic response and the development of a hypoxic environment which favors the survival and proliferation of cancer stem cells. Part of the survival strategy of cancer stem cells may manifested by alterations in cell metabolism. Once tumors appear, growth and metastasis may be supported by overproduction of appropriate hormones (in hormonally dependent cancers), by promoting angiogenesis, by undergoing epithelial to mesenchymal transition, by triggering autophagy, and by taking cues from surrounding stromal cells. A number of natural compounds (e.g., curcumin, resveratrol, indole-3-carbinol, brassinin, sulforaphane, epigallocatechin-3-gallate, genistein, ellagitannins, lycopene and quercetin) have been found to inhibit one or more pathways that contribute to proliferation (e.g., hypoxia inducible factor 1, nuclear factor kappa B, phosphoinositide 3 kinase/Akt, insulin-like growth factor receptor 1, Wnt, cell cycle associated proteins, as well as androgen and estrogen receptor signaling). These data, in combination with bioinformatics analyses, will be very important for identifying signaling pathways and molecular targets that may provide early diagnostic markers and/or critical targets for the development of new drugs or drug combinations that block tumor formation and progression.
Collapse
Affiliation(s)
- Mark A Feitelson
- Department of Biology, Temple University, Philadelphia, PA, United States.
| | - Alla Arzumanyan
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Rob J Kulathinal
- Department of Biology, Temple University, Philadelphia, PA, United States
| | - Stacy W Blain
- Department of Pediatrics, State University of New York, Downstate Medical Center, Brooklyn, NY, United States
| | - Randall F Holcombe
- Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, United States
| | - Jamal Mahajna
- MIGAL-Galilee Technology Center, Cancer Drug Discovery Program, Kiryat Shmona, Israel
| | - Maria Marino
- Department of Science, University Roma Tre, V.le G. Marconi, 446, 00146 Rome, Italy
| | - Maria L Martinez-Chantar
- Metabolomic Unit, CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Technology Park of Bizkaia, Bizkaia, Spain
| | - Roman Nawroth
- Department of Urology, Klinikum rechts der Isar der Technischen Universität München, Munich, Germany
| | - Isidro Sanchez-Garcia
- Experimental Therapeutics and Translational Oncology Program, Instituto de Biología Molecular y Celular del Cáncer, CSIC/Universidad de Salamanca, Salamanca, Spain
| | - Dipali Sharma
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Neeraj K Saxena
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, United States
| | - Neetu Singh
- Tissue and Cell Culture Unit, CSIR-Central Drug Research Institute, Council of Scientific & Industrial Research, Lucknow, India
| | | | - Shanchun Guo
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara 634-8521, Japan
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara 634-8521, Japan
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Zografou 15780, Athens, Greece
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, UK
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, UAE University, Al-Ain, United Arab Emirates
| | - S Salman Ashraf
- Department of Chemistry, College of Science, UAE University, Al-Ain, United Arab Emirates
| | - Chandra S Boosani
- Department of BioMedical Sciences, Creighton University, Omaha, NE, United States
| | - Gunjan Guha
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Maria Rosa Ciriolo
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Sophie Chen
- Department of Research and Development, Ovarian and Prostate Cancer Research Trust Laboratory, Guildford, Surrey GU2 7YG, United Kingdom
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - Asfar S Azmi
- Department of Pathology, Karmonas Cancer Institute, Wayne State University School of Medicine, Detroit, MI, United States
| | - Dipita Bhakta
- School of Chemical and Bio Technology, SASTRA University, Thanjavur, India
| | - Dorota Halicka
- Brander Cancer Research Institute, Department of Pathology, New York Medical College, Valhalla, NY, United States
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, UK
| | - Somaira Nowsheen
- Mayo Graduate School, Mayo Medical School, Mayo Clinic Medical Scientist Training Program, Rochester, MN, United States
| |
Collapse
|
177
|
Xiao H, Liu Y, Tan H, Liang P, Wang B, Su L, Wang S, Gao J. A pilot study using low-dose Spectral CT and ASIR (Adaptive Statistical Iterative Reconstruction) algorithm to diagnose solitary pulmonary nodules. BMC Med Imaging 2015; 15:54. [PMID: 26576676 PMCID: PMC4647278 DOI: 10.1186/s12880-015-0096-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 10/25/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Lung cancer is the most common cancer which has the highest mortality rate. With the development of computed tomography (CT) techniques, the case detection rates of solitary pulmonary nodules (SPN) has constantly increased and the diagnosis accuracy of SPN has remained a hot topic in clinical and imaging diagnosis. The aim of this study was to evaluate the combination of low-dose spectral CT and ASIR (Adaptive Statistical Iterative Reconstruction) algorithm in the diagnosis of solitary pulmonary nodules (SPN). METHODS 62 patients with SPN (42 cases of benign SPN and 20 cases of malignant SPN, pathology confirmed) were scanned by spectral CT with a dual-phase contrast-enhanced method. The iodine and water concentration (IC and WC) of the lesion and the artery in the image that had the same density were measured by the GSI (Gemstone Spectral Imaging) software. The normalized iodine and water concentration (NIC and NWC) of the lesion and the normalized iodine and water concentration difference (ICD and WCD) between the arterial and venous phases (AP and VP) were also calculated. The spectral HU (Hounsfield Unit ) curve was divided into 3 sections based on the energy (40-70, 70-100 and 100-140 keV) and the slopes (λHU) in both phases were calculated. The ICAP, ICVP, WCAP and WCVP, NIC and NWC, and the λHU in benign and malignant SPN were compared by independent sample t-test. RESULTS The iodine related parameters (ICAP, ICVP, NICAP, NICVP, and the ICD) of malignant SPN were significantly higher than that of benign SPN (t = 3.310, 1.330, 2.388, 1.669 and 3.251, respectively, P <0.05). The 3 λHU values of venous phase in malignant SPN were higher than that of benign SPN (t = 3.803, 2.846 and 3.205, P <0.05). The difference of water related parameters (WCAP, WCVP, NWCAP, NWCVP and WCD) between malignant and benign SPN were not significant (t = 0.666, 0.257, 0.104, 0.550 and 0.585, P > 0.05). CONCLUSIONS The iodine related parameters and the slope of spectral curve are useful markers to distinguish the benign from the malignant lung diseases, and its application is extremely feasible in clinical applications.
