51
|
Wang J, Li T, Yue C, Zhong S, Yang X, Li J, Li Y. Preparation of nanoparticles of β-cyclodextrin-loaded scutellarein anti-tumor activity research by targeting integrin αvβ3. Cancer Nanotechnol 2021. [DOI: 10.1186/s12645-021-00102-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Abstract
Background
The problems associated with the poor water solubility of anticancer drugs are one of the most important challenges in achieving effective cancer therapy. The present study was designed to evaluate the effect of scutellarein on human colon cancer cells in vitro by using a target αvβ3 novel scutellarein (Scu)-loaded niosome nanoparticle (β-CD-CL-Scu-cRGD).
Results
β-CD-CL-Scu-cRGD has a diameter of 140.2 nm and zeta potential of − 11.3 mV with constant physicochemical stability. The MTT assay showed both Scu and β-CD-CL-Scu-cRGD caused a decrease in cell proliferation and viability of LoVo, but β-CD-CL-Scu-cRGD showed better activity in vitro. Colony formation assay and flow cytometry assay showed that β-CD-CL-Scu-cRGD has a better effect on cell proliferation and apoptosis. In vivo, animal experimental results showed that β-CD-CL-Scu-cRGD can significantly inhibit tumor growth, and the bodyweight of mice decreases during the treatment of scutellarein and its derivatives. β-CD-CL-Scu-cRGD could inhibit the protein levels of Ki67 and αvβ3, thereby inhibiting tumor growth.
Conclusions
Although further in vitro and in vivo studies are necessary, our results suggested that β-CD-CL-Scu-cRGD could be an outstanding carrier to deliver Scu for potential therapeutic approaches into colon cancer.
Collapse
|
52
|
Ghalamfarsa F, Khatami SH, Vakili O, Taheri-Anganeh M, Tajbakhsh A, Savardashtaki A, Fazli Y, Uonaki LR, Shabaninejad Z, Movahedpour A, Ghalamfarsa G. Bispecific antibodies in colorectal cancer therapy: recent insights and emerging concepts. Immunotherapy 2021; 13:1355-1367. [PMID: 34641708 DOI: 10.2217/imt-2021-0107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Colorectal cancer (CRC) is identified as a life-threatening malignancy. Despite several efforts and proceedings available for CRC therapy, it is still a health concern. Among a vast array of novel therapeutic procedures, employing bispecific antibodies (BsAbs) is currently considered to be a promising approach for cancer therapy. BsAbs, as a large family of molecules designed to realize two distinct epitopes or antigens, can be beneficial microgadgets to target the tumor-associated antigen pairs. On the other hand, applying the immune system's capabilities to attack malignant cells has been proven as a tremendous development in cancer therapeutic projects. The current study has attempted to overview some of the approved BsAbs in CRC therapy and those under clinical trials. For this purpose, reputable scientific search engines and databases, such as PubMed, ScienceDirect, Google Scholar, Scopus, etc., were explored using the keywords 'bispecific antibodies', 'colorectal cancer', 'immunotherapy' and 'tumor markers'.
Collapse
Affiliation(s)
- Farideh Ghalamfarsa
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyyed Hossein Khatami
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy & Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mortaza Taheri-Anganeh
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amir Savardashtaki
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yousef Fazli
- Dena Clinical Diagnostic Laboratory, Yasuj, Iran
| | - Leila Rezaei Uonaki
- Department of Biotechnology, School of Science, Shahrekord University, Shahrekord, Iran
| | - Zahra Shabaninejad
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ahmad Movahedpour
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.,Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ghasem Ghalamfarsa
- Department of Microbiology & Immunology, School of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| |
Collapse
|
53
|
Effect of Peptide Receptor Radionuclide Therapy in Combination with Temozolomide against Tumor Angiogenesis in a Glioblastoma Model. Cancers (Basel) 2021; 13:cancers13195029. [PMID: 34638512 PMCID: PMC8507696 DOI: 10.3390/cancers13195029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/27/2021] [Accepted: 10/04/2021] [Indexed: 11/16/2022] Open
Abstract
Cell adhesion receptor integrin αvβ3 is a promising biomarker for developing tumor-angiogenesis targeted theranostics. In this study, we aimed to examine the therapeutic potential of peptide receptor radionuclide therapy (PRRT) with 188Re-IDA-D-[c(RGDfK)]2 (11.1 MBq). The results showed that the tumor volume was significantly decreased by 81% compared with the vehicle-treated group in U87-MG xenografts. The quantitative in vivo anti-angiogenic responses of PRRT were obtained using 99mTc-IDA-D-[c(RGDfK)]2 SPECT and corresponded to the measured tumor volume. PRRT combined with temozolomide (TMZ) resulted in a 93% reduction in tumor volume, which was markedly greater than that of each agent used individually. In addition, histopathological characterization showed that PRRT combined with TMZ was superior to PRRT or TMZ alone, even when TMZ was used at half dose. Overall, our results indicated that integrin-targeted PRRT and TMZ combined therapy might be a new medical tool for the effective treatment of glioblastoma.
Collapse
|
54
|
Ewert KK, Scodeller P, Simón-Gracia L, Steffes VM, Wonder EA, Teesalu T, Safinya CR. Cationic Liposomes as Vectors for Nucleic Acid and Hydrophobic Drug Therapeutics. Pharmaceutics 2021; 13:1365. [PMID: 34575441 PMCID: PMC8465808 DOI: 10.3390/pharmaceutics13091365] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/09/2021] [Accepted: 08/21/2021] [Indexed: 12/15/2022] Open
Abstract
Cationic liposomes (CLs) are effective carriers of a variety of therapeutics. Their applications as vectors of nucleic acids (NAs), from long DNA and mRNA to short interfering RNA (siRNA), have been pursued for decades to realize the promise of gene therapy, with approvals of the siRNA therapeutic patisiran and two mRNA vaccines against COVID-19 as recent milestones. The long-term goal of developing optimized CL-based NA carriers for a broad range of medical applications requires a comprehensive understanding of the structure of these vectors and their interactions with cell membranes and components that lead to the release and activity of the NAs within the cell. Structure-activity relationships of lipids for CL-based NA and drug delivery must take into account that these lipids act not individually but as components of an assembly of many molecules. This review summarizes our current understanding of how the choice of the constituting lipids governs the structure of their CL-NA self-assemblies, which constitute distinct liquid crystalline phases, and the relation of these structures to their efficacy for delivery. In addition, we review progress toward CL-NA nanoparticles for targeted NA delivery in vivo and close with an outlook on CL-based carriers of hydrophobic drugs, which may eventually lead to combination therapies with NAs and drugs for cancer and other diseases.
Collapse
Affiliation(s)
- Kai K. Ewert
- Materials, Physics, and Molecular, Cellular, and Developmental Biology Departments, and Biomolecular Science and Engineering Program, University of California at Santa Barbara, Santa Barbara, CA 93106, USA; (V.M.S.); (E.A.W.)
| | - Pablo Scodeller
- Laboratory of Precision- and Nanomedicine, Institute of Biomedicine and Translational Medicine, Centre of Excellence for Translational Medicine, University of Tartu, Ravila 14b, 50411 Tartu, Estonia; (P.S.); (L.S.-G.)
| | - Lorena Simón-Gracia
- Laboratory of Precision- and Nanomedicine, Institute of Biomedicine and Translational Medicine, Centre of Excellence for Translational Medicine, University of Tartu, Ravila 14b, 50411 Tartu, Estonia; (P.S.); (L.S.-G.)
| | - Victoria M. Steffes
- Materials, Physics, and Molecular, Cellular, and Developmental Biology Departments, and Biomolecular Science and Engineering Program, University of California at Santa Barbara, Santa Barbara, CA 93106, USA; (V.M.S.); (E.A.W.)
| | - Emily A. Wonder
- Materials, Physics, and Molecular, Cellular, and Developmental Biology Departments, and Biomolecular Science and Engineering Program, University of California at Santa Barbara, Santa Barbara, CA 93106, USA; (V.M.S.); (E.A.W.)
| | - Tambet Teesalu
- Laboratory of Precision- and Nanomedicine, Institute of Biomedicine and Translational Medicine, Centre of Excellence for Translational Medicine, University of Tartu, Ravila 14b, 50411 Tartu, Estonia; (P.S.); (L.S.-G.)
- Center for Nanomedicine and Department of Cell, Molecular and Developmental Biology, University of California at Santa Barbara, Santa Barbara, CA 93106, USA
| | - Cyrus R. Safinya
- Materials, Physics, and Molecular, Cellular, and Developmental Biology Departments, and Biomolecular Science and Engineering Program, University of California at Santa Barbara, Santa Barbara, CA 93106, USA; (V.M.S.); (E.A.W.)
| |
Collapse
|
55
|
Tarvirdipour S, Skowicki M, Schoenenberger CA, Palivan CG. Peptide-Assisted Nucleic Acid Delivery Systems on the Rise. Int J Mol Sci 2021; 22:9092. [PMID: 34445799 PMCID: PMC8396486 DOI: 10.3390/ijms22169092] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/13/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022] Open
Abstract
Concerns associated with nanocarriers' therapeutic efficacy and side effects have led to the development of strategies to advance them into targeted and responsive delivery systems. Owing to their bioactivity and biocompatibility, peptides play a key role in these strategies and, thus, have been extensively studied in nanomedicine. Peptide-based nanocarriers, in particular, have burgeoned with advances in purely peptidic structures and in combinations of peptides, both native and modified, with polymers, lipids, and inorganic nanoparticles. In this review, we summarize advances on peptides promoting gene delivery systems. The efficacy of nucleic acid therapies largely depends on cell internalization and the delivery to subcellular organelles. Hence, the review focuses on nanocarriers where peptides are pivotal in ferrying nucleic acids to their site of action, with a special emphasis on peptides that assist anionic, water-soluble nucleic acids in crossing the membrane barriers they encounter on their way to efficient function. In a second part, we address how peptides advance nanoassembly delivery tools, such that they navigate delivery barriers and release their nucleic acid cargo at specific sites in a controlled fashion.
Collapse
Affiliation(s)
- Shabnam Tarvirdipour
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- Department of Biosystem Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Michal Skowicki
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- NCCR-Molecular Systems Engineering, BPR1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Cora-Ann Schoenenberger
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- NCCR-Molecular Systems Engineering, BPR1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| | - Cornelia G. Palivan
- Department of Chemistry, University of Basel, Mattenstrasse 24a, 4058 Basel, Switzerland; (S.T.); (M.S.)
- NCCR-Molecular Systems Engineering, BPR1095, Mattenstrasse 24a, 4058 Basel, Switzerland
| |
Collapse
|
56
|
Conrado FO, Sharkey LC, Cudney SE, Paul AL, Matthews MH, Robinson NA. Pathology in Practice. J Am Vet Med Assoc 2021; 259:261-264. [PMID: 34242074 DOI: 10.2460/javma.259.3.261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
57
|
Bui TM, Yalom LK, Sumagin R. Tumor-associated neutrophils: orchestrating cancer pathobiology and therapeutic resistance. Expert Opin Ther Targets 2021; 25:573-583. [PMID: 34236924 DOI: 10.1080/14728222.2021.1954162] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Introduction: Neutrophils or polymorphonuclear cells (PMNs) account for a considerable portion of the tumor immune stroma. Emerging single-cell transcriptomic analyses have elucidated the striking cellular heterogeneity of PMNs during homeostasis and pathologic conditions and have established their diverse roles in cancer. PMNs have emerged as important players in cancer pathobiology and therapeutic resistance. Tumor-associated neutrophils (TANs) effector functions influence tumor development and resistance or response to therapy.Areas covered: This review focuses on PMN heterogeneity and functional diversity in the context of carcinogenesis. TANs, by activating diverse signaling pathways, contribute to cancer progression and resistance to therapies. Mechanisms by which TANs impact therapeutic resistance include alterations of the tumoral DNA damage response, angiogenesis, reactivation of cancer dormancy, enhancement of tumor cell proliferation/survival and immune evasion.Expert opinion: With the emerging phenotypic and function heterogeneity of TANs, targeting specific TAN functions in developing tumors can lead to translatable therapeutic approaches and limit drug resistance. We propose that combining specific targeting of TAN activity with standard cancer therapy can help patients achieving a complete response and prevent cancer relapse.
