Purpose
The selective elimination of cancer stem cells (CSCs) in tumor patients is a crucial goal because CSCs cause drug refractory relapse. To improve the current conventional bispecific immune-engager platform, a 16133 bispecific natural killer (NK) cell engager (BiKE), consisting of scFvs binding FcγRIII (CD16) on NK cells and CD133 on carcinoma cells, was first synthesized and a modified interleukin (IL)-15 crosslinker capable of stimulating NK effector cells was introduced.
Materials and Methods
DNA shuffling and ligation techniques were used to assemble and synthesize the 1615133 trispecific NK cell engager (TriKE). The construct was tested for its specificity using flow cytometry, cytotoxic determinations using chromium release assays, and lytic degranulation. IL-15–mediated expansion was measured using flow-based proliferation assays. The level of interferon (IFN)-γ release was measured because of its importance in the anti-cancer response.
Results
1615133 TriKE induced NK cell–mediated cytotoxicity and NK expansion far greater than that achieved with BiKE devoid of IL-15. The drug binding and induction of cytotoxic degranulation was CD133+ specific and the anti-cancer activity was improved by integrating the IL-15 cross linker. The NK cell–related cytokine release measured by IFN-γ detection was higher than that of BiKE. NK cytokine release studies showed that although the IFN-γ levels were elevated, they did not approach the levels achieved with IL-12/IL-18, indicating that release was not at the supraphysiologic level.
Conclusion
1615133 TriKE enhances the NK cell anti-cancer activity and provides a self-sustaining mechanism via IL-15 signaling. By improving the NK cell performance, the new TriKE represents a highly active drug against drug refractory relapse mediated by CSCs.
Citations
Citations to this article as recorded by
CD56brightcytokine-induced memory-like NK cells and NK-cell engagers synergize against non-small cell lung cancer cancer-stem cells Maria L Guevara Lopez, Ann Gebo, Monica Parodi, Stefano Persano, Josephine Maus-Conn, Maria Cristina Mingari, Fabrizio Loiacono, Paola Orecchia, Simona Sivori, Claudia Cantoni, Marco Gentili, Federica Facchinetti, Riccardo Ferracini, Daniel A Vallera, Mar Journal for ImmunoTherapy of Cancer.2025; 13(2): e010205. CrossRef
New immune cell engagers for cancer immunotherapy Aurore Fenis, Olivier Demaria, Laurent Gauthier, Eric Vivier, Emilie Narni-Mancinelli Nature Reviews Immunology.2024; 24(7): 471. CrossRef
Natural killer cell therapies Eric Vivier, Lucas Rebuffet, Emilie Narni-Mancinelli, Stéphanie Cornen, Rob Y. Igarashi, Valeria R. Fantin Nature.2024; 626(8000): 727. CrossRef
Tri-specific killer engager: unleashing multi-synergic power against cancer Peeranut Winidmanokul, Aussara Panya, Seiji Okada Exploration of Targeted Anti-tumor Therapy.2024; 5(2): 432. CrossRef
Bridging the gap with multispecific immune cell engagers in cancer and infectious diseases Camille Rolin, Jacques Zimmer, Carole Seguin-Devaux Cellular & Molecular Immunology.2024; 21(7): 643. CrossRef
Natural killer cells in neuroblastoma: immunological insights and therapeutic perspectives Magdalena Rados, Anna Landegger, Lukas Schmutzler, Kimberlie Rabidou, Sabine Taschner-Mandl, Irfete S. Fetahu Cancer and Metastasis Reviews.2024; 43(4): 1401. CrossRef
Implications of glycosylation for the development of selected cytokines and their derivatives for medical use Giulia Scapin, Ece Cagdas, Lise Marie Grav, Nathan E Lewis, Steffen Goletz, Lise Hafkenscheid Biotechnology Advances.2024; 77: 108467. CrossRef