Collapse
Affiliation(s)
- Huijuan Xiao
- The Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1, East Jianshe Road, Zhengzhou, Henan Province, 450052, China.
| | - Yihe Liu
- The No.7 People's Hospital of Zhengzhou, 17 Jingnan 5th Road, Zhengzhou Economic and Technological Development Zone, Zhengzhou, Henan Province, 450000, China.
| | - Hongna Tan
- The Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1, East Jianshe Road, Zhengzhou, Henan Province, 450052, China.
| | - Pan Liang
- The Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1, East Jianshe Road, Zhengzhou, Henan Province, 450052, China.
| | - Bo Wang
- The Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1, East Jianshe Road, Zhengzhou, Henan Province, 450052, China.
| | - Lei Su
- The Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1, East Jianshe Road, Zhengzhou, Henan Province, 450052, China.
| | - Suya Wang
- The Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1, East Jianshe Road, Zhengzhou, Henan Province, 450052, China.
| | - Jianbo Gao
- The Department of Radiology, The First Affiliated Hospital of Zhengzhou University, No.1, East Jianshe Road, Zhengzhou, Henan Province, 450052, China.
| |
Collapse
|
178
|
Kim JH, Graef AJ, Dickerson EB, Modiano JF. Pathobiology of Hemangiosarcoma in Dogs: Research Advances and Future Perspectives. Vet Sci 2015; 2:388-405. [PMID: 29061949 PMCID: PMC5644642 DOI: 10.3390/vetsci2040388] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Revised: 10/05/2015] [Accepted: 10/20/2015] [Indexed: 12/12/2022] Open
Abstract
Hemangiosarcoma (HSA) is an aggressive and common cancer in dogs. While cutaneous masses are often treatable by tumor excision, visceral tumors are almost always incurable. Treatment advances for this disease have been limited due to a poor understanding of the overall tumor biology. Based upon its histological appearance, HSA has been presumed to originate from transformed endothelial cells; however, accumulating data now suggest a pluripotent bone marrow progenitor as the cell of origin for this disease. More recently, the identification of a novel subclassification of HSAs has provided a foundation to further our understanding of the cellular characteristics of HSA tumor cells, along with those of the cells comprising the tumor microenvironment. These discoveries hold promise for the development of new approaches to improve treatments for canine HSA, as well as to establish the utility of this disease as a spontaneous model to understand the pathogenesis and develop new treatments for vascular tumors of humans. In this review, we will provide a brief historical perspective and pathobiology of canine HSA, along with a focus on the recent advances in the molecular and cellular understanding of these tumors. In addition, future directions that should continue to improve our understanding of HSA pathogenesis will be discussed.
Collapse
Affiliation(s)
- Jong-Hyuk Kim
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA.
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Ashley J Graef
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Erin B Dickerson
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA.
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Jaime F Modiano
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA.
- Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA.
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
179
|
Schmithals C, Köberle V, Korkusuz H, Pleli T, Kakoschky B, Augusto EA, Ibrahim AA, Arencibia JM, Vafaizadeh V, Groner B, Korf HW, Kronenberger B, Zeuzem S, Vogl TJ, Waidmann O, Piiper A. Improving Drug Penetrability with iRGD Leverages the Therapeutic Response to Sorafenib and Doxorubicin in Hepatocellular Carcinoma. Cancer Res 2015; 75:3147-54. [PMID: 26239478 DOI: 10.1158/0008-5472.can-15-0395] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
iRGD is a derivative of the integrin-binding peptide RGD, which selectively increases the penetrability of tumor tissue to various coadministered substances in several preclinical models. In this study, we investigated the ability of iRGD to improve the delivery of sorafenib and doxorubicin therapy in hepatocellular carcinoma (HCC) using established mouse models of the disease. A contrast-enhanced MRI method was developed in parallel to assess the in vivo effects of iRGD in this setting. We found that iRGD improved the delivery of marker substances to the tumors of HCC-bearing mice about three-fold without a parallel increase in normal tissues. Control peptides lacking the critical CendR motif had no effect. Similarly, iRGD also selectively increased the signal intensity from tumors in Gd-DTPA-enhanced MRI. In terms of antitumor efficacy, iRGD coadministration significantly augmented the individual inhibitory effects of sorafenib and doxorubicin without increasing systemic toxicity. Overall, our results offered a preclinical proof of concept for the use of iRGD coadministration as a strategy to widen the therapeutic window for HCC chemotherapy, as monitored by Gd-DTPA-enhanced MRI as a noninvasive, clinically applicable method to identify iRGD-reactive tumors.
Collapse
Affiliation(s)
| | - Verena Köberle
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | - Hüdayi Korkusuz
- Department of Nuclear Medicine, University Hospital Frankfurt, Frankfurt, Germany
| | - Thomas Pleli
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | - Bianca Kakoschky
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | | | - Ahmed Atef Ibrahim
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany. The Immunology and Infectious Diseases Laboratory, Therapeutic Chemistry Department, The National Research Center, Dokki, Cairo, Egypt
| | - Jose M Arencibia
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | | | | | - Horst-Werner Korf
- Institute of Anatomy 2, University Hospital Frankfurt, Frankfurt, Germany
| | - Bernd Kronenberger
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | - Stefan Zeuzem
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | - Thomas J Vogl
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Frankfurt, Germany
| | - Oliver Waidmann
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany
| | - Albrecht Piiper
- Department of Medicine 1, University Hospital Frankfurt, Frankfurt, Germany.
| |
Collapse
|
180
|
Borjan B, Steiner N, Karbon S, Kern J, Francesch A, Hermann M, Willenbacher W, Gunsilius E, Untergasser G. The Aplidin analogs PM01215 and PM02781 inhibit angiogenesis in vitro and in vivo. BMC Cancer 2015; 15:738. [PMID: 26483043 PMCID: PMC4615365 DOI: 10.1186/s12885-015-1729-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 10/08/2015] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Novel synthesized analogs of Aplidin, PM01215 and PM02781, were tested for antiangiogenic effects on primary human endothelial cells in vitro and for inhibition of angiogenesis and tumor growth in vivo. METHODS Antiangiogenic activity of both derivatives was evaluated by real-time cell proliferation, capillary tube formation and vascular endothelial growth factor (VEGF)-induced spheroid sprouting assays. Distribution of endothelial cells in the different phases of the cell cycle was analyzed by flow cytometry. Aplidin analogs were tested in vivo in chicken chorioallantoic membrane (CAM) assays. RESULTS Both derivatives inhibited angiogenic capacities of human endothelial cells (HUVECs) in vitro at low nanomolar concentrations. Antiangiogenic effects of both analogs were observed in the CAM. In addition, growth of human multiple myeloma xenografts in vivo in CAM was significantly reduced after application of both analogs. On the molecular level, both derivatives induced cell cycle arrest in G1 phase. This growth arrest of endothelial cells correlated with induction of the cell cycle inhibitor p16(INK4A) and increased senescence-associated beta galactosidase activity. In addition, Aplidin analogs induced oxidative stress and decreased production of the vascular maturation factors Vasohibin-1 and Dickkopf-3. CONCLUSIONS From these findings we conclude that both analogs are promising agents for the development of antiangiogenic drugs acting independent on classical inhibition of VEGF signaling.