Collapse
Affiliation(s)
- Triet M Bui
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lenore K Yalom
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ronen Sumagin
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
58
|
Belhabib I, Zaghdoudi S, Lac C, Bousquet C, Jean C. Extracellular Matrices and Cancer-Associated Fibroblasts: Targets for Cancer Diagnosis and Therapy? Cancers (Basel) 2021; 13:3466. [PMID: 34298680 PMCID: PMC8303391 DOI: 10.3390/cancers13143466] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/25/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Solid cancer progression is dictated by neoplastic cell features and pro-tumoral crosstalks with their microenvironment. Stroma modifications, such as fibroblast activation into cancer-associated fibroblasts (CAFs) and extracellular matrix (ECM) remodeling, are now recognized as critical events for cancer progression and as potential therapeutic or diagnostic targets. The recent appreciation of the key, complex and multiple roles of the ECM in cancer and of the CAF diversity, has revolutionized the field and raised innovative but challenging questions. Here, we rapidly present CAF heterogeneity in link with their specific ECM remodeling features observed in cancer, before developing each of the impacts of such ECM modifications on tumor progression (survival, angiogenesis, pre-metastatic niche, chemoresistance, etc.), and on patient prognosis. Finally, based on preclinical studies and recent results obtained from clinical trials, we highlight key mechanisms or proteins that are, or may be, used as potential therapeutic or diagnostic targets, and we report and discuss benefits, disappointments, or even failures, of recently reported stroma-targeting strategies.
Collapse
Affiliation(s)
| | | | | | | | - Christine Jean
- Centre de Recherche en Cancérologie de Toulouse (CRCT), INSERM U1037, Université Toulouse III Paul Sabatier, ERL5294 CNRS, 31037 Toulouse, France; (I.B.); (S.Z.); (C.L.); (C.B.)
| |
Collapse
|
59
|
Liu M, Wu C, Ke L, Li Z, Wu YL. Emerging Biomaterials-Based Strategies for Inhibiting Vasculature Function in Cancer Therapy. SMALL METHODS 2021; 5:e2100347. [PMID: 34927997 DOI: 10.1002/smtd.202100347] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/20/2021] [Indexed: 06/14/2023]
Abstract
The constant feeding of oxygen and nutrients through the blood vasculature has a vital role in maintaining tumor growth. Interestingly, recent endeavors have shown that nanotherapeutics with the strategy to block tumor blood vessels feeding nutrients and oxygen for starvation therapy can be helpful in cancer treatment. However, this field has not been detailed. Hence, this review will present an exhaustive summary of the existing biomaterial based strategies to disrupt tumor vascular function for effective cancer treatment, including hydrogel or nanogel-mediated local arterial embolism, thrombosis activator loaded nano-material-mediated vascular occlusion and anti-vascular drugs that block tumor vascular function, which may be beneficial to the design of anti-cancer nanomedicine by targeting the tumor vascular system.
Collapse
Affiliation(s)
- Minting Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Caisheng Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Lingjie Ke
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Zhiguo Li
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
60
|
Balberova OV, Bykov EV, Shnayder NA, Petrova MM, Gavrilyuk OA, Kaskaeva DS, Soloveva IA, Petrov KV, Mozheyko EY, Medvedev GV, Nasyrova RF. The "Angiogenic Switch" and Functional Resources in Cyclic Sports Athletes. Int J Mol Sci 2021; 22:ijms22126496. [PMID: 34204341 PMCID: PMC8234968 DOI: 10.3390/ijms22126496] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 12/22/2022] Open
Abstract
Regular physical activity in cyclic sports can influence the so-called “angiogenic switch”, which is considered as an imbalance between proangiogenic and anti-angiogenic molecules. Disruption of the synthesis of angiogenic molecules can be caused by local changes in tissues under the influence of excessive physical exertion and its consequences, such as chronic oxidative stress and associated hypoxia, metabolic acidosis, sports injuries, etc. A review of publications on signaling pathways that activate and inhibit angiogenesis in skeletal muscles, myocardium, lung, and nervous tissue under the influence of intense physical activity in cyclic sports. Materials: We searched PubMed, SCOPUS, Web of Science, Google Scholar, Clinical keys, and e-LIBRARY databases for full-text articles published from 2000 to 2020, using keywords and their combinations. Results: An important aspect of adaptation to training loads in cyclic sports is an increase in the number of capillaries in muscle fibers, which improves the metabolism of skeletal muscles and myocardium, as well as nervous and lung tissue. Recent studies have shown that myocardial endothelial cells not only respond to hemodynamic forces and paracrine signals from neighboring cells, but also take an active part in heart remodeling processes, stimulating the growth and contractility of cardiomyocytes or the production of extracellular matrix proteins in myofibroblasts. As myocardial vascularization plays a central role in the transition from adaptive heart hypertrophy to heart failure, further study of the signaling mechanisms involved in the regulation of angiogenesis in the myocardium is important in sports practice. The study of the “angiogenic switch” problem in the cerebrovascular and cardiovascular systems allows us to claim that the formation of new vessels is mediated by a complex interaction of all growth factors. Although the lungs are one of the limiting systems of the body in cyclic sports, their response to high-intensity loads and other environmental stresses is often overlooked. Airway epithelial cells are the predominant source of several growth factors throughout lung organogenesis and appear to be critical for normal alveolarization, rapid alveolar proliferation, and normal vascular development. There are many controversial questions about the role of growth factors in the physiology and pathology of the lungs. The presented review has demonstrated that when doing sports, it is necessary to give a careful consideration to the possible positive and negative effects of growth factors on muscles, myocardium, lung tissue, and brain. Primarily, the “angiogenic switch” is important in aerobic sports (long distance running). Conclusions: Angiogenesis is a physiological process of the formation of new blood capillaries, which play an important role in the functioning of skeletal muscles, myocardium, lung, and nervous tissue in athletes. Violation of the “angiogenic switch” as a balance between proangiogenic and anti-angiogenic molecules can lead to a decrease in the functional resources of the nervous, musculoskeletal, cardiovascular, and respiratory systems in athletes and, as a consequence, to a decrease in sports performance.
Collapse
Affiliation(s)
- Olga V. Balberova
- Research Institute of Olympic Sports, Ural State University of Physical Culture, 454091 Chelyabinsk, Russia;
- Correspondence: (O.V.B.); (N.A.S.); (R.F.N.)
| | - Evgeny V. Bykov
- Research Institute of Olympic Sports, Ural State University of Physical Culture, 454091 Chelyabinsk, Russia;
| | - Natalia A. Shnayder
- V.M. Bekhterev National Medical Research Center for Neurology and Psychiatry, Department of Personalized Psychiatry and Neurology, 192019 Saint Petersburg, Russia
- Department of Outpatient Therapy and Family Medicine with a Postgraduate Course, Shared Core Facilities Molecular and Cell Technologies, Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (M.M.P.); (D.S.K.)
- Correspondence: (O.V.B.); (N.A.S.); (R.F.N.)
| | - Marina M. Petrova
- Department of Outpatient Therapy and Family Medicine with a Postgraduate Course, Shared Core Facilities Molecular and Cell Technologies, Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (M.M.P.); (D.S.K.)
| | - Oksana A. Gavrilyuk
- The Department of Polyclinic Therapy and Family Medicine and Healthy Lifesttyle with a Course of PE, V. F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia;
| | - Daria S. Kaskaeva
- Department of Outpatient Therapy and Family Medicine with a Postgraduate Course, Shared Core Facilities Molecular and Cell Technologies, Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (M.M.P.); (D.S.K.)
| | - Irina A. Soloveva
- Department of Hospital Therapy and Immunology with a Postgraduate Course, Shared Core Facilities Molecular and Cell Technologies, Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia;
| | - Kirill V. Petrov
- Department of Physical and Rehabilitation Medicine with a Postgraduate Course, Shared Core Facilities Molecular and Cell Technologies, Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (K.V.P.); (E.Y.M.)
| | - Elena Y. Mozheyko
- Department of Physical and Rehabilitation Medicine with a Postgraduate Course, Shared Core Facilities Molecular and Cell Technologies, Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia; (K.V.P.); (E.Y.M.)
| | - German V. Medvedev
- R. R. Vreden National Medical Research Center for Traumatology and Orthopedics, Department of Hand Surgery with Microsurgical Equipment, 195427 Saint-Petersburg, Russia;
| | - Regina F. Nasyrova
- V.M. Bekhterev National Medical Research Center for Neurology and Psychiatry, Department of Personalized Psychiatry and Neurology, 192019 Saint Petersburg, Russia
- Correspondence: (O.V.B.); (N.A.S.); (R.F.N.)
| |
Collapse
|
61
|
Tsai WC, Chang KM, Kao KJ. Dynamic Contrast Enhanced MRI and Intravoxel Incoherent Motion to Identify Molecular Subtypes of Breast Cancer with Different Vascular Normalization Gene Expression. Korean J Radiol 2021; 22:1021-1033. [PMID: 34047501 PMCID: PMC8236363 DOI: 10.3348/kjr.2020.0760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 02/01/2021] [Accepted: 02/04/2021] [Indexed: 11/15/2022] Open
Abstract
Objective To assess the expression of vascular normalization genes in different molecular subtypes of breast cancer and to determine whether molecular subtypes with a higher vascular normalization gene expression can be identified using dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) and intravoxel incoherent motion (IVIM) diffusion-weighted imaging (DWI). Materials and Methods This prospective study evaluated 306 female (mean age ± standard deviation, 50 ± 10 years), recruited between January 2014 and August 2017, who had de novo breast cancer larger than 1 cm in diameter (308 tumors). DCE MRI followed by IVIM DWI studies using 11 different b-values (0 to 1200 s/mm2) were performed on a 1.5T MRI system. The Tofts model and segmented biexponential IVIM analysis were used. For each tumor, the molecular subtype (according to six [I-VI] subtypes and PAM50 subtypes), expression profile of genes for vascular normalization, pericytes, and normal vascular signatures were determined using freshly frozen tissue. Statistical associations between imaging parameters and molecular subtypes were examined using logistic regression or linear regression with a significance level of p = 0.05. Results Breast cancer subtypes III and VI and PAM50 subtypes luminal A and normal-like exhibited a higher expression of genes for vascular normalization, pericyte markers, and normal vessel function signature (p < 0.001 for all) compared to other subtypes. Subtypes III and VI and PAM50 subtypes luminal A and normal-like, versus the remaining subtypes, showed significant associations with Ktrans, kep, vp, and IAUGCBN90 on DEC MRI, with relatively smaller values in the former. The subtype grouping was significantly associated with D, with relatively less restricted diffusion in subtypes III and VI and PAM50 subtypes luminal A and normal-like. Conclusion DCE MRI and IVIM parameters may identify molecular subtypes of breast cancers with a different vascular normalization gene expression.
Collapse
Affiliation(s)
- Wan Chen Tsai
- Department of Radiology, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan.,School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan.
| | - Kai Ming Chang
- Department of Research, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan
| | - Kuo Jang Kao
- Department of Research, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan
| |
Collapse
|
62
|
Carrion CC, Nasrollahzadeh M, Sajjadi M, Jaleh B, Soufi GJ, Iravani S. Lignin, lipid, protein, hyaluronic acid, starch, cellulose, gum, pectin, alginate and chitosan-based nanomaterials for cancer nanotherapy: Challenges and opportunities. Int J Biol Macromol 2021; 178:193-228. [PMID: 33631269 DOI: 10.1016/j.ijbiomac.2021.02.123] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/07/2021] [Accepted: 02/16/2021] [Indexed: 12/11/2022]
Abstract
Although nanotechnology-driven drug delivery systems are relatively new, they are rapidly evolving since the nanomaterials are deployed as effective means of diagnosis and delivery of assorted therapeutic agents to targeted intracellular sites in a controlled release manner. Nanomedicine and nanoparticulate drug delivery systems are rapidly developing as they play crucial roles in the development of therapeutic strategies for various types of cancer and malignancy. Nevertheless, high costs, associated toxicity and production of complexities are some of the critical barriers for their applications. Green nanomedicines have continually been improved as one of the viable approaches towards tumor drug delivery, thus making a notable impact on which considerably affect cancer treatment. In this regard, the utilization of natural and renewable feedstocks as a starting point for the fabrication of nanosystems can considerably contribute to the development of green nanomedicines. Nanostructures and biopolymers derived from natural and biorenewable resources such as proteins, lipids, lignin, hyaluronic acid, starch, cellulose, gum, pectin, alginate, and chitosan play vital roles in the development of cancer nanotherapy, imaging and management. This review uncovers recent investigations on diverse nanoarchitectures fabricated from natural and renewable feedstocks for the controlled/sustained and targeted drug/gene delivery systems against cancers including an outlook on some of the scientific challenges and opportunities in this field. Various important natural biopolymers and nanomaterials for cancer nanotherapy are covered and the scientific challenges and opportunities in this field are reviewed.
