1
|
Xiao H, Chen H, Zhang L, Duolikun M, Zhen B, Kuerban S, Li X, Wang Y, Chen L, Lin J. Cytoskeletal gene alterations linked to sorafenib resistance in hepatocellular carcinoma. World J Surg Oncol 2024; 22:152. [PMID: 38849867 PMCID: PMC11157844 DOI: 10.1186/s12957-024-03417-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/20/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Although sorafenib has been consistently used as a first-line treatment for advanced hepatocellular carcinoma (HCC), most patients will develop resistance, and the mechanism of resistance to sorafenib needs further study. METHODS Using KAS-seq technology, we obtained the ssDNA profiles within the whole genome range of SMMC-7721 cells treated with sorafenib for differential analysis. We then intersected the differential genes obtained from the analysis of hepatocellular carcinoma patients in GSE109211 who were ineffective and effective with sorafenib treatment, constructed a PPI network, and obtained hub genes. We then analyzed the relationship between the expression of these genes and the prognosis of hepatocellular carcinoma patients. RESULTS In this study, we identified 7 hub ERGs (ACTB, CFL1, ACTG1, ACTN1, WDR1, TAGLN2, HSPA8) related to drug resistance, and these genes are associated with the cytoskeleton. CONCLUSIONS The cytoskeleton is associated with sorafenib resistance in hepatocellular carcinoma. Using KAS-seq to analyze the early changes in tumor cells treated with drugs is feasible for studying the drug resistance of tumors, which provides reference significance for future research.
Collapse
Affiliation(s)
- Hong Xiao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Hainan, China
| | - Hangyu Chen
- Department of Pharmacy, Peking University Third Hospital, 49 Huayuan North Rd, Haidian District, Beijing, 100191, China
| | - Lei Zhang
- Department of Pharmacy, Peking University Third Hospital, 49 Huayuan North Rd, Haidian District, Beijing, 100191, China
| | - Maimaitiyasen Duolikun
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Hainan, China
| | - Baixin Zhen
- Department of Pharmacology, Xinjiang Medical University, Urumqi, China
| | - Subinuer Kuerban
- Department of Pharmacology, Xinjiang Medical University, Urumqi, China
| | - Xuehui Li
- Department of Pharmacology, Xinjiang Medical University, Urumqi, China
| | - Yuxi Wang
- Department of Pharmacy, Peking University Third Hospital, 49 Huayuan North Rd, Haidian District, Beijing, 100191, China
| | - Long Chen
- Department of Pharmacy, Peking University Third Hospital, 49 Huayuan North Rd, Haidian District, Beijing, 100191, China.
- Peking University, Third Hospital Cancer Center, 49 Huayuan North Rd, Haidian District, Beijing, 100191, China.
| | - Jian Lin
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Hainan, China.
- Department of Pharmacy, Peking University Third Hospital, 49 Huayuan North Rd, Haidian District, Beijing, 100191, China.
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Peking University, 49 Huayuan North Rd, Haidian District, Beijing, 100191, China.
- Peking University, Third Hospital Cancer Center, 49 Huayuan North Rd, Haidian District, Beijing, 100191, China.
