1
|
Tanaka R, Yoshinouchi S, Karouji K, Tanaka Y, Tominari T, Hirata M, Matsumoto C, Itoh Y, Miyaura C, Inada M. A mouse model of lung cancer induced via intranasal injection for anticancer drug screening and evaluation of pathology. FEBS Open Bio 2022; 13:51-59. [PMID: 36102619 PMCID: PMC9810119 DOI: 10.1002/2211-5463.13486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 02/17/2022] [Accepted: 09/12/2022] [Indexed: 01/07/2023] Open
Abstract
The pathologies and lethality of lung cancers are associated with smoking, lifestyle, and genomic factors. Several experimental mouse models of lung cancer, including those induced via intrapulmonary injection and intratracheal injection, have been reported for evaluating the pharmacological effect of drugs. However, these models are not sufficient for evaluating the efficacy of drugs during screening, as these direct injection models ignore the native processes of cancer progression in vivo, resulting in the inadequate pathological formation of lung cancer. In the present study, we developed a novel intranasal injection model of lung cancer simulating the native lung cancer pathology for anticancer drug screening. A mouse lung cancer cell line (Lewis lung carcinoma; LCC) was intranasally injected into mouse lungs, and injected cell number-dependent cancer proliferation was apparent in both the left and right lungs. Human non-small-cell lung cancer (NCI-H460) cells were also intranasally injected into nude mice and similarly showed injected cell number-dependent cancer growth. For the pharmacological evaluation of cisplatin, two different treatment frequencies were tested four times per month and twice a month. The intranasal injection model confirmed that cisplatin suppressed lung cancer progression to a greater extent under the more frequent treatment condition. In conclusion, these results indicated that our intranasal injection model is a powerful tool for investigating lung cancer pathology and may aid in the development of new anti-lung cancer agents.
Collapse
Affiliation(s)
- Ryo Tanaka
- Cooperative Major of Advanced Health ScienceTokyo University of Agriculture and TechnologyJapan,Testing and Research LaboratoriesHAMLI Co., Ltd.IbarakiJapan
| | - Shosei Yoshinouchi
- Cooperative Major of Advanced Health ScienceTokyo University of Agriculture and TechnologyJapan
| | - Kento Karouji
- Department of Biotechnology and Life ScienceTokyo University of Agriculture and TechnologyJapan
| | - Yuki Tanaka
- Department of Biotechnology and Life ScienceTokyo University of Agriculture and TechnologyJapan
| | - Tsukasa Tominari
- Department of Biotechnology and Life ScienceTokyo University of Agriculture and TechnologyJapan
| | - Michiko Hirata
- Department of Biotechnology and Life ScienceTokyo University of Agriculture and TechnologyJapan
| | - Chiho Matsumoto
- Department of Biotechnology and Life ScienceTokyo University of Agriculture and TechnologyJapan
| | - Yoshifumi Itoh
- Institute of Global Innovation ResearchTokyo University of Agriculture and TechnologyJapan,Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal SciencesUniversity of OxfordUK
| | - Chisato Miyaura
- Cooperative Major of Advanced Health ScienceTokyo University of Agriculture and TechnologyJapan,Department of Biotechnology and Life ScienceTokyo University of Agriculture and TechnologyJapan,Institute of Global Innovation ResearchTokyo University of Agriculture and TechnologyJapan
| | - Masaki Inada
- Cooperative Major of Advanced Health ScienceTokyo University of Agriculture and TechnologyJapan,Department of Biotechnology and Life ScienceTokyo University of Agriculture and TechnologyJapan,Institute of Global Innovation ResearchTokyo University of Agriculture and TechnologyJapan
| |
Collapse
|
2
|
Ahmed Khalil A, Rauf A, Alhumaydhi FA, Aljohani ASM, Javed MS, Khan MA, Khan IA, El-Esawi MA, Bawazeer S, Bouyahya A, Rebezov M, Shariati MA, Thiruvengadam M. Recent Developments and Anticancer Therapeutics of Paclitaxel: An Update. Curr Pharm Des 2022; 28:3363-3373. [PMID: 36330627 DOI: 10.2174/1381612829666221102155212] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/19/2022] [Accepted: 09/26/2022] [Indexed: 11/06/2022]
Abstract
Plants are a source of diverse classes of secondary metabolites with anticancer properties. Paclitaxel (Taxol) is an anticancer drug isolated from various Taxus species and is used as a chemotherapeutic agent against various cancers. The biosynthesis of paclitaxel is a complex pathway, making its total chemical synthesis commercially non-viable; hence, alternative novel sources - like plant cell culture and heterologous expression systems, are being investigated to overcome this issue. Advancements in the field of genetic engineering, microbial fermentation engineering, and recombinant techniques have significantly increased the achievable yields of paclitaxel. Indeed, paclitaxel selectively targets microtubules and causes cell cycle arrest in the G2/M phase, inducing a cytotoxic effect in a concentration and time-dependent manner. Innovative drug delivery formulations, like the development of albumin-bound nanoparticles, nano-emulsions, nano-suspensions, liposomes, and polymeric micelles, have been applied to enhance the delivery of paclitaxel to tumor cells. This review focuses on the production, biosynthesis, mechanism of action, and anticancer effects of paclitaxel.
Collapse
Affiliation(s)
- Anees Ahmed Khalil
- University Institute of Diet and Nutritional Sciences, Faculty of Allied Health Sciences, The University of Lahore, Pakistan
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Anbar-23561, K.P.K, Pakistan
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Abdullah S M Aljohani
- Department of Veterinary Medicine, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Muhammad Sameem Javed
- Institute of Food Science and Nutrition, Bahauddin Zakariya University, Multan, Pakistan
| | | | - Imtiaz Ali Khan
- Department of Entomology, University of Peshawar, KP, Pakistan
| | - Mohamed A El-Esawi
- Botany Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Sami Bawazeer
- Department of Pharmacognosy, Faculty of Pharmacy, Umm Al-Qura University, Makkah, P.O. Box 42, Saudi Arabia
| | - Abdelhakim Bouyahya
- Department of Biology, Laboratory of Human Pathologies Biology, Faculty of Sciences, and Genomic Center of Human Pathologies, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, 10106 Morocco
| | - Maksim Rebezov
- V.M. Gorbatov Federal Research Center for Food Systems of Russian Academy of Sciences, Moscow, Russian Federation.,Prokhorov General Physics Institute, Russian Academy of Sciences, Moscow, Russian Federation
| | - Mohammad Ali Shariati
- K.G. Razumovsky Moscow State University of Technologies and management (the First Cossack University), Moscow, Russian Federation
| | - Muthu Thiruvengadam
- Department of Applied Bioscience, College of Life and Environmental Sciences, Konkuk University, Seoul 05029, South Korea.,Department of Microbiology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 600077, India
| |
Collapse
|
3
|
Milic M, Mondini M, Deutsch E. How to Improve SBRT Outcomes in NSCLC: From Pre-Clinical Modeling to Successful Clinical Translation. Cancers (Basel) 2022; 14:cancers14071705. [PMID: 35406477 PMCID: PMC8997119 DOI: 10.3390/cancers14071705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/14/2022] [Accepted: 03/22/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Despite major research and clinical efforts, lung cancer remains the leading cause of cancer-related death. Stereotactic body radiotherapy (SBRT) has emerged as a major treatment modality for lung cancer in the last decade. Additional research is needed to elucidate underlying mechanisms of resistance and to develop improved therapeutic strategies. Clinical progress relies on accurate preclinical modelling of human disease in order to yield clinically meaningful results; however, successful translation of pre-clinical research is still lagging behind. In this review, we summarize the major clinical developments of radiation therapy for non-small-cell lung cancer (NSCLC), and we discuss the pre-clinical research models at our disposal, highlighting ongoing translational challenges and future perspectives. Abstract Despite major research and clinical efforts, lung cancer remains the leading cause of cancer-related death. While the delivery of conformal radiotherapy and image guidance of stereotactic body radiotherapy (SBRT) have revolutionized the treatment of early-stage non-small-cell lung cancer (NSCLC), additional research is needed to elucidate underlying mechanisms of resistance and identify novel therapeutic combinations. Clinical progress relies on the successful translation of pre-clinical work, which so far has not always yielded expected results. Improved clinical modelling involves characterizing the preclinical models and selecting appropriate experimental designs that faithfully mimic precise clinical scenarios. Here, we review the current role of SBRT and the scope of pre-clinical armamentarium at our disposal to improve successful clinical translation of pre-clinical research in the radiation oncology of NSCLC.
Collapse
Affiliation(s)
- Marina Milic
- Gustave Roussy, Université Paris-Saclay, INSERM U1030, F-94805 Villejuif, France;
| | - Michele Mondini
- Gustave Roussy, Université Paris-Saclay, INSERM U1030, F-94805 Villejuif, France;
- Correspondence: (M.M.); (E.D.)
| | - Eric Deutsch
- Gustave Roussy, Université Paris-Saclay, INSERM U1030, F-94805 Villejuif, France;
- Gustave Roussy, Département d’Oncologie-Radiothérapie, F-94805 Villejuif, France
- Correspondence: (M.M.); (E.D.)
| |
Collapse
|
4
|
Kozlowski MR, Kozlowski RE. A novel, small peptide with activity against human pancreatic cancer. Am J Cancer Res 2020; 10:1356-1365. [PMID: 32509384 PMCID: PMC7269787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 04/30/2020] [Indexed: 06/11/2023] Open
Abstract
KTH-222 is a novel, 8-amino acid length peptide. It is derived from a motif identified in a group of peptides that are related to atrial natriuretic peptide and that are able to inhibit cancer cell growth. We report here that KTH-222 inhibits the attachment, proliferation, and development of an invasive morphology in cultured human pancreatic tumor cells (MIA PaCa-2 and HPAC). At a biochemical level, it inhibits tubulin polymerization which may underlie these cellular effects. We further report that KTH-222 reduces the rate of tumor growth and prolongs survival in mice implanted with MIA PaCa-2 cells. In this model system, KTH-222 is more effective than gemcitabine, a drug commonly used in the treatment of pancreatic cancer. Furthermore, KTH-222 does not decrease the rate of weight gain in the treated mice, suggesting the absence of gross toxicity. These activities of KTH-222 suggest that it may be useful in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Michael R Kozlowski
- Arizona College of Optometry, Midwestern UniversityGlendale, Arizona, United States of America
| | - Roni E Kozlowski
- Kalos Therapeutics IncorporatedPhoenix, Arizona, United States of America
| |
Collapse
|
5
|
Nerome K, Ito-Kureha T, Paganini T, Fukuda T, Igarashi Y, Ashitomi H, Ikematsu S, Yamamoto T. Potent and broad anticancer activities of leaf extracts from Melia azedarach L. of the subtropical Okinawa islands. Am J Cancer Res 2020; 10:581-594. [PMID: 32195029 PMCID: PMC7061759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 01/23/2020] [Indexed: 06/10/2023] Open
Abstract
Plant extracts have been traditionally used for various therapeutic applications. By conducting an initial screening of several subtropical plants, in this study, we evaluated the anticancer activities of Melia azedarach L. The extract from Melia azedarach L. leaves (MLE) show high cytotoxic effects on cancer cells and in vivo mouse and dog tumor models. During the initial screening, MLE showed strong antiproliferative activity against HT-29 colon, A549 lung, and MKN1 gastric cancer cells. In subsequent tests, using 39 human tumor cell lines, we confirmed the potent anticancer activities of MLE. The anticancer activity of MLE was also confirmed in vivo. MLE markedly inhibited the growth of transplanted gastric MKN1 cancer xenografts in mice. To elucidate the mechanism underlying the anticancer effects of MLE, MLE-treated MKN1 cells were observed using an electron microscope; MLE treatment induced autophagy. Furthermore, western blot analysis of proteins in lysates of MLE-treated cells revealed induction of light chain 3 (LC3)-II autophagosomal proteins. Thus, MLE appeared to suppress MKN1 cell proliferation by inducing autophagy. In addition, in the mouse macrophage cell line J774A.1, MLE treatment induced TNF-α production, which might play a role in tumor growth suppression in vivo. We also performed a preclinical evaluation of MLE treatment on dogs with various cancers in veterinary hospitals. Dogs with various types of cancers showed a mean recovery of 76% when treated with MLE. Finally, we tried to identify the active substances present in MLE. All the active fractions obtained by reverse-phase chromatography contained azedarachin B-related moieties, such as 3-deacetyl-12-hydroxy-amoorastatin, 12-hydroxy-amoorastatin, and 12-hydroxyamoorastaton. In conclusion, MLE contains substances with promising anticancer effects, suggesting their future use as safe and effective anticancer agents.