Collapse
Affiliation(s)
- Carolina Carrillo Carrion
- Department of Organic Chemistry, University of Córdoba, Campus de Rabanales, Edificio Marie Curie, Ctra Nnal IV-A Km. 396, E-14014 Cordoba, Spain
| | | | - Mohaddeseh Sajjadi
- Department of Chemistry, Faculty of Science, University of Qom, Qom 37185-359, Iran
| | - Babak Jaleh
- Department of Physics, Bu-Ali Sina University, 65174 Hamedan, Iran
| | | | - Siavash Iravani
- Faculty of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
63
|
Ayo A, Laakkonen P. Peptide-Based Strategies for Targeted Tumor Treatment and Imaging. Pharmaceutics 2021; 13:pharmaceutics13040481. [PMID: 33918106 PMCID: PMC8065807 DOI: 10.3390/pharmaceutics13040481] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 02/03/2023] Open
Abstract
Cancer is one of the leading causes of death worldwide. The development of cancer-specific diagnostic agents and anticancer toxins would improve patient survival. The current and standard types of medical care for cancer patients, including surgery, radiotherapy, and chemotherapy, are not able to treat all cancers. A new treatment strategy utilizing tumor targeting peptides to selectively deliver drugs or applicable active agents to solid tumors is becoming a promising approach. In this review, we discuss the different tumor-homing peptides discovered through combinatorial library screening, as well as native active peptides. The different structure–function relationship data that have been used to improve the peptide’s activity and conjugation strategies are highlighted.
Collapse
Affiliation(s)
- Abiodun Ayo
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
| | - Pirjo Laakkonen
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
- Laboratory Animal Center, HiLIFE—Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
- Correspondence: ; Tel.: +358-50-4489100
| |
Collapse
|
64
|
Chen Q, Song H, Yu J, Kim K. Current Development and Applications of Super-Resolution Ultrasound Imaging. SENSORS 2021; 21:s21072417. [PMID: 33915779 PMCID: PMC8038018 DOI: 10.3390/s21072417] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023]
Abstract
Abnormal changes of the microvasculature are reported to be key evidence of the development of several critical diseases, including cancer, progressive kidney disease, and atherosclerotic plaque. Super-resolution ultrasound imaging is an emerging technology that can identify the microvasculature noninvasively, with unprecedented spatial resolution beyond the acoustic diffraction limit. Therefore, it is a promising approach for diagnosing and monitoring the development of diseases. In this review, we introduce current super-resolution ultrasound imaging approaches and their preclinical applications on different animals and disease models. Future directions and challenges to overcome for clinical translations are also discussed.
Collapse
Affiliation(s)
- Qiyang Chen
- Department of Bioengineering, School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- Center for Ultrasound Molecular Imaging and Therapeutics, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Hyeju Song
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Korea;
| | - Jaesok Yu
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Korea;
- DGIST Robotics Research Center, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Korea
- Correspondence: (J.Y.); (K.K.)
| | - Kang Kim
- Department of Bioengineering, School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- Center for Ultrasound Molecular Imaging and Therapeutics, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Cardiology, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Mechanical Engineering and Materials Science, School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Correspondence: (J.Y.); (K.K.)
| |
Collapse
|
65
|
Li B, Chu T, Wei J, Zhang Y, Qi F, Lu Z, Gao C, Zhang T, Jiang E, Xu J, Xu J, Li S, Nie G. Platelet-Membrane-Coated Nanoparticles Enable Vascular Disrupting Agent Combining Anti-Angiogenic Drug for Improved Tumor Vessel Impairment. NANO LETTERS 2021; 21:2588-2595. [PMID: 33650872 DOI: 10.1021/acs.nanolett.1c00168] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Compared with traditional chemotherapeutics, vascular disruption agents (VDAs) have the advantages of rapidly blocking the supply of nutrients and starving tumors to death. Although the VDAs are effective under certain scenarios, this treatment triggers angiogenesis in the later stage of therapy that frequently leads to tumor recurrence and treatment failure. Additionally, the nonspecific tumor targeting and considerable side effects also impede the clinical applications of VDAs. Here we develop a customized strategy that combines a VDA with an anti-angiogenic drug (AAD) using mesoporous silica nanoparticles (MSNs) coated with platelet membrane for the self-assembled tumor targeting accumulation. The tailor-made nanoparticles accumulate in tumor tissues through the targeted adhesion of platelet membrane surface to damaged vessel sites, resulting in significant vascular disruption and efficient anti-angiogenesis in animal models. This study demonstrates the promising potential of combining VDA and AAD in a single nanoplatform for tumor eradication.
Collapse
Affiliation(s)
- Bozhao Li
- College of Pharmaceutical Science, Jilin University, Changchun 130021, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Tianjiao Chu
- College of Pharmaceutical Science, Jilin University, Changchun 130021, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Jingyan Wei
- College of Pharmaceutical Science, Jilin University, Changchun 130021, China
| | - Yinlong Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Feilong Qi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Zefang Lu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chao Gao
- College of Pharmaceutical Science, Jilin University, Changchun 130021, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Tianjiao Zhang
- College of Pharmaceutical Science, Jilin University, Changchun 130021, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Ershuai Jiang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junchao Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiaqi Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Suping Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
- The GBA National Institute for Nanotechnology Innovation, Guangzhou 510530, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
- The GBA National Institute for Nanotechnology Innovation, Guangzhou 510530, China
| |
Collapse
|
66
|
Kotowski K, Rosik J, Machaj F, Supplitt S, Wiczew D, Jabłońska K, Wiechec E, Ghavami S, Dzięgiel P. Role of PFKFB3 and PFKFB4 in Cancer: Genetic Basis, Impact on Disease Development/Progression, and Potential as Therapeutic Targets. Cancers (Basel) 2021; 13:909. [PMID: 33671514 PMCID: PMC7926708 DOI: 10.3390/cancers13040909] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 12/11/2022] Open
Abstract
Glycolysis is a crucial metabolic process in rapidly proliferating cells such as cancer cells. Phosphofructokinase-1 (PFK-1) is a key rate-limiting enzyme of glycolysis. Its efficiency is allosterically regulated by numerous substances occurring in the cytoplasm. However, the most potent regulator of PFK-1 is fructose-2,6-bisphosphate (F-2,6-BP), the level of which is strongly associated with 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase activity (PFK-2/FBPase-2, PFKFB). PFK-2/FBPase-2 is a bifunctional enzyme responsible for F-2,6-BP synthesis and degradation. Four isozymes of PFKFB (PFKFB1, PFKFB2, PFKFB3, and PFKFB4) have been identified. Alterations in the levels of all PFK-2/FBPase-2 isozymes have been reported in different diseases. However, most recent studies have focused on an increased expression of PFKFB3 and PFKFB4 in cancer tissues and their role in carcinogenesis. In this review, we summarize our current knowledge on all PFKFB genes and protein structures, and emphasize important differences between the isoenzymes, which likely affect their kinase/phosphatase activities. The main focus is on the latest reports in this field of cancer research, and in particular the impact of PFKFB3 and PFKFB4 on tumor progression, metastasis, angiogenesis, and autophagy. We also present the most recent achievements in the development of new drugs targeting these isozymes. Finally, we discuss potential combination therapies using PFKFB3 inhibitors, which may represent important future cancer treatment options.
Collapse
Affiliation(s)
- Krzysztof Kotowski
- Department of Histology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (K.K.); (K.J.)
| | - Jakub Rosik
- Department of Pathology, Pomeranian Medical University, 71-252 Szczecin, Poland; (J.R.); (F.M.)
| | - Filip Machaj
- Department of Pathology, Pomeranian Medical University, 71-252 Szczecin, Poland; (J.R.); (F.M.)
| | - Stanisław Supplitt
- Department of Genetics, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Daniel Wiczew
- Department of Biochemical Engineering, Wroclaw University of Science and Technology, 50-370 Wroclaw, Poland;
- Laboratoire de physique et chimie théoriques, Université de Lorraine, F-54000 Nancy, France
| | - Karolina Jabłońska
- Department of Histology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (K.K.); (K.J.)
| | - Emilia Wiechec
- Department of Biomedical and Clinical Sciences (BKV), Division of Cell Biology, Linköping University, Region Östergötland, 581 85 Linköping, Sweden;
- Department of Otorhinolaryngology in Linköping, Anesthetics, Operations and Specialty Surgery Center, Region Östergötland, 581 85 Linköping, Sweden
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Research Institute in Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Piotr Dzięgiel
- Department of Histology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland; (K.K.); (K.J.)
- Department of Physiotherapy, Wroclaw University School of Physical Education, 51-612 Wroclaw, Poland
| |
Collapse
|
67
|
Hao SY, Qi ZY, Wang S, Wang XR, Chen SW. Synthesis and bioevaluation of N-(3,4,5-trimethoxyphenyl)-1H-pyrazolo[3,4-b]pyridin-3-amines as tubulin polymerization inhibitors with anti-angiogenic effects. Bioorg Med Chem 2021; 31:115985. [DOI: 10.1016/j.bmc.2020.115985] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/21/2020] [Accepted: 12/24/2020] [Indexed: 02/08/2023]
|
68
|
Piscatelli JA, Ban J, Lucas AT, Zamboni WC. Complex Factors and Challenges that Affect the Pharmacology, Safety and Efficacy of Nanocarrier Drug Delivery Systems. Pharmaceutics 2021; 13:114. [PMID: 33477395 PMCID: PMC7830329 DOI: 10.3390/pharmaceutics13010114] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/01/2021] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
Major developments in nanomedicines, such as nanoparticles (NPs), nanosomes, and conjugates, have revolutionized drug delivery capabilities over the past four decades. Although nanocarrier agents provide numerous advantages (e.g., greater solubility and duration of systemic exposure) compared to their small-molecule counterparts, there is considerable inter-patient variability seen in the systemic disposition, tumor delivery and overall pharmacological effects (i.e., anti-tumor efficacy and unwanted toxicity) of NP agents. This review aims to provide a summary of fundamental factors that affect the disposition of NPs in the treatment of cancer and why they should be evaluated during preclinical and clinical development. Furthermore, this chapter will highlight some of the translational challenges associated with elements of NPs and how these issues can only be addressed by detailed and novel pharmacology studies.
Collapse
Affiliation(s)
- Joseph A. Piscatelli
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; (J.A.P.); (J.B.); (W.C.Z.)
| | - Jisun Ban
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; (J.A.P.); (J.B.); (W.C.Z.)
| | - Andrew T. Lucas
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; (J.A.P.); (J.B.); (W.C.Z.)
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Carolina Center of Cancer Nanotechnology Excellence, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - William C. Zamboni
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA; (J.A.P.); (J.B.); (W.C.Z.)