Collapse
Affiliation(s)
- Bojana Borjan
- Department of Internal Medicine V, Innsbruck Medical University, Innrain 66, 6020, Innsbruck, Austria.
| | - Normann Steiner
- Department of Internal Medicine V, Innsbruck Medical University, Innrain 66, 6020, Innsbruck, Austria.
| | - Silvia Karbon
- Department of Internal Medicine V, Innsbruck Medical University, Innrain 66, 6020, Innsbruck, Austria.
| | - Johann Kern
- Oncotyrol GmbH, Karl Kapfererstrasse 5, 6020, Innsbruck, Austria.
| | - Andrés Francesch
- Pharmamar, R&D Department, Avda de los Reyes 1, 28770, Colmenar Viejo, Madrid, Spain.
| | - Martin Hermann
- Department of Anesthesiology & Critical Care Medicine, Innsbruck Medical University, Innsbruck, Austria.
| | - Wolfgang Willenbacher
- Department of Internal Medicine V, Innsbruck Medical University, Innrain 66, 6020, Innsbruck, Austria.
| | - Eberhard Gunsilius
- Department of Internal Medicine V, Innsbruck Medical University, Innrain 66, 6020, Innsbruck, Austria.
| | - Gerold Untergasser
- Department of Internal Medicine V, Innsbruck Medical University, Innrain 66, 6020, Innsbruck, Austria. .,Tyrolean Cancer Research Institute, 6020, Innsbruck, Austria.
| |
Collapse
|
181
|
Mao X, Liu J, Gong Z, Zhang H, Lu Y, Zou H, Yu Y, Chen Y, Sun Z, Li W, Li B, Gao J, Zhong Y. iRGD-conjugated DSPE-PEG2000 nanomicelles for targeted delivery of salinomycin for treatment of both liver cancer cells and cancer stem cells. Nanomedicine (Lond) 2015; 10:2677-95. [PMID: 26355733 DOI: 10.2217/nnm.15.106] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIMS To develop novel iRGD (internalizing Arg-Gly-Asp peptide)-conjugated DSPE-PEG2000 nanomicelles (M-SAL-iRGD) for delivery of salinomycin to both liver cancer cells and cancer stem cells (CSCs). MATERIALS & METHODS The characterization, antitumor activity and mechanism of action of M-SAL-iRGD were evaluated. RESULTS & CONCLUSION M-SAL-iRGD possessed a small size of around 10 nm, and drug encapsulation efficacy higher than 90%. M-SAL-iRGD showed significantly increased cytotoxic effect toward both nontargeted M-SAL (salinomycin-loaded DSPE-PEG2000 nanomicelles) and salinomycin in both liver cancer cells and CSCs. The tissue distribution and antitumor assays in mice bearing liver cancer xenograft confirmed the superior penetration tumor efficacy and antitumor activity of M-SAL-iRGD. M-SAL-iRGD represent a potential effective nanomedicine against liver cancer.
Collapse
Affiliation(s)
- Xiaoli Mao
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China.,School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian, China
| | - Junjie Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Zhirong Gong
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - He Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Ying Lu
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China.,School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian, China
| | - Hao Zou
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Yuan Yu
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Yan Chen
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Zhiguo Sun
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Wei Li
- International Joint Cancer Institute, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Bohua Li
- International Joint Cancer Institute, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Jie Gao
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Yanqiang Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy, Second Military Medical University, 325 Guohe Road, Shanghai 200433, China.,School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fujian, China
| |
Collapse
|
182
|
Liu D, Auguste DT. Cancer targeted therapeutics: From molecules to drug delivery vehicles. J Control Release 2015; 219:632-643. [PMID: 26342659 DOI: 10.1016/j.jconrel.2015.08.041] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 08/19/2015] [Accepted: 08/20/2015] [Indexed: 02/07/2023]
Abstract
The pitfall of all chemotherapeutics lies in drug resistance and the severe side effects experienced by patients. One way to reduce the off-target effects of chemotherapy on healthy tissues is to alter the biodistribution of drug. This can be achieved in two ways: Passive targeting utilizes shape, size, and surface chemistry to increase particle circulation and tumor accumulation. Active targeting employs either chemical moieties (e.g. peptides, sugars, aptamers, antibodies) to selectively bind to cell membranes or responsive elements (e.g. ultrasound, magnetism, light) to deliver its cargo within a local region. This article will focus on the systemic administration of anti-cancer agents and their ability to home to tumors and, if relevant, distant metastatic sites.
Collapse
Affiliation(s)
- Daxing Liu
- Department of Biomedical Engineering, The City College of New York, New York, NY 10031, United States
| | - Debra T Auguste
- Department of Biomedical Engineering, The City College of New York, New York, NY 10031, United States.
| |
Collapse
|
183
|
Hamilton AM, Aidoudi-Ahmed S, Sharma S, Kotamraju VR, Foster PJ, Sugahara KN, Ruoslahti E, Rutt BK. Nanoparticles coated with the tumor-penetrating peptide iRGD reduce experimental breast cancer metastasis in the brain. J Mol Med (Berl) 2015; 93:991-1001. [PMID: 25869026 PMCID: PMC4807972 DOI: 10.1007/s00109-015-1279-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 02/25/2015] [Accepted: 03/30/2015] [Indexed: 01/01/2023]
Abstract
UNLABELLED Metastasis is the main killer in cancer; consequently, there is great interest in novel approaches to prevent and treat metastatic disease. Brain metastases are particularly deadly, as the protection of the blood-brain barrier obstructs the passage of common anticancer drugs. This study used magnetic resonance imaging (MRI) to investigate the therapeutic effects of nanoparticles coated with a tumor-penetrating peptide (iRGD) against a preclinical model of breast cancer brain metastasis. Single doses of iRGD nanoparticle were administered intravenously, and the effect on tumor growth was observed over time. iRGD nanoparticles, when applied in the early stages of metastasis development, strongly inhibited tumor progression. Overall, this study demonstrated for the first time that a single dose of iRGD nanoparticle can have a significant effect on metastatic tumor progression and nonproliferative cancer cell retention when applied early in course of tumor development. These data suggest that iRGD nanoparticles may be useful in preventatively reducing metastasis after a cancer diagnosis has been established. KEY MESSAGES bSSFP MRI can be used to track nonproliferative iron-labeled cells and tumor development over time. iRGD-NW, when applied early, has a significant effect on metastatic tumor progression. Retained signal voids represent a subpopulation of nonproliferating tumor cells. Reduced cell retention and tumor burden show a role for iRGD-NW in metastasis prevention. iRGD target is universally expressed; thus, iRGD-NW should be clinically translatable.