Collapse
Affiliation(s)
- Kuniaki Nerome
- The Institute of Biological Resources893-2, Nakayama, Nago-shi, Okinawa 905-0004, Japan
| | - Taku Ito-Kureha
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate UniversityOnna, Okinawa 904-0495, Japan
| | - Tiziana Paganini
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate UniversityOnna, Okinawa 904-0495, Japan
| | - Takao Fukuda
- Biotechnology Research Center and Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University5180 Kurokawa, Imizu, Toyama 939-0398, Japan
| | - Yasuhiro Igarashi
- Biotechnology Research Center and Department of Biotechnology, Faculty of Engineering, Toyama Prefectural University5180 Kurokawa, Imizu, Toyama 939-0398, Japan
| | - Hiroto Ashitomi
- National Institute of Technology, Okinawa CollegeHenoko 905, Nago-shi, Okinawa 905-2192, Japan
| | - Shinya Ikematsu
- National Institute of Technology, Okinawa CollegeHenoko 905, Nago-shi, Okinawa 905-2192, Japan
| | - Tadashi Yamamoto
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate UniversityOnna, Okinawa 904-0495, Japan
| |
Collapse
|
6
|
Li MY, Liu LZ, Li W, Ng CSH, Liu Y, Kong AWY, Zhao Z, Wang S, Qi H, Jia H, Yang S, Du J, Long X, Ho RLK, Chak ECW, Wan IYP, Mok TSK, Underwood MJ, Gali NK, Ning Z, Chen GG. Ambient fine particulate matter inhibits 15-lipoxygenases to promote lung carcinogenesis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:359. [PMID: 31420013 PMCID: PMC6697918 DOI: 10.1186/s13046-019-1380-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 08/12/2019] [Indexed: 12/21/2022]
Abstract
Background Epidemiological observations have demonstrated that ambient fine particulate matter with dp < 2.5 μm (PM2.5) as the major factor responsible for the increasing incidence of lung cancer in never-smokers. However, there are very limited experimental data to support the association of PM2.5 with lung carcinogenesis and to compare PM2.5 with smoking carcinogens. Methods To study whether PM2.5 can contribute to lung tumorigenesis in a way similar to smoking carcinogen 4-methylnitrosamino-l-3-pyridyl-butanone (NNK) via 15-lipoxygenases (15-LOXs) reduction, normal lung epithelial cells and cancer cells were treated with NNK or PM2.5 and then epigenetically and post-translationally examined the cellular and molecular profiles of the cells. The data were verified in lung cancer samples and a mouse lung tumor model. Results We found that similar to smoking carcinogen NNK, PM2.5 significantly enhanced cell proliferation, migration and invasion, but reduced the levels of 15-lipoxygenases-1 (15-LOX1) and 15-lipoxygenases-2 (15-LOX2), both of which were also obviously decreased in lung cancer tissues. 15-LOX1/15-LOX2 overexpression inhibited the oncogenic cell functions induced by PM2.5/NNK. The tumor formation and growth were significantly higher/faster in mice implanted with PM2.5- or NNK-treated NCI-H23 cells, accompanied with a reduction of 15-LOX1/15-LOX2. Moreover, 15-LOX1 expression was epigenetically regulated at methylation level by PM2.5/NNK, while both 15-LOX1 and 15-LOX2 could be significantly inhibited by a set of PM2.5/NNK-mediated microRNAs. Conclusion Collectively, PM2.5 can function as the smoking carcinogen NNK to induce lung tumorigenesis by inhibiting 15-LOX1/15-LOX2. Electronic supplementary material The online version of this article (10.1186/s13046-019-1380-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ming-Yue Li
- Department of Surgery, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T, Hong Kong. .,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, China.
| | - Li-Zhong Liu
- Faculty of Medicine, Shenzhen University Health Science Center, Shenzhen University, Shenzhen, China
| | - Wende Li
- Guangdong Key Laboratory of Laboratory Animal, Guangdong Laboratory Animals Monitoring Institute, Guangzhou, China
| | - Calvin S H Ng
- Department of Surgery, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T, Hong Kong
| | - Yi Liu
- Department of Surgery, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T, Hong Kong.,Guangdong Medical College, Zhangjiang, Guangdong, China
| | - Angel W Y Kong
- Department of Surgery, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T, Hong Kong
| | - Zhili Zhao
- Department of Surgery, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T, Hong Kong
| | - Shanshan Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T, Hong Kong
| | - Haolong Qi
- Department of Surgery, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T, Hong Kong
| | - Hao Jia
- Department of Surgery, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T, Hong Kong
| | - Shucai Yang
- Department of Clinical Laboratory, Pingshan District People's Hospital of Shenzhen, Shenzhen, China
| | - Jing Du
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Xiang Long
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Rocky L K Ho
- Department of Surgery, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T, Hong Kong
| | - Ernest C W Chak
- Department of Surgery, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T, Hong Kong
| | - Innes Y P Wan
- Department of Surgery, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T, Hong Kong
| | - Tony S K Mok
- Department of Clinical Oncology, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T, Hong Kong
| | - Malcolm J Underwood
- Department of Surgery, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T, Hong Kong
| | - Nirmal Kumar Gali
- Division of Environment and Sustainability, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong
| | - Zhi Ning
- Division of Environment and Sustainability, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong.
| | - George G Chen
- Department of Surgery, Head and Neck Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, N.T, Hong Kong. .,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, China.
| |
Collapse
|
7
|
Chronic Obstructive Pulmonary Disease and Lung Cancer: Underlying Pathophysiology and New Therapeutic Modalities. Drugs 2019; 78:1717-1740. [PMID: 30392114 DOI: 10.1007/s40265-018-1001-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) and lung cancer are major lung diseases affecting millions worldwide. Both diseases have links to cigarette smoking and exert a considerable societal burden. People suffering from COPD are at higher risk of developing lung cancer than those without, and are more susceptible to poor outcomes after diagnosis and treatment. Lung cancer and COPD are closely associated, possibly sharing common traits such as an underlying genetic predisposition, epithelial and endothelial cell plasticity, dysfunctional inflammatory mechanisms including the deposition of excessive extracellular matrix, angiogenesis, susceptibility to DNA damage and cellular mutagenesis. In fact, COPD could be the driving factor for lung cancer, providing a conducive environment that propagates its evolution. In the early stages of smoking, body defences provide a combative immune/oxidative response and DNA repair mechanisms are likely to subdue these changes to a certain extent; however, in patients with COPD with lung cancer the consequences could be devastating, potentially contributing to slower postoperative recovery after lung resection and increased resistance to radiotherapy and chemotherapy. Vital to the development of new-targeted therapies is an in-depth understanding of various molecular mechanisms that are associated with both pathologies. In this comprehensive review, we provide a detailed overview of possible underlying factors that link COPD and lung cancer, and current therapeutic advances from both human and preclinical animal models that can effectively mitigate this unholy relationship.
Collapse
|
8
|
Singh AP, Adrianzen Herrera D, Zhang Y, Perez-Soler R, Cheng H. Mouse models in squamous cell lung cancer: impact for drug discovery. Expert Opin Drug Discov 2018; 13:347-358. [PMID: 29394493 DOI: 10.1080/17460441.2018.1437137] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Squamous cell lung cancer (SQCLC) is the second most common subtype of non-small cell lung cancer (NSCLC) and has limited therapeutic options. Its development is likely a result of a multistep process in response to chronic tobacco exposure, involving sequential metaplasia, dysplasia and invasive carcinoma. Its complex genomic landscape has recently been revealed but no driver mutations have been validated that could lead to molecularly targeted therapy as have emerged in lung adenocarcinoma. Few preclinical murine models exist for testing and developing novel therapeutics in SQCLC. Areas covered: This review discusses the pathophysiology and molecular underpinnings of SQCLC that have limited the development of animal models. It then explores the advantages and limitations of a variety of existing mouse models and illustrates their potential application in drug discovery and chemoprevention. Expert opinion: There are several challenges in the development of mouse models for SQCLC, such as lack of validated driver genetic alterations, unclear cell of origin, and difficulty in reproducing the sophisticated tumor microenvironment of human disease. Nevertheless, several successful SQCLC murine models have emerged, especially Patient Derived Xenografts (PDXs) and Genetically Engineered Mouse Models (GEMMs). Continued efforts are needed to generate more SQCLC animal models to better understand its carcinogenesis and metastasis and to further test novel therapeutic strategies.
Collapse
Affiliation(s)
- Aditi P Singh
- a Department of Oncology , Montefiore Medical Center/Albert Einstein College of Medicine , Bronx , NY , USA
| | - Diego Adrianzen Herrera
- a Department of Oncology , Montefiore Medical Center/Albert Einstein College of Medicine , Bronx , NY , USA
| | - Yifei Zhang
- b Department of Medicine , Montefiore Medical Center/Albert Einstein College of Medicine , Bronx , NY , USA
| | - Roman Perez-Soler
- a Department of Oncology , Montefiore Medical Center/Albert Einstein College of Medicine , Bronx , NY , USA
| | - Haiying Cheng
- a Department of Oncology , Montefiore Medical Center/Albert Einstein College of Medicine , Bronx , NY , USA
| |
Collapse
|
9
|
Ohashi Y, Okamura M, Katayama R, Fang S, Tsutsui S, Akatsuka A, Shan M, Choi HW, Fujita N, Yoshimatsu K, Shiina I, Yamori T, Dan S. Targeting the Golgi apparatus to overcome acquired resistance of non-small cell lung cancer cells to EGFR tyrosine kinase inhibitors. Oncotarget 2017; 9:1641-1655. [PMID: 29416720 PMCID: PMC5788588 DOI: 10.18632/oncotarget.22895] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 11/17/2017] [Indexed: 11/25/2022] Open
Abstract
Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (EGFR-TKIs) were demonstrated to provide survival benefit in patients with non-small cell lung cancer (NSCLC) harboring activating mutations of EGFR; however, emergence of acquired resistance to EGFR-TKIs has been shown to cause poor outcome. To overcome the TKI resistance, drugs with different mode of action are required. We previously reported that M-COPA (2-methylcoprophilinamide), a Golgi disruptor, suppressed the growth of gastric cancers overexpressing receptor tyrosine kinases (RTKs) such as hepatocyte growth factor receptor (MET) via downregulating their cell surface expression. In this study, we examined the antitumor effect of M-COPA on NSCLC cells with TKI resistance. As a result, M-COPA effectively downregulated cell surface EGFR and its downstream signals, and finally exerted in vivo antitumor effect in NSCLC cells harboring secondary (T790M/del19) and tertiary (C797S/T790M/del19) mutated EGFR, which exhibit acquired resistance to first- and third generation EGFR-TKIs, respectively. M-COPA also downregulated MET expression potentially involved in the acquired resistance to EGFR-TKIs via bypassing the EGFR pathway blockade. These results provide the first evidence that targeting the Golgi apparatus might be a promising therapeutic strategy to overcome the vicious cycle of TKI resistance in EGFR-mutated NSCLC cells via downregulating cell surface RTK expression.