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Carolina Center of Cancer Nanotechnology Excellence, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
69
|
Gao J, Wang S, Dong X, Wang Z. RGD-expressed bacterial membrane-derived nanovesicles enhance cancer therapy via multiple tumorous targeting. Theranostics 2021; 11:3301-3316. [PMID: 33537088 PMCID: PMC7847689 DOI: 10.7150/thno.51988] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/09/2020] [Indexed: 12/19/2022] Open
Abstract
Background: A tumor microenvironment is a complicated multicellular system comprised of tumor cells, immune cells and blood vessels. Blood vessels are the barriers for drug tissue penetration. Effectively treating a cancer requires drug delivery systems to overcome biological barriers present in tumor microenvironments (TMEs). Methods: We designed a drug delivery system made of bacterial (Escherichia coli) double layer membrane-derived nanovesicles (DMVs) with the expression of RGD peptides and endogenous targeting ligands of bacteria. The physical and biological characteristics of DMVs were assessed by cryogenic transmission electron microscopy, western blotting, flow cytometry and confocal microscopy. Doxorubicin (DOX) was loaded in DMVs via a pH gradient driven drug loading method. Therapeutical effects of DOX-loaded DMVs were studied in a melanoma xenograft mouse model. Results:In vitro and in vivo experiments showed that DMVs can target neutrophils and monocytes that mediated the transport of DMVs across blood vessel barriers and they can also directly target tumor vasculature and tumor cells, resulting in enhanced delivery of therapeutics to TMEs. Furthermore, we developed a remote drug loading approach to efficiently encapsulate DOX inside DMVs, and the drug loading was 12% (w/w). In the B16-F10 melanoma mouse model, we showed that DOX-RGD-DMVs significantly inhibited the tumor growth compared to several controls. Conclusion: Our studies reveal that DMVs are a powerful tool to simultaneously target multiple cells in TMEs, thus increasing drug delivery for improved cancer therapies.
Collapse
|
70
|
Sonju JJ, Dahal A, Singh SS, Jois SD. Peptide-functionalized liposomes as therapeutic and diagnostic tools for cancer treatment. J Control Release 2021; 329:624-644. [PMID: 33010333 PMCID: PMC8082750 DOI: 10.1016/j.jconrel.2020.09.055] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 12/26/2022]
Abstract
Clinically efficacious medication in anticancer therapy has been successfully designed with liposome-based nanomedicine. The liposomal formulation in cancer drug delivery can be facilitated with a functionalized peptide that mediates the specific drug delivery opportunities with increased drug penetrability, specific accumulation in the targeted site, and enhanced therapeutic efficacy. This review aims to focus on recent advances in peptide-functionalized liposomal formulation techniques in cancer diagnosis and treatment regarding recently published literature. It also will highlight different aspects of novel liposomal formulation techniques that incorporate surface functionalization with peptides for better anticancer effect and current challenges in peptide-functionalized liposomal drug formulation.
Collapse
Affiliation(s)
- Jafrin Jobayer Sonju
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Achyut Dahal
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Sitanshu S Singh
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Seetharama D Jois
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA.
| |
Collapse
|
71
|
Abstract
Tumor-homing peptides are widely used for improving tumor selectivity of anticancer drugs and imaging agents. The goal is to increase tumor uptake and reduce accumulation at nontarget sites. Here, we describe current approaches for tumor-homing peptide identification and validation, and provide comprehensive overview of classes of tumor-homing peptides undergoing preclinical and clinical development. We focus on unique mechanistic features and applications of a recently discovered class of tumor-homing peptides, tumor-penetrating C-end Rule (CendR) peptides, that can be used for tissue penetrative targeting of extravascular tumor tissue. Finally, we discuss unanswered questions and future directions in the field of development of peptide-guided smart drugs and imaging agents.
Collapse
|
72
|
Fakhri KU, Sultan A, Mushtaque M, Hasan MR, Nafees S, Hafeez ZB, Zafaryab M, Rizwanullah M, Sharma D, Bano F, AlMalki WH, Ahmad FJ, Rizvi MMA. Obstructions in Nanoparticles Conveyance, Nano-Drug Retention, and EPR Effect in Cancer Therapies. HANDBOOK OF RESEARCH ON ADVANCEMENTS IN CANCER THERAPEUTICS 2021. [DOI: 10.4018/978-1-7998-6530-8.ch026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In this chapter, the authors first review nano-devices that are mixtures of biologic molecules and synthetic polymers like nano-shells and nano-particles for the most encouraging applications for different cancer therapies. Nano-sized medications additionally spill especially into tumor tissue through penetrable tumor vessels and are then held in the tumor bed because of diminished lymphatic drainage. This procedure is known as the enhanced penetrability and retention (EPR) impact. Nonetheless, while the EPR impact is generally held to improve conveyance of nano-medications to tumors, it in certainty offers not exactly a 2-overlay increment in nano-drug conveyance contrasted with basic ordinary organs, bringing about medication concentration that is not adequate for restoring most malignant growths. In this chapter, the authors likewise review different obstructions for nano-sized medication conveyance and to make the conveyance of nano-sized medications to tumors progressively successful by expanding on the EPR impact..
Collapse
Affiliation(s)
| | | | | | | | | | | | - Md Zafaryab
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Md Rizwanullah
- School of Pharmaceutical Education and Research, Jamia Hamdard, India
| | - Deepti Sharma
- Institute of Nuclear Medicine and Allied Sciences, India
| | - Farhad Bano
- National Institute of Immunology, New Delhi, India
| | | | - Farhan Jalees Ahmad
- School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | | |
Collapse
|
73
|
Adityan S, Tran M, Bhavsar C, Wu SY. Nano-therapeutics for modulating the tumour microenvironment: Design, development, and clinical translation. J Control Release 2020; 327:512-532. [DOI: 10.1016/j.jconrel.2020.08.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 08/08/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022]
|
74
|
Mehrotra N, Kharbanda S, Singh H. Peptide-based combination nanoformulations for cancer therapy. Nanomedicine (Lond) 2020; 15:2201-2217. [PMID: 32914691 DOI: 10.2217/nnm-2020-0220] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Research in cancer therapy is moving towards the use of biomolecules in combination with conventional approaches for improved disease outcome. Among the biomolecules explored, peptides are strong contenders due to their small size, high specificity, low systemic toxicity and wide inter/intracellular targets. The use of nanoformulations for such combination approaches can lead to further improvement in efficacy by reducing off-target cytotoxicity, increasing circulation time, tumor penetration and accumulation. This review focuses on nanodelivery systems for peptide-based combinations with chemo, immuno, radiation and hormone therapy. It gives an overview of the latest therapeutic research being conducted using combination nanoformulations with anticancer peptides, cell penetrating/tumor targeting peptides, peptide nanocarriers, peptidomimetics, peptide-based hormones and peptide vaccines. The challenges hindering clinical translation are also discussed.
Collapse
Affiliation(s)
- Neha Mehrotra
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Surender Kharbanda
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Harpal Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| |
Collapse
|
75
|
Keam S, Gill S, Ebert MA, Nowak AK, Cook AM. Enhancing the efficacy of immunotherapy using radiotherapy. Clin Transl Immunology 2020; 9:e1169. [PMID: 32994997 PMCID: PMC7507442 DOI: 10.1002/cti2.1169] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/04/2020] [Accepted: 08/02/2020] [Indexed: 12/12/2022] Open
Abstract
Recent clinical breakthroughs in cancer immunotherapy, especially with immune checkpoint blockade, offer great hope for cancer sufferers - and have greatly changed the landscape of cancer treatment. However, whilst many patients achieve clinical responses, others experience minimal benefit or do not respond to immune checkpoint blockade at all. Researchers are therefore exploring multimodal approaches by combining immune checkpoint blockade with conventional cancer therapies to enhance the efficacy of treatment. A growing body of evidence from both preclinical studies and clinical observations indicates that radiotherapy could be a powerful driver to augment the efficacy of immune checkpoint blockade, because of its ability to activate the antitumor immune response and potentially overcome resistance. In this review, we describe how radiotherapy induces DNA damage and apoptosis, generates immunogenic cell death and alters the characteristics of key immune cells in the tumor microenvironment. We also discuss recent preclinical work and clinical trials combining radiotherapy and immune checkpoint blockade in thoracic and other cancers. Finally, we discuss the scheduling of immune checkpoint blockade and radiotherapy, biomarkers predicting responses to combination therapy, and how these novel data may be translated into the clinic.
Collapse
Affiliation(s)
- Synat Keam
- National Centre for Asbestos Related DiseasesPerthWAAustralia
- School of MedicineThe University of Western AustraliaPerthWAAustralia
| | - Suki Gill
- Department of Radiation OncologySir Charles Gairdner HospitalPerthWAAustralia
| | - Martin A Ebert
- Department of Radiation OncologySir Charles Gairdner HospitalPerthWAAustralia
- School of Physics, Mathematics and ComputingThe University of Western AustraliaPerthWAAustralia
| | - Anna K Nowak
- National Centre for Asbestos Related DiseasesPerthWAAustralia
- School of MedicineThe University of Western AustraliaPerthWAAustralia
- Department of Medical OncologySir Charles Gairdner HospitalNedlands, PerthWAAustralia
| | - Alistair M Cook
- National Centre for Asbestos Related DiseasesPerthWAAustralia
- School of MedicineThe University of Western AustraliaPerthWAAustralia
| |
Collapse
|
76
|
Yao Y, Zhou Y, Liu L, Xu Y, Chen Q, Wang Y, Wu S, Deng Y, Zhang J, Shao A. Nanoparticle-Based Drug Delivery in Cancer Therapy and Its Role in Overcoming Drug Resistance. Front Mol Biosci 2020; 7:193. [PMID: 32974385 DOI: 10.3389/fmolb.2020.00193/bibtex] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/21/2020] [Indexed: 05/26/2023] Open
Abstract
Nanotechnology has been extensively studied and exploited for cancer treatment as nanoparticles can play a significant role as a drug delivery system. Compared to conventional drugs, nanoparticle-based drug delivery has specific advantages, such as improved stability and biocompatibility, enhanced permeability and retention effect, and precise targeting. The application and development of hybrid nanoparticles, which incorporates the combined properties of different nanoparticles, has led this type of drug-carrier system to the next level. In addition, nanoparticle-based drug delivery systems have been shown to play a role in overcoming cancer-related drug resistance. The mechanisms of cancer drug resistance include overexpression of drug efflux transporters, defective apoptotic pathways, and hypoxic environment. Nanoparticles targeting these mechanisms can lead to an improvement in the reversal of multidrug resistance. Furthermore, as more tumor drug resistance mechanisms are revealed, nanoparticles are increasingly being developed to target these mechanisms. Moreover, scientists have recently started to investigate the role of nanoparticles in immunotherapy, which plays a more important role in cancer treatment. In this review, we discuss the roles of nanoparticles and hybrid nanoparticles for drug delivery in chemotherapy, targeted therapy, and immunotherapy and describe the targeting mechanism of nanoparticle-based drug delivery as well as its function on reversing drug resistance.
Collapse
Affiliation(s)
- Yihan Yao
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lihong Liu
- Department of Radiation Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanyan Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Qiang Chen
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yali Wang
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shijie Wu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongchuan Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
77
|
Yao Y, Zhou Y, Liu L, Xu Y, Chen Q, Wang Y, Wu S, Deng Y, Zhang J, Shao A. Nanoparticle-Based Drug Delivery in Cancer Therapy and Its Role in Overcoming Drug Resistance. Front Mol Biosci 2020; 7:193. [PMID: 32974385 PMCID: PMC7468194 DOI: 10.3389/fmolb.2020.00193] [Citation(s) in RCA: 483] [Impact Index Per Article: 120.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022] Open
Abstract
Nanotechnology has been extensively studied and exploited for cancer treatment as nanoparticles can play a significant role as a drug delivery system. Compared to conventional drugs, nanoparticle-based drug delivery has specific advantages, such as improved stability and biocompatibility, enhanced permeability and retention effect, and precise targeting. The application and development of hybrid nanoparticles, which incorporates the combined properties of different nanoparticles, has led this type of drug-carrier system to the next level. In addition, nanoparticle-based drug delivery systems have been shown to play a role in overcoming cancer-related drug resistance. The mechanisms of cancer drug resistance include overexpression of drug efflux transporters, defective apoptotic pathways, and hypoxic environment. Nanoparticles targeting these mechanisms can lead to an improvement in the reversal of multidrug resistance. Furthermore, as more tumor drug resistance mechanisms are revealed, nanoparticles are increasingly being developed to target these mechanisms. Moreover, scientists have recently started to investigate the role of nanoparticles in immunotherapy, which plays a more important role in cancer treatment. In this review, we discuss the roles of nanoparticles and hybrid nanoparticles for drug delivery in chemotherapy, targeted therapy, and immunotherapy and describe the targeting mechanism of nanoparticle-based drug delivery as well as its function on reversing drug resistance.