Collapse
Affiliation(s)
- Amanda M Hamilton
- Robarts Research Institute, University of Western Ontario, 1151 Richmond St N, London, ON, N6A 5B7, Canada.
| | | | - Shweta Sharma
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Venkata R Kotamraju
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Paula J Foster
- Robarts Research Institute, University of Western Ontario, 1151 Richmond St N, London, ON, N6A 5B7, Canada
| | - Kazuki N Sugahara
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
| | - Erkki Ruoslahti
- Cancer Research Center, Sanford-Burnham Medical Research Institute, La Jolla, CA, USA
- Center for Nanomedicine and Department of Cell, Molecular and Developmental Biology, University of California, Santa Barbara, CA, USA
| | - Brian K Rutt
- Radiological Sciences Laboratory, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
184
|
Sugahara KN, Scodeller P, Braun GB, de Mendoza TH, Yamazaki CM, Kluger MD, Kitayama J, Alvarez E, Howell SB, Teesalu T, Ruoslahti E, Lowy AM. A tumor-penetrating peptide enhances circulation-independent targeting of peritoneal carcinomatosis. J Control Release 2015; 212:59-69. [PMID: 26071630 PMCID: PMC4508207 DOI: 10.1016/j.jconrel.2015.06.009] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 06/01/2015] [Accepted: 06/09/2015] [Indexed: 01/09/2023]
Abstract
Peritoneal carcinomatosis is a major source of morbidity and mortality in patients with advanced abdominal neoplasms. Intraperitoneal chemotherapy (IPC) is an area of intense interest given its efficacy in ovarian cancer. However, IPC suffers from poor drug penetration into peritoneal tumors. As such, extensive cytoreductive surgery is required prior to IPC. Here, we explore the utility of iRGD, a tumor-penetrating peptide, for improved tumor-specific penetration of intraperitoneal compounds and enhanced IPC in mice. Intraperitoneally administered iRGD significantly enhanced penetration of an attached fluorescein into disseminated peritoneal tumor nodules. The penetration was tumor-specific, circulation-independent, and mediated by the neuropilin-binding RXXK tissue-penetration peptide motif of iRGD. Q-iRGD, which fluoresces upon cleavage, including the one that leads to RXXK activation, specifically labeled peritoneal metastases displaying different growth patterns in mice. Importantly, iRGD enhanced intratumoral entry of intraperitoneally co-injected dextran to approximately 300% and doxorubicin to 250%. Intraperitoneal iRGD/doxorubicin combination therapy inhibited the growth of bulky peritoneal tumors and reduced systemic drug toxicity. iRGD delivered attached fluorescein and co-applied nanoparticles deep into fresh human peritoneal metastasis explants. These results indicate that intraperitoneal iRGD co-administration serves as a simple and effective strategy to facilitate tumor detection and improve the therapeutic index of IPC for peritoneal carcinomatosis.
Collapse
Affiliation(s)
- Kazuki N Sugahara
- Cancer Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA; Department of Surgery, Columbia University College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, 1130St Nicholas Avenue, New York, NY 10032, USA.
| | - Pablo Scodeller
- Cancer Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Gary B Braun
- Cancer Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA; Center of Nanomedicine and Department of Cell, Molecular and Developmental Biology, 2203 Life Sciences Building, MCDB, University of California, Santa Barbara, CA 93106-9625, USA.
| | - Tatiana Hurtado de Mendoza
- Cancer Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Chisato M Yamazaki
- Department of Surgery, Columbia University College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, 1130St Nicholas Avenue, New York, NY 10032, USA.
| | - Michael D Kluger
- Department of Surgery, Columbia University College of Physicians and Surgeons, Herbert Irving Comprehensive Cancer Center, 1130St Nicholas Avenue, New York, NY 10032, USA.
| | - Joji Kitayama
- Department of Surgical Oncology, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| | - Edwin Alvarez
- Department of Reproductive Medicine, Division of Gynecologic Oncology, Moores Cancer Center, 3855 Health Sciences Drive, La Jolla, CA 92093-0987, USA.
| | - Stephen B Howell
- Department of Medicine, Moores Cancer Center, 3855 Health Sciences Drive, La Jolla, CA 92093-0819, USA.
| | - Tambet Teesalu
- Cancer Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA; Center of Nanomedicine and Department of Cell, Molecular and Developmental Biology, 2203 Life Sciences Building, MCDB, University of California, Santa Barbara, CA 93106-9625, USA; Institute of Biomedicine and Translational Medicine, Centre of Excellence for Translational Medicine, University of Tartu, Ravila 14b, Tartu 50411, Estonia.
| | - Erkki Ruoslahti
- Cancer Research Center, Sanford-Burnham Medical Research Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA; Center of Nanomedicine and Department of Cell, Molecular and Developmental Biology, 2203 Life Sciences Building, MCDB, University of California, Santa Barbara, CA 93106-9625, USA.
| | - Andrew M Lowy
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, 3855 Health Sciences Drive, La Jolla, CA 92093-0987, USA.
| |
Collapse
|
185
|
Reppas AI, Alfonso JCL, Hatzikirou H. In silico tumor control induced via alternating immunostimulating and immunosuppressive phases. Virulence 2015; 7:174-86. [PMID: 26305801 DOI: 10.1080/21505594.2015.1076614] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Despite recent advances in the field of Oncoimmunology, the success potential of immunomodulatory therapies against cancer remains to be elucidated. One of the reasons is the lack of understanding on the complex interplay between tumor growth dynamics and the associated immune system responses. Toward this goal, we consider a mathematical model of vascularized tumor growth and the corresponding effector cell recruitment dynamics. Bifurcation analysis allows for the exploration of model's dynamic behavior and the determination of these parameter regimes that result in immune-mediated tumor control. In this work, we focus on a particular tumor evasion regime that involves tumor and effector cell concentration oscillations of slowly increasing and decreasing amplitude, respectively. Considering a temporal multiscale analysis, we derive an analytically tractable mapping of model solutions onto a weakly negatively damped harmonic oscillator. Based on our analysis, we propose a theory-driven intervention strategy involving immunostimulating and immunosuppressive phases to induce long-term tumor control.
Collapse
Affiliation(s)
- A I Reppas
- a Center for Advancing Electronics; Technische Universität Dresden ; Dresden , Germany
| | - J C L Alfonso
- a Center for Advancing Electronics; Technische Universität Dresden ; Dresden , Germany
| | - H Hatzikirou
- a Center for Advancing Electronics; Technische Universität Dresden ; Dresden , Germany
| |
Collapse
|
186
|
Sun L, Wu Q, Peng F, Liu L, Gong C. Strategies of polymeric nanoparticles for enhanced internalization in cancer therapy. Colloids Surf B Biointerfaces 2015; 135:56-72. [PMID: 26241917 DOI: 10.1016/j.colsurfb.2015.07.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 06/19/2015] [Accepted: 07/07/2015] [Indexed: 02/05/2023]
Abstract
In order to achieve long circulation time and high drug accumulation in the tumor sites via the EPR effects, anticancer drugs have to be protected by non-fouling polymers such as poly(ethylene glycol) (PEG), poly(ethylene oxide) (PEO), dextran, and poly(acrylic acid) (PAA). However, the dense layer of stealth polymer also prohibits efficient uptake of anticancer drugs by target cancer cells. For cancer therapy, it is often more desirable to accomplish rapid cellular uptake after anticancer drugs arriving at the pathological site, which could on one hand maximize the therapeutic efficacy and on the other hand reduce probability of drug resistance in cells. In this review, special attention will be focused on the recent potential strategies that can enable drug-loaded polymeric nanoparticles to rapidly recognize cancer cells, leading to enhanced internalization.
