1
|
Tenchov R, Hughes KJ, Ganesan M, Iyer KA, Ralhan K, Lotti Diaz LM, Bird RE, Ivanov JM, Zhou QA. Transforming Medicine: Cutting-Edge Applications of Nanoscale Materials in Drug Delivery. ACS NANO 2025; 19:4011-4038. [PMID: 39823199 PMCID: PMC11803921 DOI: 10.1021/acsnano.4c09566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/19/2025]
Abstract
Since their inception in the early 1960s, the development and use of nanoscale materials have progressed tremendously, and their roles in diverse fields ranging from human health to energy and electronics are undeniable. The application of nanotechnology inventions has revolutionized many aspects of everyday life including various medical applications and specifically drug delivery systems, maximizing the therapeutic efficacy of the contained drugs by means of bioavailability enhancement or minimization of adverse effects. In this review, we utilize the CAS Content Collection, a vast repository of scientific information extracted from journal and patent publications, to analyze trends in nanoscience research relevant to drug delivery in an effort to provide a comprehensive and detailed picture of the use of nanotechnology in this field. We examine the publication landscape in the area to provide insights into current knowledge advances and developments. We review the major classes of nanosized drug delivery systems, their delivery routes, and targeted diseases. We outline the most discussed concepts and assess the advantages of various nanocarriers. The objective of this review is to provide a broad overview of the evolving landscape of current knowledge regarding nanosized drug delivery systems, to outline challenges, and to evaluate growth opportunities. The merit of the review stems from the extensive, wide-ranging coverage of the most up-to-date scientific information, allowing unmatched breadth of landscape analysis and in-depth insights.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Kevin J. Hughes
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | - Kavita A. Iyer
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | - Leilani M. Lotti Diaz
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Robert E. Bird
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Julian M. Ivanov
- CAS,
a division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
2
|
Wang M, Yu F, Zhang Y. Present and future of cancer nano-immunotherapy: opportunities, obstacles and challenges. Mol Cancer 2025; 24:26. [PMID: 39827147 PMCID: PMC11748575 DOI: 10.1186/s12943-024-02214-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/25/2024] [Indexed: 01/22/2025] Open
Abstract
Clinically, multimodal therapies are adopted worldwide for the management of cancer, which continues to be a leading cause of death. In recent years, immunotherapy has firmly established itself as a new paradigm in cancer care that activates the body's immune defense to cope with cancer. Immunotherapy has resulted in significant breakthroughs in the treatment of stubborn tumors, dramatically improving the clinical outcome of cancer patients. Multiple forms of cancer immunotherapy, including immune checkpoint inhibitors (ICIs), adoptive cell therapy and cancer vaccines, have become widely available. However, the effectiveness of these immunotherapies is not much satisfying. Many cancer patients do not respond to immunotherapy, and disease recurrence appears to be unavoidable because of the rapidly evolving resistance. Moreover, immunotherapies can give rise to severe off-target immune-related adverse events. Strategies to remove these hindrances mainly focus on the development of combinatorial therapies or the exploitation of novel immunotherapeutic mediations. Nanomaterials carrying anticancer agents to the target site are considered as practical approaches for cancer treatment. Nanomedicine combined with immunotherapies offers the possibility to potentiate systemic antitumor immunity and to facilitate selective cytotoxicity against cancer cells in an effective and safe manner. A myriad of nano-enabled cancer immunotherapies are currently under clinical investigation. Owing to gaps between preclinical and clinical studies, nano-immunotherapy faces multiple challenges, including the biosafety of nanomaterials and clinical trial design. In this review, we provide an overview of cancer immunotherapy and summarize the evidence indicating how nanomedicine-based approaches increase the efficacy of immunotherapies. We also discuss the key challenges that have emerged in the era of nanotechnology-based cancer immunotherapy. Taken together, combination nano-immunotherapy is drawing increasing attention, and it is anticipated that the combined treatment will achieve the desired success in clinical cancer therapy.
Collapse
Affiliation(s)
- Man Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao, 266021, China.
| | - Fei Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao, 266021, China
| | - Yuan Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao, 266021, China
| |
Collapse
|
3
|
Zhang H, Li S, Ma X. Transforming Healthcare with Nanomedicine: A SWOT Analysis of Drug Delivery Innovation. Drug Des Devel Ther 2024; 18:3499-3521. [PMID: 39132625 PMCID: PMC11314449 DOI: 10.2147/dddt.s470210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 07/11/2024] [Indexed: 08/13/2024] Open
Abstract
Objective Nanomedicine represents a transformative approach in biomedical applications. This study aims to delineate the application of nanomedicine in the biomedical field through the strengths, weaknesses, opportunities, and threats (SWOT) analysis to evaluate its efficacy and potential in clinical applications. Methods The SWOT analysis framework was employed to systematically review and assess the internal strengths and weaknesses, along with external opportunities and threats of nanomedicine. This method provides a balanced consideration of the potential benefits and challenges. Results Findings from the SWOT analysis indicate that nanomedicine presents significant potential in drug delivery, diagnostic imaging, and tissue engineering. Nonetheless, it faces substantial hurdles such as safety issues, environmental concerns, and high development costs. Critical areas for development were identified, particularly concerning its therapeutic potential and the uncertainties surrounding long-term effects. Conclusion Nanomedicine holds substantial promise in driving medical innovation. However, successful clinical translation requires addressing safety, cost, and regulatory challenges. Interdisciplinary collaboration and comprehensive strategic planning are crucial for the safe and effective application of nanomedicine.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Nuclear Medicine, Affiliated Hospital of North Sichuan Medical College North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Suping Li
- Department of Nuclear Medicine, Affiliated Hospital of North Sichuan Medical College North Sichuan Medical College, Nanchong, 637000, People’s Republic of China
| | - Xingming Ma
- School of Health Management, Xihua University, Chengdu, 610039, People’s Republic of China
| |
Collapse
|
4
|
Kocyigit E, Kocaadam-Bozkurt B, Bozkurt O, Ağagündüz D, Capasso R. Plant Toxic Proteins: Their Biological Activities, Mechanism of Action and Removal Strategies. Toxins (Basel) 2023; 15:356. [PMID: 37368657 PMCID: PMC10303728 DOI: 10.3390/toxins15060356] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/11/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
Plants evolve to synthesize various natural metabolites to protect themselves against threats, such as insects, predators, microorganisms, and environmental conditions (such as temperature, pH, humidity, salt, and drought). Plant-derived toxic proteins are often secondary metabolites generated by plants. These proteins, including ribosome-inactivating proteins, lectins, protease inhibitors, α-amylase inhibitors, canatoxin-like proteins and ureases, arcelins, antimicrobial peptides, and pore-forming toxins, are found in different plant parts, such as the roots, tubers, stems, fruits, buds, and foliage. Several investigations have been conducted to explore the potential applications of these plant proteins by analyzing their toxic effects and modes of action. In biomedical applications, such as crop protection, drug development, cancer therapy, and genetic engineering, toxic plant proteins have been utilized as potentially useful instruments due to their biological activities. However, these noxious metabolites can be detrimental to human health and cause problems when consumed in high amounts. This review focuses on different plant toxic proteins, their biological activities, and their mechanisms of action. Furthermore, possible usage and removal strategies for these proteins are discussed.