Collapse
Affiliation(s)
- Yihan Yao
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lihong Liu
- Department of Radiation Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yanyan Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, China.,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Qiang Chen
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yali Wang
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shijie Wu
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongchuan Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
78
|
|
79
|
Tang Y, Chen M, Xie Q, Li L, Zhu L, Ma Q, Gao S. Construction and evaluation of hyaluronic acid-based copolymers as a targeted chemotherapy drug carrier for cancer therapy. NANOTECHNOLOGY 2020; 31:305702. [PMID: 32272454 DOI: 10.1088/1361-6528/ab884d] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Melanoma (MM) is a highly aggressive skin cancer with limited treatment options. Although chemotherapy has been using for advanced melanoma treatment, the lack of targetability, the poor biocompatibility and the severe side effects still hamper the wide applications of chemotherapy agents in MM management. Herein, a biocompatible and biodegradable polymeric hyaluronic acid nanoparticle (HANP) encapsulated with Paclitaxel (PTX) was developed for MM targeted therapy. Our results showed that PTX at 37 ± 2.1% (w/w) was able to be loaded into HANP with over 5 d of stability under physiological conditions. In vitro, HANP/PTX presented hyaluronidase-dependent drug release. Compared to free PTX, HANP/PTX demonstrated a 6-75 times higher growth inhibition in five different cancer cells, while only presenting minimum toxicity to normal cells. After intravenous administration at a 10 mg kg-1 equivalent dose of PTX, HANP/PTX significantly ablated MM tumor growth in a mouse model. As confirmed by 18F-fluoro-2-deoxy-D-glucose (FDG) positron emission tomography (PET) imaging, the tumors started to respond to the HANP/PTX as early as 7 d after the initial treatment, which will significantly benefit for personalized treatment. In conclusion, the HANP/PTX nanocomplex demonstrated great promise as a translational nanomedicine for cancer chemotherapy.
Collapse
Affiliation(s)
- Yuting Tang
- Department of Nuclear Medicine, China-Japan Union Hospital, Jilin University, Changchun 130033, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
80
|
Wu J, Ning P, Gao R, Feng Q, Shen Y, Zhang Y, Li Y, Xu C, Qin Y, Plaza GR, Bai Q, Fan X, Li Z, Han Y, Lesniak MS, Fan H, Cheng Y. Programmable ROS-Mediated Cancer Therapy via Magneto-Inductions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1902933. [PMID: 32596106 PMCID: PMC7312334 DOI: 10.1002/advs.201902933] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/06/2020] [Indexed: 05/05/2023]
Abstract
Reactive oxygen species (ROS), a group of oxygen derived radicals and derivatives, can induce cancer cell death via elevated oxidative stress. A spatiotemporal approach with safe and deep-tissue penetration capabilities to elevate the intracellular ROS level is highly desirable for precise cancer treatment. Here, a mechanical-thermal induction therapy (MTIT) strategy is developed for a programmable increase of ROS levels in cancer cells via assembly of magnetic nanocubes integrated with alternating magnetic fields. The magneto-based mechanical and thermal stimuli can disrupt the lysosomes, which sequentially induce the dysfunction of mitochondria. Importantly, intracellular ROS concentrations are responsive to the magneto-triggers and play a key role for synergistic cancer treatment. In vivo experiments reveal the effectiveness of MTIT for efficient eradication of glioma and breast cancer. By remote control of the force and heat using magnetic nanocubes, MTIT is a promising physical approach to trigger the biochemical responses for precise cancer treatment.
Collapse
Affiliation(s)
- Jiaojiao Wu
- Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East HospitalTongji University School of Medicine1800 Yuntai RoadShanghai200123China
- Collaborative Innovation Center for Brain ScienceTongji UniversityShanghai200092China
| | - Peng Ning
- Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East HospitalTongji University School of Medicine1800 Yuntai RoadShanghai200123China
| | - Rui Gao
- Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East HospitalTongji University School of Medicine1800 Yuntai RoadShanghai200123China
| | - Qishuai Feng
- Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East HospitalTongji University School of Medicine1800 Yuntai RoadShanghai200123China
| | - Yajing Shen
- Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East HospitalTongji University School of Medicine1800 Yuntai RoadShanghai200123China
| | - Yifan Zhang
- College of Chemistry and Materials ScienceNorthwest UniversityXi'an710127China
| | - Yingze Li
- Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East HospitalTongji University School of Medicine1800 Yuntai RoadShanghai200123China
| | - Chang Xu
- Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East HospitalTongji University School of Medicine1800 Yuntai RoadShanghai200123China
| | - Yao Qin
- Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East HospitalTongji University School of Medicine1800 Yuntai RoadShanghai200123China
| | - Gustavo R. Plaza
- Center for Biomedical TechnologyUniversidad Politécnica de MadridPozuelo de Alarcón28223Spain
| | - Qianwen Bai
- Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East HospitalTongji University School of Medicine1800 Yuntai RoadShanghai200123China
| | - Xing Fan
- Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East HospitalTongji University School of Medicine1800 Yuntai RoadShanghai200123China
| | - Zhenguang Li
- Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East HospitalTongji University School of Medicine1800 Yuntai RoadShanghai200123China
| | - Yu Han
- Feinberg School of MedicineNorthwestern University676 North Saint Clair Street, Suite 2210ChicagoIL60611USA
| | - Maciej S. Lesniak
- Feinberg School of MedicineNorthwestern University676 North Saint Clair Street, Suite 2210ChicagoIL60611USA
| | - Haiming Fan
- College of Chemistry and Materials ScienceNorthwest UniversityXi'an710127China
| | - Yu Cheng
- Institute for Regenerative Medicine, Institute for Translational Nanomedicine, Shanghai East HospitalTongji University School of Medicine1800 Yuntai RoadShanghai200123China
- Collaborative Innovation Center for Brain ScienceTongji UniversityShanghai200092China
| |
Collapse
|
81
|
Fan Q, Cui X, Guo H, Xu Y, Zhang G, Peng B. Application of rare earth-doped nanoparticles in biological imaging and tumor treatment. J Biomater Appl 2020; 35:237-263. [DOI: 10.1177/0885328220924540] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Rare earth-doped nanoparticles have been widely used in disease diagnosis, drug delivery, tumor therapy, and bioimaging. Among various bioimaging methods, the fluorescence imaging technology based on the rare earth-doped nanoparticles can visually display the cell activity and lesion evolution in living animals, which is a powerful tool in biological technology and has being widely applied in medical and biological fields. Especially in the band of near infrared (700–1700 nm), the emissions show the characteristics of deep penetration due to low absorption, low photon scattering, and low autofluorescence interference. Furthermore, the rare earth-doped nanoparticles can be endowed with the water solubility, biocompatibility, drug-loading ability, and the targeting ability for different tumors by surface functionalization. This confirms its potential in the cancer diagnosis and treatment. In this review, we summarized the recent progress in the application of rare earth-doped nanoparticles in the field of bioimaging and tumor treatment. The luminescent mechanism, properties, and structure design were also discussed.
Collapse
Affiliation(s)
- Qi Fan
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Science (CAS), Xi’an, Shaanxi, PR China
- University of Chinese Academy of Sciences (UCAS), Beijing, PR China
| | - Xiaoxia Cui
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Science (CAS), Xi’an, Shaanxi, PR China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, PR China
| | - Haitao Guo
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Science (CAS), Xi’an, Shaanxi, PR China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, PR China
| | - Yantao Xu
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Science (CAS), Xi’an, Shaanxi, PR China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, PR China
| | - Guangwei Zhang
- Zhejiang Fountain Aptitude Technology Inc., Hangzhou, Zhejiang, PR China
| | - Bo Peng
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Science (CAS), Xi’an, Shaanxi, PR China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, PR China
| |
Collapse
|
82
|
Azizi M, Dianat-Moghadam H, Salehi R, Farshbaf M, Iyengar D, Sau S, Iyer AK, Valizadeh H, Mehrmohammadi M, Hamblin MR. Interactions Between Tumor Biology and Targeted Nanoplatforms for Imaging Applications. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1910402. [PMID: 34093104 PMCID: PMC8174103 DOI: 10.1002/adfm.201910402] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Indexed: 05/04/2023]
Abstract
Although considerable efforts have been conducted to diagnose, improve, and treat cancer in the past few decades, existing therapeutic options are insufficient, as mortality and morbidity rates remain high. Perhaps the best hope for substantial improvement lies in early detection. Recent advances in nanotechnology are expected to increase the current understanding of tumor biology, and will allow nanomaterials to be used for targeting and imaging both in vitro and in vivo experimental models. Owing to their intrinsic physicochemical characteristics, nanostructures (NSs) are valuable tools that have received much attention in nanoimaging. Consequently, rationally designed NSs have been successfully employed in cancer imaging for targeting cancer-specific or cancer-associated molecules and pathways. This review categorizes imaging and targeting approaches according to cancer type, and also highlights some new safe approaches involving membrane-coated nanoparticles, tumor cell-derived extracellular vesicles, circulating tumor cells, cell-free DNAs, and cancer stem cells in the hope of developing more precise targeting and multifunctional nanotechnology-based imaging probes in the future.
Collapse
Affiliation(s)
- Mehdi Azizi
- Proteomics Research Centre, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran
| | - Hassan Dianat-Moghadam
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz 5165665621, Iran
| | - Roya Salehi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht Street, Tabriz 516615731, Iran
| | - Masoud Farshbaf
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 6581151656, Iran
| | - Disha Iyengar
- U-BiND Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Samaresh Sau
- U-BiND Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Arun K Iyer
- U-BiND Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Hadi Valizadeh
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Golgasht Street, Tabriz 516615731, Iran
| | | | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
83
|
Yang PP, Zhang K, He PP, Fan Y, Gao XJ, Gao X, Chen ZM, Hou DY, Li Y, Yi Y, Cheng DB, Zhang JP, Shi L, Zhang XZ, Wang L, Wang H. A biomimetic platelet based on assembling peptides initiates artificial coagulation. SCIENCE ADVANCES 2020; 6:eaaz4107. [PMID: 32766439 PMCID: PMC7385434 DOI: 10.1126/sciadv.aaz4107] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 03/18/2020] [Indexed: 05/16/2023]
Abstract
Platelets play a critical role in the regulation of coagulation, one of the essential processes in life, attracting great attention. However, mimicking platelets for in vivo artificial coagulation is still a great challenge due to the complexity of the process. Here, we design platelet-like nanoparticles (pNPs) based on self-assembled peptides that initiate coagulation and form clots in blood vessels. The pNPs first bind specifically to a membrane glycoprotein (i.e., CD105) overexpressed on angiogenetic endothelial cells in the tumor site and simultaneously transform into activated platelet-like nanofibers (apNFs) through ligand-receptor interactions. Next, the apNFs expose more binding sites and recruit and activate additional pNPs, forming artificial clots in both phantom and animal models. The pNPs are proven to be safe in mice without systemic coagulation. The self-assembling peptides mimic platelets and achieve artificial coagulation in vivo, thus providing a promising therapeutic strategy for tumors.
