15301
|
Patel K, Siraj S, Smith C, Nair M, Vishwanatha JK, Basha R. Pancreatic Cancer: An Emphasis on Current Perspectives in Immunotherapy. Crit Rev Oncog 2019; 24:105-118. [PMID: 31679206 PMCID: PMC8038975 DOI: 10.1615/critrevoncog.2019031417] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Pancreatic cancer affects both male and female individuals with higher incidences and death rates among the male population. Detection of this malignancy is delayed due to the lack of symptoms in the early-stage cancer, which makes it extremely difficult to treat. Identifying effective strategies has been a challenge for improving the survival rates in pancreatic cancer patients. Resistance to chemotherapy is often developed in pancreatic cancer treatment. Although many strategies are under clinical trials to target certain markers associated with cancer, immunotherapeutic approaches are currently gaining importance. Immunotherapy for pancreatic cancer is in the limelight after preclinical research showed some promise. Immunotherapy approaches were tested along with other treatment options to enhance the treatment effect. Adoptive cell transfer and immune checkpoint inhibitors are currently in clinical trials. The Food and Drug Administration approved pembrolizumab in a fast-tracked review for advanced pancreatic cancer patients. Pembrolizumab blocks the checkpoint protein, programmed cell death protein 1 (PD-1), on T cells to boost the response of the immune system against cancer cells, thereby shrinking tumors. The recent developments in immunotherapy and the early success in other cancers are encouraging to further test immunotherapy in pancreatic cancer. The combination of pembrolizumab and pelareorep, an isolate of human reovirus, is in phase II clinical study in metastatic disease. Depending on the results of current clinical trials and testing, the strategies in the pipeline are expected to increase the use of immunotherapy in the clinical testing setting. Success in immunotherapy is urgently needed to address the side-effects, treating patients with advanced disease and reducing metastasis for increasing the survival rate in pancreatic cancer patients.
Collapse
Affiliation(s)
| | | | - Chloe Smith
- Old Dominion University, Norfolk, Virginia 23529
| | - Maya Nair
- Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, Texas 76107
| | - Jamboor K. Vishwanatha
- Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, Texas 76107
| | | |
Collapse
|
15302
|
Tang W, Fan W, Lau J, Deng L, Shen Z, Chen X. Emerging blood–brain-barrier-crossing nanotechnology for brain cancer theranostics. Chem Soc Rev 2019; 48:2967-3014. [DOI: 10.1039/c8cs00805a] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The advancements, perspectives, and challenges in blood–brain-barrier (BBB)-crossing nanotechnology for effective brain tumor delivery and highly efficient brain cancer theranostics.
Collapse
Affiliation(s)
- Wei Tang
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN)
- National Institute of Biomedical Imaging and Bioengineering (NIBIB)
- National Institutes of Health (NIH)
- Bethesda
- USA
| | - Wenpei Fan
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN)
- National Institute of Biomedical Imaging and Bioengineering (NIBIB)
- National Institutes of Health (NIH)
- Bethesda
- USA
| | - Joseph Lau
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN)
- National Institute of Biomedical Imaging and Bioengineering (NIBIB)
- National Institutes of Health (NIH)
- Bethesda
- USA
| | - Liming Deng
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN)
- National Institute of Biomedical Imaging and Bioengineering (NIBIB)
- National Institutes of Health (NIH)
- Bethesda
- USA
| | - Zheyu Shen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN)
- National Institute of Biomedical Imaging and Bioengineering (NIBIB)
- National Institutes of Health (NIH)
- Bethesda
- USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN)
- National Institute of Biomedical Imaging and Bioengineering (NIBIB)
- National Institutes of Health (NIH)
- Bethesda
- USA
| |
Collapse
|
15303
|
Cartledge Wolf DM, Langhans SA. Moving Myeloid Leukemia Drug Discovery Into the Third Dimension. Front Pediatr 2019; 7:314. [PMID: 31417884 PMCID: PMC6682595 DOI: 10.3389/fped.2019.00314] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/11/2019] [Indexed: 12/12/2022] Open
Abstract
The development of therapies aimed at leukemia has progressed substantially in the past years but childhood acute myeloid leukemia (AML) remains one of the most challenging cancers to treat. Genomic profiling of AML has greatly enhanced our understanding of the genetic and epigenetic landscape of this high-risk leukemia. With it comes the opportunity to develop targeted therapies that are expected to be more effective and less toxic than current treatment regimens. Nevertheless, often overlooked in leukemia drug discovery are the dynamic interactions between leukemic cells and the bone marrow environment. The interplay between leukemic cells, stromal cells and the extracellular matrix plays critical roles in the development, progression and relapse of AML as well as in drug response and the development of resistance. Here we will review pediatric leukemia with a special focus on acute myeloid disease in children, and discuss the tumor microenvironment in the context of drug resistance and leukemia stem cell survival. We will emphasize how three-dimensional (3D) cell-based drug discovery may offer hope for both the identification and advancement of more effective treatment options for patients suffering from this devastating disease.