Collapse
Affiliation(s)
- Yoshimi Ohashi
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Mutsumi Okamura
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryohei Katayama
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Siyang Fang
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Saki Tsutsui
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Akinobu Akatsuka
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Mingde Shan
- Eisai AiM Institute, Eisai Inc., Andover, MA, USA
| | | | - Naoya Fujita
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | | | - Isamu Shiina
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science, Tokyo, Japan
| | - Takao Yamori
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.,Present address: Center for Product Evaluation, Pharmaceuticals and Medical Devices Agency, Tokyo, Japan
| | - Shingo Dan
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
10
|
Hureaux J, Lacoeuille F, Lagarce F, Rousselet MC, Contini A, Saulnier P, Benoit JP, Urban T. Absence of lung fibrosis after a single pulmonary delivery of lipid nanocapsules in rats. Int J Nanomedicine 2017; 12:8159-8170. [PMID: 29184405 PMCID: PMC5687496 DOI: 10.2147/ijn.s146740] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lipid nanocapsules (LNCs) are potential drug carriers for pulmonary delivery since they can be nebulized without any structural or functional changes, and the aerosols produced are highly compatible with pulmonary drug delivery in human beings. The alveolar surface tension, in vitro cytotoxicity, biodistribution and pulmonary toxicity in rats of a single endotracheal spray of LNCs or paclitaxel-loaded LNCs were studied. In vitro cytotoxicity of LNCs after a spray remained unchanged. Biodistribution study showed a homogeneous repartition in the lungs in rats with an improvement in lung retention of the radiolabeled tracer loaded in LNCs compared to the absence of LNCs with a lung half-time of 8.8±0.7 hours. Bronchoalveolar fluid analysis revealed transient 7-day alveolar inflammation, reaching a maximum between days 2 and 4, characterized by a peak of granulocytes at day 1 followed by a peak of lymphocytes at day 3. Alveolar protein levels were increased at days 3 and 7. Acute inflammation was increased with paclitaxel-loaded LNCs in comparison with blank LNCs but dropped out at day 7. No histological pulmonary lesion was observed at day 60. LNCs lowered surface tension to a greater degree than Curosurf® in a physicochemical model of the pulmonary alveolus. A single pulmonary delivery of LNCs induces a short-term alveolar inflammation with no residual lesions in rats at day 60. These data permit to start the study of LNCs in surfactant replacement therapy.
Collapse
Affiliation(s)
- José Hureaux
- Unité Micro et Nanomédecines Biomimétiques (MINT), Université d'Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire.,Université d'Angers, CHU, Pôle Hippocrate, Service de Pneumologie
| | - Franck Lacoeuille
- Université d'Angers, CHU, Pôle Signal Image Stérilisation, Service de Médecine nucléaire.,CRCINA, Université Nantes Université Angers
| | - Frédéric Lagarce
- Unité Micro et Nanomédecines Biomimétiques (MINT), Université d'Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire.,Université d'Angers, CHU, Pôle Hippocrate, Pharmacie
| | - Marie-Christine Rousselet
- Université d'Angers, CHU, Pôle de Biologie-Pathologie, Département de Cytologie et d'Histologie Pathologique
| | - Aurélien Contini
- Université d'Angers, CHU, Pôle Signal Image Stérilisation, Service de Médecine nucléaire.,CRCINA, Université Nantes Université Angers
| | - Patrick Saulnier
- Unité Micro et Nanomédecines Biomimétiques (MINT), Université d'Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire.,Université d'Angers, CHU, Service la Recherche Clinique et Innovation, Angers, France
| | - Jean-Pierre Benoit
- Unité Micro et Nanomédecines Biomimétiques (MINT), Université d'Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire.,Université d'Angers, CHU, Pôle Hippocrate, Pharmacie
| | - Thierry Urban
- Unité Micro et Nanomédecines Biomimétiques (MINT), Université d'Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire.,Université d'Angers, CHU, Pôle Hippocrate, Service de Pneumologie
| |
Collapse
|
11
|
Konaka K, Moriyama K, Sakurada T, Okada N, Imanishi M, Zamami Y, Kawazoe K, Fushitani S, Ishizawa K. Kamishoyosan and Shakuyakukanzoto promote recovery from paclitaxel-induced neurite retraction in PC12 cells. J Pharm Health Care Sci 2017; 3:20. [PMID: 28748102 PMCID: PMC5521132 DOI: 10.1186/s40780-017-0090-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 07/14/2017] [Indexed: 12/01/2022] Open
Abstract
Background In chemotherapy, the full round of treatment must be completed as scheduled to achieve the strongest therapeutic effect. However, peripheral neuropathy, a severe side effect of the chemotherapeutic agent paclitaxel, can force the premature discontinuation of treatment. As some kampo practitioners have suggested that it may be possible to counteract such side effects, we analyzed the effects of Kamishoyosan, Shakuyakukanzoto, and Goshajinkigan in an in vitro model of paclitaxel-induced peripheral neuropathy. Methods Paclitaxel-treated PC12 cells were assessed for neurite length and performed Western blot analysis for growth-associated protein-43 (GAP-43) and light neurofilament protein (NF-L) levels in the presence of nerve growth factor (NGF); they were re-assessed, with additional testing for acetylcholinesterase levels, after application of one of the kampo. We also compared phosphorylation of extracellular signal-regulated kinase (Erk)1/2 and Akt via Western blot analysis. About effect of kampo to anticancer efficacy, we confirmed cell cytotoxicity in A549 cells using MTT assay. Results Addition of Kamishoyosan or Shakuyakukanzoto, but not Goshajinkigan, significantly improved neurite length and GAP-43 and NF-L levels from paclitaxel-treated PC12 cells, relative to those of only NGF-treated PC12 cells. The promoting effect of Kamishoyosan and Shakuyakukanzoto in neurite outgrowth is confirmed when NGF promoted neurite outgrowth, and it was inhibited partially when Erk1/2 and Akt were blocked by Erk1/2 inhibitor or Akt inhibitor alone. Furthermore, neurite outgrowth induced by TJ24 and TJ68 was inhibited more strongly when Erk1/2 inhibitor and Akt inhibitor were treated at the same time. NGF with Kamishoyosan or Shakuyakukanzoto promoted the proportion of phosphorylated Erk1/2 and phosphorylated Akt compare with NGF only. On the other hand, Kamishoyosan or Shakuyakukanzoto didn’t influence cytotoxicity of paclitaxel in A549 cells. Conclusions Kamishoyosan or Shakuyakukanzoto promotes neurite outgrowth with NGF via increasing the proportion of phosphorylated Erk1/2 and phosphorylated Akt in PC12 cells. The effect applies to recovery from paclitaxel-induced axonal involvement and might promote recovery from paclitaxel-induced neuropathy without influence of anticancer effect of paclitaxel.
Collapse
Affiliation(s)
- Ken Konaka
- Department of Pharmacy, Tokushima Municipal Hospital, 2-34 Kitajyosanjima, Tokushima, 770-0812 Japan
| | - Kota Moriyama
- Department of Pharmacy, Shimane University Hospital, 89-1 Enya, Izumo, 693-0021 Japan
| | - Takumi Sakurada
- Department of Pharmacy, Tokushima University Hospital, 2-50-1 Kuramoto, Tokushima, 770-8503 Japan
| | - Naoto Okada
- Department of Pharmacy, Tokushima University Hospital, 2-50-1 Kuramoto, Tokushima, 770-8503 Japan
| | - Masaki Imanishi
- Department of Pharmacy, Tokushima University Hospital, 2-50-1 Kuramoto, Tokushima, 770-8503 Japan
| | - Yoshito Zamami
- Department of Pharmacy, Tokushima University Hospital, 2-50-1 Kuramoto, Tokushima, 770-8503 Japan.,Department of Clinical Pharmacology and Therapeutics, Institute of Biomedical Sciences, Tokushima University Graduate School, 2-50-1 Kuramoto, Tokushima, 770-8503 Japan
| | - Kazuyoshi Kawazoe
- Department of Clinical Pharmacy Practice Pedagogy Institute of Biomedical Sciences, Tokushima University Graduate School, 1-78-1 Shomachi, Tokushima, 770-8505 Japan
| | - Shuji Fushitani
- Department of Pharmacy, Tokushima Municipal Hospital, 2-34 Kitajyosanjima, Tokushima, 770-0812 Japan
| | - Keisuke Ishizawa
- Department of Pharmacy, Tokushima University Hospital, 2-50-1 Kuramoto, Tokushima, 770-8503 Japan.,Department of Clinical Pharmacology and Therapeutics, Institute of Biomedical Sciences, Tokushima University Graduate School, 2-50-1 Kuramoto, Tokushima, 770-8503 Japan
| |
Collapse
|
12
|
Pan A, Zhang H, Li Y, Lin TY, Wang F, Lee J, Cheng M, Dall'Era M, Li T, deVere White R, Pan CX, Lam KS. Disulfide-crosslinked nanomicelles confer cancer-specific drug delivery and improve efficacy of paclitaxel in bladder cancer. NANOTECHNOLOGY 2016; 27:425103. [PMID: 27640312 PMCID: PMC6522262 DOI: 10.1088/0957-4484/27/42/425103] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Chemotherapy commonly used in the treatment of advanced bladder cancer is only moderately effective and associated with significant toxicity. There has been no appreciable improvement in overall survival over the last three decades. The goal of this project is to develop and characterize bladder cancer-specific nanometer-scale micelles loaded with the chemotherapeutic drug paclitaxel (PTX) and determine the anti-tumor activity and toxicity. Micelle-building-material telodendrimers were synthesized through the stepwise conjugation of eight cholic acid units at one terminus of polyethylene glycol (PEG) and a bladder cancer-specific targeting peptide named PLZ4 at the other terminus. To synthesize disulfide-crosslinked PLZ4 nanomicelles (DC-PNM), cysteine was introduced between the cholic acid and PEG. DC-PNM-PTX was synthesized through the evaporation method by loading PTX in the core. The loading capacity of PTX in DC-PNM was 25% (W/W). The loading efficiency was over 99%. DC-PNM-PTX was spherical with the median size of 25 nm. The stability of DC-PNM-PTX was determined in a solution containing sodium docecyl sulfate (SDS). It was stable in a SDS solution, but dissolved within 5 min after the addition of glutathione at the physiological intracellular concentration of 10 mM. In vivo targeting and anti-tumor activity were determined in immunodeficient mice carrying patient-derived bladder cancer xenografts (PDXs). After intravenous administration, DC-PNM specifically targeted the bladder cancer PDXs, but very little to the lung cancer xenografts in the same mice (p < 0.001). DC-PNM loaded with PTX overcame cisplatin resistance, and improved the median survival from 55 d with free PTX to 69.5 d (p = 0.03) of mice carrying PDXs. In conclusion, DC-PNM remained stable in the SDS solution, specifically targeted the bladder cancer xenografts in vivo, and improved the anti-cancer efficacy of PTX.