Collapse
Affiliation(s)
- Lu Sun
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Qinjie Wu
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Feng Peng
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Lei Liu
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Changyang Gong
- Department of Medical Oncology, Cancer Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
187
|
An S, Jiang X, Shi J, He X, Li J, Guo Y, Zhang Y, Ma H, Lu Y, Jiang C. Single-component self-assembled RNAi nanoparticles functionalized with tumor-targeting iNGR delivering abundant siRNA for efficient glioma therapy. Biomaterials 2015; 53:330-40. [DOI: 10.1016/j.biomaterials.2015.02.084] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 02/16/2015] [Accepted: 02/19/2015] [Indexed: 11/26/2022]
|
188
|
Climent M, Quintavalle M, Miragoli M, Chen J, Condorelli G, Elia L. TGFβ Triggers miR-143/145 Transfer From Smooth Muscle Cells to Endothelial Cells, Thereby Modulating Vessel Stabilization. Circ Res 2015; 116:1753-64. [PMID: 25801897 DOI: 10.1161/circresaha.116.305178] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 03/23/2015] [Indexed: 12/30/2022]
Abstract
RATIONALE The miR-143/145 cluster is highly expressed in smooth muscle cells (SMCs), where it regulates phenotypic switch and vascular homeostasis. Whether it plays a role in neighboring endothelial cells (ECs) is still unknown. OBJECTIVE To determine whether SMCs control EC functions through passage of miR-143 and miR-145. METHODS AND RESULTS We used cocultures of SMCs and ECs under different conditions, as well as intact vessels to assess the transfer of miR-143 and miR-145 from one cell type to another. Imaging of cocultured cells transduced with fluorescent miRNAs suggested that miRNA transfer involves membrane protrusions known as tunneling nanotubes. Furthermore, we show that miRNA passage is modulated by the transforming growth factor (TGF) β pathway because both a specific transforming growth factor-β (TGFβ) inhibitor (SB431542) and an shRNA against TGFβRII suppressed the passage of miR-143/145 from SMCs to ECs. Moreover, miR-143 and miR-145 modulated angiogenesis by reducing the proliferation index of ECs and their capacity to form vessel-like structures when cultured on matrigel. We also identified hexokinase II (HKII) and integrin β 8 (ITGβ8)-2 genes essential for the angiogenic potential of ECs-as targets of miR-143 and miR-145, respectively. The inhibition of these genes modulated EC phenotype, similarly to miR-143 and miR-145 overexpression in ECs. These findings were confirmed by ex vivo and in vivo approaches, in which it was shown that TGFβ and vessel stress, respectively, triggered miR-143/145 transfer from SMCs to ECs. CONCLUSIONS Our results demonstrate that miR-143 and miR-145 act as communication molecules between SMCs and ECs to modulate the angiogenic and vessel stabilization properties of ECs.
Collapse
Affiliation(s)
- Montserrat Climent
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.)
| | - Manuela Quintavalle
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.)
| | - Michele Miragoli
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.)
| | - Ju Chen
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.)
| | - Gianluigi Condorelli
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.).
| | - Leonardo Elia
- From IRCCS MultiMedica, Milan, Italy (M.C.); Humanitas Clinical and Research Center, Rozzano (MI), Italy (M.Q., M.M., G.C., L.E.); Milan Unit of the Institute of Genetic and Biomedical Research, Rozzano (MI), Italy (G.C., L.E.); Department of Cardiovascular Diseases, University of Milan, Rozzano (MI), Italy (G.C.); Department of Clinical and Experimental Medicine, Center of Excellence for Toxicological Research (CERT), University of Parma, Parma, Italy (M.M.); and Department of Medicine, University of California, San Diego (J.C.).
| |
Collapse
|
189
|
Combined effects of pericytes in the tumor microenvironment. Stem Cells Int 2015; 2015:868475. [PMID: 26000022 PMCID: PMC4427118 DOI: 10.1155/2015/868475] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 02/28/2015] [Indexed: 12/25/2022] Open
Abstract
Pericytes are multipotent perivascular cells whose involvement in vasculature development is well established. Evidences in the literature also suggest that pericytes display immune properties and that these cells may serve as an in vivo reservoir of stem cells, contributing to the regeneration of diverse tissues. Pericytes are also capable of tumor homing and are important cellular components of the tumor microenvironment (TME). In this review, we highlight the contribution of pericytes to some classical hallmarks of cancer, namely, tumor angiogenesis, growth, metastasis, and evasion of immune destruction, and discuss how collectively these hallmarks could be tackled by therapies targeting pericytes, providing a rationale for cancer drugs aiming at the TME.
Collapse
|
190
|
Wang CF, Sarparanta MP, Mäkilä EM, Hyvönen ML, Laakkonen PM, Salonen JJ, Hirvonen JT, Airaksinen AJ, Santos HA. Multifunctional porous silicon nanoparticles for cancer theranostics. Biomaterials 2015; 48:108-18. [DOI: 10.1016/j.biomaterials.2015.01.008] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 12/29/2014] [Accepted: 01/20/2015] [Indexed: 02/07/2023]
|
191
|
Nanomedicine to overcome radioresistance in glioblastoma stem-like cells and surviving clones. Trends Pharmacol Sci 2015; 36:236-52. [PMID: 25799457 DOI: 10.1016/j.tips.2015.02.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 01/28/2015] [Accepted: 02/03/2015] [Indexed: 12/14/2022]
Abstract
Radiotherapy is one of the standard treatments for glioblastoma, but its effectiveness often encounters the phenomenon of radioresistance. This resistance was recently attributed to distinct cell contingents known as glioblastoma stem-like cells (GSCs) and dominant clones. It is characterized in particular by the activation of signaling pathways and DNA repair mechanisms. Recent advances in the field of nanomedicine offer new possibilities for radiosensitizing these cell populations. Several strategies have been developed in this direction, the first consisting of encapsulating a contrast agent or synthesizing metal-based nanocarriers to concentrate the dose gradient at the level of the target tissue. In the second strategy the physicochemical properties of the vectors are used to encapsulate a wide range of pharmacological agents which act in synergy with the ionizing radiation to destroy the cancerous cells. This review reports on the various molecular anomalies present in GSCs and the predominant role of nanomedicines in the development of radiosensitization strategies.
Collapse
|
192
|
Yan B, Qiu F, Ren L, Dai H, Fang W, Zhu H, Wang F. 99mTc-3P-RGD2 molecular imaging targeting integrin αvβ3 in head and neck squamous cancer xenograft. J Radioanal Nucl Chem 2015. [PMID: 26224987 PMCID: PMC4514642 DOI: 10.1007/s10967-015-3928-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
99mTc-3P-RGD2 and SPECT/CT were valuable tools for selecting patient likely benefit from integrin αvβ3 blocking therapy. To evaluate the feasibility of 99mTc-3P-RGD2 imaging to detect head and neck squamous cell carcinoma, 99mTc-3P-RGD2 was prepared and the relationship between its accumulation and integrin αvβ3 expression in nude mice bearing HEP-2 or CNE-1 carcinoma xenograft were analyzed. This study demonstrated that 99mTc-3P-RGD2, with high affinity to integrin αvβ3, will provide basis for αvβ3 involved individual therapy.