Collapse
Affiliation(s)
- Donna M Cartledge Wolf
- Nemours Center for Childhood Cancer Research, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| | - Sigrid A Langhans
- Nemours Center for Childhood Cancer Research, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| |
Collapse
|
15304
|
Costa S, Reagan MR. Therapeutic Irradiation: Consequences for Bone and Bone Marrow Adipose Tissue. Front Endocrinol (Lausanne) 2019; 10:587. [PMID: 31555210 PMCID: PMC6727661 DOI: 10.3389/fendo.2019.00587] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 08/09/2019] [Indexed: 12/28/2022] Open
Abstract
Radiotherapy continues to be one of the most accepted medical treatments for cancer. Localized irradiation is the most common treatment for prostate, pancreatic, rectal, cervical and endometrial malignancies. Conventional localized fractions are total doses of 30-62Gy at 1.8-2Gy per fraction, with administration of ~60Gy often used for tumor ablation. However, even the lowest dose of localized irradiation exposure can result in adverse complications to adjacent organs, tissues, and vessels, which absorb a portion of the treatment. Skeletal complications are common amongst cancer patients undergoing these localized treatments. Irradiation exposure causes deterioration to the overall quantity and quality of bone by interfering with the trabecular architecture through increased osteoclast activity and decreased osteoblast activity. Irradiation-induced bone damage parallels adipocyte infiltration of the bone marrow (BM) resulting in compositional alterations of the microenvironment that may further affect bone quality and disease state. There may also be direct effects of irradiation on the BM adipocyte/pre-adipocyte, although in vitro findings do not always agree and cellular response is dependent on irradiation dosage. Hematopoietic cells also become apoptotic upon irradiation, which causes a range of skeletal effects. Bone loss leaves patients at a greater risk for osteopenia, osteoporosis, osteonecrosis, and skeletal fractures that drastically reduce quality of life. Osteoanabolic agents stimulate bone formation and reduce fracture risk in patients with low bone density; thus, osteoanabolic or anti-resorptive agents may be useful co-treatments with irradiation. This review discusses these topics and proposes further research directions using novel or combination therapies to enhance bone health during irradiation.
Collapse
Affiliation(s)
- Samantha Costa
- Center for Clinical and Translational Research, Maine Medical Center Research Institute, Scarborough, ME, United States
- University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, United States
- Tufts University School of Medicine, Boston, MA, United States
| | - Michaela R. Reagan
- Center for Clinical and Translational Research, Maine Medical Center Research Institute, Scarborough, ME, United States
- University of Maine Graduate School of Biomedical Science and Engineering, Orono, ME, United States
- Tufts University School of Medicine, Boston, MA, United States
- *Correspondence: Michaela R. Reagan
| |
Collapse
|
15305
|
Sridharan S, Howard CM, Tilley AMC, Subramaniyan B, Tiwari AK, Ruch RJ, Raman D. Novel and Alternative Targets Against Breast Cancer Stemness to Combat Chemoresistance. Front Oncol 2019. [PMID: 31681564 DOI: 10.3389/fonc.2019.01003.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2023] Open
Abstract
Breast cancer stem cells (BCSCs) play a vital role in tumor progression and metastasis. They are heterogeneous and inherently radio- and chemoresistant. They have the ability to self-renew and differentiate into non-BCSCs. These determinants of BCSCs including the plasticity between the mesenchymal and epithelial phenotypes often leads to minimal residual disease (MRD), tumor relapse, and therapy failure. By studying the resistance mechanisms in BCSCs, a combinatorial therapy can be formulated to co-target BCSCs and bulk tumor cells. This review addresses breast cancer stemness and molecular underpinnings of how the cancer stemness can lead to pharmacological resistance. This might occur through rewiring of signaling pathways and modulated expression of various targets that support survival and self-renewal, clonogenicity, and multi-lineage differentiation into heterogeneous bulk tumor cells following chemotherapy. We explore emerging novel and alternative molecular targets against BC stemness and chemoresistance involving survival, drug efflux, metabolism, proliferation, cell migration, invasion, and metastasis. Strategic targeting of such vulnerabilities in BCSCs may overcome the chemoresistance and increase the longevity of the metastatic breast cancer patients.
Collapse
Affiliation(s)
- Sangita Sridharan
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | - Cory M Howard
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | - Augustus M C Tilley
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | | | - Amit K Tiwari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH, United States
| | - Randall J Ruch
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| | - Dayanidhi Raman
- Department of Cancer Biology, University of Toledo, Toledo, OH, United States
| |
Collapse
|
15306
|
Abstract
Cancer is a cumulative manifestation of several complicated disease states that affect multiple organs. Over the last few decades, the fruit fly Drosophila melanogaster, has become a successful model for studying human cancers. The genetic simplicity and vast arsenal of genetic tools available in Drosophila provides a unique opportunity to address questions regarding cancer initiation and progression that would be extremely challenging in other model systems. In this chapter we provide a historical overview of Drosophila as a model organism for cancer research, summarize the multitude of genetic tools available, offer a brief comparison between different model organisms and cell culture platforms used in cancer studies and briefly discuss some of the latest models and concepts in recent Drosophila cancer research.
Collapse
|
15307
|
Zhang Y, Huang H, Fu H, Zhao M, Wu Z, Dong Y, Li H, Duan Y, Sun Y. Dual-mode US/MRI nanoparticles delivering siRNA and Pt(iv) for ovarian cancer treatment. RSC Adv 2019; 9:33302-33309. [PMID: 35529112 PMCID: PMC9073344 DOI: 10.1039/c9ra03681d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 10/06/2019] [Indexed: 12/11/2022] Open
Abstract
Phase-shifted dual-mode US/MRI nanoparticles (PFH/siRNA/Fe3O4@Pt(iv) NPs-cRGD) delivering si-survivin and Pt(iv) prodrug for enhancing ovarian cancer treatment and realizing real-time monitoring.