Collapse
Affiliation(s)
- Emine Kocyigit
- Department of Nutrition and Dietetics, Ordu University, Cumhuriyet Yerleşkesi, 52200 Ordu, Turkey;
| | - Betul Kocaadam-Bozkurt
- Department of Nutrition and Dietetics, Erzurum Technical University, Yakutiye, 25100 Erzurum, Turkey; (B.K.-B.); (O.B.)
| | - Osman Bozkurt
- Department of Nutrition and Dietetics, Erzurum Technical University, Yakutiye, 25100 Erzurum, Turkey; (B.K.-B.); (O.B.)
| | - Duygu Ağagündüz
- Department of Nutrition and Dietetics, Gazi University, Faculty of Health Sciences, Emek, 06490 Ankara, Turkey;
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, 80055 Portici, Italy
| |
Collapse
|
5
|
Koczurkiewicz-Adamczyk P, Grabowska K, Karnas E, Piska K, Wnuk D, Klaś K, Galanty A, Wójcik-Pszczoła K, Michalik M, Pękala E, Fuchs H, Podolak I. Saponin Fraction CIL1 from Lysimachia ciliata L. Enhances the Effect of a Targeted Toxin on Cancer Cells. Pharmaceutics 2023; 15:pharmaceutics15051350. [PMID: 37242592 DOI: 10.3390/pharmaceutics15051350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/22/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Saponins are plant metabolites that possess multidirectional biological activities, among these is antitumor potential. The mechanisms of anticancer activity of saponins are very complex and depend on various factors, including the chemical structure of saponins and the type of cell they target. The ability of saponins to enhance the efficacy of various chemotherapeutics has opened new perspectives for using them in combined anticancer chemotherapy. Co-administration of saponins with targeted toxins makes it possible to reduce the dose of the toxin and thus limit the side effects of overall therapy by mediating endosomal escape. Our study indicates that the saponin fraction CIL1 of Lysimachia ciliata L. can improve the efficacy of the EGFR-targeted toxin dianthin (DE). We investigated the effect of cotreatment with CIL1 + DE on cell viability in a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, on proliferation in a crystal violet assay (CV) and on pro-apoptotic activity using Annexin V/7 Actinomycin D (7-AAD) staining and luminescence detection of caspase levels. Cotreatment with CIL1 + DE enhanced the target cell-specific cytotoxicity, as well as the antiproliferative and proapoptotic properties. We found a 2200-fold increase in both the cytotoxic and antiproliferative efficacy of CIL1 + DE against HER14-targeted cells, while the effect on control NIH3T3 off-target cells was less profound (6.9- or 5.4-fold, respectively). Furthermore, we demonstrated that the CIL1 saponin fraction has a satisfactory in vitro safety profile with a lack of cytotoxic and mutagenic potential.
Collapse
Affiliation(s)
- Paulina Koczurkiewicz-Adamczyk
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Kraków, Poland
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
| | - Karolina Grabowska
- Department of Pharmacognosy, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Kraków, Poland
| | - Elżbieta Karnas
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Kamil Piska
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Kraków, Poland
| | - Dawid Wnuk
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Katarzyna Klaś
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Kraków, Poland
| | - Agnieszka Galanty
- Department of Pharmacognosy, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Kraków, Poland
| | - Katarzyna Wójcik-Pszczoła
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Kraków, Poland
| | - Marta Michalik
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland
| | - Elżbieta Pękala
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Kraków, Poland
| | - Hendrik Fuchs
- Institute of Diagnostic Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
| | - Irma Podolak
- Department of Pharmacognosy, Faculty of Pharmacy, Jagiellonian University Medical College, 30-688 Kraków, Poland
| |
Collapse
|
6
|
Thapa RK, Kim JO. Nanomedicine-based commercial formulations: current developments and future prospects. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2023; 53:19-33. [PMID: 36568502 PMCID: PMC9761651 DOI: 10.1007/s40005-022-00607-6] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022]
Abstract
Background In recent decades, there has been a considerable increase in the number of nanomedicine-based formulations, and their advantages, including controlled/targeted drug delivery with increased efficacy and reduced toxicity, make them ideal candidates for therapeutic delivery in the treatment of complex and difficult-to-treat diseases, such as cancer. Areas covered This review focuses on nanomedicine-based formulation development, approved and marketed nanomedicines, and the challenges faced in nanomedicine development as well as their future prospects. Expert opinion To date, the Food and Drug Administration and the European Medicines Agency have approved several nanomedicines, which are now commercially available. However, several critical challenges, including reproducibility, proper characterization, and biological evaluation, e.g., via assays, are still associated with their use. Therefore, rigorous studies alongside stringent guidelines for effective and safe nanomedicine development and use are still warranted. In this study, we provide an overview of currently available nanomedicine-based formulations. Thus, the findings here reported may serve as a basis for further studies regarding the use of these formulations for therapeutic purposes in near future.
Collapse
Affiliation(s)
- Raj Kumar Thapa
- Pharmacy Program, Gandaki University, Gyankunja, Pokhara-32, Kaski, Nepal
| | - Jong Oh Kim
- grid.413028.c0000 0001 0674 4447College of Pharmacy, Yeungnam University, 214-1 Dae-dong, Gyeongsan, 712-749 Republic of Korea
| |
Collapse
|
7
|
Ding YN, Xue M, Tang QS, Wang LJ, Ding HY, Li H, Gao CC, Yu WP. Immunotherapy-based novel nanoparticles in the treatment of gastrointestinal cancer: Trends and challenges. World J Gastroenterol 2022; 28:5403-5419. [PMID: 36312831 PMCID: PMC9611702 DOI: 10.3748/wjg.v28.i37.5403] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/27/2022] [Accepted: 09/15/2022] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal cancer (GIC) is the most common cancer with a poor prognosis. Currently, surgery is the main treatment for GIC. However, the high rate of postoperative recurrence leads to a low five-year survival rate. In recent years, immunotherapy has received much attention. As the only immunotherapy drugs approved by the Food and Drug Administration (FDA), immune checkpoint blockade (ICB) drugs have great potential in cancer therapy. Nevertheless, the efficacy of ICB treatment is greatly limited by the low immunogenicity and immunosuppressive microenvironment of GIC. Therefore, the targets of immunotherapy have expanded from ICB to increasing tumor immunogenicity, increasing the recruitment and maturation of immune cells and reducing the proportion of inhibitory immune cells, such as M2-like macrophages, regulatory T cells and myeloid-derived suppressor cells. Moreover, with the development of nanotechnology, a variety of nanoparticles have been approved by the FDA for clinical therapy, so novel nanodrug delivery systems have become a research focus for anticancer therapy. In this review, we summarize recent advances in the application of immunotherapy-based nanoparticles in GICs, such as gastric cancer, hepatocellular carcinoma, colorectal cancer and pancreatic cancer, and described the existing challenges and future trends.
Collapse
Affiliation(s)
- Yi-Nan Ding
- Department of Pathophysiology, College of Medicine, Southeast University, Nanjing 210000, Jiangsu Province, China
| | - Ming Xue
- Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210000, Jiangsu Province, China
| | - Qiu-Sha Tang
- Department of Pathophysiology, College of Medicine, Southeast University, Nanjing 210000, Jiangsu Province, China
| | - Li-Jun Wang
- Department of Pathophysiology, College of Medicine, Southeast University, Nanjing 210000, Jiangsu Province, China
| | - Hui-Yan Ding
- Department of Pathophysiology, College of Medicine, Southeast University, Nanjing 210000, Jiangsu Province, China
| | - Han Li
- Department of Tuberculosis, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing 210000, Jiangsu Province, China
| | - Cheng-Cheng Gao
- Department of Radiology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Wei-Ping Yu
- Medical School, Southeast University, Nanjing 210009, Jiangsu Province, China
| |
Collapse
|
8
|
Ding Y, Wang L, Li H, Miao F, Zhang Z, Hu C, Yu W, Tang Q, Shao G. Application of lipid nanovesicle drug delivery system in cancer immunotherapy. J Nanobiotechnology 2022; 20:214. [PMID: 35524277 PMCID: PMC9073823 DOI: 10.1186/s12951-022-01429-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/20/2022] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy has gradually emerged as the most promising anticancer therapy. In addition to conventional anti-PD-1/PD-L1 therapy, anti-CTLA-4 therapy, CAR-T therapy, etc., immunotherapy can also be induced by stimulating the maturation of immune cells or inhibiting negative immune cells, regulating the tumor immune microenvironment and cancer vaccines. Lipid nanovesicle drug delivery system includes liposomes, cell membrane vesicles, bacterial outer membrane vesicles, extracellular vesicles and hybrid vesicles. Lipid nanovesicles can be used as functional vesicles for cancer immunotherapy, and can also be used as drug carriers to deliver immunotherapy drugs to the tumor site for cancer immunotherapy. Here, we review recent advances in five kinds of lipid nanovesicles in cancer immunotherapy and assess the clinical application prospects of various lipid nanovesicles, hoping to provide valuable information for clinical translation in the future.