Natural killer cell engagers: From bi‐specific to tri‐specific and tetra‐specific engagers for enhanced cancer immunotherapy An Zhu, Yu Bai, Yanyang Nan, Dianwen Ju Clinical and Translational Medicine.2024;[Epub] CrossRef
Emerging Antibodies in Cancer Therapy Yaping Sun, Jian Xu Advanced NanoBiomed Research.2023;[Epub] CrossRef
Mapping the interplay between NK cells and HIV: therapeutic implications Renee R Anderko, Robbie B Mailliard Journal of Leukocyte Biology.2023; 113(2): 109. CrossRef
Generation of a symmetrical trispecific NK cell engager based on a two-in-one antibody Julia Harwardt, Stefania C. Carrara, Jan P. Bogen, Katrin Schoenfeld, Julius Grzeschik, Björn Hock, Harald Kolmar Frontiers in Immunology.2023;[Epub] CrossRef
Bispecific NK-cell engager targeting BCMA elicits stronger antitumor effects and produces less proinflammatory cytokines than T-cell engager Xinghui Xiao, Ying Cheng, Xiaodong Zheng, Yuhang Fang, Yu Zhang, Rui Sun, Zhigang Tian, Haoyu Sun Frontiers in Immunology.2023;[Epub] CrossRef
NK cell defects: implication in acute myeloid leukemia Selma Z. D’Silva, Meenakshi Singh, Andrea S. Pinto Frontiers in Immunology.2023;[Epub] CrossRef
Understanding NK cell biology for harnessing NK cell therapies: targeting cancer and beyond Eunju Shin, Seong Ho Bak, Taeho Park, Jin Woo Kim, Suk-Ran Yoon, Haiyoung Jung, Ji-Yoon Noh Frontiers in Immunology.2023;[Epub] CrossRef
Natural Killer Cell Engagers (NKCEs): a new frontier in cancer immunotherapy Minchuan Zhang, Kong-Peng Lam, Shengli Xu Frontiers in Immunology.2023;[Epub] CrossRef
Principles and current clinical landscape of NK cell engaging bispecific antibody against cancer Tian Huan, Bugao Guan, Hongbo Li, Xiu Tu, Chi Zhang, Bin Tang Human Vaccines & Immunotherapeutics.2023;[Epub] CrossRef
Emerging new cell therapies/immune therapies in B-cell non-Hodgkin's lymphoma Titas Banerjee, Anusha Vallurupalli Current Problems in Cancer.2022; 46(1): 100825. CrossRef
The use of supercytokines, immunocytokines, engager cytokines, and other synthetic cytokines in immunotherapy Xiaohu Zheng, Yaqi Wu, Jiacheng Bi, Yingying Huang, Ying Cheng, Yangyang Li, Yuwei Wu, Guoshuai Cao, Zhigang Tian Cellular & Molecular Immunology.2022; 19(2): 192. CrossRef
Engineering interferons and interleukins for cancer immunotherapy Patrick G. Holder, Shion A. Lim, Christine S. Huang, Preeti Sharma, Yavuz S. Dagdas, Beyza Bulutoglu, Jonathan T. Sockolosky Advanced Drug Delivery Reviews.2022; 182: 114112. CrossRef
iPSC-Derived Natural Killer Cell Therapies - Expansion and Targeting Benjamin H. Goldenson, Pooja Hor, Dan S. Kaufman Frontiers in Immunology.2022;[Epub] CrossRef
Natural killer cell dysfunction in cancer and new strategies to utilize NK cell potential for cancer immunotherapy Wanze Zhang, Zhenghua Zhao, Fan Li Molecular Immunology.2022; 144: 58. CrossRef
Development of Bispecific Antibody for Cancer Immunotherapy: Focus on T Cell Engaging Antibody Dain Moon, Nara Tae, Yunji Park, Seung-Woo Lee, Dae Hee Kim Immune Network.2022;[Epub] CrossRef
Harnessing natural killer cells for cancer immunotherapy: dispatching the first responders Nicholas A. Maskalenko, Dmitry Zhigarev, Kerry S. Campbell Nature Reviews Drug Discovery.2022; 21(8): 559. CrossRef
Innovative Strategies to Improve the Clinical Application of NK Cell-Based Immunotherapy Mubin Tarannum, Rizwan Romee, Roman M. Shapiro Frontiers in Immunology.2022;[Epub] CrossRef