Collapse
Affiliation(s)
- Yanhua Zhang
- State Key Laboratory of Oncogenes and Related Genes
- Shanghai Cancer Institute
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
| | - Hui Huang
- State Key Laboratory of Oncogenes and Related Genes
- Shanghai Cancer Institute
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
| | - Hao Fu
- State Key Laboratory of Oncogenes and Related Genes
- Shanghai Cancer Institute
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
| | - Meng Zhao
- State Key Laboratory of Oncogenes and Related Genes
- Shanghai Cancer Institute
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
| | - Zhihua Wu
- State Key Laboratory of Oncogenes and Related Genes
- Shanghai Cancer Institute
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
| | - Yang Dong
- State Key Laboratory of Oncogenes and Related Genes
- Shanghai Cancer Institute
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
| | - He Li
- Traditional Chinese Medicine Department
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
- Shanghai 200127
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes
- Shanghai Cancer Institute
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
| | - Ying Sun
- State Key Laboratory of Oncogenes and Related Genes
- Shanghai Cancer Institute
- Renji Hospital
- School of Medicine
- Shanghai Jiao Tong University
| |
Collapse
|
15308
|
Chu Y, Liao J, Li J, Wang Y, Yu X, Wang J, Xu X, Xu L, Zheng L, Xu J, Li L. CD103 + tumor-infiltrating lymphocytes predict favorable prognosis in patients with esophageal squamous cell carcinoma. J Cancer 2019; 10:5234-5243. [PMID: 31602274 PMCID: PMC6775603 DOI: 10.7150/jca.30354] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 07/18/2019] [Indexed: 02/05/2023] Open
Abstract
As an indispensable factor in preventing the recirculation of tissue lymphocytes to the lymphatic and blood systems, the integrin CD103 has enabled the characterization of lymphocyte populations in non-lymphoid tissues and organs. However, the expression, distribution, and clinical significance of CD103+ tumor-infiltrating lymphocytes (TILs) in esophageal squamous cell carcinoma (ESCC) remain unclear. In the present study, we included tumor and adjacent non-tumor tissue specimens from 198 patients with ESCC who had undergone surgical resection. Immunohistochemistry and immunofluorescence were used to detect CD103+ TIL distribution, as well as the co-expression of CD103 and T cell markers and functional molecules. Kaplan-Meier analysis and the Cox proportional hazards model were used to estimate the prognostic value of CD103+ TILs. The results showed that CD103+ TILs were predominantly located in adjacent non-tumor tissues compared with tumor tissues (P < 0.0001). Immunofluorescence double staining revealed that CD8+ T cells, but not CD4+ T cells, comprised the majority of CD103-expressing cells. Most of these CD103-expressing cells co-expressed CTLA-4 and granzyme B rather than the exhaustion marker PD-1. High density of intratumoral CD103+ TIL is associated with longer overall survival (OS) and disease-free survival (DFS) in both the internal (OS, P = 0.0004 and DFS, P = 0.0002) and external (OS, P = 0.038 and DFS, P = 0.12) cohorts. Multivariate Cox analysis showed the density of CD103+ TILs was an independent positive prognostic factor for OS (hazards ratio [HR] = 0.406; P = 0.0003 in the internal cohort; HR = 0.328, P = 0.01, in the external cohort) and DFS (HR = 0.385; P = 0.0002 in the internal cohort; HR = 0.270, P = 0.003, in the external cohort). Our findings indicate that CD103+ TILs might play an important role in the tumor microenvironment, and intratumoral CD103+ TILs could serve as a promising prognostic marker in ESCC.
Collapse
Affiliation(s)
- Yifan Chu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Jing Liao
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Jinqing Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Yongchun Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Xingjuan Yu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Junfeng Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
| | - Xiue Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
| | - Liyan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
| | - Limin Zheng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Jing Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, P. R. China
- ✉ Corresponding authors: Lian Li, School of Life Sciences, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou 510275, P. R. China, Tel: 86-20-84115531, E-mail: ; Jing Xu, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, No. 651, Dongfeng East Road, Guangzhou 510060, P. R. China, E-mail:
| | - Lian Li
- MOE Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou 510275, P. R. China
- ✉ Corresponding authors: Lian Li, School of Life Sciences, Sun Yat-sen University, No. 135, Xingang Xi Road, Guangzhou 510275, P. R. China, Tel: 86-20-84115531, E-mail: ; Jing Xu, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, No. 651, Dongfeng East Road, Guangzhou 510060, P. R. China, E-mail:
| |
Collapse
|
15309
|
Zhang R, Liu Q, Li T, Liao Q, Zhao Y. Role of the complement system in the tumor microenvironment. Cancer Cell Int 2019; 19:300. [PMID: 31787848 PMCID: PMC6858723 DOI: 10.1186/s12935-019-1027-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/11/2019] [Indexed: 12/17/2022] Open
Abstract
The complement system has traditionally been considered a component of innate immunity against invading pathogens and "nonself" cells. Recent studies have demonstrated the immunoregulatory functions of complement activation in the tumor microenvironment (TME). The TME plays crucial roles in tumorigenesis, progression, metastasis and recurrence. Imbalanced complement activation and the deposition of complement proteins have been demonstrated in many types of tumors. Plasma proteins, receptors, and regulators of complement activation regulate several biological functions of stromal cells in the TME and promote the malignant biological properties of tumors. Interactions between the complement system and cancer cells contribute to the proliferation, epithelial-mesenchymal transition, migration and invasion of tumor cells. In this review, we summarize recent advances related to the function of the complement system in the TME and discuss the therapeutic potential of targeting complement-mediated immunoregulation in cancer immunotherapy.