| |
Collapse
|
2
|
Liu Y, Zhou Y, Chen P. Lung cancer organoids: models for preclinical research and precision medicine. Front Oncol 2023; 13:1293441. [PMID: 37941550 PMCID: PMC10628480 DOI: 10.3389/fonc.2023.1293441] [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: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 11/10/2023] Open
Abstract
Lung cancer is a malignancy with high incidence and mortality rates globally, and it has a 5-year survival rate of only 10%-20%. The significant heterogeneity in clinical presentation, histological features, multi-omics findings, and drug sensitivity among different lung cancer patients necessitate the development of personalized treatment strategies. The current precision medicine for lung cancer, primarily based on pathological and genomic multi-omics testing, fails to meet the needs of patients with clinically refractory lung cancer. Lung cancer organoids (LCOs) are derived from tumor cells within tumor tissues and are generated through three-dimensional tissue culture, enabling them to faithfully recapitulate in vivo tumor characteristics and heterogeneity. The establishment of a series of LCOs biobanks offers promising platforms for efficient screening and identification of novel targets for anti-tumor drug discovery. Moreover, LCOs provide supplementary decision-making factors to enhance the current precision medicine for lung cancer, thereby addressing the limitations associated with pathology-guided approaches in managing refractory lung cancer. This article presents a comprehensive review on the construction methods and potential applications of LCOs in both preclinical and clinical research. It highlights the significance of LCOs in biomarker exploration, drug resistance investigation, target identification, clinical precision drug screening, as well as microfluidic technology-based high-throughput drug screening strategies. Additionally, it discusses the current limitations and future prospects of this field.
Collapse
Affiliation(s)
- Yajing Liu
- School of Pharmacy, Qingdao University, Qingdao, China
- Research and Development Department, NanoPeptide (Qingdao) Biotechnology Ltd., Qingdao, China
| | - Yanbing Zhou
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Pu Chen
- Research and Development Department, NanoPeptide (Qingdao) Biotechnology Ltd., Qingdao, China
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
3
|
Fulton MD, Najahi-Missaoui W. Liposomes in Cancer Therapy: How Did We Start and Where Are We Now. Int J Mol Sci 2023; 24:ijms24076615. [PMID: 37047585 PMCID: PMC10095497 DOI: 10.3390/ijms24076615] [Citation(s) in RCA: 41] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
Since their first discovery in the 1960s by Alec Bangham, liposomes have been shown to be effective drug delivery systems for treating various cancers. Several liposome-based formulations received approval by the U.S. Food and Drug Administration (FDA) and European Medicines Agency (EMA), with many others in clinical trials. Liposomes have several advantages, including improved pharmacokinetic properties of the encapsulated drug, reduced systemic toxicity, extended circulation time, and targeted disposition in tumor sites due to the enhanced permeability and retention (EPR) mechanism. However, it is worth noting that despite their efficacy in treating various cancers, liposomes still have some potential toxicity and lack specific targeting and disposition. This explains, in part, why their translation into the clinic has progressed only incrementally, which poses the need for more research to focus on addressing such translational limitations. This review summarizes the main properties of liposomes, their current status in cancer therapy, and their limitations and challenges to achieving maximal therapeutic efficacy.
Collapse
Affiliation(s)
- Melody D. Fulton
- Department of Chemistry, College of Arts and Sciences, Washington State University, Pullman, WA 99164, USA
| | - Wided Najahi-Missaoui
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
4
|
Mehta A. Tracking the Development of Cancer Care After 75 Years of Independence: India's Fight Against Cancer Since 1947. Indian J Surg Oncol 2022; 13:12-26. [PMID: 36691502 PMCID: PMC9859970 DOI: 10.1007/s13193-022-01689-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 12/12/2022] [Indexed: 01/13/2023] Open
Abstract
India is one of the fastest developing countries with tremendous growth in industrialization and healthcare facilities. Research and development in the field of healthcare improved the quality of life and well-being of our population. Despite the availability of healthcare facilities and infrastructure, we are still facing considerable challenges in the prevention, diagnosis, and treatment of cancer. The present review focuses on the history and development of cancer care facilities since independence. The advances in cancer diagnostics for early detection of cancer and developments in the field of conventional surgery, including laparoscopic and robotic surgeries, chemotherapy, and radiation therapy, are reviewed. Immunotherapy, vaccines, and selective targeting of tumor cells using nanotechnology are emerging areas in the field of cancer research.