Collapse
Affiliation(s)
- Yinan Ding
- Medical School of Southeast University, Nanjing, 210009, China
| | - Luhong Wang
- Medical School of Southeast University, Nanjing, 210009, China
| | - Han Li
- Department of Tuberculosis, the Second Affiliated Hospital of Southeast University (the Second Hospital of Nanjing), Nanjing, 210009, China
| | - Fengqin Miao
- Medical School of Southeast University, Nanjing, 210009, China
| | - Zhiyuan Zhang
- Department of Neurosurgery, Nanjing Jinling Hospital, Nanjing University, Nanjing, 210002, China
| | - Chunmei Hu
- Department of Tuberculosis, the Second Affiliated Hospital of Southeast University (the Second Hospital of Nanjing), Nanjing, 210009, China
| | - Weiping Yu
- Medical School of Southeast University, Nanjing, 210009, China.
| | - Qiusha Tang
- Medical School of Southeast University, Nanjing, 210009, China.
| | - Guoliang Shao
- Department of Interventional Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
9
|
Wu CC, Wang YA, Livingston JA, Zhang J, Futreal PA. Prediction of biomarkers and therapeutic combinations for anti-PD-1 immunotherapy using the global gene network association. Nat Commun 2022; 13:42. [PMID: 35013211 PMCID: PMC8748689 DOI: 10.1038/s41467-021-27651-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 12/02/2021] [Indexed: 01/05/2023] Open
Abstract
Owing to a lack of response to the anti-PD1 therapy for most cancer patients, we develop a network approach to infer genes, pathways, and potential therapeutic combinations that are associated with tumor response to anti-PD1. Here, our prediction identifies genes and pathways known to be associated with anti-PD1, and is further validated by 6 CRISPR gene sets associated with tumor resistance to cytotoxic T cells and targets of the 36 compounds that have been tested in clinical trials for combination treatments with anti-PD1. Integration of our top prediction and TCGA data identifies hundreds of genes whose expression and genetic alterations that could affect response to anti-PD1 in each TCGA cancer type, and the comparison of these genes across cancer types reveals that the tumor immunoregulation associated with response to anti-PD1 would be tissue-specific. In addition, the integration identifies the gene signature to calculate the MHC I association immunoscore (MIAS) that shows a good correlation with patient response to anti-PD1 for 411 melanoma samples complied from 6 cohorts. Furthermore, mapping drug target data to the top genes in our association prediction identifies inhibitors that could potentially enhance tumor response to anti-PD1, such as inhibitors of the encoded proteins of CDK4, GSK3B, and PTK2.
Collapse
Affiliation(s)
- Chia-Chin Wu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Y Alan Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - J Andrew Livingston
- Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - P Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
10
|
Khirehgesh MR, Sharifi J, Safari F, Akbari B. Immunotoxins and nanobody-based immunotoxins: review and update. J Drug Target 2021; 29:848-862. [PMID: 33615933 DOI: 10.1080/1061186x.2021.1894435] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Immunotoxins (ITs) are protein-based drugs that compose of targeting and cytotoxic moieties. After binding the IT to the specific cell-surface antigen, the IT internalises into the target cell and kills it. Targeting and cytotoxic moieties usually include monoclonal antibodies and protein toxins with bacterial or plant origin, respectively. ITs have been successful in haematologic malignancies treatment. However, ITs penetrate poorly into solid tumours because of their large size. Use of camelid antibody fragments known as nanobodies (Nbs) as a targeting moiety may overcome this problem. Nbs are the smallest fragment of antibodies with excellent tumour tissue penetration. The ability to recognise cryptic (immuno-evasive) target antigens, low immunogenicity, and high-affinity are other fundamental characteristics of Nbs that make them suitable candidates in targeted therapy. Here, we reviewed and discussed the structure and function of ITs, Nbs, and nanobody-based ITs. To gain sound insight into the issue at hand, we focussed on nanobody-based ITs.
Collapse
Affiliation(s)
- Mohammad Reza Khirehgesh
- Department of Medical Biotechnology, School of Medical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Jafar Sharifi
- Department of Medical Biotechnology, School of Medical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Fatemeh Safari
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahman Akbari
- Department of Medical Biotechnology, School of Medical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
11
|
Nanomedicines accessible in the market for clinical interventions. J Control Release 2021; 330:372-397. [DOI: 10.1016/j.jconrel.2020.12.034] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 12/17/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023]
|
12
|
Niesler N, Arndt J, Silberreis K, Fuchs H. Generation of a soluble and stable apoptin-EGF fusion protein, a targeted viral protein applicable for tumor therapy. Protein Expr Purif 2020; 175:105687. [PMID: 32681952 DOI: 10.1016/j.pep.2020.105687] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 06/04/2020] [Accepted: 06/09/2020] [Indexed: 11/18/2022]
Abstract
A promising candidate for tumor targeted toxins is the chicken anemia-derived protein apoptin that induces tumor-specific apoptosis. It was aimed to design a novel apoptin-based targeted toxin by genetic fusion of apoptin with the tumor-directed ligand epidermal growth factor (EGF) using Escherichia coli as expression host. However, apoptin is highly hydrophobic and tends to form insoluble aggregates. Therefore, three different apoptin-EGF variants were generated. The fusion protein hexa-histidine (His)-apoptin-EGF (HAE) was expressed in E. coli and purified under denaturing conditions due to inclusion bodies. The protein solubility was improved by maltose-binding protein (MBP) or glutathione S-transferase. The protein MBP-apoptin-EGFHis (MAEH) was found favorable as a targeted toxin regarding final yield (4-6 mg/L) and stability. MBP was enzymatically removed using clotting factor Xa, which resulted in low yield and poor separation. MAEH was tested on target and non-target cell lines. The targeted tumor cell line A431 showed significant toxicity with an IC50 of 69.55 nM upon incubation with MAEH while fibroblasts and target receptor-free cells remained unaffected. Here we designed a novel EGF receptor targeting drug with high yield, purity and stability.
Collapse
Affiliation(s)
- Nicole Niesler
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Augustenburger Platz 1, 13353, Berlin, Germany; Berlin Institute of Health (BIH), Anna-Louisa-Karsch-Str. 2, 10178, Berlin, Germany
| | - Janine Arndt
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Kim Silberreis
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Augustenburger Platz 1, 13353, Berlin, Germany; Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195, Berlin, Germany
| | - Hendrik Fuchs
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health; Institut für Laboratoriumsmedizin, Klinische Chemie und Pathobiochemie, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
13
|
Arlotta KJ, Owen SC. Antibody and antibody derivatives as cancer therapeutics. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1556. [DOI: 10.1002/wnan.1556] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 02/20/2019] [Accepted: 03/10/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Keith J. Arlotta
- Department of Biomedical Engineering University of Utah Salt Lake City Utah
| | - Shawn C. Owen
- Department of Biomedical Engineering University of Utah Salt Lake City Utah
- Department of Pharmaceutics and Pharmaceutical Chemistry University of Utah Salt Lake City Utah
| |
Collapse
|
14
|
Hunjan MK, Nowsheen S, Ramos-Rodriguez AJ, Bridges AG, Lehman JS, Hashmi SK, El-Azhary RA. Chemotherapy-induced skin toxicity and capillary leak syndrome. Int J Dermatol 2019; 58:856-860. [PMID: 30847892 DOI: 10.1111/ijd.14392] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/28/2018] [Accepted: 01/17/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND The occurrence of chemotherapy-related adverse cutaneous reactions in the setting of capillary leak syndrome (CLS) is quite rare. Our objective was to identify the type of skin reactions associated with CLS. METHODS Leukemia or hematopoietic stem cell transplant patients between January 2010 and December 2017 were identified, and medical records were reviewed for a dermatology consultation occurring concomitantly with CLS. RESULTS Five patients were identified, two with a diagnosis of toxic erythema of chemotherapy (TEC) and three others with a skin diagnosis of toxic epidermal necrolysis (TEN). Pathology of all patients was available for clinical-pathologic confirmation. CONCLUSIONS Although TEC is generally self-limited, both TEC and TEN can present with severe adverse skin manifestations during CLS secondary to toxicity from chemotherapy.