Bispecific antibodies for immune cell retargeting against cancer Rebecca P Chen, Kenta Shinoda, Pragya Rampuria, Fang Jin, Tin Bartholomew, Chunxia Zhao, Fan Yang, Javier Chaparro-Riggers Expert Opinion on Biological Therapy.2022; 22(8): 965. CrossRef
The Dynamic Role of NK Cells in Liver Cancers: Role in HCC and HBV Associated HCC and Its Therapeutic Implications Muhammad Sajid, Lianxin Liu, Cheng Sun Frontiers in Immunology.2022;[Epub] CrossRef
Emerging Trends in Immunotherapy for Cancer Alok K. Mishra, Amjad Ali, Shubham Dutta, Shahid Banday, Sunil K. Malonia Diseases.2022; 10(3): 60. CrossRef
Drug-Tolerant Persister Cells in Cancer Therapy Resistance Pavan Kumar Dhanyamraju, Todd D. Schell, Shantu Amin, Gavin P. Robertson Cancer Research.2022; 82(14): 2503. CrossRef
Natural killer cells in antitumour adoptive cell immunotherapy Tamara J. Laskowski, Alexander Biederstädt, Katayoun Rezvani Nature Reviews Cancer.2022; 22(10): 557. CrossRef
TEM8 Tri-specific Killer Engager binds both tumor and tumor stroma to specifically engage natural killer cell anti-tumor activity Michael F Kaminski, Laura Bendzick, Rachel Hopps, Marissa Kauffman, Behiye Kodal, Yvette Soignier, Peter Hinderlie, Joshua T Walker, Todd R Lenvik, Melissa A Geller, Jeffrey S Miller, Martin Felices Journal for ImmunoTherapy of Cancer.2022; 10(9): e004725. CrossRef
The soldiers needed to be awakened: Tumor-infiltrating immune cells Wang Yaping, Wang Zhe, Chu Zhuling, Li Ruolei, Fan Pengyu, Guo Lili, Ji Cheng, Zhang Bo, Liu Liuyin, Hou Guangdong, Wang Yaoling, Hou Niuniu, Ling Rui Frontiers in Genetics.2022;[Epub] CrossRef
Gene-edited and CAR-NK cells: Opportunities and challenges with engineering of NK cells for immunotherapy Xinyu Wu, Sandro Matosevic Molecular Therapy - Oncolytics.2022; 27: 224. CrossRef
Multiplexed engineering and precision gene editing in cellular immunotherapy Alexander Biederstädt, Gohar Shahwar Manzar, May Daher Frontiers in Immunology.2022;[Epub] CrossRef
NK cells and ILCs in tumor immunotherapy Simona Sivori, Daniela Pende, Linda Quatrini, Gabriella Pietra, Mariella Della Chiesa, Paola Vacca, Nicola Tumino, Francesca Moretta, Maria Cristina Mingari, Franco Locatelli, Lorenzo Moretta Molecular Aspects of Medicine.2021; 80: 100870. CrossRef
Mechanisms of NK cell dysfunction in the tumor microenvironment and current clinical approaches to harness NK cell potential for immunotherapy Raynier Devillier, Anne-Sophie Chrétien, Thomas Pagliardini, Nassim Salem, Didier Blaise, Daniel Olive Journal of Leukocyte Biology.2021; 109(6): 1071. CrossRef
Cancer stem cells and strategies for targeted drug delivery Jin Cao, Shubhmita Bhatnagar, Jiawei Wang, Xueyong Qi, Swayam Prabha, Jayanth Panyam Drug Delivery and Translational Research.2021; 11(5): 1779. CrossRef
The role of natural killer cell in gastrointestinal cancer: killer or helper Feixue Wang, Jennie Ka Ching Lau, Jun Yu Oncogene.2021; 40(4): 717. CrossRef
Next-Generation Immunotherapies to Improve Anticancer Immunity Yaoyao Shi, Katarzyna Tomczak, June Li, Joshua K. Ochieng, Younghee Lee, Cara Haymaker Frontiers in Pharmacology.2021;[Epub] CrossRef
Engineered Multifunctional Nano‐ and Biological Materials for Cancer Immunotherapy Anthony Brouillard, Nilesh Deshpande, Ashish A. Kulkarni Advanced Healthcare Materials.2021;[Epub] CrossRef
Harnessing CD16-Mediated NK Cell Functions to Enhance Therapeutic Efficacy of Tumor-Targeting mAbs Cristina Capuano, Chiara Pighi, Simone Battella, Davide De Federicis, Ricciarda Galandrini, Gabriella Palmieri Cancers.2021; 13(10): 2500. CrossRef