Collapse
Affiliation(s)
- Ashok Mehta
- Nanavati Max Super Speciality Hospital, Mumbai, India
- L S Raheja Hospital, Mumbai, India
- HCG Cancer Centre Colaba, Mumbai, India
| |
Collapse
|
5
|
Furrer D, Dragic D, Chang SL, Fournier F, Droit A, Jacob S, Diorio C. Association between genome-wide epigenetic and genetic alterations in breast cancer tissue and response to HER2-targeted therapies in HER2-positive breast cancer patients: new findings and a systematic review. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 5:995-1015. [PMID: 36627894 PMCID: PMC9771759 DOI: 10.20517/cdr.2022.63] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/25/2022] [Accepted: 10/08/2022] [Indexed: 01/12/2023]
Abstract
Recent evidence suggests that genetic and epigenetic mechanisms might be associated with acquired resistance to cancer therapies. The aim of this study was to assess the association of genome-wide genetic and epigenetic alterations with the response to anti-HER2 agents in HER2-positive breast cancer patients. PubMed was screened for articles published until March 2021 on observational studies investigating the association of genome-wide genetic and epigenetic alterations, measured in breast cancer tissues or blood, with the response to targeted treatment in HER2-positive breast cancer patients. Sixteen studies were included in the review along with ours, in which we compared the genome-wide DNA methylation pattern in breast tumor tissues of patients who acquired resistance to treatment (case group, n = 6) to that of patients who did not develop resistance (control group, n = 6). Among genes identified as differentially methylated between the breast cancer tissue of cases and controls, one of them, PRKACA, was also reported as differentially expressed in two studies included in the review. Although included studies were heterogeneous in terms of methodology and study population, our review suggests that genes of the PI3K pathway may play an important role in developing resistance to anti-HER2 agents in breast cancer patients. Genome-wide genetic and epigenetic alterations measured in breast cancer tissue or blood might be promising markers of resistance to anti-HER2 agents in HER2-positive breast cancer patients. Further studies are needed to confirm these data.
Collapse
Affiliation(s)
- Daniela Furrer
- Centre de Recherche sur le cancer de l’Université Laval, 1050 Avenue de la Médecine, Québec, QC G1V 0A6, Canada.,Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1S 4L8, Canada. ,Département de médecine sociale et préventive, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Dzevka Dragic
- Centre de Recherche sur le cancer de l’Université Laval, 1050 Avenue de la Médecine, Québec, QC G1V 0A6, Canada.,Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1S 4L8, Canada. ,Département de médecine sociale et préventive, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada.,Université Paris-Saclay, UVSQ, Inserm, CESP U1018, Exposome and Heredity Team, Gustave Roussy, Villejuif 94807, France
| | - Sue-Ling Chang
- Centre de Recherche sur le cancer de l’Université Laval, 1050 Avenue de la Médecine, Québec, QC G1V 0A6, Canada.,Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1S 4L8, Canada
| | - Frédéric Fournier
- Centre de Recherche sur le cancer de l’Université Laval, 1050 Avenue de la Médecine, Québec, QC G1V 0A6, Canada.,Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1S 4L8, Canada. ,Département de médecine moléculaire, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Arnaud Droit
- Centre de Recherche sur le cancer de l’Université Laval, 1050 Avenue de la Médecine, Québec, QC G1V 0A6, Canada.,Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1S 4L8, Canada. ,Département de médecine moléculaire, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Simon Jacob
- Centre de Recherche sur le cancer de l’Université Laval, 1050 Avenue de la Médecine, Québec, QC G1V 0A6, Canada.,Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1S 4L8, Canada. ,Département de biologie moléculaire, de biochimie médicale et de pathologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada.,Centre des Maladies du Sein, Hôpital du Saint-Sacrement, Québec, QC G1S 4L8, Canada
| | - Caroline Diorio