Collapse
Affiliation(s)
- Ronghua Zhang
- 0000 0001 0662 3178grid.12527.33Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| | - Qiaofei Liu
- 0000 0001 0662 3178grid.12527.33Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| | - Tong Li
- 0000 0001 0662 3178grid.12527.33Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| | - Quan Liao
- 0000 0001 0662 3178grid.12527.33Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| | - Yupei Zhao
- 0000 0001 0662 3178grid.12527.33Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, 1# Shuai Fu Yuan, Dong Dan District, Beijing, 100730 China
| |
Collapse
|
15310
|
Zhang G, Zhu Y, Wang Y, Wei D, Wu Y, Zheng L, Bai H, Xiao H, Zhang Z. pH/redox sensitive nanoparticles with platinum(iv) prodrugs and doxorubicin enhance chemotherapy in ovarian cancer. RSC Adv 2019; 9:20513-20517. [PMID: 35515556 PMCID: PMC9065746 DOI: 10.1039/c9ra04034j] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 06/27/2019] [Indexed: 11/21/2022] Open
Abstract
pH/redox sensitive, dual drug loaded nanoparticles were prepared from poly(ethylene glycol)-block-poly(l-lysine) (PEG-b-PLL) for improving cancer therapy.
Collapse
Affiliation(s)
- Guyu Zhang
- Beijing Chaoyang Hospital Affiliated to Capital Medical University
- Department of Gynaecological and Obstetric
- Beijing 100000
- P. R. China
| | - Yimin Zhu
- Affiliated Guangdong Medical University
- Department of Oncology
- Zhanjiang 524000
- P. R. China
| | - Yushu Wang
- Key Laboratory of Bio-based Material Science and Technology Ministry of Education
- Northeast Forestry University
- Harbin 150040
- P. R. China
| | - Dengshuai Wei
- Beijing National Laboratory for Molecular Sciences
- State Key Laboratory of Polymer Physics and Chemistry
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing 100190
| | - Yixin Wu
- College of Life Science and Technology
- Beijing University of Chemical Technology
- Beijing 100029
- P. R. China
| | - Liuchun Zheng
- Beijing National Laboratory for Molecular Sciences
- State Key Laboratory of Polymer Physics and Chemistry
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing 100190
| | - Huimin Bai
- Beijing Chaoyang Hospital Affiliated to Capital Medical University
- Department of Gynaecological and Obstetric
- Beijing 100000
- P. R. China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences
- State Key Laboratory of Polymer Physics and Chemistry
- Institute of Chemistry
- Chinese Academy of Sciences
- Beijing 100190
| | - Zhenyu Zhang
- Beijing Chaoyang Hospital Affiliated to Capital Medical University
- Department of Gynaecological and Obstetric
- Beijing 100000
- P. R. China
| |
Collapse
|
15311
|
Tan X, Zhou Y, Shen L, Jia H, Tan X. A mitochondria-targeted delivery system of doxorubicin and evodiamine for the treatment of metastatic breast cancer. RSC Adv 2019; 9:37067-37078. [PMID: 35539080 PMCID: PMC9075594 DOI: 10.1039/c9ra07096f] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 10/28/2019] [Indexed: 01/06/2023] Open
Abstract
For mitochondria-targeted nano-drug delivery systems against cancer, effectively targeting and releasing the drug into mitochondria are the keys to improve the therapeutic effect. In this study, mitochondria-targeted and reduction-sensitive micelles were developed to co-deliver doxorubicin (DOX) and evodiamine (EVO) for the treatment of metastatic breast cancer. After entering cancer cells, the micelles first targeted mitochondria through triphenylphosphonium cations. Then, the disulfide bonds of the micelles were cleaved by GSH, and both DOX and EVO were released near the mitochondria. The released EVO subsequently destroyed the mitochondrial membrane, resulting in a large amount of DOX entering the mitochondria and improving the anti-tumor effect of DOX. These mitochondria-targeted and reduction-sensitive micelles loaded with doxorubicin and evodiamine showed significant inhibition of the tumor cell growth both in vitro and in vivo. For mitochondria-targeted nano-drug delivery systems against cancer, effectively targeting and releasing the drug into mitochondria are the keys to improve the therapeutic effect.![]()
Collapse
Affiliation(s)
- Xiaoyan Tan
- Chongqing Anti-tumor Natural Drug Engineering Technology Research Center
- Chongqing Three Gorges Medical College
- 404120 P. R. China
| | - Yanlin Zhou
- Chongqing Anti-tumor Natural Drug Engineering Technology Research Center
- Chongqing Three Gorges Medical College
- 404120 P. R. China
| | - Li Shen
- Chongqing Anti-tumor Natural Drug Engineering Technology Research Center
- Chongqing Three Gorges Medical College
- 404120 P. R. China
| | - Han Jia
- Chongqing Anti-tumor Natural Drug Engineering Technology Research Center
- Chongqing Three Gorges Medical College
- 404120 P. R. China
| | - Xiaorong Tan
- Chongqing Anti-tumor Natural Drug Engineering Technology Research Center
- Chongqing Three Gorges Medical College
- 404120 P. R. China
| |
Collapse
|
15312
|
Rahman MF, Rahman MR, Islam T, Zaman T, Shuvo MAH, Hossain MT, Islam MR, Karim MR, Moni MA. A bioinformatics approach to decode core genes and molecular pathways shared by breast cancer and endometrial cancer. INFORMATICS IN MEDICINE UNLOCKED 2019. [DOI: 10.1016/j.imu.2019.100274] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
15313
|
Lixue ZMD, Xiaojuan ZMD, Yuxiu GMD, Zhaoyan DMD, Haiyang YMD, Cheng ZMD. Progress in Imaging Diagnosis and Image-guided Puncture Biopsy of Prostate Cancer. ADVANCED ULTRASOUND IN DIAGNOSIS AND THERAPY 2019. [DOI: 10.37015/audt.2019.191223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
15314
|
Dahal KS, Gamagedara S, Nuwan Perera UD, Lavine BK. Analysis of gentisic acid and related renal cell carcinoma biomarkers using reversed-phase liquid chromatography with water-rich mobile phases. J LIQ CHROMATOGR R T 2019; 42:681-687. [PMID: 33013156 DOI: 10.1080/10826076.2019.1666275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The problem of longer retention times using water-rich mobile phases in reversed phase liquid chromatography (RPLC) has been addressed using hydrophobic alcohols such as butanol in very low quantities (approximately 0.1%) as the organic modifier. Advantages of water-rich mobile phases in RPLC for the separation of water-soluble and weakly retained compounds are improved separation of congeners and better tuning of RPLC separations. This is demonstrated in the separation of gentisic acid and related renal cell carcinoma (RCC) biomarkers in urine with a Zorbax C18 column and a mobile phase of 0.1% (volume/volume) butanol in water with 0.6% (volume/volume) acetic acid. Calibration curves for the RCC biomarkers were linear over the concentration range investigated (5 ppm to 1000 ppm). Detection limits for the RCC biomarkers were 0.85ppm (quinolinic acid), 1.75ppm (gentisic acid), and 1.25ppm (4-hydroxybenzoic acid). Recovery tests using synthetic urine samples containing 20 ppm, 100 ppm, and 700 pm of each RCC biomarker were successful for all compounds.