Natural killer cell engagers in cancer immunotherapy: Next generation of immuno‐oncology treatments Olivier Demaria, Laurent Gauthier, Guilhaume Debroas, Eric Vivier European Journal of Immunology.2021; 51(8): 1934. CrossRef
Bispecific Antibodies: A Smart Arsenal for Cancer Immunotherapies Gihoon You, Jonghwa Won, Yangsoon Lee, Dain Moon, Yunji Park, Sang Hoon Lee, Seung-Woo Lee Vaccines.2021; 9(7): 724. CrossRef
Bi-specific and Tri-specific NK Cell Engagers: The New Avenue of Targeted NK Cell Immunotherapy Shee Kwan Phung, Jeffrey S. Miller, Martin Felices Molecular Diagnosis & Therapy.2021; 25(5): 577. CrossRef
Challenges and Recent Advances in NK Cell-Targeted Immunotherapies in Solid Tumors Guangyu Lian, Thomas Shiu-Kwong Mak, Xueqing Yu, Hui-Yao Lan International Journal of Molecular Sciences.2021; 23(1): 164. CrossRef
Targeting Natural Killer Cells for Tumor Immunotherapy Cai Zhang, Yuan Hu, Chongdeng Shi Frontiers in Immunology.2020;[Epub] CrossRef
Unleashing Natural Killer Cells in the Tumor Microenvironment–The Next Generation of Immunotherapy? Aviad Ben-Shmuel, Guy Biber, Mira Barda-Saad Frontiers in Immunology.2020;[Epub] CrossRef
Boosting Cytotoxic Antibodies against Cancer Laurent Gauthier, Eric Vivier Cell.2020; 180(5): 822. CrossRef
Bispecific, T-Cell-Recruiting Antibodies in B-Cell Malignancies Margaux Lejeune, Murat Cem Köse, Elodie Duray, Hermann Einsele, Yves Beguin, Jo Caers Frontiers in Immunology.2020;[Epub] CrossRef
NK-Cell-Mediated Targeting of Various Solid Tumors Using a B7-H3 Tri-Specific Killer Engager In Vitro and In Vivo Daniel A. Vallera, Soldano Ferrone, Behiye Kodal, Peter Hinderlie, Laura Bendzick, Brianna Ettestad, Caroline Hallstrom, Nicholas A. Zorko, Arpit Rao, Naomi Fujioka, Charles J. Ryan, Melissa A. Geller, Jeffrey S. Miller, Martin Felices Cancers.2020; 12(9): 2659. CrossRef
Exploiting NK Cell Surveillance Pathways for Cancer Therapy Alexander Barrow, Marco Colonna Cancers.2019; 11(1): 55. CrossRef
Overcoming Resistance to Natural Killer Cell Based Immunotherapies for Solid Tumors Gaurav Nayyar, Yaya Chu, Mitchell S. Cairo Frontiers in Oncology.2019;[Epub] CrossRef
Natural Killer Cells in Cancer Immunotherapy Jeffrey S. Miller, Lewis L. Lanier Annual Review of Cancer Biology.2019; 3(1): 77. CrossRef
Novel CD19-targeted TriKE restores NK cell function and proliferative capacity in CLL Martin Felices, Behiye Kodal, Peter Hinderlie, Michael F. Kaminski, Sarah Cooley, Daniel J. Weisdorf, Daniel A. Vallera, Jeffrey S. Miller, Veronika Bachanova Blood Advances.2019; 3(6): 897. CrossRef
Concise Review: Targeting Cancer Stem Cells and Their Supporting Niche Using Oncolytic Viruses Mathieu J.F. Crupi, John C. Bell, Ragunath Singaravelu Stem Cells.2019; 37(6): 716. CrossRef
Redirected optimized cell killing (ROCK®): A highly versatile multispecific fit-for-purpose antibody platform for engaging innate immunity Kristina Ellwanger, Uwe Reusch, Ivica Fucek, Susanne Wingert, Thorsten Ross, Thomas Müller, Ute Schniegler-Mattox, Torsten Haneke, Erich Rajkovic, Joachim Koch, Martin Treder, Michael Tesar mAbs.2019; 11(5): 899. CrossRef
T Cell–Activating Bispecific Antibodies in Cancer Therapy Asaad Trabolsi, Artavazd Arumov, Jonathan H Schatz The Journal of Immunology.2019; 203(3): 585. CrossRef
Les anticorps etscaffoldbispécifiques, des médicaments innovants en oncologie impliquant le ciblage des cellules immunitaires Patrick Chames, Thierry Wurch médecine/sciences.2019; 35(12): 1072. CrossRef
Emerging trends in bispecific antibody and scaffold protein therapeutics Petra Verdino, Shane Atwell, Stephen J Demarest Current Opinion in Chemical Engineering.2018; 19: 107. CrossRef