- Centre de Recherche sur le cancer de l’Université Laval, 1050 Avenue de la Médecine, Québec, QC G1V 0A6, Canada.,Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1S 4L8, Canada. ,Département de médecine sociale et préventive, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada.,Centre des Maladies du Sein, Hôpital du Saint-Sacrement, Québec, QC G1S 4L8, Canada.,Correspondence to: Prof. Caroline Diorio, Axe Oncologie, Centre de Recherche du CHU de Québec-Université Laval, 1050 chemin Ste-Foy, Québec, QC G1S 4L8, Canada. E-mail:
| |
Collapse
|
6
|
Network Biology and Artificial Intelligence Drive the Understanding of the Multidrug Resistance Phenotype in Cancer. Drug Resist Updat 2022; 60:100811. [DOI: 10.1016/j.drup.2022.100811] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 02/07/2023]
|
7
|
Cao L, Zhu Y, Wang W, Wang G, Zhang S, Cheng H. Emerging Nano-Based Strategies Against Drug Resistance in Tumor Chemotherapy. Front Bioeng Biotechnol 2021; 9:798882. [PMID: 34950650 PMCID: PMC8688801 DOI: 10.3389/fbioe.2021.798882] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/19/2021] [Indexed: 02/05/2023] Open
Abstract
Drug resistance is the most significant causes of cancer chemotherapy failure. Various mechanisms of drug resistance include tumor heterogeneity, tumor microenvironment, changes at cellular levels, genetic factors, and other mechanisms. In recent years, more attention has been paid to tumor resistance mechanisms and countermeasures. Nanomedicine is an emerging treatment platform, focusing on alternative drug delivery and improved therapeutic effectiveness while reducing side effects on normal tissues. Here, we reviewed the principal forms of drug resistance and the new possibilities that nanomaterials offer for overcoming these therapeutic barriers. Novel nanomaterials based on tumor types are an excellent modality to equalize drug resistance that enables gain more rational and flexible drug selectivity for individual patient treatment. With the emergence of advanced designs and alternative drug delivery strategies with different nanomaterials, overcome of multidrug resistance shows promising and opens new horizons for cancer therapy. This review discussed different mechanisms of drug resistance and recent advances in nanotechnology-based therapeutic strategies to improve the sensitivity and effectiveness of chemotherapeutic drugs, aiming to show the advantages of nanomaterials in overcoming of drug resistance for tumor chemotherapy, which could accelerate the development of personalized medicine.
Collapse
Affiliation(s)
- Lei Cao
- Department of Pathology, Quanzhou Women’s and Children’s Hospital, Quanzhou, China
| | - Yuqin Zhu
- Department of Pathology, Quanzhou Women’s and Children’s Hospital, Quanzhou, China
| | - Weiju Wang
- Department of Pathology, Qingyuan Maternal and Child Health Hospital, Qingyuan, China
| | - Gaoxiong Wang
- Department of Pathology, Quanzhou Women’s and Children’s Hospital, Quanzhou, China
| | - Shuaishuai Zhang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Hongwei Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| |
Collapse
|
8
|
Xue G, Wang Z, Zheng N, Fang J, Mao C, Li X, Jin G, Ming X, Lu Y. Elimination of acquired resistance to PD-1 blockade via the concurrent depletion of tumour cells and immunosuppressive cells. Nat Biomed Eng 2021; 5:1306-1319. [PMID: 34725506 PMCID: PMC8595849 DOI: 10.1038/s41551-021-00799-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 08/26/2021] [Indexed: 12/12/2022]
Abstract
Antigen release resulting from the death of tumour cells induced by chemotherapies and targeted therapies can augment the antitumour responses induced by immune checkpoint blockade (ICB). However, tumours responding to ICB therapies often become resistant to them. Here we show that the specific targeting of tumour cells promotes the growth of tumour-cell variants that are resistant to ICB, and that the acquired resistance can be overcome via the concurrent depletion of tumour cells and of major types of immunosuppressive cell via a monoclonal antibody binding the enzyme CD73, which we identified as highly expressed on tumour cells and on regulatory T cells, myeloid-derived suppressor cells and tumour-associated macrophages, but not on cytolytic T lymphocytes, natural killer cells and dendritic cells. In mice with murine tumours, the systemic administration of anti-PD1 antibodies and anti-CD73 antibodies conjugated to a near-infrared dye prevented near-infrared-irradiated tumours from acquiring resistance to ICB and resulted in the eradication of advanced tumours. The elimination of immunosuppressive cells may overcome acquired resistance to ICB across a range of tumour types and combination therapies.