Collapse
Affiliation(s)
- Pei-Pei Yang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
| | - Kuo Zhang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
- Faculty of Chemistry, Northeast Normal University, Changchun 130024, P. R. China
| | - Ping-Ping He
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
| | - Yu Fan
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
| | - Xuejiao J. Gao
- Key Laboratory of Functional Small Organic Molecule, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang 330022, P. R. China
| | - Xingfa Gao
- Key Laboratory of Functional Small Organic Molecule, College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang 330022, P. R. China
| | - Zi-Ming Chen
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
- Faculty of Chemistry, Northeast Normal University, Changchun 130024, P. R. China
| | - Da-Yong Hou
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
| | - Yuan Li
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
| | - Yu Yi
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
| | - Dong-Bing Cheng
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
| | - Jing-Ping Zhang
- Faculty of Chemistry, Northeast Normal University, Changchun 130024, P. R. China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, P. R. China
| | - Lei Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing 100190, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
84
|
Mello FV, de Moraes GN, Maia RC, Kyeremateng J, Iram SH, Santos-Oliveira R. The Effect of Nanosystems on ATP-Binding Cassette Transporters: Understanding the Influence of Nanosystems on Multidrug Resistance Protein-1 and P-glycoprotein. Int J Mol Sci 2020; 21:E2630. [PMID: 32290047 PMCID: PMC7178121 DOI: 10.3390/ijms21072630] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/06/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023] Open
Abstract
The cancer multidrug resistance is involved in the failure of several treatments during cancer treatment. It is a phenomenon that has been receiving great attention in the last years due to the sheer amount of mechanisms discovered and involved in the process of resistance which hinders the effectiveness of many anti-cancer drugs. Among the mechanisms involved in the multidrug resistance, the participation of ATP-binding cassette (ABC) transporters is the main one. The ABC transporters are a group of plasma membrane and intracellular organelle proteins involved in the process of externalization of substrates from cells, which are expressed in cancer. They are involved in the clearance of intracellular metabolites as ions, hormones, lipids and other small molecules from the cell, affecting directly and indirectly drug absorption, distribution, metabolism and excretion. Other mechanisms responsible for resistance are the signaling pathways and the anti- and pro-apoptotic proteins involved in cell death by apoptosis. In this study we evaluated the influence of three nanosystem (Graphene Quantum Dots (GQDs), mesoporous silica (MSN) and poly-lactic nanoparticles (PLA)) in the main mechanism related to the cancer multidrug resistance such as the Multidrug Resistance Protein-1 and P-glycoprotein. We also evaluated this influence in a group of proteins involved in the apoptosis-related resistance including cIAP-1, XIAP, Bcl-2, BAK and Survivin proteins. Last, colonogenic and MTT (3-(4,5-dimethylthiazol-2-yl)- 2,5-diphenyltetrazolium bromide) assays have also been performed. The results showed, regardless of the concentration used, GQDs, MSN and PLA were not cytotoxic to MDA-MB-231 cells and showed no impairment in the colony formation capacity. In addition, it has been observed that P-gp membrane expression was not significantly altered by any of the three nanomaterials. The results suggest that GQDs nanoparticles would be suitable for the delivery of other multidrug resistance protein 1 (MRP1) substrate drugs that bind to the transporter at the same binding pocket, while MSN can strongly inhibit doxorubicin efflux by MRP1. On the other hand, PLA showed moderate inhibition of doxorubicin efflux by MRP1 suggesting that this nanomaterial can also be useful to treat MDR (Multidrug resistance) due to MRP1 overexpression.
Collapse
Affiliation(s)
- Francisco V.C. Mello
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rua Helio de Almeida 75, Ilha do Fundão, CEP 21941-614 Rio de Janeiro, Brazil;
| | - Gabriela N. de Moraes
- Laboratory of Cellular and Molecular Hemato-Oncology, Program of Molecular Hemato-Oncology, Brazilian National Cancer Institute (INCA), CEP 20230130 Rio de Janeiro, Brazil; (G.N.d.M.); (R.C.M.)
| | - Raquel C. Maia
- Laboratory of Cellular and Molecular Hemato-Oncology, Program of Molecular Hemato-Oncology, Brazilian National Cancer Institute (INCA), CEP 20230130 Rio de Janeiro, Brazil; (G.N.d.M.); (R.C.M.)
| | - Jennifer Kyeremateng
- Department of Chemistry & Biochemistry, College of Natural Sciences, South Dakota State University, Brookings, SD 57007, USA; (J.K.); (S.H.I.)
| | - Surtaj Hussain Iram
- Department of Chemistry & Biochemistry, College of Natural Sciences, South Dakota State University, Brookings, SD 57007, USA; (J.K.); (S.H.I.)
| | - Ralph Santos-Oliveira
- Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rua Helio de Almeida 75, Ilha do Fundão, CEP 21941-614 Rio de Janeiro, Brazil;
- Laboratory of Radiopharmacy and Nanoradiopharmaceuticals, Zona Oeste State University, Campo Grande, CEP 23070200 Rio de Janeiro, Brazil
| |
Collapse
|
85
|
Coleman MF, Cozzo AJ, Pfeil AJ, Etigunta SK, Hursting SD. Cell Intrinsic and Systemic Metabolism in Tumor Immunity and Immunotherapy. Cancers (Basel) 2020; 12:cancers12040852. [PMID: 32244756 PMCID: PMC7225951 DOI: 10.3390/cancers12040852] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy has shown extraordinary promise at treating cancers otherwise resistant to treatment. However, for ICI therapy to be effective, it must overcome the metabolic limitations of the tumor microenvironment. Tumor metabolism has long been understood to be highly dysregulated, with potent immunosuppressive effects. Moreover, T cell activation and longevity within the tumor microenvironment are intimately tied to T cell metabolism and are required for the long-term efficacy of ICI therapy. We discuss in this review the intersection of metabolic competition in the tumor microenvironment, T cell activation and metabolism, the roles of tumor cell metabolism in immune evasion, and the impact of host metabolism in determining immune surveillance and ICI therapy outcomes. We also discussed the effects of obesity and calorie restriction—two important systemic metabolic perturbations that impact intrinsic metabolic pathways in T cells as well as cancer cells.
Collapse
Affiliation(s)
- Michael F. Coleman
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27516, USA; (M.F.C.); (A.J.C.); (A.J.P.); (S.K.E.)
| | - Alyssa J. Cozzo
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27516, USA; (M.F.C.); (A.J.C.); (A.J.P.); (S.K.E.)
- Department of Medicine, Duke University, Durham, NC 27705, USA
| | - Alexander J. Pfeil
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27516, USA; (M.F.C.); (A.J.C.); (A.J.P.); (S.K.E.)
| | - Suhas K. Etigunta
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27516, USA; (M.F.C.); (A.J.C.); (A.J.P.); (S.K.E.)
| | - Stephen D. Hursting
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27516, USA; (M.F.C.); (A.J.C.); (A.J.P.); (S.K.E.)
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27516, USA
- Correspondence:
| |
Collapse
|
86
|
Liu H, Kitano S, Irie S, Levato R, Matsusaki M. Collagen Microfibers Induce Blood Capillary Orientation and Open Vascular Lumen. ACTA ACUST UNITED AC 2020; 4:e2000038. [DOI: 10.1002/adbi.202000038] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 02/15/2020] [Accepted: 02/21/2020] [Indexed: 01/09/2023]
Affiliation(s)
- Hao Liu
- Department of Applied Chemistry Graduate School of Engineering Osaka University 2‐1 Yamadaoka Suita Osaka 565‐0871 Japan
| | - Shiro Kitano
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry Graduate School of Engineering Osaka University 2‐1 Yamadaoka Suita Osaka 565‐0871 Japan
| | - Shinji Irie
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry Graduate School of Engineering Osaka University 2‐1 Yamadaoka Suita Osaka 565‐0871 Japan
| | - Riccardo Levato
- Regenerative Medicine Center Utrecht and Department of Orthopaedics University Medical Center Utrecht Heidelberglaan 100 Utrecht 3584 CX The Netherlands
| | - Michiya Matsusaki
- Department of Applied Chemistry Graduate School of Engineering Osaka University 2‐1 Yamadaoka Suita Osaka 565‐0871 Japan
| |
Collapse
|
87
|
Peptide-enhanced tumor accumulation of upconversion nanoparticles for sensitive upconversion luminescence/magnetic resonance dual-mode bioimaging of colorectal tumors. Acta Biomater 2020; 104:167-175. [PMID: 31923719 DOI: 10.1016/j.actbio.2020.01.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/31/2019] [Accepted: 01/02/2020] [Indexed: 12/13/2022]
Abstract
Currently, it is still a great challenge to develop tumor targeting nanoparticles with high sensitivity and high resolution for improving the non-invasive detection ability of colorectal cancer (CRC) at an early stage. In this study, NaErF4:Yb@NaGdF4:Yb core@shell upconversion nanoparticles (UCNPs) were prepared with high upconversion luminescence (UCL) emission in red light region through adjusting the doping ratios of Er and Yb elements in the core. For biomedical applications, the carboxyl-terminated silica shell was introduced to transfer the as-prepared UCNPs from the organic phase to the aqueous phase, and allowed conjugation with peptide ligands derived from the l-SP5 peptide (i.e., l-SP5-H and l-SP5-C), respectively. Due to the tumor-targeting affinity of the PSP motif in the peptide ligands, the as-prepared peptide functionalized UCNPs (UCNP@SiO2-l-SP5-H and UCNP@SiO2-l-SP5-C) can be used as an active tumor targeting contrast agents for UCL/T1-weighted magnetic resonance (MR) dual-mode imaging. Both the in vitro and in vivo experimental results demonstrated that UCNP@SiO2-l-SP5-C has relatively high affinity for the HCT116 CRC subtype. Moreover, UCNP@SiO2-l-SP5-C can visualize ultra-small subcutaneous xenografted HCT116 tumors (c.a. 13 mm3 in volume) by in vivo UCL imaging. STATEMENT OF SIGNIFICANCE: 1. High red emission UCNPs were synthesized for tumor-targeting dual-mode bioimaging. 2. With tumor-binding affinity peptide, UCNP@SiO2-l-SP5-C shows high HCT116 tumor targeting ability. 3. UCNP@SiO2-l-SP5-C successfully achieves sensitive detection of ultrasmall HCT116 tumors.
Collapse
|
88
|
Phillips AM, Pombeiro AJ. Transition Metal-Based Prodrugs for Anticancer Drug Delivery. Curr Med Chem 2020; 26:7476-7519. [DOI: 10.2174/0929867326666181203141122] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 11/08/2018] [Accepted: 11/12/2018] [Indexed: 12/14/2022]
Abstract
:
Transition metal complexes, of which the platinum(II) complex cisplatin is an example,
have been used in medicine to treat cancer for more than 40 years. Although many successes have
been achieved, there are problems associated with the use of these drugs, such as side effects and
drug resistance. Converting them into prodrugs, to make them more inert, so that they can travel to
the tumour site unchanged and release the drug in its active form only there, is a strategy which is
the subject of much research nowadays. The new prodrugs may be activated and release the cytotoxic
agent by differences in oxygen concentration or in pH, by the action of overexpressed enzymes,
by differences in metabolic rates, etc., which characteristically distinguish cancer cells from
normal ones, or even by the input of radiation, which can be visible light. Converting a metal complex
into a prodrug may also be used to improve its pharmacological properties. In some cases, the
metal complex is a carrier which transports the active drug as a ligand. Some platinum prodrugs
have reached clinical trials. So far platinum, ruthenium and cobalt have been the most studied metals.
This review presents the recent developments in this area, including the types of complexes
used, the mechanisms of drug action and in some cases the techniques applied to monitor drug delivery
to cells.
Collapse
Affiliation(s)
- Ana M.F. Phillips
- Centro de Química Estrutural, Complexo I, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| | - Armando J.L. Pombeiro
- Centro de Química Estrutural, Complexo I, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|
89
|
Zhang M, Ye JJ, Xia Y, Wang ZY, Li CX, Wang XS, Yu W, Song W, Feng J, Zhang XZ. Platelet-Mimicking Biotaxis Targeting Vasculature-Disrupted Tumors for Cascade Amplification of Hypoxia-Sensitive Therapy. ACS NANO 2019; 13:14230-14240. [PMID: 31714733 DOI: 10.1021/acsnano.9b07330] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Tumorous vasculature plays key roles in sustaining tumor growth. Vascular disruption is accompanied by internal coagulation along with platelet recruitment and the resulting suppression of oxygen supply. We intend to artificially create this physiological process to establish the mutual feedback between vascular disruption and platelet-mimicking biotaxis for the cascade amplification of hypoxia-dependent therapy. To prove this concept, mesoporous silica nanoparticles are co-loaded with a hypoxia-activated prodrug (HAP) and a vessel-disruptive agent and then coated with platelet membranes. Upon entering into tumors, our nanotherapeutic can disrupt local vasculature for tumor inhibition. This platelet membrane-coated nanoplatform shares the hemorrhage-tropic function with parental platelets and can be persistently recruited by the vasculature-disrupted tumors. In this way, the intratumoral vascular disruption and tumor targeting are biologically interdependent and mutually reinforced. Relying on this mutual feedback, tumorous hypoxia was largely promoted by more than 20-fold, accounting for the effective recovery of the HAP's cytotoxicity. Consequently, our bioinspired nanodesign has demonstrated highly specific and effective antitumor potency via the biologically driven cooperation among intratumoral vascular disruption, platelet-mimicking biotaxis, cascade hypoxia amplification, and hypoxia-sensitive chemotherapy. This study offers a paradigm of correlating the therapeutic design with the physiologically occurring events to achieve better therapy performance.