Strategies to activate NK cells to prevent relapse and induce remission following hematopoietic stem cell transplantation Sarah Cooley, Peter Parham, Jeffrey S. Miller Blood.2018; 131(10): 1053. CrossRef
Bispecific antibodies in cancer immunotherapy Eva Dahlén, Niina Veitonmäki, Per Norlén Therapeutic Advances in Vaccines and Immunotherapy.2018; 6(1): 3. CrossRef
Through the barricades: overcoming the barriers to effective antibody-based cancer therapeutics Martin Dalziel, Stephen A Beers, Mark S Cragg, Max Crispin Glycobiology.2018; 28(9): 697. CrossRef
The role of CD133 in cancer: a concise review Paige M. Glumac, Aaron M. LeBeau Clinical and Translational Medicine.2018;[Epub] CrossRef
Advances and challenges in therapeutic monoclonal antibodies drug development Mariana Lopes dos Santos, Wagner Quintilio, Tania Maria Manieri, Lilian Rumi Tsuruta, Ana Maria Moro Brazilian Journal of Pharmaceutical Sciences.2018;[Epub] CrossRef
Redirecting T cells to hematological malignancies with bispecific antibodies Mireya Paulina Velasquez, Challice L. Bonifant, Stephen Gottschalk Blood.2018; 131(1): 30. CrossRef
Developmental and Functional Control of Natural Killer Cells by Cytokines Yang Wu, Zhigang Tian, Haiming Wei Frontiers in Immunology.2017;[Epub] CrossRef
Natural killer cells unleashed: Checkpoint receptor blockade and BiKE/TriKE utilization in NK-mediated anti-tumor immunotherapy Zachary B. Davis, Daniel A. Vallera, Jeffrey S. Miller, Martin Felices Seminars in Immunology.2017; 31: 64. CrossRef
Ivan Ho Yuen Pun, Dessy Chan, Sau Hing Chan, Po Yee Chung, Yuan Yuan Zhou, Simon Law, Alfred King Yin Lam, Chung Hin Chui, Albert Sun Chi Chan, Kim Hung Lam, Johnny Cheuk On Tang
Cancer Res Treat. 2017;49(1):219-229. Published online July 18, 2016
Purpose
83b1 is a novel quinoline derivative that has been shown to inhibit cancer growth in human esophageal squamous cell carcinoma (ESCC). This study was conducted to comprehensively evaluate the cytotoxic effects of 83b1 on a series of ESCC cell lines and investigate the mechanisms by which 83b1 suppresses cancer growth based on molecular docking analysis.
Materials and Methods
A series of ESCC and nontumor immortalized cell lines were exposed to 83b1 and cisplatin (CDDP) in a dose-dependent manner, and the cytotoxicity was examined by a MTS assay kit. Prediction of the molecular targets of 83b1 was conducted by molecular docking analysis. Expression of cyclooxygenase 2 (COX-2) mRNA and COX-2–derived prostaglandin E2 (PGE2) were measured by quantitative real-time polymerase chain reaction and enzymelinked immuno-sorbent assay, respectively. In vivo anti-tumor effect was determined using a nude mice xenografted model transplanted with an ESCC cell line, KYSE-450.
Results
83b1 showed the significant anti-cancer effects on all ESCC cell lines compared to CDDP; however, 83b1 revealed much lower toxic effects on non-tumor cell lines than CDDP. The predicted molecular target of 83b1 is peroxisome proliferator-activated receptor delta (PPARδ), which is a widely known oncoprotein. Additionally the expression of COX-2 mRNA and COX-2–derived PGE2 were down-regulated by 83b1 in a dose-dependent manner in ESCC cell lines. Furthermore, 83b1 was shown to significantly reduce the tumor size in nude mice xenograft.
Conclusion
The results of this study suggest that the potential anti-cancer effects of 83b1 on human esophageal cancers occur through the possible oncotarget, PPARδ, and down-regulation of the cancer related genes and molecules.