Collapse
Affiliation(s)
- Gang Xue
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC, USA, 27157; Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27101
| | - Ziyu Wang
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC, USA, 27157; Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27101
| | - Ningbo Zheng
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC, USA, 27157; Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27101
| | - Jing Fang
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC, USA, 27157; Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27101
| | - Chengqiong Mao
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC, USA, 27157; Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27157
| | - Xiaoyin Li
- Department of Mathematics and Statistics, St. Cloud State University, St Cloud, MN, USA, 56301
| | - Guangxu Jin
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC, USA, 27157; Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27157
| | - Xin Ming
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC, USA, 27157; Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27157
| | - Yong Lu
- Comprehensive Cancer Center, Wake Forest Baptist Health, Winston-Salem, NC, USA, 27157; Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC, USA, 27101,Corresponding authors, , Correspondence and requests for materials should be addressed to Y.L
| |
Collapse
|
9
|
miR-21 modulates cisplatin resistance of gastric cancer cells by inhibiting autophagy via the PI3K/Akt/mTOR pathway. Anticancer Drugs 2021; 31:385-393. [PMID: 31913198 DOI: 10.1097/cad.0000000000000886] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Resistance to cisplatin (DDP) remains a major obstacle in the control of gastric cancer (GC) progression. A previous study revealed that microRNA-21 (miR-21) contributes to DDP resistance in GC cells via the phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) pathway. The aim of the current study was to explore the mechanisms underlying the cytoprotective function of miR-21. In this study, DDP-resistant GC cells were obtained by continuous exposure of human gastric adenocarcinoma cells to increasing concentrations of DDP. Western blot analysis was used to evaluate activation of the PI3K/Akt/mechanistic target of rapamycin kinase (mTOR) pathway. The level of miR-21 was altered by transfection of miR-21 mimic and inhibitor. Autophagy was assessed by detecting autophagosome formation, Beclin-1 and LC3 expression. An Annexin V-propidium iodide assay was performed to estimate the survival and death of GC cells. GC cells became refractory to the growth inhibition and apoptosis induced by DDP treatment, activation of Akt and mTOR were increased in DDP-resistant GC cells. Inhibition of autophagy decreased the sensitivity of GC cells to DDP, and autophagy induction produced the opposite effect. DDP-resistant GC cells expressed higher levels of miR-21 compared with the parent cells. Transfection of GC cells with miR-21 mimics contributed to restored DDP resistance by suppressing autophagy, while miR-21 inhibitor sensitized DDP-resistant GC cells by promoting autophagy. In conclusion, the results demonstrated that miR-21 is associated with DDP resistance in GC cells by inhibiting autophagy via the PI3K/Akt/mTOR pathway, and autophagy inducers could be therapeutic targets for the effective treatment of DDP resistance in GC.