Collapse
Affiliation(s)
- Bing Yan
- Department of Nuclear Medicine, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730 China
| | - Fan Qiu
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006 China
| | - Ling Ren
- Department of Nuclear Medicine, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730 China
| | - Haojie Dai
- Department of Nuclear Medicine, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730 China
| | - Wei Fang
- Cardiovascular Institute & Fu Wai Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100037 China
| | - Haibo Zhu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100050 China
| | - Feng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006 China
| |
Collapse
|
193
|
Fletcher JW, Logan TF, Eitel JA, Mathias CJ, Ng Y, Lacy JL, Hutchins GD, Green MA. Whole-body PET/CT evaluation of tumor perfusion using generator-based 62Cu-ethylglyoxal bis(thiosemicarbazonato)copper(II): validation by direct comparison to 15O-water in metastatic renal cell carcinoma. J Nucl Med 2015; 56:56-62. [PMID: 25525184 PMCID: PMC11384506 DOI: 10.2967/jnumed.114.148106] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED This study was undertaken to demonstrate the feasibility of whole-body (62)Cu-ethylglyoxal bis(thiosemicarbazonato)copper(II) ((62)Cu-ETS) PET/CT tumor perfusion imaging in patients with metastatic renal carcinoma and to validate (62)Cu-ETS as a quantitative marker of tumor perfusion by direct comparison with (15)O-water perfusion imaging. METHODS PET/CT imaging of 10 subjects with stage IV renal cell cancer was performed after intravenous administration of (15)O-water (10-min dynamic list-mode study) with the heart and at least 1 tumor in the PET field of view, followed 10 min later by intravenous (62)Cu-ETS (6-min list-mode study). Whole-body (62)Cu imaging was then performed from 6 to 20 min at 2-3 min/bed position. Blood flow (K1) was quantified with both agents for normal and malignant tissues in the 21.7-cm dynamic field of view. The required arterial input functions were derived from the left atrium and, in the case of (62)Cu-ETS, corrected for partial decomposition of the agent by blood with data from an in vitro analysis using a sample of each patient's blood. This imaging protocol was repeated at an interval of 3-4 wk after initiation of a standard clinical treatment course of the antiangiogenic agent sunitinib. RESULTS All subjects received the scheduled (62)Cu-ETS doses for the dynamic and subsequent whole-body PET/CT scans, but technical issues resulted in no baseline (15)O-water data for 2 subjects. Direct comparisons of the perfusion estimates for normal tissues and tumor metastases were made in 18 paired baseline and treatment studies (10 subjects; 8 baseline studies, 10 repeated studies during treatment). There was an excellent correlation between the blood flow estimates made with (62)Cu-ETS and (15)O-water for normal tissues (muscle, thyroid, myocardium) and malignant lesions (pulmonary nodules, bone lesions); the regression line was y = 0.85x + 0.15, R(2) = 0.83, for the 88 regions analyzed. CONCLUSION (62)Cu-ETS provided high-quality whole-body PET/CT images, and (62)Cu-ETS measures of blood flow were highly and linearly correlated with (15)O-water-derived K1 values (mL(-1) ⋅ min(-1) ⋅ g). This tracer is suitable for use as a PET tracer of tumor perfusion in patients with metastatic renal cell carcinoma.
Collapse
Affiliation(s)
- James W Fletcher
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana
| | - Theodore F Logan
- The Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana; and
| | - Jacob A Eitel
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana
| | - Carla J Mathias
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana
| | - Yen Ng
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana
| | | | - Gary D Hutchins
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana
| | - Mark A Green
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
194
|
Saha A, Mohapatra S, Kurkute P, Jana B, Sarkar J, Mondal P, Ghosh S. Targeted delivery of a novel peptide–docetaxel conjugate to MCF-7 cells through neuropilin-1 receptor: reduced toxicity and enhanced efficacy of docetaxel. RSC Adv 2015. [DOI: 10.1039/c5ra16741h] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A novel peptide docetaxel conjugate selectively kills NRP-1 overexpressing breast cancer cells, enhances anticancer activity of docetaxel without affecting the NRP-1 negative cells.
Collapse
Affiliation(s)
- Abhijit Saha
- Organic & Medicinal Chemistry Division
- CSIR-Indian Institute of Chemical Biology
- Kolkata-700032
- India
| | - Saswat Mohapatra
- Academy of Scientific and Innovative Research (AcSIR)
- CSIR-Indian Institute of Chemical Biology Campus
- Kolkata 700 032
- India
| | - Prashant Kurkute
- Organic & Medicinal Chemistry Division
- CSIR-Indian Institute of Chemical Biology
- Kolkata-700032
- India
| | - Batakrishna Jana
- Organic & Medicinal Chemistry Division
- CSIR-Indian Institute of Chemical Biology
- Kolkata-700032
- India
| | - Jayita Sarkar
- Organic & Medicinal Chemistry Division
- CSIR-Indian Institute of Chemical Biology
- Kolkata-700032
- India
| | - Prasenjit Mondal
- Academy of Scientific and Innovative Research (AcSIR)
- CSIR-Indian Institute of Chemical Biology Campus
- Kolkata 700 032
- India
| | - Surajit Ghosh
- Organic & Medicinal Chemistry Division
- CSIR-Indian Institute of Chemical Biology
- Kolkata-700032
- India
- Academy of Scientific and Innovative Research (AcSIR)
| |
Collapse
|
195
|
Miao W, Zheng S, Dai H, Wang F, Jin X, Zhu Z, Jia B. Comparison of 99mTc-3PRGD2 integrin receptor imaging with 99mTc-MDP bone scan in diagnosis of bone metastasis in patients with lung cancer: a multicenter study. PLoS One 2014; 9:e111221. [PMID: 25338281 PMCID: PMC4206469 DOI: 10.1371/journal.pone.0111221] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 09/15/2014] [Indexed: 11/19/2022] Open
Abstract
PURPOSE 99mTc-3PRGD2, a promising tracer targeting integrin receptor, may serve as a novel tumor-specific agent for single photon emission computed tomography (SPECT). A multi-center study was prospectively designed to evaluate the diagnostic accuracy of 99mTc-3PRGD2 imaging for bone metastasis in patients with lung cancer in comparison with the conventional 99mTc-MDP bone scan. METHODS The patients underwent whole-body scan and chest tomography successively at both 1 h and 4 h after intravenous injection of 11.1 MBq/Kg 99mTc-3PRGD2. 99mTc-MDP whole-body bone scan was routinely performed within 1 week for comparison. Three experienced nuclear medicine physicians blindly read the 99mTc-3PRGD2 and 99mTc-MDP images. The final diagnosis was established based on the comprehensive assessment of all available data. RESULTS A total of 44 patients (29 male, 59±10 years old) with suspected lung cancer were recruited from 4 centers. Eighty-nine bone lesions in 18 patients were diagnosed as metastases and 23 bone lesions in 9 patients were benign. In a lesion-based analysis, 99mTc-3PRGD2 imaging demonstrated a sensitivity, specificity, and accuracy of 92.1%, 91.3%, and 92.0%, respectively. The corresponding diagnostic values for 99mTc-MDP bone scan were 87.6%, 60.9%, and 82.1%, respectively in the same patients. 99mTc-MDP bone scan had better contrast in most lesions, whereas the 99mTc-3PRGD2 imaging seemed to be more effective to exclude pseudo-positive lesions and detect bone metastases without osteogenesis. CONCLUSION 99mTc-3PRGD2 is a novel tumor-specific agent based on SPECT technology with a promising value in diagnosis of bone metastasis in patients with lung cancer. TRIAL REGISTRATION ClinicalTrials.gov NCT01737112.