Citations
Citations to this article as recorded by
Arachidonic acid metabolism as a novel pathogenic factor in gastrointestinal cancers Weiqin Lu, Aihemaitijiang Aihaiti, Paziliya Abudukeranmu, Yajun Liu, Huihui Gao Molecular and Cellular Biochemistry.2025; 480(2): 1225. CrossRef
Synthesis and biological evaluation of novel 2-morpholino-4-anilinoquinoline derivatives as antitumor agents against HepG2 cell line Ahmed Al-Sheikh, Malak A. Jaber, Hana'a Khalaf, Nour AlKhawaja, Duaa Abuarqoub RSC Advances.2024; 14(5): 3304. CrossRef
Natural alkaloids as potential treatments for esophageal squamous-cell carcinoma: A comprehensive review Eugene Jamot Ndebia, Gabriel Tchuente Kamsu Gastroenterology & Endoscopy.2024; 2(3): 131. CrossRef
Downregulation of chemokine (C‑C motif) ligand 5 induced by a novel 8‑hydroxyquinoline derivative (91b1) suppresses tumor invasiveness in esophageal carcinoma Johnny Tang, Dessy Chan, Po-Yee Chung, Yijiang Liu, Alfred Lam, Simon Law, Wolin Huang, Albert Chan, Kim-Hung Lam, Yuanyuan Zhou International Journal of Molecular Medicine.2024;[Epub] CrossRef
Peroxisome proliferator-activated receptors regulate the progression and treatment of gastrointestinal cancers Min Zhang, Shujie He Frontiers in Pharmacology.2023;[Epub] CrossRef
Roles of Nuclear Receptors in Esophageal Cancer Lihao Deng, Jiaxuan Liu, Wei-Dong Chen, Yan-Dong Wang Current Pharmaceutical Biotechnology.2023; 24(12): 1489. CrossRef
Synthesis and biological activities of new phthalimide and thiazolidine derivatives Flaviana A. Santos, Marcel L. Almeida, Vitor A. S. Silva, Douglas C. F. Viana, Michelly C. Pereira, André S. L. Lucena, Maira G. R. Pitta, Marina R. Galdino-Pitta, Moacyr J. B. de Melo Rêgo, Ivan da Rocha Pitta Medicinal Chemistry Research.2022; 31(1): 108. CrossRef
Role of Prostaglandin E2 in the Progression of Gastrointestinal Cancer David Jay Wilson, Raymond N. DuBois Cancer Prevention Research.2022; 15(6): 355. CrossRef
The Anticancer Effect of a Novel Quinoline Derivative 91b1 through Downregulation of Lumican Yuanyuan Zhou, Zhongguo Zhou, Dessy Chan, Po yee Chung, Yongqi Wang, Albert Sun chi Chan, Simon Law, Kim hung Lam, Johnny Cheuk On Tang International Journal of Molecular Sciences.2022; 23(21): 13181. CrossRef
Feasibility of Repurposing Clioquinol for Cancer Therapy Raheel Khan, Harras Khan, Yassen Abdullah, Q. Ping Dou Recent Patents on Anti-Cancer Drug Discovery.2020; 15(1): 14. CrossRef
DFIQ, a Novel Quinoline Derivative, Shows Anticancer Potential by Inducing Apoptosis and Autophagy in NSCLC Cell and In Vivo Zebrafish Xenograft Models Hurng-Wern Huang, Yung-Ding Bow, Chia-Yih Wang, Yen-Chun Chen, Pei-Rong Fu, Kuo-Feng Chang, Tso-Wen Wang, Chih-Hua Tseng, Yeh-Long Chen, Chien-Chih Chiu Cancers.2020; 12(5): 1348. CrossRef
Reverse Screening Methods to Search for the Protein Targets of Chemopreventive Compounds Hongbin Huang, Guigui Zhang, Yuquan Zhou, Chenru Lin, Suling Chen, Yutong Lin, Shangkang Mai, Zunnan Huang Frontiers in Chemistry.2018;[Epub] CrossRef
The roles of the COX2/PGE2/EP axis in therapeutic resistance Dali Tong, Qiuli Liu, Lin-ang Wang, Qiubo Xie, Jian Pang, Yiqiang Huang, Luofu Wang, Gaolei Liu, Dianzheng Zhang, Weihua Lan, Jun Jiang Cancer and Metastasis Reviews.2018; 37(2-3): 355. CrossRef
Expression of Insulin-Like Growth Factor Binding Protein-5 (IGFBP5) Reverses Cisplatin-Resistance in Esophageal Carcinoma Dessy Chan, Yuanyuan Zhou, Chung Hin Chui, Kim Hung Lam, Simon Law, Albert Sun-chi Chan, Xingshu Li, Alfred King-yin Lam, Johnny Cheuk On Tang Cells.2018; 7(10): 143. CrossRef
Targeting DNA Binding for NF-κB as an Anticancer Approach in Hepatocellular Carcinoma Po Chung, Pik Lam, Yuanyuan Zhou, Jessica Gasparello, Alessia Finotti, Adriana Chilin, Giovanni Marzaro, Roberto Gambari, Zhaoxiang Bian, Wai Kwok, Wai Wong, Xi Wang, Alfred Lam, Albert Chan, Xingshu Li, Jessica Ma, Chung Chui, Kim Lam, Johnny Tang Cells.2018; 7(10): 177. CrossRef