Collapse
Affiliation(s)
| | | | | | - Barry K Lavine
- Department of Chemistry, Oklahoma State University, Stillwater, OK 74078
| |
Collapse
|
15315
|
Lin CY, Wang SS, Yang CK, Li JR, Chen CS, Hung SC, Chiu KY, Cheng CL, Ou YC, Yang SF. Impact of GAS5 genetic polymorphism on prostate cancer susceptibility and clinicopathologic characteristics. Int J Med Sci 2019; 16:1424-1429. [PMID: 31673232 PMCID: PMC6818208 DOI: 10.7150/ijms.38080] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 08/09/2019] [Indexed: 12/21/2022] Open
Abstract
Down-regulation of Growth arrest-specific 5 (GAS5) is correlated with enhanced cell proliferation and poorer prognosis of prostate cancer. We aimed to investigate the effect of variant rs145204276 of GAS5 on the prostate cancer susceptibility and clinicopathologic characteristics. In this study, 579 prostate cancer patients who underwent robot-assisted radical prostatectomy and 579 healthy controls were included. The frequency of the allele del of rs145204276 were compared between the patients and the controls to evaluate the impact of tumor susceptibility and the correlation of clinicopathological variables. The results shown that patients who carries genotype ins/del or del/del at SNP rs145204276 showed decreased risk of pathological lymph node metastasis disease (OR=0.545, p=0.043) and risk of seminal vesicle invasion (OR=0.632, p=0.022) comparing to with genotype ins/ins. In the subgroup analysis of age, more significant risk reduction effects were noted over lymph node metastasis disease (OR=0.426, p=0.032) and lymphovascular invasion (OR=0.521, p=0.025). In conclusion, the rs145204276 polymorphic genotype of GAS5 can predict the risk of lymph node metastasis. This is the first study to report the correlation between GAS5 gene polymorphism and prostate cancer prognosis.
Collapse
Affiliation(s)
- Chia-Yen Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan.,Division of Surgical Critical Care, Department of Critical Care Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Shian-Shiang Wang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Cheng-Kuang Yang
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Jian-Ri Li
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Medicine and Nursing, Hungkuang University, Taichung, Taiwan
| | - Chuan-Shu Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Sheng-Chun Hung
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Kun-Yuan Chiu
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Chen-Li Cheng
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yen-Chuan Ou
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung, Taiwan.,Department of Urology, Tung's Taichung MetroHarbor Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
15316
|
Noronha V, Rajendra A, Joshi A, Patil V, Menon N, Prabhash K. Epidermal growth factor receptor-mutated non-small-cell lung cancer: A primer on contemporary management. CANCER RESEARCH, STATISTICS, AND TREATMENT 2019. [DOI: 10.4103/crst.crst_51_19] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
15317
|
Yan W, Zheng H, Dong J, Liu C, Zuo Z, Liu X. MicroRNA-30b is involved in the pathological process of diabetes mellitus induced by pancreatic cancer by regulating plasminogen activator inhibitor-1. BIOTECHNOL BIOTEC EQ 2019. [DOI: 10.1080/13102818.2019.1698977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Affiliation(s)
- Wei Yan
- Department of Anatomy, Histology and Embryology, Guangxi Medical University, Nanning, P.R. China
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, P.R. China
| | - He Zheng
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, P.R. China
| | - Jun Dong
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, P.R. China
| | - Chao Liu
- Department of Genetics, Jinzhou Medical University, Jinzhou, P.R. China
| | - Zhongfu Zuo
- Department of Anatomy, Histology and Embryology, Guangxi Medical University, Nanning, P.R. China
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, P.R. China
| | - Xuezheng Liu
- Department of Anatomy, Histology and Embryology, Guangxi Medical University, Nanning, P.R. China
- Department of Anatomy, Histology and Embryology, Jinzhou Medical University, Jinzhou, P.R. China
| |
Collapse
|
15318
|
Sridharan S, Robeson M, Bastihalli-Tukaramrao D, Howard CM, Subramaniyan B, Tilley AMC, Tiwari AK, Raman D. Targeting of the Eukaryotic Translation Initiation Factor 4A Against Breast Cancer Stemness. Front Oncol 2019; 9:1311. [PMID: 31867270 PMCID: PMC6909344 DOI: 10.3389/fonc.2019.01311] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Accepted: 11/11/2019] [Indexed: 12/14/2022] Open
Abstract
Breast cancer stem cells (BCSCs) are intrinsically chemoresistant and capable of self-renewal. Following chemotherapy, patients can develop minimal residual disease due to BCSCs which can repopulate into a relapsed tumor. Therefore, it is imperative to co-target BCSCs along with the bulk tumor cells to achieve therapeutic success and prevent recurrence. So, it is vital to identify actionable molecular targets against both BCSCs and bulk tumor cells. Previous findings from our lab and others have demonstrated that inhibition of the emerging drug target eIF4A with Rocaglamide A (RocA) was efficacious against triple-negative breast cancer cells (TNBC). RocA specifically targets the pool of eIF4A bound to the oncogenic mRNAs that requires its helicase activity for their translation. This property enables specific targeting of tumor cells. The efficacy of RocA against BCSCs is unknown. In this study, we postulated that eIF4A could be a vulnerable node in BCSCs. In order to test this, we generated a paclitaxel-resistant TNBC cell line which demonstrated an elevated level of eIF4A along with increased levels of cancer stemness markers (ALDH activity and CD44), pluripotency transcription factors (SOX2, OCT4, and NANOG) and drug transporters (ABCB1, ABCG2, and ABCC1). Furthermore, genetic ablation of eIF4A resulted in reduced expression of ALDH1A1, pluripotency transcription factors and drug transporters. This pointed out that eIF4A is likely associated with selected set of proteins that are critical to BCSCs, and hence targeting eIF4A may eliminate BCSCs. Therefore, we isolated BCSCs from two TNBC cell lines: MDA-Bone-Un and SUM-159PT. Following RocA treatment, the self-renewal ability of the BCSCs was significantly reduced as determined by the efficiency of the formation of primary and secondary mammospheres. This was accompanied by a reduction in the levels of NANOG, OCT4, and drug transporters. Exposure to RocA also induced cell death of the BCSCs as evaluated by DRAQ7 and cell viability assays. RocA treatment induced apoptosis with increased levels of cleaved caspase-3. Overall, we identified that RocA is effective in targeting BCSCs, and eIF4A is an actionable molecular target in both BCSCs and bulk tumor cells. Therefore, anti-eIF4A inhibitors could potentially be combined synergistically with existing chemo-, radio- and/or immunotherapies.
Collapse
Affiliation(s)
- Sangita Sridharan
- Department of Cancer Biology, University of Toledo Health Science Campus, Toledo, OH, United States
| | - Megan Robeson
- Department of Cancer Biology, University of Toledo Health Science Campus, Toledo, OH, United States
| | - Diwakar Bastihalli-Tukaramrao
- Department of Pharmacology & Experimental Therapeutics, University of Toledo Health Science Campus, Toledo, OH, United States
| | - Cory M. Howard
- Department of Cancer Biology, University of Toledo Health Science Campus, Toledo, OH, United States
| | - Boopathi Subramaniyan
- Department of Cancer Biology, University of Toledo Health Science Campus, Toledo, OH, United States
| | - Augustus M. C. Tilley
- Department of Cancer Biology, University of Toledo Health Science Campus, Toledo, OH, United States
| | - Amit K. Tiwari
- Department of Pharmacology & Experimental Therapeutics, University of Toledo Health Science Campus, Toledo, OH, United States
| | - Dayanidhi Raman
- Department of Cancer Biology, University of Toledo Health Science Campus, Toledo, OH, United States
- *Correspondence: Dayanidhi Raman
| |
Collapse
|
15319
|
Van Cleave JH, Fu MR, Bennett AV, Persky MS, Li Z, Jacobson A, Hu KS, Most A, Concert C, Kamberi M, Mojica J, Peyser A, Riccobene A, Tran A, Persky MJ, Savitski J, Liang E, Egleston BL. The development, usability, and reliability of the Electronic Patient Visit Assessment (ePVA) for head and neck cancer. Mhealth 2019; 5:21. [PMID: 31463307 PMCID: PMC6691072 DOI: 10.21037/mhealth.2019.06.05] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 06/24/2019] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Annually, over 65,000 persons are diagnosed with head and neck cancer in the United States. During treatment, up to 50% of patients become severely symptomatic with pain, fatigue, mouth sores, and inability to eat. Long term complications are lymphedema, fibrosis, dysphagia, and musculoskeletal impairment. Patients' ability to perform daily activities and to interact socially may be impaired, resulting in poor quality of life. A pragmatic, clinically useful assessment is needed to ensure early detection and intervention for patients to report symptoms and functional limitations over time. We developed the Electronic Patient Visit Assessment (ePVA) that enables patients to report 42 symptoms related to head and neck cancer and 17 limitations of functional status. This manuscript reports (I) the development of the ePVA, (II) the content validity of the ePVA, and (III) the usability and reliability of the ePVA. METHODS Usability was evaluated using the "Think Aloud" technique to guide the iterative process to refine the ePVA based on participants' evaluations. After signing the informed consent, 30 participants with head and neck cancer completed the ePVA using digital tablet devices while thinking aloud about ease of use. All patient conversations were recorded and professionally transcribed. Reliability of the ePVA symptom and functional limitation measures was estimated using the Kuder-Richardson test. Convergent validity of the ePVA was evaluated using the European Organization for Research and Treatment of Cancer (EORTC) QLQ-C30 global QoL/health scale. Transcribed qualitative data were analyzed using directed content analysis approach. Quantitative analyses consisted of descriptive statistics and correlation analyses. RESULTS Among participants, 90% strongly agreed or agreed that the ePVA system was easy to use and 80% were very satisfied. Only minor usability problems were reported due to formatting and software "bugs". Reporting of usability problems decreased in frequency over the study period and no usability problems were reported by the last 3 participants who completed the ePVA. Based on participants' suggestions during the iterative process, refinement of the ePVA included increased touch sensitivity of the touch screen technology and customized error messages to improve ease of use. The ePVA also recorded patient reported symptoms (mouth symptoms: 93%, fibrosis: 60%, fatigue: 60%). The ePVA demonstrated acceptable reliability (alpha =0.82-0.85) and convergent validity (ePVA total number of reported symptoms and function limitations was negatively correlated with EORTC QLQ-C30 global QOL/health scale: r=-0.55038, P<0.01). CONCLUSIONS The ePVA was rigorously developed, accepted by patients with satisfaction, and demonstrated acceptable reliability and convergent validity. Future research will use data generated by the ePVA to determine the impact of symptom trajectories on functional status, treatment interruptions and terminations, and health resource use in head and neck cancer.