Collapse
Affiliation(s)
- Weibing Miao
- Department of Nuclear Medicine, the 1st Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Shan Zheng
- Department of Nuclear Medicine, the 1st Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Haojie Dai
- Department of Nuclear Medicine, Beijing Tongren Hospital, Beijing, China
| | - Feng Wang
- Department of Nuclear Medicine, the 1st Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaona Jin
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhaohui Zhu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bing Jia
- Medical Isotopes Research Center, Peking University, Beijing, China
| |
Collapse
|
196
|
Ma Q, Chen B, Gao S, Ji T, Wen Q, Song Y, Zhu L, Xu Z, Liu L. 99mTc-3P4-RGD2 scintimammography in the assessment of breast lesions: comparative study with 99mTc-MIBI. PLoS One 2014; 9:e108349. [PMID: 25250628 PMCID: PMC4176966 DOI: 10.1371/journal.pone.0108349] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 08/19/2014] [Indexed: 12/03/2022] Open
Abstract
Purpose To compare the potential application of 99mTc-3P-Arg-Gly-Asp (99mTc-3P4-RGD2) scintimammography (SMM) and 99mTc-methoxyisobutylisonitrile (99mTc-MIBI) SMM for the differentiation of malignant from benign breast lesions. Method Thirty-six patients with breast masses on physical examination and/or suspicious mammography results that required fine needle aspiration cytology biopsy (FNAB) were included in the study. 99mTc-3P4-RGD2 and 99mTc-MIBI SMM were performed with single photon emission computed tomography (SPECT) at 60 min and 20 min respectively after intravenous injection of 738±86 MBq radiotracers on a separate day. Images were evaluated by the tumor to non-tumor localization ratios (T/NT). Receiver operating characteristic (ROC) curve analysis was performed on each radiotracer to calculate the cut-off values of quantitative indices and to compare the diagnostic performance for the ability to differentiate malignant from benign diseases. Results The mean T/NT ratio of 99mTc-3P4-RGD2 in malignant lesions was significantly higher than that in benign lesions (3.54±1.51 vs. 1.83±0.98, p<0.001). The sensitivity, specificity, and accuracy of 99mTc-3P4-RGD2 SMM were 89.3%, 90.9% and 89.7%, respectively, with a T/NT cut-off value of 2.40. The mean T/NT ratio of 99mTc-MIBI in malignant lesions was also significantly higher than that in benign lesions (2.86±0.99 vs. 1.51±0.61, p<0.001). The sensitivity, specificity and accuracy of 99mTc-MIBI SMM were 87.5%, 72.7% and 82.1%, respectively, with a T/NT cut-off value of 1.45. According to the ROC analysis, the area under the curve for 99mTc-3P4-RGD2 SMM (area = 0.851) was higher than that for 99mTc-MIBI SMM (area = 0.781), but the statistical difference was not significant. Conclusion 99mTc-3P4-RGD2 SMM does not provide any significant advantage over the established 99mTc-MIBI SMM for the detection of primary breast cancer. The T/NT ratio of 99mTc-3P4-RGD2 SMM was significantly higher than that of 99mTc-MIBI SMM. Both tracers could offer an alternative method for elucidating non-diagnostic mammograms.
Collapse
Affiliation(s)
- Qingjie Ma
- China-Japan Union Hospital, Jilin University, Changchun, China
| | - Bin Chen
- China-Japan Union Hospital, Jilin University, Changchun, China
| | - Shi Gao
- China-Japan Union Hospital, Jilin University, Changchun, China
| | - Tiefeng Ji
- China-Japan Union Hospital, Jilin University, Changchun, China
| | - Qiang Wen
- China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yan Song
- China-Japan Union Hospital, Jilin University, Changchun, China
| | - Lei Zhu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Zheli Xu
- China-Japan Union Hospital, Jilin University, Changchun, China
| | - Lin Liu
- China-Japan Union Hospital, Jilin University, Changchun, China
- * E-mail:
| |
Collapse
|
197
|
Lao X, Li B, Liu M, Chen J, Gao X, Zheng H. Increased antitumor activity of tumor-specific peptide modified thymopentin. Biochimie 2014; 107 Pt B:277-85. [PMID: 25236717 DOI: 10.1016/j.biochi.2014.09.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 09/08/2014] [Indexed: 11/29/2022]
Abstract
Thymopoietin pentapeptide (thymopentin, TP5), an immunomodulatory peptide, has been successfully used as an immune system enhancer for treating immune deficiency, cancer, and infectious diseases. However, poor penetration into tumors remains a key limitation to the efficacy and application of TP5. iRGD (CRGDK/RGPD/EC) has been introduced to certain anticancer agents, and increased specific tumor penetrability of drugs and cell internalization have been observed. In the present study, we fused this iRGD fragment with the C-terminal of TP5 to yield a new product, TP5-iRGD. Cell attachment assay showed that TP5-iRGD exhibits more extensive attachment to the melanoma cell line B16F10 than wild-type TP5. Tumor cell viability assay showed that iRGD conjugation with the TP5 C-terminus increases the basal antiproliferative activity of the pentapeptide against the melanoma cell line B16F10, the human lung cancer cell line H460, and the human breast cancer cell line MCF-7. Subsequent injections of TP5-iRGD inhibited in vivo melanoma progression more efficiently than the native TP5. Murine spleen lymphocyte proliferation assay also showed that TP5-iRGD and the parent pentapeptide feature nearly identical spleen lymphocyte proliferation activities. We built an integrin αvβ3 and TP5-iRGD computational binding model to investigate the mechanism by which TP5-iRGD promotes increased activity further. Conjugation with iRGD promotes binding to integrin αvβ3, thereby increasing the tumor-homing efficiency of the resultant peptide. These experimental and computational observations of increased TP5-iRGD activity help broaden the usage of TP5 and reflect the great application potential of the peptide as an anticancer agent.