PURPOSE Platinum coordination complex (cisplatin) has been currently used as one of the most effective compound in the treatment of various solid tumors. However, its use has been limited by severe side effects such as renal toxicity. Our platinum-based drug discovery program has been aimed at developing drugs capable of diminishing toxicity and selective cytotoxicity. MATERIALS AND METHODS A new series of highly water soluble platinum (II) complexes Pt (II) [1,3-Bis (phenylthio) propane) (trans-l-1,2-diaminocyclohexane) (PC-1) and Pt (II) [1,3-Bis-(phenylthio) (propane)]-1,2-diaminocyclohexane (PC-2) were synthesized and characterized by their elemental analysis and by various spectroscopic techniques [infrared (.IR), ""C-nuclear magnetic resonance (NMR)]. In vitro antitumor activity and nephrotoxi -cities of new Pt (II) complexes were tested against MKN-45 human gastric cancer cell- lines and normal kidney cells using colorimetric MTT[3-(4,5-dimethyl thiazol-2-yl)-2,5- diphenyl tetrazolium bromide] assay for cell survival and proliferation. RESULTS PC-1 showed activity against MKN-45/P and MKN-45/CDDP human gastric adenocarcinoma cells, and the antitumor activity of this compound was comparable or superior to that of PC-2 and cisplatin. The nephrotoxicity of PC-1 and PC-2 were found quite less than that of cisplatin using MTT, [H] thymidine uptake and glucose consumption tests in rabbit proximal tubule cells, human kidney cortical cells and human renal cortical tissues. CONCLUSION Based on these results, this novel platinum (II) complex compound (PC-1) represent a valuable lead in the development of a new anticancer chemotherapeutic agent capable of improving antitumor activity and low nephrotoxicity.
PURPOSE The goal of this study is to understand the activation processes that take place within the liposomal formulation of lipophilic diaminocyclohexane platinum (DACH-Pt) complexes, to identify the activated species of this class of compounds, and to use that information to develop a reproducible liposomal formulation of DACH-Pt complexes. MATERIALS AND METHODS Liposomal DACH-Pt complexes were prepared by lyophilization-rehydration method using PC, PG and PA. Their intraliposomal stability and biological activity were determined by HPLC and in vitro/in vivo experiments. RESULTS DACH-Pt complexes in a liposomal formulation have shown significant promise in preclinical studies and clinical phase I, II trials. Interestingly, they are prodrugs which converts into one or more undetennined activated platinum species within the liposomes ex vivo. Our studies have shown that the stability of liposomal DACH-Pt complexes is inversely related with the antitumor activity of those complexes. The configuratian of leaving group in the complexes and pH of the liposome suspension affect significantly the degradation/activation process that takes place within the liposomes. DACH-Pt complexes with linear (L10) leaving groups are more stable than complexes with branched ones (B10 and NDDP), but also significantly less potent. The presence of PG and PA in the liposome is a prerequisite for the degradation/activation process of DACH-Pt complexes. As PG and PA formulation gave more dramatic changes of the original complexes than PC alone due to lower pH, the cytotoxicity and antitumor activity at those fonnulations increased against PC alone. DACH-Pt complexes are very stable in liposomes containing PC alone but inactive in vitro/in vivo experiments. CONCLUSION These results also support that the active species produced within the liposomal DACH-Pt complexes is DACH-Pt-Cl2.
PURPOSE Cytotoxicity of the bile acids on colon cancer cell lines was studied to know which bile acid was most cytotoxic to colonic mucosal epithelium. We performed agarose gel electrophoresis whether this toxicity was caused by detergent effect of the bile acids or by apoptotic pathway. MATERIALS AND METHODS HT29, LoVo, SW620 colon cancer cell lines were exposed to lithocholate, cholate, deoxycholate and chenodeoxycholate with 50, 100, 150, 200, 250, 300 pM as final concentration in DMEM culture media for short time (for 2 hours) and for long time (for 5 days). Agarose gel electrophoresis was performed on each colon cancer cell lines (HT29, LoVo, SW620, SW480) after 1, 2, 3, 4, 5 days exposure to deoxycholate with 150 pM concentration to detect intemucleosomal fragmentation. RESULTS There was no toxicity after short time exposure in all bile acids concentration and in all colon cancer cell lines. Of the bile acids, deoxycholate was most toxic for all colon cancer cell lines. And DNA fragmentation was noticed after 2 days exposure with deoxycholate. Only LoVo cell line showed apoptotic DNA pattern after 4 days of exposure with deoxycholate. CONCLUSION Bile acids (especially deoxycholate) are suggested to be possible agents to cause apoptosis in colonic mucosal epithelium.