Collapse
Affiliation(s)
- Manrup K Hunjan
- Sandwell and West Birmingham Hospital Trusts, Birmingham, UK
| | - Somaira Nowsheen
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic Alix School of Medicine and Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, USA
| | | | - Alina G Bridges
- Department of Dermatology, Mayo Clinic, Rochester, MN, USA.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Julia S Lehman
- Department of Dermatology, Mayo Clinic, Rochester, MN, USA.,Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Shahrukh K Hashmi
- Department of Hematology and Oncology, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
15
|
Co-Solvents Effects on the Stability of Recombinant Immunotoxin Denileukin Diftitox: Structure and Function Assessment. IRANIAN JOURNAL OF SCIENCE AND TECHNOLOGY, TRANSACTIONS A: SCIENCE 2019. [DOI: 10.1007/s40995-019-00676-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
16
|
Empowering dendritic cell cancer vaccination: the role of combinatorial strategies. Cytotherapy 2018; 20:1309-1323. [PMID: 30360963 DOI: 10.1016/j.jcyt.2018.09.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 01/22/2023]
Abstract
Dendritic cells (DCs) are bone marrow-derived immune cells that play a crucial role in inducing the adaptive immunity and supporting the innate immune response independently from T cells. In the last decade, DCs have become a hopeful instrument for cancer vaccines that aims at re-educating the immune system, leading to a potent anti-cancer immune response able to overcome the immunosuppressive tumor microenvironment (TME). Although several studies have indicated that DC-based vaccines are feasible and safe, the clinical advantages of DC vaccination as monotherapy for most of the neoplasms remain a distant target. Recently, many reports and clinical trials have widely used innovative combinatorial therapeutic strategies to normalize the immune function in the TME and synergistically enhance DC function. This review will describe the most relevant and updated evidence of the anti-cancer combinatorial approaches to boost the clinical potency of DC-based vaccines.
Collapse
|
17
|
Kuan SL, Fischer S, Hafner S, Wang T, Syrovets T, Liu W, Tokura Y, Ng DYW, Riegger A, Förtsch C, Jäger D, Barth TFE, Simmet T, Barth H, Weil T. Boosting Antitumor Drug Efficacy with Chemically Engineered Multidomain Proteins. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2018; 5:1701036. [PMID: 30128225 PMCID: PMC6097141 DOI: 10.1002/advs.201701036] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/01/2018] [Indexed: 05/05/2023]
Abstract
A facile chemical approach integrating supramolecular chemistry, site-selective protein chemistry, and molecular biology is described to engineer synthetic multidomain protein therapeutics that sensitize cancer cells selectively to significantly enhance antitumor efficacy of existing chemotherapeutics. The desired bioactive entities are assembled via supramolecular interactions at the nanoscale into structurally ordered multiprotein complexes comprising a) multiple copies of the chemically modified cyclic peptide hormone somatostatin for selective targeting and internalization into human A549 lung cancer cells expressing SST-2 receptors and b) a new cysteine mutant of the C3bot1 (C3) enzyme from Clostridium botulinum, a Rho protein inhibitor that affects and influences intracellular Rho-mediated processes like endothelial cell migration and blood vessel formation. The multidomain protein complex, SST3-Avi-C3, retargets C3 enzyme into non-small cell lung A549 cancer cells and exhibits exceptional tumor inhibition at a concentration ≈100-fold lower than the clinically approved antibody bevacizumab (Avastin) in vivo. Notably, SST3-Avi-C3 increases tumor sensitivity to a conventional chemotherapeutic (doxorubicin) in vivo. These findings show that the integrated approach holds vast promise to expand the current repertoire of multidomain protein complexes and can pave the way to important new developments in the area of targeted and combination cancer therapy.
Collapse
Affiliation(s)
- Seah Ling Kuan
- Max‐Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
- Institute of Inorganic Chemistry IUlm UniversityAlbert‐Einstein‐Allee 1189081UlmGermany
| | - Stephan Fischer
- Institute of Inorganic Chemistry IUlm UniversityAlbert‐Einstein‐Allee 1189081UlmGermany
- Institute of Pharmacology and ToxicologyUniversity of Ulm Medical CenterAlbert‐Einstein‐Allee 1189081UlmGermany
| | - Susanne Hafner
- Institute of Pharmacology of Natural Products and Clinical PharmacologyUlm UniversityHelmholtzstraße 2089081UlmGermany
| | - Tao Wang
- Institute of Inorganic Chemistry IUlm UniversityAlbert‐Einstein‐Allee 1189081UlmGermany
- School of Materials Science and EngineeringSouthwest Jiaotong University610031ChengduP. R. China
| | - Tatiana Syrovets
- Institute of Pharmacology of Natural Products and Clinical PharmacologyUlm UniversityHelmholtzstraße 2089081UlmGermany
| | - Weina Liu
- Max‐Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
- Institute of Inorganic Chemistry IUlm UniversityAlbert‐Einstein‐Allee 1189081UlmGermany
| | - Yu Tokura
- Max‐Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
- Institute of Inorganic Chemistry IUlm UniversityAlbert‐Einstein‐Allee 1189081UlmGermany
| | - David Yuen Wah Ng
- Max‐Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
- Institute of Inorganic Chemistry IUlm UniversityAlbert‐Einstein‐Allee 1189081UlmGermany
| | - Andreas Riegger
- Institute of Inorganic Chemistry IUlm UniversityAlbert‐Einstein‐Allee 1189081UlmGermany
| | - Christina Förtsch
- Institute of Pharmacology and ToxicologyUniversity of Ulm Medical CenterAlbert‐Einstein‐Allee 1189081UlmGermany
| | - Daniela Jäger
- Institute of PathologyUlm UniversityAlbert‐Einstein‐Allee 2389070UlmGermany
| | - Thomas F. E. Barth
- Institute of PathologyUlm UniversityAlbert‐Einstein‐Allee 2389070UlmGermany
| | - Thomas Simmet
- Institute of Pharmacology of Natural Products and Clinical PharmacologyUlm UniversityHelmholtzstraße 2089081UlmGermany
| | - Holger Barth
- Institute of Pharmacology and ToxicologyUniversity of Ulm Medical CenterAlbert‐Einstein‐Allee 1189081UlmGermany
| | - Tanja Weil
- Max‐Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
- Institute of Inorganic Chemistry IUlm UniversityAlbert‐Einstein‐Allee 1189081UlmGermany
| |
Collapse
|
18
|
Manoilov K, Palladin Institute of Biochemistry of the National Academy of Sciences of Ukraine. BIOLOGICAL PROPERTIES AND MEDICAL APPLICATION OF DIPHTHERIA TOXIN DERIVATIVES. BIOTECHNOLOGIA ACTA 2018. [DOI: 10.15407/biotech11.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
19
|
|
20
|
Lewis DJ, Dao H, Nagarajan P, Duvic M. Primary cutaneous anaplastic large-cell lymphoma: Complete remission for 13 years after denileukin diftitox. JAAD Case Rep 2017; 3:501-504. [PMID: 29296637 PMCID: PMC5728491 DOI: 10.1016/j.jdcr.2017.06.031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Affiliation(s)
- Daniel J Lewis
- School of Medicine, Baylor College of Medicine, Houston, Texas.,Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Harry Dao
- Department of Dermatology, Baylor College of Medicine, Houston, Texas
| | | | - Madeleine Duvic
- Department of Dermatology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
21
|
Cellular Entry of the Diphtheria Toxin Does Not Require the Formation of the Open-Channel State by Its Translocation Domain. Toxins (Basel) 2017; 9:toxins9100299. [PMID: 28937631 PMCID: PMC5666346 DOI: 10.3390/toxins9100299] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/20/2017] [Accepted: 09/20/2017] [Indexed: 12/30/2022] Open
Abstract
Cellular entry of diphtheria toxin is a multistage process involving receptor targeting, endocytosis, and translocation of the catalytic domain across the endosomal membrane into the cytosol. The latter is ensured by the translocation (T) domain of the toxin, capable of undergoing conformational refolding and membrane insertion in response to the acidification of the endosomal environment. While numerous now classical studies have demonstrated the formation of an ion-conducting conformation-the Open-Channel State (OCS)-as the final step of the refolding pathway, it remains unclear whether this channel constitutes an in vivo translocation pathway or is a byproduct of the translocation. To address this question, we measure functional activity of known OCS-blocking mutants with H-to-Q replacements of C-terminal histidines of the T-domain. We also test the ability of these mutants to translocate their own N-terminus across lipid bilayers of model vesicles. The results of both experiments indicate that translocation activity does not correlate with previously published OCS activity. Finally, we determined the topology of TH5 helix in membrane-inserted T-domain using W281 fluorescence and its depth-dependent quenching by brominated lipids. Our results indicate that while TH5 becomes a transbilayer helix in a wild-type protein, it fails to insert in the case of the OCS-blocking mutant H322Q. We conclude that the formation of the OCS is not necessary for the functional translocation by the T-domain, at least in the histidine-replacement mutants, suggesting that the OCS is unlikely to constitute a translocation pathway for the cellular entry of diphtheria toxin in vivo.