Collapse
Affiliation(s)
- Amy Pan
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Ohashi Y, Okamura M, Hirosawa A, Tamaki N, Akatsuka A, Wu KM, Choi HW, Yoshimatsu K, Shiina I, Yamori T, Dan S. M-COPA, a Golgi Disruptor, Inhibits Cell Surface Expression of MET Protein and Exhibits Antitumor Activity against MET-Addicted Gastric Cancers. Cancer Res 2016; 76:3895-903. [PMID: 27197184 DOI: 10.1158/0008-5472.can-15-2220] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 03/28/2016] [Indexed: 12/31/2022]
Abstract
The Golgi apparatus is responsible for transporting, processing, and sorting numerous proteins in the cell, including cell surface-expressed receptor tyrosine kinases (RTK). The small-molecule compound M-COPA [2-methylcoprophilinamide (AMF-26)] disrupts the Golgi apparatus by inhibiting the activation of Arf1, resulting in suppression of tumor growth. Here, we report an evaluation of M-COPA activity against RTK-addicted cancers, focusing specifically on human gastric cancer (GC) cells with or without MET amplification. As expected, the MET-addicted cell line MKN45 exhibited a better response to M-COPA than cell lines without MET amplification. Upon M-COPA treatment, cell surface expression of MET was downregulated with a concurrent accumulation of its precursor form. M-COPA also reduced levels of the phosphorylated form of MET along with the downstream signaling molecules Akt and S6. Similar results were obtained in additional GC cell lines with amplification of MET or the FGF receptor FGFR2 MKN45 murine xenograft experiments demonstrated the antitumor activity of M-COPA in vivo Taken together, our results offer an initial preclinical proof of concept for the use of M-COPA as a candidate treatment option for MET-addicted GC, with broader implications for targeting the Golgi apparatus as a novel cancer therapeutic approach. Cancer Res; 76(13); 3895-903. ©2016 AACR.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Apoptosis/drug effects
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Case-Control Studies
- Cell Proliferation/drug effects
- Female
- Follow-Up Studies
- Golgi Apparatus/drug effects
- Golgi Apparatus/metabolism
- Golgi Apparatus/pathology
- Humans
- Immunoenzyme Techniques
- Mice
- Mice, Inbred C57BL
- Mice, Nude
- Naphthols/pharmacology
- Neoplasm Staging
- Phosphorylation/drug effects
- Prognosis
- Proto-Oncogene Proteins c-met/genetics
- Proto-Oncogene Proteins c-met/metabolism
- Pyridines/pharmacology
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Signal Transduction/drug effects
- Stomach Neoplasms/drug therapy
- Stomach Neoplasms/metabolism
- Stomach Neoplasms/pathology
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Yoshimi Ohashi
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Mutsumi Okamura
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Asaka Hirosawa
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Naomi Tamaki
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Akinobu Akatsuka
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kuo-Ming Wu
- Next Generation Systems, Eisai Inc., Andover, Massachusetts
| | | | | | - Isamu Shiina
- Department of Applied Chemistry, Faculty of Science, Tokyo University of Science, Tokyo, Japan
| | - Takao Yamori
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shingo Dan
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.
| |
Collapse
|
14
|
Zhao S, Chu Z, Blanco VM, Nie Y, Hou Y, Qi X. SapC-DOPS nanovesicles as targeted therapy for lung cancer. Mol Cancer Ther 2015; 14:491-8. [PMID: 25670331 DOI: 10.1158/1535-7163.mct-14-0661] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Lung cancer is the deadliest type of cancer for both men and women. In this study, we evaluate the in vitro and in vivo efficacy of a biotherapeutic agent composed of a lysosomal protein (Saposin C, SapC) and a phospholipid (dioleoylphosphatidylserine, DOPS), which can be assembled into nanovesicles (SapC-DOPS) with selective antitumor activity. SapC-DOPS targets phosphatidylserine, an anionic phospholipid preferentially exposed in the surface of cancer cells and tumor-associated vasculature. Because binding of SapC to phosphatidylserine is favored at acidic pHs, and the latter characterizes the milieu of many solid tumors, we tested the effect of pH on the binding capacity of SapC-DOPS to lung tumor cells. Results showed that SapC-DOPS binding to cancer cells was more pronounced at low pH. Viability assays on a panel of human lung tumor cells showed that SapC-DOPS cytotoxicity was positively correlated with cell surface phosphatidylserine levels, whereas mitochondrial membrane potential measurements were consistent with apoptosis-related cell death. Using a fluorescence tracking method in live mice, we show that SapC-DOPS specifically targets human lung cancer xenografts, and that systemic therapy with SapC-DOPS induces tumor apoptosis and significantly inhibits tumor growth. These results suggest that SapC-DOPS nanovesicles are a promising treatment option for lung cancer.
Collapse
Affiliation(s)
- Shuli Zhao
- State Key Laboratory of Reproductive Medicine, Central Laboratory of Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhengtao Chu
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio. Division of Human Genetics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Victor M Blanco
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Yunzhong Nie
- Immunology and Reproductive Biology Laboratory, Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, China
| | - Yayi Hou
- Immunology and Reproductive Biology Laboratory, Medical School and State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, Jiangsu, China
| | - Xiaoyang Qi
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio. Division of Human Genetics, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| |
Collapse
|
15
|
Callmann CE, Barback CV, Thompson MP, Hall DJ, Mattrey RF, Gianneschi NC. Therapeutic Enzyme-Responsive Nanoparticles for Targeted Delivery and Accumulation in Tumors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:4611-5. [PMID: 26178920 PMCID: PMC4699560 DOI: 10.1002/adma.201501803] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 05/31/2015] [Indexed: 05/21/2023]
Abstract
An enzyme-responsive, paclitaxel-loaded nanoparticle is described and assessed in vivo in a human fibrosarcoma murine xenograft. This work represents a proof-of-concept study demonstrating the utility of enzyme-responsive nanoscale drug carriers capable of targeted accumulation and retention in tumor tissue in response to overexpressed endogenous enzymes.
Collapse
Affiliation(s)
- Cassandra E. Callmann
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | | | - Matthew P. Thompson
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| | - David J. Hall
- Department of Radiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Robert F. Mattrey
- Department of Radiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Nathan C. Gianneschi
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
16
|
Preclinical Murine Models for Lung Cancer: Clinical Trial Applications. BIOMED RESEARCH INTERNATIONAL 2015; 2015:621324. [PMID: 26064932 PMCID: PMC4433653 DOI: 10.1155/2015/621324] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 11/24/2014] [Indexed: 12/18/2022]
Abstract
Murine models for the study of lung cancer have historically been the backbone of preliminary preclinical data to support early human clinical trials. However, the availability of multiple experimental systems leads to debate concerning which model, if any, is best suited for a particular therapeutic strategy. It is imperative that these models accurately predict clinical benefit of therapy. This review provides an overview of the current murine models used to study lung cancer and the advantages and limitations of each model, as well as a retrospective evaluation of the uses of each model with respect to accuracy in predicting clinical benefit of therapy. A better understanding of murine models and their uses, as well as their limitations may aid future research concerning the development and implementation of new targeted therapies and chemotherapeutic agents for lung cancer.
Collapse
|
17
|
Miwa T, Kokuryo T, Yokoyama Y, Yamaguchi J, Nagino M. Therapeutic potential of targeting protein for Xklp2 silencing for pancreatic cancer. Cancer Med 2015; 4:1091-100. [PMID: 25914189 PMCID: PMC4529347 DOI: 10.1002/cam4.453] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 02/17/2015] [Accepted: 02/18/2015] [Indexed: 11/24/2022] Open
Abstract
The targeting protein for Xklp2 (TPX2) is a microtubule- and, cell cycle-associated protein who’s overexpression has been reported in various malignancies. In this study, we verified the overexpression of TPX2 in both surgically resected specimens of pancreatic cancer and multiple pancreatic cancer cell lines. Subsequently, we found that TPX2 siRNA effectively suppressed the proliferation of pancreatic cancer cells in culture, and the direct injection of TPX2 siRNA into subcutaneously implanted pancreatic cancer cells in nude mice revealed antiproliferative effects. These results implied a therapeutic potential of TPX2 siRNA in pancreatic cancer. Among 56 angiogenesis-related factors examined using angiogenesis arrays, the average protein levels of insulin-like growth factor-binding protein-3 (IGFBP-3) were significantly higher in TPX2 siRNA-treated tumors than in the Control siRNA-treated tumors. Moreover, we demonstrated that CD34-positive microvessels were significantly reduced in tumors treated with TPX2 siRNA compared to tumors that treated with Control siRNA. The attenuated expression of CD34 in TPX2 siRNA-treated tumors coincided with the overexpression of IGFBP-3. These results indicated that TPX2 has an impact on tumor angiogenesis in pancreatic cancer. The results also implied that the antiangiogenic effect observed in TPX2 siRNA-treated pancreatic cancer cells may be partly explained by the upregulation of IGFBP-3.
Collapse
Affiliation(s)
- Tomohiro Miwa
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Toshio Kokuryo
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yukihiro Yokoyama
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Junpei Yamaguchi
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Masato Nagino
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
18
|
Razi SS, Rehmani S, Li X, Park K, Schwartz GS, Latif MJ, Bhora FY. Antitumor activity of paclitaxel is significantly enhanced by a novel proapoptotic agent in non–small cell lung cancer. J Surg Res 2015; 194:622-630. [DOI: 10.1016/j.jss.2014.11.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 07/26/2014] [Accepted: 11/04/2014] [Indexed: 11/24/2022]
|
19
|
Wang LH, Li Y, Yang SN, Wang FY, Hou Y, Cui W, Chen K, Cao Q, Wang S, Zhang TY, Wang ZZ, Xiao W, Yang JY, Wu CF. Gambogic acid synergistically potentiates cisplatin-induced apoptosis in non-small-cell lung cancer through suppressing NF-κB and MAPK/HO-1 signalling. Br J Cancer 2013; 110:341-52. [PMID: 24300974 PMCID: PMC3899775 DOI: 10.1038/bjc.2013.752] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Revised: 10/23/2013] [Accepted: 10/29/2013] [Indexed: 12/20/2022] Open
Abstract
Background: Gambogic acid (GA) has been reported to have potent anticancer activity and is authorised to be tested in phase II clinical trials for treatment of non-small-cell lung cancer (NSCLC). The present study aims to investigate whether GA would be synergistic with cisplatin (CDDP) against the NSCLC. Methods: 1-(4,5-Dimethylthiazol-2-yl)-3,5-diphenylformazan (MTT), combination index (CI) isobologram, western blot, quantitative PCR, flow cytometry, electrophoretic mobility shift assay, xenograft tumour models and terminal deoxynucleotide transferase-mediated dUTP nick-end labelling analysis were used in this study. Results: The cell viability results showed that sequential CDDP-GA treatment resulted in a strong synergistic action in A549, NCI-H460, and NCI-H1299 cell lines, whereas the reverse sequence and simultaneous treatments led to a slight synergistic or additive action. Increased sub-G1 phase cells and enhanced PARP cleavage demonstrated that the sequence of CDDP-GA treatment markedly increased apoptosis in comparison with other treatments. Furthermore, the sequential combination could enhance the activation of caspase-3, -8, and 9, increase the expression of Fas and Bax, and decrease the expression of Bcl-2, survivin and X-inhibitor of apoptosis protein (X-IAP) in A549 and NCI-H460 cell lines. In addition, increased apoptosis was correlated with enhanced reactive oxygen species generation. Importantly, it was found that, followed by CDDP treatment, GA could inhibit NF-κB and mitogen-activated protein kinase (MAPK)/heme oxygenase-1 (HO-1) signalling pathways, which have been validated to reduce ROS release and confer CDDP resistance. The roles of NF-κB and MAPK pathways were further confirmed by using specific inhibitors, which significantly increased ROS release and apoptosis induced by the sequential combination of CDDP and GA. Moreover, our results indicated that the combination of CDDP and GA exerted increased antitumour effects on A549 xenograft models through inhibiting NF-κB, HO-1, and subsequently inducing apoptosis. Conclusion: Gambogic acid sensitises lung cancer cells to CDDP in vitro and in vivo in NSCLC through inactivation of NF-κB and MAPK/HO-1 signalling pathways, providing a rationale for the combined use of CDDP and GA in lung cancer chemotherapy.