PURPOSE The anti-tumor effect of the complex of acriflavine and guanosine (AG60) was investigated. MATERIALS AND METHODS In vitro cytotoxicity of AG60 was measured using SRB assay, and in vivo antitumor activity of AG60 was examined in CDF1 mice intraperitoneally inoculated with the P388 leukemic cells and in ICR mice inguinally implanted with S-180 cells. Tumor size and mean survival time were determined. RESULTS AG60 and acriflavine showed strong anti-tumor effect in vitro on lung cancer (A549), renal cancer (UO-31) and colon cancer (COLO205) cells. However, AG60 did not show the cytotoxicity against normal cell line, 3T3. The range of the IC50 of AG60 to the various tumor cell lines was 0.09 microgram/ml through 1.94 microgram/ml. The treatment of ascitic tumor bearing CDF1 mice with AG60 resulted in over 160% increases in the mean survival time. The most effective dose of AG60 was 30 mg/kg body weight in tumor implanted mice. In solid tumor bearing ICR mice tumor growth and progression were suppressed in response to the different doses at 30 days; 69.8% suppression of tumor size in response to acriflavine, 16.0% to guanosine, 87.7% to AG60 and 78.5% to doxorubicin. In addition, 35% increases were observed in the means survival time of AG60 treated group compared with control group. CONCLUSION The prominant anti-tumor effects of AG60 shown in this report would represent the possibility of the clinical trials.
To investigate the possibility of increased cytotoxicity of anticancer drugs, we treated human leukemia cells with combinations of anticancer drugs and biological response modifiers (BRMs). Using the colorimetric [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] (MTT) assay, we evaluated the chemosensitivity of 8 anticancer drugs(vincristine, vinblastine, adria- mycin, cisplatin, etoposide, cytosine arabinoside, bleomycin, and cyclophosphamide), and the anticancer effects of 3 BRMs(interleukin-2, alpha interferon, and gamma interferon) combined with these anticancer drugs against human leukemia HL-60 and KG-1 cells. The results were as follows; in the chemosensitivity of 8 anticancer drugs, VCR, VBL, ADR, and CPDD were effective, while VP-16, ara-C, Bleo, and CYC produced less than 50% inhibition of HL-60 and KG-1 cells lines. Among 3 BRMs investigated, all of them showed less than 20% inhibition of KG-1 cell lines and none were effective against HL-60 cells. In the anticancer effects of 3 BRMs, all of them showed about 20% inhibitory effects against KG-1 cells, but there were not any effects against HL-60 cells. All of the anticancer drugs markedly increased cytotoxic effects when they were combined with BRM. Especially, the ID values of VCR, VBL, and ADR when combined with BRMs decreased to 67~3%. These results demonstrate that some BRMs can markedly increase the cytotoxicity of VCR, VBL, ADR, CPDD, VP-16, ara-C, Bleo, and CYC and suggest possible clinical usefulwess.
Although there is still controversy to the relationship between c-erbB-2 protein expression and prognosis of the gastric carcinoma, there were many reports about the poor prognosis with the expression of c-erbB-2 in gastric carcinoma. The mechanisms underlying this phenomenon are not known. However several possibilities such as acquired resistance to 5-Fluorouracil(5-FU) and resistance to the host immune system were suggested. So we performed this study to evaluate whether c-erbB-2 expression can alter the natural killeriNK) cell cytotoxic activity. Using single cell suspensions from primary gastric cancer tissues and malignant ascites due to stomach cancer, the immunohistochemical reactivity to c-cerB-2 protein was examined and we performed the tests for NK cell cytotoxic activity. The c-erbB-2(+) cancer cells were significantly more resistant to NK cell cytotoxic activity than c-erbB-2( ) cancer cells. However there was no significant difference in the resiatance to NK cell cytotoxic activity according to their immunohistochemical staining intensities. These results suggest that the resistance to the NK cel1 cytotoxicity may be one of the possi- ble mechanisms of the poorer prognosis of the c-erbB-2(+ ) gastric cancers.