Collapse
|
22
|
Bhopale MK, Hilliard B, Constantinescu CS, Phillips SM, Rostami A. DAB 389IL-2 recombinant fusion toxin effect on lymphocyte- and macrophage-producing cytokine subpopulation cells in experimentally induced demyelinating disease in mice. Immunopharmacol Immunotoxicol 2017; 39:318-329. [PMID: 28929835 DOI: 10.1080/08923973.2017.1369099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
CONTEXT We have reported previously that DAB389IL-2 recombinant fusion toxin targets IL-2R bearing CD4+ cells, and suppresses demyelinating disease in acute (A) - and chronic (C) - experimental autoimmune encephalomyelitis (EAE) animal models of multiple sclerosis. OBJECTIVES The present study was undertaken to investigate the effect of DAB389IL-2 treatment on various cytokine-secreting cell populations in A-EAE and C-EAE mice. MATERIALS AND METHODS The effects of DAB389IL-2 at doses of 200-, 800-, or 1600 kU administered i.v. on days 11-13 and 15 on the clinical score and cytokine-secreting cell populations were examined using flow cytometry. RESULTS C-EAE mice treated with 1600kU DAB389IL-2, but not A-EAE mice treated with 800 kU had significantly reduced disease. The CD3+CD25+ sub-population in spleens and spinal cords of A-EAE mice treated with 800 kU DAB389IL-2 a was increased, whereas in C-EAE mice treated with 1600 kU this population was increased. DAB389IL-2 treatment reduced CD3+CD4+, CD3+CD8+, CD4+CD8+, CD3+IL-2+, CD3+IFN-γ+ and CD3+TNF-α+ T cell subpopulations in the spinal cord in A-EAE, and C-EAE mice on day 16. CD11b+ macrophages that were IL-2-, IFN-γ-, and TNF-α- positive were reduced in A-EAE mice. DAB389IL-2 treatment reduced CD19+ B-cells positive for IL-2 or CD11b+ in the spinal cord in acute and chronic disease. DAB389IL-2 treatment also reduced lymph node CD3+CD8+, CD4+CD8+, CD3+CD25+ populations on day 16, and lymph node CD3+IL-10+ and peripheral blood CD3+CD25+ populations on day 24. DISCUSSION AND CONCLUSIONS Our study demonstrates that DAB389IL-2 fusion toxin suppresses EAE in a dose-dependent manner, and alters inflammatory cell sub-populations during disease development.
Collapse
Affiliation(s)
- Mahendra K Bhopale
- a Department of Radiology , Temple University School of Medicine , Philadelphia , PA , USA
| | - Brendan Hilliard
- b Department of Medicine , Temple University School of Medicine , Philadelphia , PA , USA
| | | | - S Michael Phillips
- d Department of Medicine , University of Pennsylvania School of Medicine , Philadelphia , PA , USA
| | - Abdolmohamad Rostami
- e Department of Neurology , Thomas Jefferson University , Philadelphia , PA , USA
| |
Collapse
|
23
|
Abstract
Drug delivery systems (DDS) are designed to improve the pharmacological and therapeutic effect. In the past few decades, there are some problems that impeded applications of particulate DDS have been resolved, with several DDS formulations of anticancer now approved for clinical use. Liposomal nanoparticles (LNs) encapsulating therapeutic agents have been recognized as one of the most advanced classes of DDS. Liposomal nanoparticles (LNs) could encapsulate both conventional anticancer drugs and the new genetic drugs with several properties such as high drug-to-lipid ratio, excellent retention of drug and a long circulation lifetime. These excellent properties of LNs have the potentials to offer new treatments in area of cancer therapy. Here, we will discuss recent advances in this field involving conventional anticancer drugs as well as the new genetic drugs.
Collapse
Affiliation(s)
- J. Zhong
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, Huzhou, Zhejiang Province, No. 198, Hongqi Road, China 313000
| | - L. C. Dai
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, Huzhou, Zhejiang Province, No. 198, Hongqi Road, China 313000
| |
Collapse
|
24
|
Dang L, Van Damme EJM. Toxic proteins in plants. PHYTOCHEMISTRY 2015; 117:51-64. [PMID: 26057229 PMCID: PMC7111729 DOI: 10.1016/j.phytochem.2015.05.020] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Revised: 05/25/2015] [Accepted: 05/27/2015] [Indexed: 05/06/2023]
Abstract
Plants have evolved to synthesize a variety of noxious compounds to cope with unfavorable circumstances, among which a large group of toxic proteins that play a critical role in plant defense against predators and microbes. Up to now, a wide range of harmful proteins have been discovered in different plants, including lectins, ribosome-inactivating proteins, protease inhibitors, ureases, arcelins, antimicrobial peptides and pore-forming toxins. To fulfill their role in plant defense, these proteins exhibit various degrees of toxicity towards animals, insects, bacteria or fungi. Numerous studies have been carried out to investigate the toxic effects and mode of action of these plant proteins in order to explore their possible applications. Indeed, because of their biological activities, toxic plant proteins are also considered as potentially useful tools in crop protection and in biomedical applications, such as cancer treatment. Genes encoding toxic plant proteins have been introduced into crop genomes using genetic engineering technology in order to increase the plant's resistance against pathogens and diseases. Despite the availability of ample information on toxic plant proteins, very few publications have attempted to summarize the research progress made during the last decades. This review focuses on the diversity of toxic plant proteins in view of their toxicity as well as their mode of action. Furthermore, an outlook towards the biological role(s) of these proteins and their potential applications is discussed.
Collapse
Affiliation(s)
- Liuyi Dang
- Ghent University, Dept. Molecular Biotechnology, Laboratory Biochemistry and Glycobiology, 9000 Gent, Belgium.
| | - Els J M Van Damme
- Ghent University, Dept. Molecular Biotechnology, Laboratory Biochemistry and Glycobiology, 9000 Gent, Belgium.
| |
Collapse
|
25
|
Duron L, Delestre F, Amoura Z, Arnaud L. [Idiopathic and secondary capillary leak syndromes: A systematic review of the literature]. Rev Med Interne 2015; 36:386-94. [PMID: 25600329 DOI: 10.1016/j.revmed.2014.11.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 11/23/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE The capillary leak syndrome (CLS) is a rare condition characterized by the onset of hypotension, edema, hemoconcentration and hypoalbuminemia. CLS can be idiopathic (Clarkson's disease) or secondary to various conditions and treatments. Here, we review the clinical and biological features, pathophysiology, causes and treatment of this rare condition. METHODS We performed a systematic review of the literature (Medline database through February 2014) to identify all articles about CLS. The relevant references were selected by two independent authors. RESULTS Secondary CLSs are mostly due to malignant hematological diseases, viral infections, and treatments such as chemotherapies and therapeutic growth factors. Diagnosis of idiopathic CLS is made by exclusion of secondary diseases, especially as a serum monoclonal immunoglobulin is present, or when there is a relapsing disease, no initial lung involvement or preserved consciousness despite low blood pressure. Acute episodes are treated with vasopressor therapy and judicious fluid replacement. Between episodes, patients with Clarkson's disease may be treated with intravenous immunoglobulins. CLS is a severe disease with significantly impaired prognosis. CONCLUSION Clarification of the pathophysiological mechanisms of CLS is essential to improve the prognosis of this rare disease with more targeted treatments.