Collapse
Affiliation(s)
- L-H Wang
- 1] Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China [2] Jiangsu Kanion Pharmaceutical Co. Ltd, Lianyungang 222001, People's Republic of China
| | - Y Li
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - S-N Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - F-Y Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Y Hou
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - W Cui
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - K Chen
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Q Cao
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - S Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - T-Y Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Z-Z Wang
- Jiangsu Kanion Pharmaceutical Co. Ltd, Lianyungang 222001, People's Republic of China
| | - W Xiao
- Jiangsu Kanion Pharmaceutical Co. Ltd, Lianyungang 222001, People's Republic of China
| | - J-Y Yang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - C-F Wu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| |
Collapse
|
20
|
The use of porous scaffold as a tumor model. Int J Biomater 2013; 2013:396056. [PMID: 24101930 PMCID: PMC3786466 DOI: 10.1155/2013/396056] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 08/12/2013] [Accepted: 08/12/2013] [Indexed: 12/30/2022] Open
Abstract
Background. Human cancer is a three-dimensional (3D) structure consisting of neighboring cells, extracellular matrix, and blood vessels. It is therefore critical to mimic the cancer cells and their surrounding environment during in vitro study. Our aim was to establish a 3D cancer model using a synthetic composite scaffold. Methods. High-density low-volume seeding was used to promote attachment of a non-small-cell lung cancer cell line (NCI-H460) to scaffolds. Growth patterns in 3D culture were compared with those of monolayers. Immunohistochemistry was conducted to compare the expression of Ki67, CD44, and carbonic anhydrase IX. Results. NCI-H460 readily attached to the scaffold without surface pretreatment at a rate of 35% from a load of 1.5 × 106 cells. Most cells grew vertically to form clumps along the surface of the scaffold, and cell morphology resembled tissue origin; 2D cultures exhibited characteristics of adherent epithelial cancer cell lines. Expression patterns of Ki67, CD44, and CA IX varied markedly between 3D and monolayer cultures. Conclusions. The behavior of cancer cells in our 3D model is similar to tumor growth in vivo. This model will provide the basis for future study using 3D cancer culture.
Collapse
|
21
|
Seki T, Kokuryo T, Yokoyama Y, Suzuki H, Itatsu K, Nakagawa A, Mizutani T, Miyake T, Uno M, Yamauchi K, Nagino M. Antitumor effects of α-bisabolol against pancreatic cancer. Cancer Sci 2011; 102:2199-205. [PMID: 21883695 DOI: 10.1111/j.1349-7006.2011.02082.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In the present study, we investigated whether α-bisabolol, a sesquiterpene alcohol present in essential oils derived from a variety of plants, has antitumor effects against pancreatic cancer. α-Bisabolol induced a decrease in cell proliferation and viability in pancreatic cancer cell lines (KLM1, KP4, Panc1, MIA Paca2), but not in pancreatic epithelial cells (ACBRI515). α-Bisabolol treatment induced apoptosis and suppressed Akt activation in pancreatic cancer cell lines. Furthermore, α-bisabolol treatment induced the overexpression of early growth response-1 (EGR1), whereas EGR1 siRNA decreased the α-bisabolol-induced cell death of KLM1 cells. Tumor growth in both subcutaneous and peritoneal xenograft nude mouse models was significantly inhibited by intragastric administration of 1000 mg/kg of α-bisabolol, once a week for three weeks. The results indicate that α-bisabolol could be a novel therapeutic option for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Takashi Seki
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Li B, Torossian A, Li W, Schleicher S, Niu K, Giacalone NJ, Kim SJ, Chen H, Gonzalez A, Moretti L, Lu B. A novel bioluminescence orthotopic mouse model for advanced lung cancer. Radiat Res 2011; 176:486-93. [PMID: 21663394 DOI: 10.1667/rr2565.1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Lung cancer is the leading cause of cancer-related death in the United States despite recent advances in our understanding of this challenging disease. An animal model for high-throughput screening of therapeutic agents for advanced lung cancer could help promote the development of more successful treatment interventions. To develop our orthotopic lung cancer model, luciferase-expressing A549 cancer cells were injected into the mediastinum of athymic nude mice. To determine whether the model would allow easy monitoring of response to therapeutic interventions, tumors were treated with 30 mg/kg Paclitaxel or were irradiated with 5 fractions of 2 Gy, and tumor burden was monitored using bioluminescence imaging. Evidence of radiation-induced lung injury was assessed using immunohistochemical staining for phospho-Smad2/3 and cleaved caspase-3. We found that tumor implantation recapitulated advanced human lung cancer as evidenced by tumor establishment and proliferation within the mediastinum. The tumor responded to Paclitaxel or radiation as shown by decreased tumor bioluminescence and improved overall survival. Immunohistochemistry revealed increased phospho-Smad2/3 and cleaved caspase-3 in irradiated lungs, consistent with radiation-induced lung injury. This orthotopic lung cancer model may help provide a method to assess therapeutic interventions in a preclinical setting that recapitulates locally advanced lung cancer.
Collapse
Affiliation(s)
- Bo Li
- Department of Radiation Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Nakamura H, Shoji A, Takeuchi A, Ban HS, Lee JD, Yamori T, Kang SO. Discovery of ortho-Carborane-Conjugated Triazines as Selective Topoisomerase I/II Inhibitors. Aust J Chem 2011. [DOI: 10.1071/ch11295] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The cell growth inhibition profile of 2,4-(2-methyl-ortho-carboranyl)-4-(dimethylamino)-1,3,5-triazine (TAZ-6) was found to be similar to that of ICRF-193, a topoisomerase II inhibitor, as revealed by COMPARE analysis (correlation coefficient (r) = 0.724). Various mono- and di-ortho-carborane-substituted 1,3,5-triazines were synthesized based on the structure of TAZ-6 and tested for their ability to inhibit cell growth and the activities of topoisomerases I and II. Among the compounds synthesized, 3c, 4c, and 4f completely inhibited topoisomerase I activity without affecting topoisomerase II activity, whereas 3a and 3d completely inhibited topoisomerase II activity without affecting topoisomerase I activity, at 100 μM.
Collapse
|
24
|
Correlation between cytotoxic activities and reduction potentials of heterocyclic quinones. Molecules 2010; 15:6559-69. [PMID: 20877243 PMCID: PMC6257668 DOI: 10.3390/molecules15096559] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Revised: 09/07/2010] [Accepted: 09/16/2010] [Indexed: 11/17/2022] Open
Abstract
To search for possible anti-tumor agents or anti-tumor promoters among natural or synthetic products, we used cyclic voltammetry to determine the reduction-oxidation potentials of heterocyclic quinones in phosphate buffer at pH 7.2. We determined the growth inhibitory- and cytotoxic activities of 12 heterocyclic quinone anti-tumor agent candidates against a panel of 39 human cancer cell lines (JFCR39). The average concentrations of the heterocyclic quinones required for 50% growth inhibition (GI50) against JFCR39 ranged from 0.045 to 13.2 μM, and the 50% lethal concentration (LC50) against JFCR39 ranged from 0.398 to 77.7 μM. The average values of GI50 or LC50 of the heterocyclic quinones correlated significantly with their reduction potentials. These results suggested that reduction-oxidation potentials could be a useful method for the discovery of novel antitumor agents.
Collapse
|
25
|
Williamson MJ, Silva MD, Terkelsen J, Robertson R, Yu L, Xia C, Hatsis P, Bannerman B, Babcock T, Cao Y, Kupperman E. The relationship among tumor architecture, pharmacokinetics, pharmacodynamics, and efficacy of bortezomib in mouse xenograft models. Mol Cancer Ther 2009; 8:3234-43. [DOI: 10.1158/1535-7163.mct-09-0239] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
26
|
Javeed A, Ashraf M, Riaz A, Ghafoor A, Afzal S, Mukhtar MM. Paclitaxel and immune system. Eur J Pharm Sci 2009; 38:283-90. [DOI: 10.1016/j.ejps.2009.08.009] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Accepted: 08/29/2009] [Indexed: 12/17/2022]
|
27
|
Suzuki N, Suzuki T, Ota Y, Nakano T, Kurihara M, Okuda H, Yamori T, Tsumoto H, Nakagawa H, Miyata N. Design, synthesis, and biological activity of boronic acid-based histone deacetylase inhibitors. J Med Chem 2009; 52:2909-22. [PMID: 19419205 DOI: 10.1021/jm900125m] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Guided by the proposed catalytic mechanism of histone deacetylases (HDACs), we designed and synthesized a series of boronic acid-based HDAC inhibitors bearing an alpha-amino acid moiety. In this series, compounds (S)-18, 20, and 21 showed potent HDAC-inhibitory activity, highlighting the significance of the (S)-amino acid moiety. In cancer cell growth inhibition assays, compounds (S)-18, 20, and 21 exerted strong activity, and the values of the ratio of the concentration causing 50% growth inhibition (GI(50)) to the concentration causing 50% enzyme inhibition (IC(50)), i.e., GI(50)/IC(50), were low. The potency of these compounds was similar to that of clinically used suberoylanilide hydroxamic acid (SAHA) (2). The results of Western blot analysis indicated that the cancer cell growth-inhibitory activity of compounds (S)-18, 20, and 21 is the result of HDAC inhibition. A molecular modeling study suggested that the hydrated boronic acid interacts with zinc ion, Tyr residue, and His residue in the active site of HDACs. Our findings indicate that these boronic acid derivatives represent an entry into a new class of HDAC inhibitors.