Collapse
Affiliation(s)
- Mingkang Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry , Wuhan University , Wuhan 430072 , People's Republic of China
| | - Jing-Jie Ye
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry , Wuhan University , Wuhan 430072 , People's Republic of China
| | - Yu Xia
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry , Wuhan University , Wuhan 430072 , People's Republic of China
| | - Zi-Yang Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry , Wuhan University , Wuhan 430072 , People's Republic of China
| | - Chu-Xin Li
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry , Wuhan University , Wuhan 430072 , People's Republic of China
| | - Xiao-Shuang Wang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry , Wuhan University , Wuhan 430072 , People's Republic of China
| | - Wuyang Yu
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry , Wuhan University , Wuhan 430072 , People's Republic of China
| | - Wen Song
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry , Wuhan University , Wuhan 430072 , People's Republic of China
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry , Wuhan University , Wuhan 430072 , People's Republic of China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry , Wuhan University , Wuhan 430072 , People's Republic of China
| |
Collapse
|
90
|
Covarrubias-Zambrano O, Yu J, Bossmann SH. Nano-Inspired Technologies for Peptide Delivery. Curr Protein Pept Sci 2019; 21:379-400. [PMID: 31793426 DOI: 10.2174/1389203720666191202112429] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/26/2019] [Accepted: 10/02/2019] [Indexed: 12/15/2022]
Abstract
Nano-inspired technologies offer unique opportunities to treat numerous diseases by using therapeutic peptides. Therapeutic peptides have attractive pharmacological profiles and can be manufactured at relatively low costs. The major advantages of using a nanodelivery approach comprises significantly lower required dosages compared to systemic delivery, and thus reduced toxicity and immunogenicity. The combination of therapeutic peptides with delivery peptides and nanoparticles or small molecule drugs offers systemic treatment approaches, instead of aiming for single biological targets or pathways. This review article discusses exemplary state-of-the-art nanosized delivery systems for therapeutic peptides and antibodies, as well as their biochemical and biophysical foundations and emphasizes still remaining challenges. The competition between using different nanoplatforms, such as liposome-, hydrogel-, polymer-, silica nanosphere-, or nanosponge-based delivery systems is still "on" and no clear frontrunner has emerged to date.
Collapse
Affiliation(s)
| | - Jing Yu
- Department of Chemistry, Kansas State University, 419 CBC Building, Manhattan, KS 66506-0401, United States.,Johns Hopkins University, Department of Radiology, Baltimore, MD, United States
| | - Stefan H Bossmann
- Department of Chemistry, Kansas State University, 419 CBC Building, Manhattan, KS 66506-0401, United States
| |
Collapse
|
91
|
Zhang Y, Ho S, Li B, Nie G, Li S. Modulating the tumor microenvironment with new therapeutic nanoparticles: A promising paradigm for tumor treatment. Med Res Rev 2019; 40:1084-1102. [DOI: 10.1002/med.21644] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/01/2019] [Accepted: 10/27/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Yinlong Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of Sciences Beijing China
| | - Shih‐Hsin Ho
- State Key Laboratory of Urban Water Resource and Environment, School of EnvironmentHarbin Institute of Technology Harbin China
| | - Bozhao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of Sciences Beijing China
- Australian Institute for Bioengineering and NanotechnologyThe University of Queensland Brisbane Australia
| | - Suping Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology Beijing China
- Center of Materials Science and Optoelectronics EngineeringUniversity of Chinese Academy of Sciences Beijing China
| |
Collapse
|
92
|
Tee JK, Yip LX, Tan ES, Santitewagun S, Prasath A, Ke PC, Ho HK, Leong DT. Nanoparticles' interactions with vasculature in diseases. Chem Soc Rev 2019; 48:5381-5407. [PMID: 31495856 DOI: 10.1039/c9cs00309f] [Citation(s) in RCA: 195] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The ever-growing use of inorganic nanoparticles (NPs) in biomedicine provides an exciting approach to develop novel imaging and drug delivery systems, owing to the ease with which these NPs can be functionalized to cater to various applications. In cancer therapeutics, nanomedicine generally relies on the enhanced permeability and retention (EPR) effect observed in tumour vasculature to deliver anti-cancer drugs across the endothelium. However, such a phenomenon is dependent on the tumour microenvironment and is not consistently observed in all tumour types, thereby limiting drug transport to the tumour site. On the other hand, there is a rise in utilizing inorganic NPs to intentionally induce endothelial leakiness, creating a window of opportunity to control drug delivery across the endothelium. While this active targeting approach creates a similar phenomenon compared to the EPR effect arising from tumour tissues, its drug delivery applications extend beyond cancer therapeutics and into other vascular-related diseases. In this review, we summarize the current findings of the EPR effect and assess its limitations in the context of anti-cancer drug delivery systems. While the EPR effect offers a possible route for drug passage, we further explore alternative uses of NPs to create controllable endothelial leakiness within short exposures, a phenomenon we coined as nanomaterial-induced endothelial leakiness (NanoEL). Furthermore, we discuss the main mechanistic features of the NanoEL effect that make it unique from conventionally established endothelial leakiness in homeostatic and pathologic conditions, as well as examine its potential applicability in vascular-related diseases, particularly cancer. Therefore, this new paradigm changes the way inorganic NPs are currently being used for biomedical applications.
Collapse
Affiliation(s)
- Jie Kai Tee
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.
| | | | | | | | | | | | | | | |
Collapse
|
93
|
Mohan V, Das A, Sagi I. Emerging roles of ECM remodeling processes in cancer. Semin Cancer Biol 2019; 62:192-200. [PMID: 31518697 DOI: 10.1016/j.semcancer.2019.09.004] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/01/2019] [Accepted: 09/07/2019] [Indexed: 02/07/2023]
Abstract
Extracellular matrix (ECM) plays a central and dynamic role in the creation of tumor microenvironment. Herein we discuss the emerging biophysical and biochemical aspects of ECM buildup and proteolysis in cancer niche formation. Dysregulated ECM remodeling by cancer cells facilitate irreversible proteolysis and crosslinking, which in turn influence cell signaling, micro environmental cues, angiogenesis and tissue biomechanics. Further, we introduce the emerging roles of cancer microbiome in aberrant tumor ECM remodeling and membrane bound nano-sized vesicles called exosomes in creation of distant pre-metastatic niches. A detailed molecular and biophysical understanding of the ECM morphologies and its components such as key enzymes, structural and signaling molecules are critical in identifying the next generation of therapeutic and diagnostic targets in cancer.
Collapse
Affiliation(s)
- Vishnu Mohan
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Alakesh Das
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
94
|
Ercetin E, Richtmann S, Delgado BM, Gomez-Mariano G, Wrenger S, Korenbaum E, Liu B, DeLuca D, Kühnel MP, Jonigk D, Yuskaeva K, Warth A, Muley T, Winter H, Meister M, Welte T, Janciauskiene S, Schneider MA. Clinical Significance of SERPINA1 Gene and Its Encoded Alpha1-antitrypsin Protein in NSCLC. Cancers (Basel) 2019; 11:cancers11091306. [PMID: 31487965 PMCID: PMC6770941 DOI: 10.3390/cancers11091306] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 08/29/2019] [Accepted: 09/02/2019] [Indexed: 12/20/2022] Open
Abstract
High expression of SERPINA1 gene encoding acute phase protein, alpha1-antitrypsin (AAT), is associated with various tumors. We sought to examine the significance of SERPINA1 and AAT protein in non-small-cell lung cancer (NSCLC) patients and NSCLC cell lines. Tumor and adjacent non-tumor lung tissues and serum samples from 351 NSCLC patients were analyzed for SERPINA1 expression and AAT protein levels. We also studied the impact of SERPINA1 expression and AAT protein on H1975 and H661 cell behavior, in vitro. Lower SERPINA1 expression in tumor but higher in adjacent non-tumor lung tissues (n = 351, p = 0.016) as well as higher serum levels of AAT protein (n = 170, p = 0.033) were associated with worse survival rates. Specifically, in NSCLC stage III patients, higher blood AAT levels (>2.66 mg/mL) correlated with a poor survival (p = 0.002). Intriguingly, levels of serum AAT do not correlate with levels of C-reactive protein, neutrophils-to-leukocyte ratio, and do not correlate with SERPINA1 expression or AAT staining in the tumor tissue. Additional experiments in vitro revealed that external AAT and/or overexpressed SERPINA1 gene significantly improve cancer cell migration, colony formation and resistance to apoptosis. SERPINA1 gene and AAT protein play an active role in the pathogenesis of lung cancer and not just reflect inflammatory reaction related to cancer development.
Collapse
Affiliation(s)
- Evrim Ercetin
- Department of Respiratory Medicine, Hannover Medical School, 30625 Hannover, Germany.
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), 30625 Hannover, Germany.
| | - Sarah Richtmann
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany.
- Translational Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany.
| | - Beatriz Martinez Delgado
- Department of Molecular Genetics. Institute of Health Carlos III. Center for Biomedical Research in the Network of Rare Diseases (CIBERER), 28220 Majadahonda (Madrid), Spain.
| | - Gema Gomez-Mariano
- Department of Molecular Genetics. Institute of Health Carlos III. Center for Biomedical Research in the Network of Rare Diseases (CIBERER), 28220 Majadahonda (Madrid), Spain.
| | - Sabine Wrenger
- Department of Respiratory Medicine, Hannover Medical School, 30625 Hannover, Germany.
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), 30625 Hannover, Germany.
| | - Elena Korenbaum
- Institute of Biophysical Chemistry and Anatomy, Hannover Medical School, 30625 Hannover, Germany.
| | - Bin Liu
- Department of Respiratory Medicine, Hannover Medical School, 30625 Hannover, Germany.
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), 30625 Hannover, Germany.
| | - David DeLuca
- Department of Respiratory Medicine, Hannover Medical School, 30625 Hannover, Germany.
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), 30625 Hannover, Germany.
| | - Mark P Kühnel
- Institute of Pathology, Hannover Medical School, 30625 Hannover, Germany.
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, 30625 Hannover, Germany.
| | - Kadriya Yuskaeva
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany.
| | - Arne Warth
- Institute of Pathology, Heidelberg University Hospital, D-69120 Heidelberg, Germany.
| | - Thomas Muley
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany.
- Translational Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany.
| | - Hauke Winter
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany.
- Department of Surgery, Thoraxklinik at Heidelberg University Hospital, D-69126 Heidelberg, Germany.
| | - Michael Meister
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany.
- Translational Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany.
| | - Tobias Welte
- Department of Respiratory Medicine, Hannover Medical School, 30625 Hannover, Germany.
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), 30625 Hannover, Germany.
| | - Sabina Janciauskiene
- Department of Respiratory Medicine, Hannover Medical School, 30625 Hannover, Germany.
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), 30625 Hannover, Germany.
| | - Marc A Schneider
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, 69126 Heidelberg, Germany.
- Translational Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), 69120 Heidelberg, Germany.
| |
Collapse
|
95
|
Pieterse Z, Sinha D, Kaur P. Pericytes in Metastasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1147:125-135. [PMID: 31147875 DOI: 10.1007/978-3-030-16908-4_5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Pericytes have long been known to contribute indirectly to tumour growth by regulating angiogenesis. Thus, remodelling tumour blood vessels to maintain blood supply is critical for continued tumour growth. A role for pericytes in restricting leakage of tumour cells through blood vessels has also become evident given that adequate pericyte coverage of these blood vessels is critical for maintaining vascular permeability. Interestingly, the relocation of pericytes from blood vessels to the tumour microenvironment results in the emergence of different properties in these cells that actively promote tumour growth and metastasis-functions not associated with their well-studied role in vascular stability and permeability. These form the focus of this review.