Collapse
Affiliation(s)
- L Duron
- Service de médecine interne 2, groupe hospitalier Pitié-Salpêtrière, AP-HP, 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | - F Delestre
- Service de médecine interne 2, groupe hospitalier Pitié-Salpêtrière, AP-HP, 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | - Z Amoura
- Service de médecine interne 2, groupe hospitalier Pitié-Salpêtrière, AP-HP, 47-83, boulevard de l'Hôpital, 75013 Paris, France; UPMC université Paris 06, Sorbonne universités, 75013 Paris, France
| | - L Arnaud
- Service de médecine interne 2, groupe hospitalier Pitié-Salpêtrière, AP-HP, 47-83, boulevard de l'Hôpital, 75013 Paris, France; UPMC université Paris 06, Sorbonne universités, 75013 Paris, France.
| |
Collapse
|
26
|
Duvic M. Choosing a systemic treatment for advanced stage cutaneous T-cell lymphoma: mycosis fungoides and Sézary syndrome. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2015; 2015:529-544. [PMID: 26637769 DOI: 10.1182/asheducation-2015.1.529] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Affiliation(s)
- Madeleine Duvic
- Department of Dermatology, University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
27
|
Sandvig K, Bergan J, Kavaliauskiene S, Skotland T. Lipid requirements for entry of protein toxins into cells. Prog Lipid Res 2014; 54:1-13. [PMID: 24462587 DOI: 10.1016/j.plipres.2014.01.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 01/15/2014] [Accepted: 01/15/2014] [Indexed: 01/05/2023]
Abstract
The plant toxin ricin and the bacterial toxin Shiga toxin both belong to a group of protein toxins having one moiety that binds to the cell surface, and another, enzymatically active moiety, that enters the cytosol and inhibits protein synthesis by inactivating ribosomes. Both toxins travel all the way from the cell surface to endosomes, the Golgi apparatus and the ER before the ribosome-inactivating moiety enters the cytosol. Shiga toxin binds to the neutral glycosphingolipid Gb3 at the cell surface and is therefore dependent on this lipid for transport into the cells, whereas ricin binds both glycoproteins and glycolipids with terminal galactose. The different steps of transport used by these toxins have specific requirements for lipid species, and with the recent developments in mass spectrometry analysis of lipids and microscopical and biochemical dissection of transport in cells, we are starting to see the complexity of endocytosis and intracellular transport. In this article we describe lipid requirements and the consequences of lipid changes for the entry and intoxication with ricin and Shiga toxin. These toxins can be a threat to human health, but can also be exploited for diagnosis and therapy, and have proven valuable as tools to study intracellular transport.
Collapse
Affiliation(s)
- Kirsten Sandvig
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Department of Biosciences, University of Oslo, Oslo, Norway.
| | - Jonas Bergan
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.
| | - Simona Kavaliauskiene
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway; Department of Biosciences, University of Oslo, Oslo, Norway.
| | - Tore Skotland
- Centre for Cancer Biomedicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Biochemistry, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
28
|
Kun L, Hernandez-Ilizaliturri FJ, Reddy NM. Novel Therapeutic Strategies for Cutaneous T-Cell Lymphoma in Advanced Stages. Semin Hematol 2014; 51:35-41. [DOI: 10.1053/j.seminhematol.2013.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
29
|
Abstract
Effective antitumor immunity requires the generation and persistence of functional tumor-specific T-cell responses. Among the critical factors that often control these responses is how the antigen is delivered and presented to T cells. The use of peptide-based vaccination has been found to be a promising means to induce antitumor T-cell responses but with limited effects even if the peptide is co-delivered with a potent adjuvant. This limited response could be due to cancer-induced dysfunction in dendritic cells (DC), which play a central role in shaping the quantity and quality of antitumor immunity. Therefore, DC-based peptide delivery of tumor antigen is becoming a potential approach in cancer immunotherapy. In this approach, autologous DC are generated from their precursors in bone marrow or peripheral blood mononuclear cells, loaded with tumor antigen(s) and then infused back to the tumor-bearing host in about 7 days. This DC-based vaccination can act as an antigen delivery vehicle as well as a potent adjuvant, resulting in measurable antitumor immunity in several cancer settings in preclinical and clinical studies. This chapter focuses on DC-based vaccination and how this approach can be more efficacious in cancer immunotherapy.Effective antitumor immunity requires the generation and persistence of functional tumor-specific T-cell responses. Among the critical factors that often control these responses is how the antigen is delivered and presented to T cells. The use of peptide-based vaccination has been found to be a promising means to induce antitumor T-cell responses but with limited effects even if the peptide is co-delivered with a potent adjuvant. This limited response could be due to cancer-induced dysfunction in dendritic cells (DC), which play a central role in shaping the quantity and quality of antitumor immunity. Therefore, DC-based peptide delivery of tumor antigen is becoming a potential approach in cancer immunotherapy. In this approach, autologous DC are generated from their precursors in bone marrow or peripheral blood mononuclear cells, loaded with tumor antigen(s) and then infused back to the tumor-bearing host in about 7 days. This DC-based vaccination can act as an antigen delivery vehicle as well as a potent adjuvant, resulting in measurable antitumor immunity in several cancer settings in preclinical and clinical studies. This chapter focuses on DC-based vaccination and how this approach can be more efficacious in cancer immunotherapy.
Collapse
Affiliation(s)
- Mohamed L Salem
- Immunology and Biotechnology Unit, Zoology Department, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
30
|
pH-triggered conformational switching along the membrane insertion pathway of the diphtheria toxin T-domain. Toxins (Basel) 2013; 5:1362-80. [PMID: 23925141 PMCID: PMC3760040 DOI: 10.3390/toxins5081362] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 07/26/2013] [Accepted: 07/26/2013] [Indexed: 11/17/2022] Open
Abstract
The translocation (T)-domain plays a key role in the action of diphtheria toxin and is responsible for transferring the catalytic domain across the endosomal membrane into the cytosol in response to acidification. Deciphering the molecular mechanism of pH-dependent refolding and membrane insertion of the T-domain, which is considered to be a paradigm for cell entry of other bacterial toxins, reveals general physicochemical principles underlying membrane protein assembly and signaling on membrane interfaces. Structure-function studies along the T-domain insertion pathway have been affected by the presence of multiple conformations at the same time, which hinders the application of high-resolution structural techniques. Here, we review recent progress in structural, functional and thermodynamic studies of the T-domain archived using a combination of site-selective fluorescence labeling with an array of spectroscopic techniques and computer simulations. We also discuss the principles of conformational switching along the insertion pathway revealed by studies of a series of T-domain mutants with substitutions of histidine residues.
Collapse
|
31
|
Chen W, Ying T, Dimitrov DS. Antibody-based candidate therapeutics against HIV-1: implications for virus eradication and vaccine design. Expert Opin Biol Ther 2013; 13:657-71. [PMID: 23293858 DOI: 10.1517/14712598.2013.761969] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The currently available anti-HIV-1 drugs can control the infection but do not eradicate the virus. Their long-term use can lead to side effects and resistance to therapy. Therefore, eradication of the virus has been a major goal of research. Biological therapeutics including broadly neutralizing monoclonal antibodies (bnAbs) are promising tools to reach this goal. They could also help design novel vaccine immunogens potentially capable of eliciting bnAbs targeting the HIV-1 envelope glycoproteins (Envs). AREAS COVERED We review HIV-1 bnAbs and their potential as candidate prophylactics and therapeutics used individually, in combination, or as bispecific fusion proteins. We also discuss their potential use in the 'activation-elimination' approach for HIV-1 eradication in infected patients receiving antiretroviral treatment as well as current vaccine design efforts based on understanding of interactions of candidate vaccine immunogens with matured bnAbs and their putative germline predecessors, and related antibody maturation pathways. EXPERT OPINION Exploration of HIV-1 bnAbs has provided and will continue to provide useful knowledge that helps develop novel types of biotherapeutics and vaccines. It is possible that bnAb-based candidate therapeutics could help eradicate HIV-1. Development of vaccine immunogens capable of eliciting potent bnAbs in humans remains a fundamental challenge.