Collapse
Affiliation(s)
| | - Mei R. Fu
- New York University Meyers College of Nursing, New York, NY, USA
| | - Antonia V. Bennett
- Gillings School of Global Public Health, University of North Carolina - Chapel Hill, Chapel Hill, NC, USA
| | - Mark S. Persky
- Department of Otolaryngology – Head and Neck Surgery, Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Zujun Li
- Department of Medicine, Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Adam Jacobson
- Department of Otolaryngology – Head and Neck Surgery, Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Kenneth S. Hu
- Department of Radiation Oncology, Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Allison Most
- Department of Otolaryngology – Head and Neck Surgery, Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Catherine Concert
- Department of Radiation Oncology, Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Maria Kamberi
- Department of Otolaryngology – Head and Neck Surgery, Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Jacqueline Mojica
- Department of Otolaryngology – Head and Neck Surgery, Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Amanda Peyser
- Department of Medicine, Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Ann Riccobene
- Department of Medicine, Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Anh Tran
- Department of Medicine, Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Michael J. Persky
- Department of Otolaryngology – Head and Neck Surgery, Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Justin Savitski
- Department of Otolaryngology – Head and Neck Surgery, Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Eva Liang
- New York University Meyers College of Nursing, New York, NY, USA
| | | |
Collapse
|
15320
|
Shi H, Imberti C, Sadler PJ. Diazido platinum(iv) complexes for photoactivated anticancer chemotherapy. Inorg Chem Front 2019. [DOI: 10.1039/c9qi00288j] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Diazido Pt(iv) complexes with a general formula [Pt(N3)2(L)(L′)(OR)(OR′)] are a new generation of anticancer prodrugs designed for use in photoactivated chemotherapy.
Collapse
Affiliation(s)
- Huayun Shi
- Department of Chemistry
- University of Warwick
- Coventry
- UK
| | | | | |
Collapse
|
15321
|
Sun G, He Z, Hao M, Zuo M, Xu Z, Hu XY, Zhu JJ, Wang L. Dual acid-responsive bola-type supramolecular vesicles for efficient intracellular anticancer drug delivery. J Mater Chem B 2019. [DOI: 10.1039/c9tb00555b] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Dual acid-responsive bola-type supramolecular vesicles have been successfully constructed for efficient intracellular anticancer drug delivery and controlled release.
Collapse
Affiliation(s)
- Guangping Sun
- Key Laboratory of Mesoscopic Chemistry of MOE
- Jiangsu Key Laboratory of Advanced Organic Materials
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing
| | - Zhimei He
- State Key Laboratory of Analytical Chemistry for Life Science
- Collaborative Innovation Center of Chemistry for Life Science
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210023
| | - Min Hao
- Key Laboratory of Mesoscopic Chemistry of MOE
- Jiangsu Key Laboratory of Advanced Organic Materials
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing
| | - Minzan Zuo
- Key Laboratory of Mesoscopic Chemistry of MOE
- Jiangsu Key Laboratory of Advanced Organic Materials
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing
| | - Zuqiang Xu
- Key Laboratory of Mesoscopic Chemistry of MOE
- Jiangsu Key Laboratory of Advanced Organic Materials
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing
| | - Xiao-Yu Hu
- Key Laboratory of Mesoscopic Chemistry of MOE
- Jiangsu Key Laboratory of Advanced Organic Materials
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing
| | - Jun-Jie Zhu
- State Key Laboratory of Analytical Chemistry for Life Science
- Collaborative Innovation Center of Chemistry for Life Science
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing 210023
| | - Leyong Wang
- Key Laboratory of Mesoscopic Chemistry of MOE
- Jiangsu Key Laboratory of Advanced Organic Materials
- School of Chemistry and Chemical Engineering
- Nanjing University
- Nanjing
| |
Collapse
|
15322
|
Abraham B, Nair MS. Automated grading of prostate cancer using convolutional neural network and ordinal class classifier. INFORMATICS IN MEDICINE UNLOCKED 2019. [DOI: 10.1016/j.imu.2019.100256] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
15323
|
Dai J, Wang K, Liu T, Wang Q, Pang Y. Retracted Article: Long non-coding RNA KCNQ1OT1 regulates cell proliferation, apoptosis and chemo-sensitivity through modulating the miR-186-5p/NCAM1 axis in acute myeloid leukemia cells. RSC Adv 2019; 9:36256-36265. [PMID: 35540579 PMCID: PMC9074956 DOI: 10.1039/c9ra06378a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 10/25/2019] [Indexed: 11/21/2022] Open
Abstract
Recent studies show that lncRNA KCNQ1OT1 and microRNA-186-5p (miR-186-5p) are involved in various human cancers. Moreover, it is reported that KCNQ1OT1 expression is upregulated in acute myeloid leukemia (AML). However, their roles in AML remain unknown. This study aimed to reveal the functional mechanism of KCNQ1OT1 and miR-186-5p in AML development. Quantitative real time polymerase chain reaction (qRT-PCR) was performed to detect the levels of genes. Cell proliferation and apoptosis were assessed by a 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay and flow cytometry analysis respectively. A western blot assay was carried out to examine the protein levels. In addition, the interaction between miR-186-5p and KCNQ1OT1 or neural cell adhesion molecule 1 (NCAM1) was predicted by bioinformatics analysis tool starbase2.0 and confirmed by the dual luciferase reporter assay. KCNQ1OT1 and NCAM1 expressions were increased and miR-186-5p expression was