Collapse
Affiliation(s)
- Xingzhen Lao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Bin Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Meng Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jiao Chen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xiangdong Gao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Heng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
198
|
Panzarini E, Dini L. Nanomaterial-induced autophagy: a new reversal MDR tool in cancer therapy? Mol Pharm 2014; 11:2527-38. [PMID: 24921216 DOI: 10.1021/mp500066v] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Most of the therapeutic strategies to counteract cancer imply killing of malignant cells. The most exploited cell death mechanism in cancer therapies is apoptosis, but recently, a lot of papers report that other mechanisms, mainly autophagy, could represent a new line of attack in the fight against cancer. One of the limitations for the effectiveness of the approved clinical treatments is the phenomenon of multidrug resistance (MDR) which enables the cancer cells to develop resistance to therapy, especially for chemotherapy. The MDR mechanisms include (a) decreased uptake of drug, (b) reduced intracellular drug concentration by efflux pumps, (c) altered cell cycle checkpoints, (d) altered drug targets, (e) increased metabolism of drugs, (f) induced emergency response genes to impair apoptotic pathway, and (g) altered drug detoxification. Great efforts have been made to reverse MDR. Currently, autophagy and nanosized drug delivery systems (DDSs) belonging to nanomaterials (NMs) provide alternative strategies to circumvent MDR. Nanosized DDSs are very promising tools to accumulate chemotherapeutics at targeting sites and control temporal and spatial drug release into tumor cells. On the other hand, autophagy could overrule drug resistance upon its activation by ensuring cell death via switching its prosurvival role to a prodeath one or by mediating the occurrence of cell death, i.e., apoptosis or necrosis. Likewise, the autophagy inhibition could counteract MDR by sensitizing the cells to anticancer molecules, i.e., Src family tyrosine kinase (SFK) inhibitors or 5-fluorouracil. Noteworthy, autophagy has been recently indicated to be a common cellular response to NMs, corroborating the fascinating idea of the exploitation of NM-induced autophagy in nanomedicine therapy. This review focuses on recently published literature about the relationship between MDR reversal and NMs or autophagy pointing to hypothesize a pivotal role of autophagy modulation induced by NMs in counteracting MDR.
Collapse
Affiliation(s)
- Elisa Panzarini
- Department of Biological and Environmental Science and Technology (Di.S.Te.B.A.), University of Salento , 73100 Lecce, Italy
| | | |
Collapse
|
199
|
Singh T, Kothapalli C, Varma D, Nicoll SB, Vazquez M. Carboxymethylcellulose hydrogels support central nervous system-derived tumor-cell chemotactic migration: Comparison with conventional extracellular matrix macromolecules. J Biomater Appl 2014; 29:433-41. [DOI: 10.1177/0885328214532969] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The local microenvironment plays an important role in maintaining the dynamics of the extracellular matrix and the cell–extracellular matrix relationship. The extracellular matrix is a complex network of macromolecules with distinct mechanical and biochemical characteristics. Disruptions in extracellular matrix homeostasis are associated with the onset of cancer. The extracellular matrix becomes highly disorganized, and the cell–matrix relationship changes, resulting in altered cell-signaling processes and metastasis. Medulloblastoma is one of the most common malignant pediatric brain tumors in the United States. In order to gain a better understanding of the interplay between cell–extracellular matrix interactions and cell-migratory responses in tumors, eight different matrix macromolecule formulations were investigated using a medulloblastoma-derived cell line: poly-d-lysine, matrigel, laminin, collagen 1, fibronectin, a 10% blend of laminin–collagen 1, a 20% blend of laminin–collagen 1, and a cellulose-derived hydrogel, carboxymethylcellulose. Over time, the average changes in cell morphology were quantified in 2D and 3D, as was migration in the presence and absence of the chemoattractant, epidermal growth factor. Data revealed that carboxymethylcellulose allowed for a cell–extracellular matrix relationship typically believed to be present in tumors, with cells exhibiting a rounded, amoeboid morphology consistent with chemotactic migration, while the other matrices promoted an elongated cell shape as well as both haptotactic and chemotactic motile processes. Therefore, carboxymethylcellulose hydrogels may serve as effective platforms for investigating central nervous system-derived tumor-cell migration in response to soluble factors.
Collapse
Affiliation(s)
- Tanya Singh
- Department of Biomedical Engineering, The City College of New York-CUNY, USA
| | | | - Devika Varma
- Department of Biomedical Engineering, The City College of New York-CUNY, USA
| | - Steven B Nicoll
- Department of Biomedical Engineering, The City College of New York-CUNY, USA
| | - Maribel Vazquez
- Department of Biomedical Engineering, The City College of New York-CUNY, USA
| |
Collapse
|
200
|
Heylman C, Sobrino A, Shirure VS, Hughes CC, George SC. A strategy for integrating essential three-dimensional microphysiological systems of human organs for realistic anticancer drug screening. Exp Biol Med (Maywood) 2014; 239:1240-54. [PMID: 24740872 DOI: 10.1177/1535370214525295] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cancer is one of the leading causes of morbidity and mortality around the world. Despite some success, traditional anticancer drugs developed to reduce tumor growth face important limitations primarily due to undesirable bone marrow and cardiovascular toxicity. Many drugs fail in clinical development after showing promise in preclinical trials, suggesting that the available in vitro and animal models are poor predictors of drug efficacy and toxicity in humans. Thus, novel models that more accurately mimic the biology of human organs are necessary for high-throughput drug screening. Three-dimensional (3D) microphysiological systems can utilize induced pluripotent stem cell technology, tissue engineering, and microfabrication techniques to develop tissue models of human tumors, cardiac muscle, and bone marrow on the order of 1 mm(3) in size. A functional network of human capillaries and microvessels to overcome diffusion limitations in nutrient delivery and waste removal can also nourish the 3D microphysiological tissues. Importantly, the 3D microphysiological tissues are grown on optically clear platforms that offer non-invasive and non-destructive image acquisition with subcellular resolution in real time. Such systems offer a new paradigm for high-throughput drug screening and will significantly improve the efficiency of identifying new drugs for cancer treatment that minimize cardiac and bone marrow toxicity.
Collapse
Affiliation(s)
- Christopher Heylman
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA 92697, USA
| | - Agua Sobrino
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| | - Venktesh S Shirure
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA 92697, USA
| | - Christopher Cw Hughes
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA 92697, USA Department of Molecular Biology and Biochemistry, University of California, Irvine, CA 92697, USA
| | - Steven C George
- Department of Biomedical Engineering, University of California, Irvine, CA 92697, USA The Edwards Lifesciences Center for Advanced Cardiovascular Technology, University of California, Irvine, CA 92697, USA Department of Chemical Engineering and Materials Science, University of California, Irvine, CA 92697, USA Department of Medicine, University of California, Irvine, CA 92697, USA
| |
Collapse
|