Collapse
Affiliation(s)
- Nobuaki Suzuki
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Shichiri M, Fukai N, Kono Y, Tanaka Y. Rifampicin as an Oral Angiogenesis Inhibitor Targeting Hepatic Cancers. Cancer Res 2009; 69:4760-8. [DOI: 10.1158/0008-5472.can-08-3417] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
29
|
Liu J, Meisner D, Kwong E, Wu XY, Johnston MR. Translymphatic Chemotherapy by Intrapleural Placement of Gelatin Sponge Containing Biodegradable Paclitaxel Colloids Controls Lymphatic Metastasis in Lung Cancer. Cancer Res 2009; 69:1174-81. [DOI: 10.1158/0008-5472.can-08-1753] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
30
|
Zhong H, Han B, Tourkova IL, Lokshin A, Rosenbloom A, Shurin MR, Shurin GV. Low-dose paclitaxel prior to intratumoral dendritic cell vaccine modulates intratumoral cytokine network and lung cancer growth. Clin Cancer Res 2007; 13:5455-62. [PMID: 17875775 PMCID: PMC2474691 DOI: 10.1158/1078-0432.ccr-07-0517] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE The main goal of this study was to provide the "proof-of-principle" that low-dose paclitaxel is able to change the tumor microenvironment and improve the outcome of intratumoral dendritic cell vaccine in a murine lung cancer model. EXPERIMENTAL DESIGN We evaluated the antitumor potential and changes in the intratumoral milieu of a combination of low-dose chemotherapy and dendritic cell vaccine in the Lewis lung carcinoma model in vivo. RESULTS The low-dose paclitaxel, which induced apoptosis in approximately 10% of tumor cells, was not toxic to bone marrow cells and dendritic cells and stimulated dendritic cell maturation and function in vitro. Although tumor cells inhibited dendritic cell differentiation in vitro, this immunosuppressive effect was abrogated by the pretreatment of tumor cells with low-dose paclitaxel. Based on these data, we next tested whether pretreatment of tumor-bearing mice with low-dose paclitaxel in vivo would improve the antitumor potential of dendritic cell vaccine administered intratumorally. Significant inhibition of tumor growth in mice treated with low-dose paclitaxel plus intratumoral dendritic cell vaccine, associated with increased tumor infiltration by CD4(+) and CD8(+) T cells and elevated tumor-specific IFN-gamma production by draining lymph node cells, was revealed. Using a novel intratumoral microdialysis technique and Luminex technology for collecting and characterizing soluble factors released within the tumor bed for several days in live freely moving animals, we showed that low-dose paclitaxel altered the cytokine network at the tumor site. CONCLUSIONS Our data indicate that low-dose chemotherapy before intratumoral delivery of dendritic cells might be associated with beneficial alterations of the intratumoral microenvironment and thus support antitumor immunity.
Collapse
Affiliation(s)
- Hua Zhong
- Shanghai Chest Hospital, Shanghai, China
| | - Baohui Han
- Shanghai Chest Hospital, Shanghai, China
| | | | - Anna Lokshin
- Departments of Medicine, University of Pittsburgh Medical Center
| | - Alan Rosenbloom
- Molecular Biosensors and Imaging Center, Carnegie Mellon University, Pittsburgh, Pennsylvania Received 3/5/07; revised 5/10/07; accepted 5/31/07
| | - Michael R. Shurin
- Departments of Pathology, University of Pittsburgh Medical Center
- Departments of Immunology, University of Pittsburgh Medical Center
| | - Galina V. Shurin
- Departments of Pathology, University of Pittsburgh Medical Center
| |
Collapse
|
31
|
NK314, a novel topoisomerase II inhibitor, induces rapid DNA double-strand breaks and exhibits superior antitumor effects against tumors resistant to other topoisomerase II inhibitors. Cancer Lett 2007; 259:99-110. [PMID: 17998154 DOI: 10.1016/j.canlet.2007.10.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2007] [Revised: 09/21/2007] [Accepted: 10/03/2007] [Indexed: 12/15/2022]
Abstract
NK314 is a novel synthetic benzo[c]phenanthridine alkaloid that shows strong antitumor activity. It inhibited topoisomerase II activity and stabilized topoisomerase II-DNA cleavable complexes. The DNA breaks occurred within 1h after treatment with NK314 even without digestion of topoisomerase II by proteinase K, whereas etoposide required digestion of the enzyme protein in cleavable complex to detect DNA breaks. Pretreatment with topoisomerase II catalytic inhibitors, ICRF-193 and suramin, reduced both cleavable complex-mediated DNA breaks and proteinase K-independent DNA breaks, but protease inhibitors and nuclease inhibitors only decreased the latter. These results indicate that NK314 might affect topoisomerase II in the different manner from cleavable complex formation and activate intracellular proteinase and nuclease to produce DNA fragmentation. As a result of this unique mechanism of DNA breakage, NK314 showed substantial growth inhibition of topoisomerase II inhibitor-resistant tumors.
Collapse
|
32
|
Ishizu K, Sunose N, Yamazaki K, Tsuruo T, Sadahiro S, Makuuchi H, Yamori T. Development and characterization of a model of liver metastasis using human colon cancer HCT-116 cells. Biol Pharm Bull 2007; 30:1779-83. [PMID: 17827739 DOI: 10.1248/bpb.30.1779] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In order to develop a model of liver metastasis of human gastrointestinal cancer cells, we examined the potential of 10 human colon and stomach cancer cell lines (HT-29, WiDr, HCT-116, HCT-15, HCC-2998, MKN7, MKN28, MKN45, MKN74 and St-4) to form liver metastases in nude mice. Among the cell lines, HCT-116 cells consistently formed gross liver metastases when injected into the spleens of nude mice. In contrast, other human colon and stomach cancer cells produced little or no liver metastasis. In order to analyze the high metastatic potential of HCT-116 cells, the adhesion potential was compared between HCT-116 cells and the other colon cancer cell lines. HCT-116 cells showed more efficient adhesion to fibronectin (FN) than other cells. Furthermore, FN enhanced haptotaxis of HCT-116 cells, but not of other colon cancer cells. The high adhesion potential to FN and enhanced haptotaxis may contribute, at least in part, to the high metastatic potential of HCT-116. To assess the value of this newly developed model of liver metastasis, we compared the ability of four anticancer drugs (fluorouracil, doxifluridine, paclitaxel and irinotecan) to inhibit the formation of liver metastases. Paclitaxel and irinotecan showed strong inhibition of liver metastasis but fluorouracil and doxifluridine showed only slight inhibition. Therefore, this model of metastasis may be useful for screening anti-liver metastatic reagents. These results indicate that the HCT-116 liver-metastasis model should be useful for analyzing the molecular mechanism of liver metastasis and for evaluating new anti-liver metastatic drugs.
Collapse
Affiliation(s)
- Kazuhiro Ishizu
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Koto-ku, Tokyo 135-8550, Japan
| | | | | | | | | | | | | |
Collapse
|
33
|
Nakamura H, Kuroda H, Saito H, Suzuki R, Yamori T, Maruyama K, Haga T. Synthesis and biological evaluation of boronic acid containing cis-stilbenes as apoptotic tubulin polymerization inhibitors. ChemMedChem 2006; 1:729-40. [PMID: 16902927 DOI: 10.1002/cmdc.200600068] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
A series of boronic acid containing cis-stilbenes as potent inhibitors of tubulin polymerization was synthesized by the introduction of boronic acid as an acceptor-type functional group into the aromatic ring B of the combretastatin framework. High cell-growth inhibition was observed with boron compounds 13 c and 13 d, in which a hydroxy group on the aromatic ring B of combretastatin A-4 was replaced with boronic acid; IC50 values toward B-16 and 1-87 cell lines are 0.48-2.1 microM. Compounds 13 c and 13 d exhibited significant inhibitory activity toward tubulin polymerization (IC50=21-22 microM). The carboxylic acid derivative 17, which can be considered as a mimic of boronic acid 13 c, did not show significant inhibition of cell growth or tubulin polymerization. According to the FACScan analysis using Jurkat cells, apoptosis was induced after incubation for 8 h with 13 c at a concentration of >10(-8) M. Growth inhibitory experiments against a panel of 39 human cancer cell lines revealed 13 c to inhibit growth differently than combretastatin A-4; the correlation coefficient (r) between the two compounds was 0.553 in the COMPARE analysis.
Collapse
Affiliation(s)
- Hiroyuki Nakamura
- Department of Chemistry, Faculty of Science, Gakushuin University, Mejiro, Toshima-ku, Tokyo 171-8588, Japan.
| | | | | | | | | | | | | |
Collapse
|
34
|
Sasaki S, Bando T, Minoshima M, Shimizu T, Shinohara KI, Takaoka T, Sugiyama H. Sequence-Specific Alkylation of Double-Strand Human Telomere Repeat Sequence by Pyrrole-Imidazole Polyamides with Indole Linkers. J Am Chem Soc 2006; 128:12162-8. [PMID: 16967966 DOI: 10.1021/ja0626584] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We designed and synthesized pyrrole (Py)-imidazole (Im) hairpin polyamide 1-(chloromethyl)-5-hydroxy-1,2-dihydro-3H-benz[e]indole (seco-CBI) conjugates 1 and 2, which target both strands of the double-stranded region of the human telomere repeat sequences, 5'-d(TTAGGG)(n)-3'/5'-d(CCCTAA)(n)-3'. High-resolution denaturing polyacrylamide gel electrophoresis demonstrated that conjugates 1 and 2 alkylated DNA at the 3' A of 5'-ACCCTA-3' and 5'-AGGGTTA-3', respectively. Cytotoxicities of conjugates 1 and 2 were evaluated using 39 human cancer cell lines; averages of log IC(50) values for conjugates 1 and 2 were -6.96 (110 nM) and -7.24 (57.5 nM), respectively. Conjugates 1 and 2 have potential as antitumor drugs capable of targeting telomere repeat sequence.
Collapse
Affiliation(s)
- Shunta Sasaki
- Department of Chemistry, Graduate School of Science, Kyoto University, Kitashirakawa-Oiwakecho, Sakyo, Kyoto, 606-8502, Japan
| | | | | | | | | | | | | |
Collapse
|
35
|
Roberts WG, Whalen PM, Soderstrom E, Moraski G, Lyssikatos JP, Wang HF, Cooper B, Baker DA, Savage D, Dalvie D, Atherton JA, Ralston S, Szewc R, Kath JC, Lin J, Soderstrom C, Tkalcevic G, Cohen BD, Pollack V, Barth W, Hungerford W, Ung E. Antiangiogenic and Antitumor Activity of a Selective PDGFR Tyrosine Kinase Inhibitor, CP-673,451. Cancer Res 2005. [DOI: 10.1158/0008-5472.957.65.3] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
CP-673,451 is a potent inhibitor of platelet-derived growth factor β-receptor (PDGFR-β) kinase- and PDGF-BB-stimulated autophosphorylation of PDGFR-β in cells (IC50 = 1 nmol/L) being more than 450-fold selective for PDGFR-β versus other angiogenic receptors (e.g., vascular endothelial growth factor receptor 2, TIE-2, and fibroblast growth factor receptor 2). Multiple models have been used to evaluate in vivo activity of CP-673,451 and to understand the pharmacology of PDGFR-β inhibition and the effect on tumor growth. These models include an ex vivo measure of PDGFR-β phosphorylation in glioblastoma tumors, a sponge model to measure inhibition of angiogenesis, and multiple models of tumor growth inhibition. Inhibition of PDGFR-β phosphorylation in tumors correlates with plasma and tumor levels of CP-673,451. A dose of 33 mg/kg was adequate to provide >50% inhibition of receptor for 4 hours corresponding to an EC50 of 120 ng/mL in plasma at Cmax. In a sponge angiogenesis model, CP-673,451 inhibited 70% of PDGF-BB-stimulated angiogenesis at a dose of 3 mg/kg (q.d. × 5, p.o., corresponding to 5.5 ng/mL at Cmax). The compound did not inhibit vascular endothelial growth factor- or basic fibroblast growth factor-induced angiogenesis at concentrations which inhibited tumor growth. The antitumor efficacy of CP-673,451 was evaluated in a number of human tumor xenografts grown s.c. in athymic mice, including H460 human lung carcinoma, Colo205 and LS174T human colon carcinomas, and U87MG human glioblastoma multiforme. Once-daily p.o. × 10 days dosing routinely inhibited tumor growth (ED50 ≤ 33 mg/kg). These data show that CP-673,451 is a pharmacologically selective PDGFR inhibitor, inhibits tumor PDGFR-β phosphorylation, selectively inhibits PDGF-BB-stimulated angiogenesis in vivo, and causes significant tumor growth inhibition in multiple human xenograft models.