decreased in AML samples and cells. The depletion of KCNQ1OT1 inhibited cell proliferation, and promoted apoptosis and chemo-sensitivity in AML. In addition, the upregulation of miR-186-5p suppressed AML cell proliferation, and induced apoptosis and chemo-sensitivity. Interestingly, KCNQ1OT1 directly downregulated miR-186-5p expression and miR-186-5p decreased NCAM1 expression by binding to the 3′ untranslated region (UTR) of NCAM1 mRNA. Furthermore, miR-186-5p knockdown or NCAM1 overexpression reversed the effects of KCNQ1OT1 depletion on AML cell progression. Our results firstly revealed a linear relationship between KCNQ1OT1, miR-186-5p, and NCAM1, and demonstrated that KCNQ1OT1 mediated AML cell progression via regulating the miR-186-5p/NCAM1 axis, revealing functional mechanisms of KCNQ1OT1 and miR-186-5p in AML development. Recent studies show that lncRNA KCNQ1OT1 and microRNA-186-5p (miR-186-5p) are involved in various human cancers.![]()
Collapse
Affiliation(s)
- Jing Dai
- Department of Hematology
- Zhoukou Central Hospital
- Zhengzhou
- China
| | - Kai Wang
- Department of Hematology
- Zhoukou Central Hospital
- Zhengzhou
- China
| | - Tao Liu
- Department of Hematology
- Zhoukou Central Hospital
- Zhengzhou
- China
| | - Qiong Wang
- Department of Hematology
- Zhoukou Central Hospital
- Zhengzhou
- China
| | - Yingxu Pang
- Department of Hematology
- Zhoukou Central Hospital
- Zhengzhou
- China
| |
Collapse
|
15324
|
De Los Santos MC, Dragomir MP, Calin GA. The role of exosomal long non-coding RNAs in cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:1178-1192. [PMID: 31867576 PMCID: PMC6924635 DOI: 10.20517/cdr.2019.74] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
One of the major challenges in oncology is drug resistance, which triggers relapse and shortens patients’ survival. In order to promote drug desensitization, cancer cells require the establishment of an ideal tumor microenvironment that accomplishes specific conditions. To achieve this objective, cellular communication is a key factor. Classically, cells were believed to restrictively communicate by ligand-receptor binding, physical cell-to-cell interactions and synapses. Nevertheless, the crosstalk between tumor cells and stroma cells has also been recently reported to be mediated through exosomes, the smallest extracellular vesicles, which transport a plethora of functionally active molecules, such as: proteins, lipids, messenger RNA, DNA, microRNA or long non-coding RNA (lncRNAs). LncRNAs are RNA molecules greater than 200 base pairs that are deregulated in cancer and other diseases. Exosomal lncRNAs are highly stable and can be found in several body fluids, being considered potential biomarkers for tumor liquid biopsy. Exosomal lncRNAs promote angiogenesis, cell proliferation and drug resistance. The role of exosomal lncRNAs in drug resistance affects the main treatment strategies in oncology: chemotherapy, targeted therapy, hormone therapy and immunotherapy. Overall, knowing the molecular mechanisms by which exosomal lncRNA induce pharmacologic resistance could improve further drug development and identify drug resistance biomarkers.
Collapse
Affiliation(s)
- Mireia Cruz De Los Santos
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Mihnea P Dragomir
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.,Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 40015, Romania.,Department of Surgery, Fundeni Clinical Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest 022328, Romania
| | - George A Calin
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.,Center for RNA Interference and Non-coding RNAs, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| |
Collapse
|
15325
|
Abstract
Higher organisms are all born with general immunity as well as with, increasingly, more specific immune systems. All immune mechanisms function with the intent of aiding the body in defense against infection. Internal and external factors alike have varying effects on the immune system, and the immune response is tailored specifically to each one. Accompanying the components of the human innate and adaptive immune systems are the other intermingling systems of the human body. Increasing understanding of the body's immune interactions with other systems has opened new avenues of study, including that of the microbiome. The microbiome has become a highly active area of research over the last 10 to 20 years since the NIH began funding the Human Microbiome Project (HMP), which was established in 2007. Several publications have focused on the characterization, functions, and complex interplay of the microbiome as it relates to the rest of the body. A dysfunction between the microbiome and the host has been linked to various diseases including cancers, metabolic deficiencies, autoimmune disorders, and infectious diseases. Further understanding of the microbiome and its interaction with the host in relation to diseases is needed in order to understand the implications of microbiome dysfunction and the possible use of microbiota in the prevention of disease. In this review, we have summarized information on the immune system, the microbiome, the microbiome's interplay with other systems, and the association of the immune system and the microbiome in diseases such as diabetes and colorectal cancer.
Collapse
Affiliation(s)
| | - Sohail Siraj
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX
| | - Krishna Patel
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX
| | - Umesh T. Sankpal
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX
| | - Stephen Mathew
- Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX
| | - Riyaz Basha
- Graduate School of Biomedical Sciences, UNT Health Science Center, Fort Worth, TX
- Texas College of Osteopathic Medicine, UNT Health Science Center, Fort Worth, TX
| |
Collapse
|