Collapse
|
10
|
Chaudhary R, Slebos RJC, Song F, McCleary-Sharpe KP, Masannat J, Tan AC, Wang X, Amaladas N, Wu W, Hall GE, Conejo-Garcia JR, Hernandez-Prera JC, Chung CH. Effects of checkpoint kinase 1 inhibition by prexasertib on the tumor immune microenvironment of head and neck squamous cell carcinoma. Mol Carcinog 2021; 60:138-150. [PMID: 33378592 PMCID: PMC7856233 DOI: 10.1002/mc.23275] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 12/20/2022]
Abstract
Prognosis for patients with recurrent and/or metastatic head and neck squamous cell carcinoma (HNSCC) remains poor. Development of more effective and less toxic targeted therapies is necessary for HNSCC patients. Checkpoint kinase 1 (CHK1) plays a vital role in cell cycle regulation and is a promising therapeutic target in HNSCC. Prexasertib, a CHK1 inhibitor, induces DNA damage and cell death, however, its effect on the tumor immune microenvironment (TIME) is largely unknown. Therefore, we evaluated a short-term and long-term effects of prexasertib in HNSCC and its TIME. Prexasertib caused increased DNA damage and cell death in vitro and significant tumor regression and improved survival in vivo. The gene expression and multiplex immunohistochemistry (mIHC) analyses of the in vivo tumors demonstrated increased expression of genes that are related to T-cell activation and increased immune cell trafficking, and decreased expression of genes that related to immunosuppression. However, increased expression of genes related to immunosuppression emerged over time suggesting evasion of immune surveillances. These findings in gene expression analyses were confirmed using mIHC which showed differential modulation of TIME in the tumor margins and as well as cores over time. These results suggest that evasion of immune surveillance, at least in part, may contribute to the acquired resistance to prexasertib in HNSCC.
Collapse
Affiliation(s)
- Ritu Chaudhary
- Department of Head and Neck-Endocrine Oncology, Moffitt
Cancer Center, Tampa, Florida, USA
| | - Robbert J. C. Slebos
- Department of Head and Neck-Endocrine Oncology, Moffitt
Cancer Center, Tampa, Florida, USA
| | - Feifei Song
- Department of Head and Neck-Endocrine Oncology, Moffitt
Cancer Center, Tampa, Florida, USA
| | | | - Jude Masannat
- Department of Head and Neck-Endocrine Oncology, Moffitt
Cancer Center, Tampa, Florida, USA
| | - Aik Choon Tan
- Department of Biostatistics and Bioinformatics, Moffitt
Cancer Center, Tampa, Florida, USA
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, Moffitt
Cancer Center, Tampa, Florida, USA
| | - Nelusha Amaladas
- Lilly Research Laboratories, Eli Lilly and Company,
Indianapolis, Indiana, USA
| | - Wenjuan Wu
- Lilly Research Laboratories, Eli Lilly and Company,
Indianapolis, Indiana, USA
| | - Gerald E. Hall
- Lilly Research Laboratories, Eli Lilly and Company,
Indianapolis, Indiana, USA
| | | | | | - Christine H. Chung
- Department of Head and Neck-Endocrine Oncology, Moffitt
Cancer Center, Tampa, Florida, USA
| |
Collapse
|
11
|
Mele L, Del Vecchio V, Liccardo D, Prisco C, Schwerdtfeger M, Robinson N, Desiderio V, Tirino V, Papaccio G, La Noce M. The role of autophagy in resistance to targeted therapies. Cancer Treat Rev 2020; 88:102043. [PMID: 32505806 DOI: 10.1016/j.ctrv.2020.102043] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/20/2020] [Accepted: 05/22/2020] [Indexed: 02/06/2023]
Abstract
Autophagy is a self-degradative cellular process, involved in stress response such as starvation, hypoxia, and oxidative stress. This mechanism balances macro-molecule recycling to regulate cell homeostasis. In cancer, autophagy play a role in the development and progression, while several studies describe it as one of the key processes in drug resistance. In the last years, in addition to standard anti-cancer treatments such as chemotherapies and irradiation, targeted therapy became one of the most adopted strategies in clinical practices, mainly due to high specificity and reduced side effects. However, similar to standard treatments, drug resistance is the main challenge in most patients. Here, we summarize recent studies that investigated the role of autophagy in drug resistance after targeted therapy in different types of cancers. We highlight positive results and limitations of pre-clinical and clinical studies in which autophagy inhibitors are used in combination with targeted therapies.