Collapse
Affiliation(s)
- W. Gregory Roberts
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - Pamela M. Whalen
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - Erik Soderstrom
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - Garrett Moraski
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | | | - Huifen-F. Wang
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - Beth Cooper
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - Deborah A. Baker
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - Douglas Savage
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - Deepak Dalvie
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - James A. Atherton
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - Sherry Ralston
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - Ruby Szewc
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - John C. Kath
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - Jing Lin
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - Cathy Soderstrom
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - George Tkalcevic
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - Bruce D. Cohen
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - Vince Pollack
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - Wayne Barth
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - Will Hungerford
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| | - Ethan Ung
- Pfizer Oncology, Pfizer Global Research and Development, Groton, Connecticut
| |
Collapse
|
36
|
Lai YY, Huang LJ, Lee KH, Xiao Z, Bastow KF, Yamori T, Kuo SC. Synthesis and biological relationships of 3′,6-substituted 2-phenyl-4-quinolone-3-carboxylic acid derivatives as antimitotic agents. Bioorg Med Chem 2005; 13:265-75. [PMID: 15582470 DOI: 10.1016/j.bmc.2004.09.041] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2004] [Accepted: 09/21/2004] [Indexed: 10/26/2022]
Abstract
As part of a continuing search for potential anticancer drug candidates in the 2-phenyl-4-quinolone series, 3',6-substituted 2-phenyl-4-quinolone-3-carboxylic acid derivatives and their salts were synthesized and evaluated. Preliminary screening showed that carboxylic acid analogs containing a m-fluoro substituted 2-phenyl group displayed the highest in vitro anticancer activity. Activity decreased significantly if a chlorine or methoxy group replaced the fluorine atom. 3'-Fluoro-6-methoxy-2-phenyl-4-quinolone-3-carboxylic acid (68) had the highest in vitro cytotoxic activity among all tested carboxylic acid derivatives and their salts. The mechanism of action may be similar, but not identical, to that of tubulin binding drugs, such as navelbine and taxol. Compound 68 merits further investigation as a novel hydrophilic antimitotic agent.
Collapse
Affiliation(s)
- Ya-Yun Lai
- Graduate Institute of Pharmaceutical Chemistry, China Medical University, Taichung, Taiwan
| | | | | | | | | | | | | |
Collapse
|
37
|
Nomura T, Shinoda S, Yamori T, Sawaki S, Nagata I, Ryoyama K, Fuke Y. Selective sensitivity to wasabi-derived 6-(methylsulfinyl)hexyl isothiocyanate of human breast cancer and melanoma cell lines studied in vitro. ACTA ACUST UNITED AC 2005; 29:155-60. [PMID: 15829375 DOI: 10.1016/j.cdp.2004.07.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2004] [Accepted: 07/27/2004] [Indexed: 11/17/2022]
Abstract
Recently, attention has focused on the anticancer properties of an aromatic component 6-(methylsulfinyl)hexyl isothiocyanate (6-MITC) in a typical Japanese spice, wasabi. In this paper, anticancer activity of 6-MITC in vitro was studied by using a human cancer cell (HCC) panel. 6-MITC directly affected the cells in the HCC panel and inhibited their growth in culture. The mean concentration required to inhibit 50% of control cell growth was 3.9 microM, which is a sufficiently low dosage for practical use. The suppression influenced not only the cell growth, but also the survival of these cells. The mean concentration to suppress cells to a 50% survival was 43.7 microM. The reduction activity of 6-MITC was differential, and it suppressed specific cells. These severely suppressed cell lines included breast cancer and melanoma cell lines. For example, one melanoma line was seriously damaged at a concentration of 0.3 microM of 6-MITC. Compared with other MITCs (2-MITC, 4-MITC and 8-MITC), 6-MITC showed the most effective suppression and with the most specific manner of the cells mentioned above. A "COMPARE" analysis using a computerized algorithm, which was based on the HCC database, suggested that the suppression mechanism of 6-MITC is unique and may be different from that of other known chemicals. The actual mechanism may not a simple one but may involve multiple pathways. On account of its sufficiently small size, 6-MITC is a new possible candidate for controlling cancer cells.
Collapse
Affiliation(s)
- Takahiro Nomura
- Division of Food and Nutritional Science, Kanazawa Gakuin College, 10 Sue-machi, Kanazawa, Ishikawa 920-1392, Japan.
| | | | | | | | | | | | | |
Collapse
|
38
|
Bai J, Kitabatake M, Toyoizumi K, Fu L, Zhang S, Dai J, Sakai J, Hirose K, Yamori T, Tomida A, Tsuruo T, Ando M. Production of biologically active taxoids by a callus culture of Taxus cuspidata. JOURNAL OF NATURAL PRODUCTS 2004; 67:58-63. [PMID: 14738387 DOI: 10.1021/np0301083] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Ten known taxoids, paclitaxel, 7-epi-taxol, taxol C, baccatin VI, taxayuntin C, taxuyunnanine C and its analogues (2-5), and yunnanxane (6), and an abietane, taxamairin A, were produced in the callus culture of Taxus cuspidata cultivated on a modified Gamborg's B5 medium in the presence of 0.5 mg/L NAA. After stimulation with 100 microM methyl jasmonate, five more taxoids, cephalomannine, 1beta-dehydroxybaccatin VI, taxinine NN-11 (1), baccatin I, and 2alpha-acetoxytaxusin, and one more abietane, taxamairin C, were found in addition to the above-mentioned compounds. It was also observed that the content of the products increased over three times. Taxinine NN-11 (1) is a new taxane whose structure was assigned as 5alpha,13alpha-diacetoxy-9alpha-cinnamoyloxy-4(20),11-taxadien-10beta-ol by analysis of its spectral data. Taxinine NN-11 (1) exhibited significant MDR reversal activity toward 2780 AD tumor cells. The results of primary screening based on 39 human cancer cell lines suggest that 1 also belongs to a new mechanistic class. Efficient production of 1 was investigated using the callus culture of T. cuspidata.
Collapse
Affiliation(s)
- Jiao Bai
- Graduate School of Science and Technology and Department of Chemistry and Chemical Engineering, Niigata University, 2-8050 Ikarashi, Niigata 950-2181, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Takahashi R, Bando T, Sugiyama H. Specific alkylation of human telomere repeats by hairpin pyrrole-imidazole polyamide. Bioorg Med Chem 2003; 11:2503-9. [PMID: 12757718 DOI: 10.1016/s0968-0896(03)00176-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A novel hairpin polyamide-cyclopropapyrroloindole (CPI) conjugate PyImImIm-gamma-PyPyPyLDu86 (conjugate 11), which targets human telomere repeats d(TTAGGG)(n)/d(CCCTAA)(n), was synthesized. High resolution denaturing polyacrylamide gel electrophoresis using 44 bp DNA fragments and HPLC product analysis of a synthetic nonanucleotide demonstrated that conjugate 11 alkylates the target adenine in the telomere repeats, 5'-CCCTAA-3'. Examination of the antitumor activity of conjugate 11 using a panel of 39 cancer cell lines demonstrated that the average concentration of conjugate 11 required for 50% growth inhibition was 5.75 microM, which is superior to pepleomycin and bleomycin and comparable to cisplatin.
Collapse
Affiliation(s)
- Ryoko Takahashi
- Division of Biofunctional Molecules, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Surugadai, Kanda, Chiyoda, Tokyo 101-0062, Japan
| | | | | |
Collapse
|
40
|
Murakami Y. Functional cloning of a tumor suppressor gene, TSLC1, in human non-small cell lung cancer. Oncogene 2002; 21:6936-48. [PMID: 12362275 DOI: 10.1038/sj.onc.1205825] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The identification of a tumor suppressor gene in non-small cell lung cancer (NSCLC) is one of the most important issues to elucidate the molecular mechanisms of this type of refractory cancer and to establish a novel strategy against it. Since NSCLC, like most other human cancers, develops as a sporadic disease, linkage analysis is not available for gene cloning. This review describes the functional cloning approaches to a tumor suppressor gene in sporadic cancers. Suppression of the malignant phenotype of cancer cells by fusion with a normal fibroblast was the first demonstration of the recessive phenotype of cancer cells in 1969. Evidence of tumor suppressor genes on the specific chromosomes was later provided by functional complementation of the cancer phenotype through microcell-mediated chromosome transfer. Further introduction of more restricted DNA fragments by YAC transfer provides a potent tool to localize the gene to a small segment, appropriate for the subsequent gene cloning. TSLC1, a novel tumor suppressor gene in NSCLC, was identified on chromosome 11q23.2 through a series of functional complementation of A549 cells in tumorigenicity. Two-hit inactivation of the TSLC1 by promoter methylation and gene deletion was observed in 40% of primary NSCLC tumors. The strong tumor suppressor activity of TSLC1, and its possible involvement in cell adhesion, suggest that the functional cloning approach could cast a new light on a group of genes that have not yet been characterized, but are important for general human carcinogenesis as well as tumor suppression.
Collapse
Affiliation(s)
- Yoshinori Murakami
- Tumor Suppression and Functional Genomics Project, National Cancer Center Research Institute, Japan.
| |
Collapse
|
41
|
Saji H, Koike M, Yamori T, Saji S, Seiki M, Matsushima K, Toi M. Significant correlation of monocyte chemoattractant protein-1 expression with neovascularization and progression of breast carcinoma. Cancer 2001; 92:1085-91. [PMID: 11571719 DOI: 10.1002/1097-0142(20010901)92:5<1085::aid-cncr1424>3.0.co;2-k] [Citation(s) in RCA: 229] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Macrophages often infiltrate into solid tumor tissues. Tumor-associated macrophages (TAMs) are known to play a crucial role in tumor progression. Monocyte chemoattractant protein-1 (MCP-1) is one of the major chemokines capable of inducing chemotactic migration of monocytes. METHODS With the objective of investigating the clinical significance of MCP-1, the authors analyzed the expression of MCP-1 and of some other molecules by immunohistochemistry in 230 samples of primary breast carcinoma tissue. MCP-1 staining was performed using an anti-MCP-1 monoclonal antibody, and it was assessed by grading the percentage of stained cells. RESULTS It was found that 117 breast tumor specimens (51%) had intensive staining in tumor cells. The expression of MCP-1 in tumor cells had a significant correlation with the expression of thymidine phosphorylase and membrane type 1-matrix metalloproteinase. In addition, MCP-1 expression tended to be associated with the accumulation of TAMs, which were counted by CD68 staining, and with microvessel density. MCP-1 expression in TAMs was correlated significantly with the histologic vessel invasion of tumor cells. CONCLUSIONS The results of this study suggest that MCP-1 may play key roles in macrophage recruitment, in the expression of angiogenic factors, and in the activation of matrix metalloproteinases in patients with breast carcinoma.