Collapse
Affiliation(s)
- Weizao Chen
- National Cancer Institute, National Institutes of Health, Frederick National Laboratory for Cancer Research, Protein Interactions Group, Miller Drive, Building 469, Room 144, Frederick, MD 21702, USA.
| | | | | |
Collapse
|
32
|
Jang MS, Kang DY, Han SH, Park JB, Kim ST, Suh KS. CD25+ folliculotropic Sézary syndrome with CD30+ large cell transformation. Australas J Dermatol 2012. [DOI: 10.1111/ajd.12000] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Min Soo Jang
- Department of Dermatology; Kosin University College of Medicine; Busan South Korea
| | - Dong Young Kang
- Department of Dermatology; Kosin University College of Medicine; Busan South Korea
| | - Sang Hwa Han
- Department of Dermatology; Kosin University College of Medicine; Busan South Korea
| | - Jong Bin Park
- Department of Dermatology; Kosin University College of Medicine; Busan South Korea
| | - Sang Tae Kim
- Department of Dermatology; Kosin University College of Medicine; Busan South Korea
| | - Kee Suck Suh
- Department of Dermatology; Kosin University College of Medicine; Busan South Korea
| |
Collapse
|
33
|
Izu-Belloso R, García-Ruiz J. Actualización terapéutica en linfomas cutáneos. ACTAS DERMO-SIFILIOGRAFICAS 2012; 103:694-707. [DOI: 10.1016/j.ad.2012.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 01/12/2012] [Accepted: 01/29/2012] [Indexed: 10/28/2022] Open
|
34
|
Izu-Belloso R, García-Ruiz J. Treatment of Cutaneous Lymphomas: an Update. ACTAS DERMO-SIFILIOGRAFICAS 2012. [DOI: 10.1016/j.adengl.2012.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
|
35
|
Talpur R, Duvic M. Pilot study of denileukin diftitox alternate dosing regimen in patients with cutaneous peripheral T-cell lymphomas. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2012; 12:180-5. [PMID: 22521504 DOI: 10.1016/j.clml.2012.01.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 01/21/2012] [Accepted: 01/30/2012] [Indexed: 11/25/2022]
Abstract
UNLABELLED Better treatment and survival outcomes are needed for the rare primary cutaneous peripheral T-cell lymphomas.Five (62.5%) of 8 patients with peripheral T-cell lymphomas enrolled in a pilot study of denileukin diftitoxat 18 μg/kg per day for 5 days followed by once weekly for 24 weeks responded, including 2 complete responses, one of which is ongoing at 8 years. PURPOSE To evaluate the safety and efficacy of an alternate dosing regimen in rare primary cutaneous peripheral T-cell lymphoma variants. METHODS This is a prospective, single center, pilot study of denileukin diftitox (Dd) in patients with persistent or recurrent cutaneous peripheral T-cell lymphomas and mycosis fungoides (MF) variants, excluding Sézary syndrome (SS). Dd was administered at 18 μg/kg per day for 5 days and once weekly for 24 weeks, with response by modified skin weighed assessment tool. RESULTS Eight patients, with a median age of 76 years (range, 44-88 years), were treated between December 2003 and July 2008. Five (62.5%) of 8 patients responded, including 3 patients with CD30(+) anaplastic large-cell lymphoma (ALCL) with 2 complete responses, one ongoing at 8 years. One patient with CD8(+) and 1 patient with natural killer T cell lymphoma (NK-T) had partial responses. Progressive disease occurred in 1 patient positive for human T-cell lymphotropic virus and 1 patient with ALCL. Vascular leak syndrome (VLS) occurred in 6 (75%) of 8 patients during or just after cycle 1. Three were grade 3, and 2 of these resulted in study withdrawal. Other adverse effects included nausea or vomiting (n = 3), fatigue (n = 1), back pain (n = 1), transaminase elevations (n = 3), and elevated creatinine (n = 1). CONCLUSIONS Dd with an alternate dosing schedule was active in this small study of primary cutaneous T-cell lymphomas.
Collapse
Affiliation(s)
- Rakhshandra Talpur
- Division of Internal Medicine, Department of Dermatology, the University of Texas, M.D. Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
36
|
Nierkens S, Janssen EM. Harnessing dendritic cells for tumor antigen presentation. Cancers (Basel) 2011; 3:2195-213. [PMID: 24212804 PMCID: PMC3757412 DOI: 10.3390/cancers3022195] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 04/14/2011] [Accepted: 04/19/2011] [Indexed: 12/11/2022] Open
Abstract
Dendritic cells (DC) are professional antigen presenting cells that are crucial for the induction of anti-tumor T cell responses. As a consequence, research has focused on the harnessing of DCs for therapeutic interventions. Although current strategies employing ex vivo-generated and tumor-antigen loaded DCs have been proven feasible, there are still many obstacles to overcome in order to improve clinical trial successes and offset the cost and complexity of customized cell therapy. This review focuses on one of these obstacles and a pivotal step for the priming of tumor-specific CD8+ and CD4+ T cells; the in vitro loading of DCs with tumor antigens.
Collapse
Affiliation(s)
- Stefan Nierkens
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Geert Grooteplein 28, Nijmegen 6525 GA, The Netherlands; E-Mail:
| | - Edith M. Janssen
- Division of Molecular Immunology, Cincinnati Children's Hospital Research Foundation, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| |
Collapse
|
37
|
Abstract
Immune cells infiltrate tumors and make up a significant component of the multicellular cancer micro-environment, yet the immune system often fails to prevent tumor formation and progression. One explanation for this paradox is the presence of tolerance-promoting regulatory T cells (Tregs) that counteract antitumor immune cells. Tregs were known to be essential for maintaining self-tolerance. Recently, Tregs have been found to promote tolerance to tumors in mouse models. Moreover, Treg infiltration in human tumors and malignant ascites is associated with worse clinical outcomes for various types of cancers. As many reviews have discussed the development and function of Tregs, this review focuses on the cellular and molecular mechanisms by which Tregs influence antitumor immune responses, and also discusses how these mechanisms might be exploited to develop innovative immune-based approaches that can improve cancer therapy.
Collapse
Affiliation(s)
- Xuefang Cao
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.
| |
Collapse
|
38
|
Berger EA, Pastan I. Immunotoxin complementation of HAART to deplete persisting HIV-infected cell reservoirs. PLoS Pathog 2010; 6:e1000803. [PMID: 20548940 PMCID: PMC2883583 DOI: 10.1371/journal.ppat.1000803] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Edward A Berger
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America.
| | | |
Collapse
|
39
|
Zhang Y, Schulte W, Pink D, Phipps K, Zijlstra A, Lewis JD, Waisman DM. Sensitivity of cancer cells to truncated diphtheria toxin. PLoS One 2010; 5:e10498. [PMID: 20463924 PMCID: PMC2864767 DOI: 10.1371/journal.pone.0010498] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Accepted: 04/14/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Diphtheria toxin (DT) has been utilized as a prospective anti-cancer agent for the targeted delivery of cytotoxic therapy to otherwise untreatable neoplasia. DT is an extremely potent toxin for which the entry of a single molecule into a cell can be lethal. DT has been targeted to cancer cells by deleting the cell receptor-binding domain and combining the remaining catalytic portion with targeting proteins that selectively bind to the surface of cancer cells. It has been assumed that "receptorless" DT cannot bind to and kill cells. In the present study, we report that "receptorless" recombinant DT385 is in fact cytotoxic to a variety of cancer cell lines. METHODS In vitro cytotoxicity of DT385 was measured by cell proliferation, cell staining and apoptosis assays. For in vivo studies, the chick chorioallantoic membrane (CAM) system was used to evaluate the effect of DT385 on angiogenesis. The CAM and mouse model system was used to evaluate the effect of DT385 on HEp3 and Lewis lung carcinoma (LLC) tumor growth, respectively. RESULTS Of 18 human cancer cell lines tested, 15 were affected by DT385 with IC(50) ranging from 0.12-2.8 microM. Furthermore, high concentrations of DT385 failed to affect growth arrested cells. The cellular toxicity of DT385 was due to the inhibition of protein synthesis and induction of apoptosis. In vivo, DT385 diminished angiogenesis and decreased tumor growth in the CAM system, and inhibited the subcutaneous growth of LLC tumors in mice. CONCLUSION DT385 possesses anti-angiogenic and anti-tumor activity and may have potential as a therapeutic agent.