Collapse
Affiliation(s)
- Zalitha Pieterse
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Devbarna Sinha
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Pritinder Kaur
- Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia.
| |
Collapse
|
96
|
Tannenbaum CS, Rayman PA, Pavicic PG, Kim JS, Wei W, Polefko A, Wallace W, Rini BI, Morris-Stiff G, Allende DS, Hamilton T, Finke JH, Diaz-Montero CM. Mediators of Inflammation-Driven Expansion, Trafficking, and Function of Tumor-Infiltrating MDSCs. Cancer Immunol Res 2019; 7:1687-1699. [PMID: 31439615 DOI: 10.1158/2326-6066.cir-18-0578] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 03/15/2019] [Accepted: 08/14/2019] [Indexed: 12/15/2022]
Abstract
Myeloid-derived suppressor cells (MDSC) are induced by and accumulate within many histologically distinct solid tumors, where they promote disease by secreting angiogenic and immunosuppressive molecules. Although IL1β can drive the generation, accumulation, and functional capacity of MDSCs, the specific IL1β-induced inflammatory mediators contributing to these activities remain incompletely defined. Here, we identified IL1β-induced molecules that expand, mobilize, and modulate the accumulation and angiogenic and immunosuppressive potencies of polymorphonuclear (PMN)-MDSCs. Unlike parental CT26 tumors, which recruited primarily monocytic (M)-MDSCs by constitutively expressing GM-CSF- and CCR2-directed chemokines, IL1β-transfected CT26 produced higher G-CSF, multiple CXC chemokines, and vascular adhesion molecules required for mediating infiltration of PMN-MDSCs with increased angiogenic and immunosuppressive properties. Conversely, CT26 tumors transfected with IL1β-inducible molecules could mobilize PMN-MDSCs, but because they lacked the ability to upregulate IL1β-inducible CXCR2-directed chemokines or vascular adhesion molecules, additional PMN-MDSCs could not infiltrate tumors. IL1β-expressing CT26 increased angiogenic and immunosuppressive factors of tumor-infiltrating MDSCs, as did CT26 tumors individually transfected with G-CSF, Bv8, CXCL1, or CXCL5, demonstrating that mediators downstream of IL1β could also modulate MDSC functional activity. Translational relevance was indicated by the finding that the same growth factors, cytokines, chemokines, and adhesion molecules responsible for the mobilization and recruitment of PMN-MDSCs into inflammatory CT26 murine tumors were also coordinately upregulated with increasing IL1β expression in human renal cell carcinoma tumors. These studies demonstrated that IL1β stimulated the components of a multifaceted inflammatory program that produces, mobilizes, chemoattracts, activates, and mediates the infiltration of PMN-MDSCs into inflammatory tumors to promote tumor progression.
Collapse
Affiliation(s)
- Charles S Tannenbaum
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Patricia A Rayman
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Paul G Pavicic
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Jin Sub Kim
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Wei Wei
- Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio
| | - Alexandra Polefko
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Wesley Wallace
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Brian I Rini
- Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio
| | | | | | - Thomas Hamilton
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - James H Finke
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - C Marcela Diaz-Montero
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
| |
Collapse
|
97
|
Atukorale PU, Raghunathan SP, Raguveer V, Moon TJ, Zheng C, Bielecki PA, Wiese ML, Goldberg AL, Covarrubias G, Hoimes CJ, Karathanasis E. Nanoparticle Encapsulation of Synergistic Immune Agonists Enables Systemic Codelivery to Tumor Sites and IFNβ-Driven Antitumor Immunity. Cancer Res 2019; 79:5394-5406. [PMID: 31431457 DOI: 10.1158/0008-5472.can-19-0381] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/14/2019] [Accepted: 08/14/2019] [Indexed: 12/17/2022]
Abstract
Effective cancer immunotherapy depends on the robust activation of tumor-specific antigen-presenting cells (APC). Immune agonists encapsulated within nanoparticles (NP) can be delivered to tumor sites to generate powerful antitumor immune responses with minimal off-target dissemination. Systemic delivery enables widespread access to the microvasculature and draining to the APC-rich perivasculature. We developed an immuno-nanoparticle (immuno-NP) coloaded with cyclic diguanylate monophosphate, an agonist of the stimulator of interferon genes pathway, and monophosphoryl lipid A, and a Toll-like receptor 4 agonist, which synergize to produce high levels of type I IFNβ. Using a murine model of metastatic triple-negative breast cancer, systemic delivery of these immuno-NPs resulted in significant therapeutic outcomes due to extensive upregulation of APCs and natural killer cells in the blood and tumor compared with control treatments. These results indicate that NPs can facilitate systemic delivery of multiple immune-potentiating cargoes for effective APC-driven local and systemic antitumor immunity. SIGNIFICANCE: Systemic administration of an immuno-nanoparticle in a murine breast tumor model drives a robust tumor site-specific APC response by delivering two synergistic immune-potentiating molecules, highlighting the potential of nanoparticles for immunotherapy.
Collapse
Affiliation(s)
- Prabhani U Atukorale
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Shruti P Raghunathan
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Vanitha Raguveer
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Taylor J Moon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Carolyn Zheng
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Peter A Bielecki
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Michelle L Wiese
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Amy L Goldberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Gil Covarrubias
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Christopher J Hoimes
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Efstathios Karathanasis
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio. .,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio.,Department of Radiology, Case Western Reserve University, Cleveland, Ohio.,Case Center for Imaging Research, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
98
|
Zanotelli MR, Reinhart-King CA. Mechanical Forces in Tumor Angiogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1092:91-112. [PMID: 30368750 PMCID: PMC6986816 DOI: 10.1007/978-3-319-95294-9_6] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A defining hallmark of cancer and cancer development is upregulated angiogenesis. The vasculature formed in tumors is structurally abnormal, not organized in the conventional hierarchical arrangement, and more permeable than normal vasculature. These features contribute to leaky, tortuous, and dilated blood vessels, which act to create heterogeneous blood flow, compression of vessels, and elevated interstitial fluid pressure. As such, abnormalities in the tumor vasculature not only affect the delivery of nutrients and oxygen to the tumor, but also contribute to creating an abnormal tumor microenvironment that further promotes tumorigenesis. The role of chemical signaling events in mediating tumor angiogenesis has been well researched; however, the relative contribution of physical cues and mechanical regulation of tumor angiogenesis is less understood. Growing research indicates that the physical microenvironment plays a significant role in tumor progression and promoting abnormal tumor vasculature. Here, we review how mechanical cues found in the tumor microenvironment promote aberrant tumor angiogenesis. Specifically, we discuss the influence of matrix stiffness and mechanical stresses in tumor tissue on tumor vasculature, as well as the mechanosensory pathways utilized by endothelial cells to respond to the physical cues found in the tumor microenvironment. We also discuss the impact of the resulting aberrant tumor vasculature on tumor progression and therapeutic treatment.
Collapse
Affiliation(s)
- Matthew R Zanotelli
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Cynthia A Reinhart-King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
99
|
iRGD: A Promising Peptide for Cancer Imaging and a Potential Therapeutic Agent for Various Cancers. JOURNAL OF ONCOLOGY 2019; 2019:9367845. [PMID: 31346334 PMCID: PMC6617877 DOI: 10.1155/2019/9367845] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 06/04/2019] [Accepted: 06/11/2019] [Indexed: 12/11/2022]
Abstract
Poor penetration into the tumor parenchyma and the reduced therapeutic efficacy of anticancer drugs and other medications are the major problems in tumor treatment. A new tumor-homing and penetrating peptide, iRGD (CRGDK/RGPD/EC), can be effectively used to combine and deliver imaging agents or anticancer drugs into tumors. The different “vascular zip codes” expressed in different tissues can serve as targets for docking-based (synaptic) delivery of diagnostic and therapeutic molecules. αv-Integrins are abundantly expressed in the tumor vasculature, where they are recognized by peptides containing the RGD integrin recognition motif. The iRGD peptide follows a multistep tumor-targeting process: First, it is proteolytically cleaved to generate the CRGDK fragment by binding to the surface of cells expressing αv integrins (αvβ3 and αvβ5). Then, the fragment binds to neuropilin-1 and penetrates the tumor parenchyma more deeply. Compared with conventional RGD peptides, the affinity of iRGD for αv integrins is in the mid to low nanomolar range, and the CRGDK fragment has a stronger affinity for neuropilin-1 than that for αv integrins because of the C-terminal exposure of a conditional C-end Rule (CendR) motif (R/KXXR/K), whose receptor proved to be neuropilin-1. Consequently, these advantages facilitate the transfer of CRGDK fragments from integrins to neuropilin-1 and consequently deeper penetration into the tumor. Due to its specific binding and strong affinity, the iRGD peptide can deliver imaging agents and anticancer drugs into tumors effectively and deeply, which is useful in detecting the tumor, blocking tumor growth, and inhibiting tumor metastasis. This review aims to focus on the role of iRGD in the imaging and treatment of various cancers.
Collapse
|
100
|
Jiao D, Li Y, Yang F, Han D, Wu J, Shi S, Tian F, Guo Z, Xi W, Li G, Zhao A, Yang AG, Qin W, Wang H, Wen W. Expression of Prostate-Specific Membrane Antigen in Tumor-Associated Vasculature Predicts Poor Prognosis in Hepatocellular Carcinoma. Clin Transl Gastroenterol 2019; 10:1-7. [PMID: 31116141 PMCID: PMC6602770 DOI: 10.14309/ctg.0000000000000041] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 03/12/2019] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Prostate-specific membrane antigen (PSMA) was originally found to be specifically expressed in normal prostate, and its expression was upregulated in almost all stages of prostate cancer. In recent years, PSMA was also found to be expressed in tumor-associated vasculature in many nonprostatic solid tumors. However, the expression pattern of PSMA in hepatocellular carcinoma (HCC) is not well studied. METHODS In this study, we examined PSMA expression in 103 HCC tissues using immunohistochemical staining and analyzed the association between PSMA expression and other clinicopathological features and prognosis. RESULTS Among the 103 cases, 27 cases (26%) showed PSMA expression in more than 50% of tumor-associated vasculature, 49 cases (48%) showed PSMA expression in less than 50% of vasculature, and 27 cases (26%) did not have detectable PSMA expression. Vascular PSMA expression was associated with several clinicopathological features, such as tumor stage, tumor differentiation, lymph node metastasis, and Ki-67 index. Furthermore, high vascular PSMA expression was also associated with poor prognosis in patients with HCC. Univariate and multivariate analyses showed that high vascular PSMA expression can be used as an independent prognostic marker for HCC. DISCUSSION Our study provides the evidence that PSMA is specifically expressed in tumor-associated vasculature of HCC, and vascular PSMA expression may be used as a novel prognostic marker and a vascular therapeutic target for HCC.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Antigens, Surface/analysis
- Antigens, Surface/metabolism
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/metabolism
- Carcinoma, Hepatocellular/blood supply
- Carcinoma, Hepatocellular/mortality
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/therapy
- Female
- Follow-Up Studies
- Glutamate Carboxypeptidase II/analysis
- Glutamate Carboxypeptidase II/antagonists & inhibitors
- Glutamate Carboxypeptidase II/metabolism
- Hepatectomy
- Humans
- Immunohistochemistry
- Kaplan-Meier Estimate
- Liver/blood supply
- Liver/pathology
- Liver/surgery
- Liver Neoplasms/blood supply
- Liver Neoplasms/mortality
- Liver Neoplasms/pathology
- Liver Neoplasms/therapy
- Lymphatic Metastasis/pathology
- Male
- Middle Aged
- Neoplasm Staging
- Neovascularization, Pathologic/mortality
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/therapy
- Prognosis
- Time Factors
- Young Adult
Collapse
Affiliation(s)
- Dian Jiao
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Yu Li
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fa Yang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Donghui Han
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jieheng Wu
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Shengjia Shi
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Feng Tian
- Department of Urology, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhangyan Guo
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Wenjin Xi
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, China
| | | | | | - An-Gang Yang
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, China
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - He Wang
- Department of Urology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Weihong Wen
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|