Collapse
MESH Headings
- Angiogenesis Inducing Agents
- Breast Neoplasms/blood supply
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carcinoma/metabolism
- Carcinoma/pathology
- Carcinoma, Ductal, Breast/blood supply
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Chemokine CCL2/metabolism
- Humans
- Immunohistochemistry
- Neovascularization, Pathologic/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- H Saji
- Department of Surgery, Breast Oncology Unit, Tokyo Metropolitan Komagome Hospital, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
42
|
Sato Y, Kashimoto S, MacDonald JR, Nakano K. In vivo antitumour efficacy of MGI-114 (6-hydroxymethylacylfulvene, HMAF) in various human tumour xenograft models including several lung and gastric tumours. Eur J Cancer 2001; 37:1419-28. [PMID: 11435075 DOI: 10.1016/s0959-8049(01)00135-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
MGI-114 (6-hydroxymethylacylfulvene, HMAF) is a semi-synthetic analogue of the cytotoxic sesquiterpenoid illudins. In the present study, the in vivo antitumour efficacy of MGI-114 was examined in a panel of human tumour xenograft models consisting mainly of human lung and gastric tumours, and compared with that of other antitumour drugs such as irinotecan, paclitaxel, cisplatin, doxorubicin, vindesine, etoposide and 5-fluorouracil (5-FU). When different administration schedules were compared, daily administration of MGI-114 was found to be more effective than intermittent administrations. In human tumour xenograft models of nasopharyngeal, breast and colon carcinoma and melanoma, MGI-114 exerted a strong antitumour activity with complete tumour regression being observed. Moreover, in four human lung and three gastric tumour xenograft models, MGI-114 showed a strong antitumour activity with complete tumour regression being observed in some of the models. The antitumour efficacy of MGI-114 was generally higher than or equivalent to that of other antitumour drugs such as irinotecan and paclitaxel. These results support the potential utility of MGI-114 in the treatment of a variety of human solid tumours.
Collapse
Affiliation(s)
- Y Sato
- Department of Pharmacology II, Discovery Research Laboratories, Dainippon Pharmaceutical Co., Ltd. Enoki 33-94, Suita, 564-0053, Osaka, Japan.
| | | | | | | |
Collapse
|
43
|
Iwasa K, Moriyasu M, Yamori T, Turuo T, Lee DU, Wiegrebe W. In vitro cytotoxicity of the protoberberine-type alkaloids. JOURNAL OF NATURAL PRODUCTS 2001; 64:896-898. [PMID: 11473418 DOI: 10.1021/np000554f] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
In vitro cytotoxic activities of 24 quaternary protoberberine alkaloids related to berberine have been evaluated using a human cancer cell line panel coupled with a drug sensitivity database. Extending the alkyl chain at position 8 or 13 strongly influenced the cytotoxic activity, that is, relative lipophilicity as well as the size of the substituent affects cytotoxicity. The highest level of activity was observed in 8- or 13-hexyl-substituted derivatives of berberine. Structure-activity relationships are described.
Collapse
Affiliation(s)
- K Iwasa
- Kobe Pharmaceutical University, 4-19-1 Motoyamakita, Higashinada-ku, Kobe 658-8558, Japan.
| | | | | | | | | | | |
Collapse
|
44
|
Kuramochi M, Fukuhara H, Nobukuni T, Kanbe T, Maruyama T, Ghosh HP, Pletcher M, Isomura M, Onizuka M, Kitamura T, Sekiya T, Reeves RH, Murakami Y. TSLC1 is a tumor-suppressor gene in human non-small-cell lung cancer. Nat Genet 2001; 27:427-30. [PMID: 11279526 DOI: 10.1038/86934] [Citation(s) in RCA: 349] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The existence of tumor-suppressor genes was originally demonstrated by functional complementation through whole-cell and microcell fusion. Transfer of chromosome 11 into a human non-small-cell lung cancer (NSCLC) cell line, A549, suppresses tumorigenicity. Loss of heterozygosity (LOH) on the long arm of chromosome 11 has been reported in NSCLC and other cancers. Several independent studies indicate that multiple tumor-suppressor genes are found in this region, including the gene PPP2R1B at 11q23-24 (ref. 7). Linkage studies of NSCLC are precluded because no hereditary forms are known. We previously identified a region of 700 kb on 11q23.2 that completely suppresses tumorigenicity of A549 human NSCLC cells. Most of this tumor-suppressor activity localizes to a 100-kb segment by functional complementation. Here we report that this region contains a single confirmed gene, TSLC1, whose expression is reduced or absent in A549 and several other NSCLC, hepatocellular carcinoma (HCC) and pancreatic cancer (PaC) cell lines. TSLC1 expression or suppression is correlated with promoter methylation state in these cell lines. Restoration of TSLC1 expression to normal or higher levels suppresses tumor formation by A549 cells in nude mice. Only 2 inactivating mutations of TSLC1 were discovered in 161 tumors and tumor cell lines, both among the 20 primary tumors with LOH for 11q23.2. Promoter methylation was observed in 15 of the other 18 primary NSCLC, HCC and PaC tumors with LOH for 11q23.2. Thus, attenuation of TSLC1 expression occurred in 85% of primary tumors with LOH. Hypermethylation of the TSLC1 promoter would seem to represent the 'second hit' in NSCLC with LOH.
Collapse
Affiliation(s)
- M Kuramochi
- Tumor Suppression & Functional Genomics Project, National Cancer Center Research Institute, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Fukushima S, Takeuchi Y, Kishimoto S, Yamashita S, Uetsuki K, Shirakawa S, Suzuki M, Furuta K, Noyori R, Sasaki H, Kikuchi Y, Kita T, Yamori T, Sawada J, Kojima M, Hazato A, Kurozumi S, Fukushima M. Antitumor activity, optimum administration method and pharmacokinetics of 13,14-dihydro-15-deoxy-deoxy-Delta7 -prostaglandin A1 methyl ester (TEI-9826) integrated in lipid microspheres (Lipo TEI-9826). Anticancer Drugs 2001; 12:221-34. [PMID: 11290870 DOI: 10.1097/00001813-200103000-00008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
13,14-Dihydro-15-deoxy-Delta7-prostaglandin A1 methyl ester (TEI-9826), an antitumor prostaglandin analog, is a candidate for clinical trial. In the present study, we examined its biological stability in vitro, antitumor activity in vitro and in vivo, and pharmacokinetics. Although TEI-9826 was rapidly hydrolyzed to the carboxylic acid form (TOK-4528), TOK-4528 as well as Delta12-prostaglandin J2 (PGJ2) were found to be stable in rat, mouse and human serum in vitro. TEI-9826 exhibited nearly identical or greater potential antitumor activity compared to Delta12-PGJ2 and Delta7-PGA1 in vitro against Colon26 tumor cells. Further evaluation of TEI-9826 using the 38 human cancer cell lines panel and COMPARE analysis suggested that its mode of action is quite different from other anticancer agents that are currently used. TEI-9826 was integrated into lipid microspheres (Lipo TEI-9826) for dosing. Growth inhibition by Lipo TEI-9826 against Colon26 tumor inoculated s.c. in mice depended on administration route, i.e. at 80 mg/kg, no growth suppressive effect was observed for daily bolus i.v., but significant growth suppressive effect was observed for daily i.p., daily s.c. every other day s.c. and 4 times a day continuous (5 min) i.v. These tumor growth-suppressive effects were cytostatic and the tumor started to regrow at the end or a few days after the end of administration. The pharmacokinetic study suggested that maintaining the blood level of TEI-9826 and/or TOK-4528 was essential for their antitumor effects. These results show that continuous i.v. infusion might be the most suitable administration method of Lipo TEI-9826 for clinical trial.
Collapse
Affiliation(s)
- S Fukushima
- Department of Pharmaceutics, Faculty of Pharmaceutical Science and High Technology Research Center, Kobe Gakuin University, Nishiku, Kobe 651-2180, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Fukushima S, Kishimoto S, Takeuchi Y, Fukushima M. Preparation and evaluation of o/w type emulsions containing antitumor prostaglandin. Adv Drug Deliv Rev 2000; 45:65-75. [PMID: 11104898 DOI: 10.1016/s0169-409x(00)00101-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Antitumor prostaglandins(PGs) such as Delta12-PGJ2 and Delta7-PGA1 possess a cyclopentenone or cross-conjugated dienone structures. Antitumor PGs are actively incorporated through cell membrane and control gene expression. Very recent studies clarified that P53 independent expression of p21 and gadd 45, activation of PPARgamma are involved in antitumor mechanism of these PGs. At the low concentration, these PGs exhibit physiological or pathological activity such as osteoblast calcification, promotion of colon cancer cell proliferation. COMPARE PROGRAM using human 38 tumor cell lines suggested that antitumor mechanism of Delta7-PGA1 and 13, 14-dihydro-15-deoxy-Delta7-PGA1 methyl ester (TEI-9826) are quite different from other anticancer agents which are clinically used. Lipid microspheres and Lipiodol formulation were examined as dosage form of the PGs and lipid microspheres were selected for further study. At first lipid microspheres integrated TEI-9038 (Lipo TEI-9038) was chosen as a candidate for clinical trial. However Lipo TEI-9038 failed to exhibit substantial antitumor effect because of its enzymatic instability and toxicity in vivo. Lipo TEI-9826 was then selected as promising candidate for clinical trial because of its stability in serum. Lipo TEI-9826 exhibited marked antitumor effect in several animal models including CDDP resistant nude mice model. Pharmacokinetic and toxicological studies using rats suggested that continuous infusion is the most suitable administration method for Lipo TEI-9826. New type emulsifier, Controlled High Pressure Process Homogenizer (De-BEE 2000 and mini De-BEE) was developed during the preclinical studies on manufacturing process of Lipo TEI-9826. These results warrant the clinical trial for Lipo TEI-9826 in CDDP resistant cancer.
Collapse
Affiliation(s)
- S Fukushima
- Department of Pharmaceutics, Faculty of Pharmaceutical Science, Kobe Gakuin University, Nishiku, 651-2180, Kobe, Japan.
| | | | | | | |
Collapse
|
47
|
Myrick D, Blackinton D, Klostergaard J, Kouttab N, Maizel A, Wanebo H, Mehta S. Paclitaxel-induced apoptosis in Jurkat, a leukemic T cell line, is enhanced by ceramide. Leuk Res 1999; 23:569-78. [PMID: 10374850 DOI: 10.1016/s0145-2126(99)00048-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We hypothesized that the lipid second messenger, ceramide, and microtubule-directed chemotherapeutic agents might engage converging pathways in inducing apoptosis. Our studies demonstrated that simultaneous treatment of Jurkat cells with paclitaxel and ceramide enhanced paclitaxel-induced cell growth inhibition. Cell cycle analysis indicated a significant increase in the hypodiploid population over that observed with paclitaxel treatment alone. Morphologic evaluation and a TUNEL assay confirmed a dramatic increase in apoptosis in Jurkat cells treated with the combination of these two agents. This is the first demonstration that paclitaxel and ceramide interact in a supra-additive manner to decrease leukemic T-cell growth, suggesting a possible application of paclitaxel and ceramide in combination therapy.
Collapse
Affiliation(s)
- D Myrick
- Department of Pathobiology, Brown University, Providence, RI, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Morihira K, Nishimori T, Kusama H, Horiguchi Y, Kuwajima I, Tsuruo T. Synthesis and evaluation of artificial taxoids with antitumor and multi-drug resistance reversing activities. Bioorg Med Chem Lett 1998; 8:2977-82. [PMID: 9873658 DOI: 10.1016/s0960-894x(98)00540-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
Artificial taxoids were synthesized and subjected to evaluation of their ability of multi-drug resistance reversing and antitumor activities. While the taxoid 4 could not increase cellular accumulation of vincristine in multi-drug resistant tumor cells, the C4-hydroxy analog 15 showed significant effect. However, these compounds showed weak activities on growth inhibition of cancer cells.
Collapse
Affiliation(s)
- K Morihira
- Department of Chemistry, Tokyo Institute of Technology, Japan
| | | | | | | | | | | |
Collapse
|