Collapse
Affiliation(s)
- Yi Zhang
- Departments of Biochemistry and Molecular Biology and Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | | | - Kyle Phipps
- Departments of Biochemistry and Molecular Biology and Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Andries Zijlstra
- Innovascreen Inc, Halifax, Nova Scotia, Canada
- Department of Pathology, Vanderbilt University, Nashville, Tennessee, Unites States of America
| | - John D. Lewis
- Innovascreen Inc, Halifax, Nova Scotia, Canada
- Department of Oncology, University of Western Ontario, London, Ontario, Canada
| | - David Morton Waisman
- Departments of Biochemistry and Molecular Biology and Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- * E-mail:
| |
Collapse
|
40
|
Protein toxins from plants and bacteria: Probes for intracellular transport and tools in medicine. FEBS Lett 2010; 584:2626-34. [DOI: 10.1016/j.febslet.2010.04.008] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Accepted: 04/07/2010] [Indexed: 01/07/2023]
|
41
|
Duvic M, Donato M, Dabaja B, Richmond H, Singh L, Wei W, Acholonu S, Khouri I, Champlin R, Hosing C. Total skin electron beam and non-myeloablative allogeneic hematopoietic stem-cell transplantation in advanced mycosis fungoides and Sezary syndrome. J Clin Oncol 2010; 28:2365-72. [PMID: 20351328 DOI: 10.1200/jco.2009.25.8301] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Transformed mycosis fungoides (MF) and Sézary syndrome (SS) are currently incurable. We studied the safety and efficacy of total skin electron beam with allogeneic hematopoietic stem-cell transplantation (HSCT) in patients with cutaneous T-cell lymphoma (CTCL). PATIENTS AND METHODS Nineteen patients with advanced CTCL (median age, 50 years; four prior therapies) underwent total skin electron beam radiation followed by allogeneic HSCT between July 2001 and July 2008. Sixteen patients were conditioned with fludarabine (125 mg/m(2)) and melphalan (140 mg/m(2)) plus thymoglobulin (for mismatched donors). Graft-versus-host disease (GVHD) prophylaxis was with tacrolimus/mini methotrexate. RESULTS Eighteen patients experienced engraftment, and one died as a result of sepsis on day 16. Median time to recovery of absolute neutrophil count (ANC) was 12 days. Fifteen achieved full donor chimerism, 12 had acute GVHD, and 12 were treated for chronic GVHD. The overall intent-to-treat response was 68%, and the complete response rate was 58%. Four of six patients died in complete remission as a result of bacterial sepsis (n = 2), chronic GVHD and fungal infection (n = 1), or lung cancer (n = 1); only two died as a result of progressive disease. Eight experienced relapse in skin; five regained complete response with reduced immunosuppression or donor lymphocyte infusions. Eleven of 13 are currently in complete remissions, with median follow-up of 19 months (range, 1.3 to 8.3 years). Median overall survival has not been reached. CONCLUSION Total skin electron beam followed by allogeneic stem-cell transplantation merits additional evaluation for a selected group of patients with refractory, advanced, cutaneous T-cell lymphoma with evidence for graft-versus-tumor effect.
Collapse
Affiliation(s)
- Madeleine Duvic
- Department of Dermatology, MD Anderson Cancer Center, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Salem ML, El-Demellawy M, El-Azm ARA. The potential use of Toll-like receptor agonists to restore the dysfunctional immunity induced by hepatitis C virus. Cell Immunol 2010; 262:96-104. [PMID: 20338549 DOI: 10.1016/j.cellimm.2010.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Accepted: 03/02/2010] [Indexed: 02/07/2023]
Abstract
Hepatitis C virus (HCV) infection is a major public health concern with approximately 3% of the world's population is infected, posing social, economical and health burden. Less than 20% of the infected individuals clear the virus during the acute infection, while the rest develop chronic infection. The treatment of choice for HCV infection is pegylated interferon-alpha (IFN-alpha) in combination with ribavarin. Despite the cost and side effects of this treatment regimen, many patients fail this therapy and develop persistent HCV infection, leading to cirrhosis and hepatocellular carcinoma. Although the mechanisms underlying the failure to resolve HCV infection are poorly understood, the incapability of patients to develop effective anti-HCV immunity is a potential cause. We hypothesize that the dysfunctional anti-HCV immunity is due to the emergence of immunosuppressive cells coinciding with a decrease in the stimulatory dendritic cells (DCs) and natural killer (NK) cells. We further hypothesize that applying agents that can correct the imbalance between the immunosuppressive cells and stimulatory cells can results in resolution of chronic HCV. In this review article, we will discuss potential approaches, focusing on the use of Toll-like receptor agonists, to block the suppressive effects of the regulatory cells and restore the stimulatory effects of DCs and NK cells.
Collapse
MESH Headings
- Adjuvants, Immunologic/therapeutic use
- Antiviral Agents/therapeutic use
- Dendritic Cells/immunology
- Enzyme Inhibitors/therapeutic use
- Hepacivirus/immunology
- Hepatitis C, Chronic/drug therapy
- Hepatitis C, Chronic/epidemiology
- Hepatitis C, Chronic/immunology
- Hepatitis C, Chronic/virology
- Humans
- Immunity, Innate
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Interferon-alpha/therapeutic use
- Killer Cells, Natural/immunology
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Ribavirin/therapeutic use
- T-Lymphocytes, Regulatory/immunology
- Toll-Like Receptors/agonists
- Toll-Like Receptors/immunology
Collapse
Affiliation(s)
- Mohamed L Salem
- Surgery Department and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA.
| | | | | |
Collapse
|
43
|
Phillips SM, Bhopale MK, Hilliard B, Zekavat SA, Ali MAR, Rostami A. Suppression of murine experimental autoimmune encephalomyelitis by interleukin-2 receptor targeted fusion toxin, DAB389IL-2. Cell Immunol 2010; 261:144-52. [DOI: 10.1016/j.cellimm.2009.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2009] [Revised: 11/30/2009] [Accepted: 12/01/2009] [Indexed: 12/16/2022]
|
44
|
White-Gilbertson S, Kurtz DT, Voelkel-Johnson C. The role of protein synthesis in cell cycling and cancer. Mol Oncol 2009; 3:402-8. [PMID: 19546037 PMCID: PMC2784195 DOI: 10.1016/j.molonc.2009.05.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2009] [Revised: 05/04/2009] [Accepted: 05/25/2009] [Indexed: 01/08/2023] Open
Abstract
Cell cycling and protein synthesis are both key physiological tasks for cancer cells. Here we present a model for how the elongation phase of protein synthesis, governed by elongation factor 2 and elongation factor 2 kinase, both modulates and responds to cell cycling. Within this framework we also discuss survivin, a protein with both pro-mitotic and anti-apoptotic roles whose persistence in the cell is tied to protein synthesis due to its short half-life. Finally, we provide a brief overview of efforts of cancer researchers to target EF2 and EF2 kinase.
Collapse
Affiliation(s)
- Shai White-Gilbertson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - David T. Kurtz
- Department of Pharmacology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Christina Voelkel-Johnson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
45
|
Kusmartsev S, Vieweg J. Enhancing the efficacy of cancer vaccines in urologic oncology: new directions. Nat Rev Urol 2009; 6:540-9. [DOI: 10.1038/nrurol.2009.177] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
46
|
Abstract
Biologics are protein molecules which are used in various diseases to target the specific points in the immunopathogenesis of the diseases. The molecules are produced by recombinant DNA technology. The molecules bind to the specific targets without interfering wtih rest of the pathogenetic pathways. Therefore the so called 'immunosuppressives' have, although, a broader broader spectrum of action on immune system, their side-effects are also equally more. The biologics, because of their spefic action on the immune system, have very little side effects. The biologics which have revolutionized the treatment of various dermatologic diseases have been discussed here.
Collapse
Affiliation(s)
- Arijit Coondoo
- From the Department of Dermatology, Vivekananda Institute of Medical Science, Kolkata, India.
| |
Collapse
|
47
|
|