Collapse
Affiliation(s)
- Luigi Mele
- Department of Experimental Medicine, University of Campania "L. Vanvitelli" Naples, Italy
| | - Vitale Del Vecchio
- Department of Experimental Medicine, University of Campania "L. Vanvitelli" Naples, Italy
| | - Davide Liccardo
- Department of Experimental Medicine, University of Campania "L. Vanvitelli" Naples, Italy
| | - Claudia Prisco
- Department of Experimental Medicine, University of Campania "L. Vanvitelli" Naples, Italy; The John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK
| | - Melanie Schwerdtfeger
- Department of Experimental Medicine, University of Campania "L. Vanvitelli" Naples, Italy; Department of Medicine IV -Division of Clinical Pharmacology-University of Munich, Germany
| | - Nirmal Robinson
- Centre for Cancer Biology, SA Pathology and University of South Australia, GPO Box 2471, Adelaide, Australia
| | - Vincenzo Desiderio
- Department of Experimental Medicine, University of Campania "L. Vanvitelli" Naples, Italy
| | - Virginia Tirino
- Department of Experimental Medicine, University of Campania "L. Vanvitelli" Naples, Italy
| | - Gianpaolo Papaccio
- Department of Experimental Medicine, University of Campania "L. Vanvitelli" Naples, Italy.
| | - Marcella La Noce
- Department of Experimental Medicine, University of Campania "L. Vanvitelli" Naples, Italy
| |
Collapse
|
12
|
|
13
|
Hippo/YAP Signaling Pathway: A Promising Therapeutic Target in Bone Paediatric Cancers? Cancers (Basel) 2020; 12:cancers12030645. [PMID: 32164350 PMCID: PMC7139637 DOI: 10.3390/cancers12030645] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/06/2020] [Accepted: 03/07/2020] [Indexed: 12/11/2022] Open
Abstract
Osteosarcoma and Ewing sarcoma are the most prevalent bone pediatric tumors. Despite intensive basic and medical research studies to discover new therapeutics and to improve current treatments, almost 40% of osteosarcoma and Ewing sarcoma patients succumb to the disease. Patients with poor prognosis are related to either the presence of metastases at diagnosis or resistance to chemotherapy. Over the past ten years, considerable interest for the Hippo/YAP signaling pathway has taken place within the cancer research community. This signaling pathway operates at different steps of tumor progression: Primary tumor growth, angiogenesis, epithelial to mesenchymal transition, and metastatic dissemination. This review discusses the current knowledge about the involvement of the Hippo signaling pathway in cancer and specifically in paediatric bone sarcoma progression.
Collapse
|
14
|
Zhou J, Liu Y, Zhang Y, Li Q, Cao Y. Modeling Tumor Evolutionary Dynamics to Predict Clinical Outcomes for Patients with Metastatic Colorectal Cancer: A Retrospective Analysis. Cancer Res 2020; 80:591-601. [PMID: 31676575 PMCID: PMC7002273 DOI: 10.1158/0008-5472.can-19-1940] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/05/2019] [Accepted: 10/28/2019] [Indexed: 01/22/2023]
Abstract
Over 50% of colorectal cancer patients develop resistance after a transient response to therapy. Understanding tumor resistance from an evolutionary perspective leads to better predictions of treatment outcomes. The objectives of this study were to develop a computational framework to analyze tumor longitudinal measurements and recapitulate the individual evolutionary dynamics in metastatic colorectal cancer (mCRC) patients. A stochastic modeling framework was developed to depict the whole spectrum of tumor evolution prior to diagnosis and during and after therapy. The evolutionary model was optimized using a nonlinear mixed effect (NLME) method based on the longitudinal measurements of liver metastatic lesions from 599 mCRC patients. The deterministic limits in the NLME model were applied to optimize the stochastic model for each patient. Cox proportional hazards models coupled with the least absolute shrinkage and selection operator (LASSO) algorithm were applied to predict patients' progression-free survival (PFS) and overall survival (OS). The stochastic evolutionary model well described the longitudinal profiles of tumor sizes. The evolutionary parameters optimized for each patient indicated substantial interpatient variability. The number of resistant subclones at diagnosis was found to be a significant predictor to survival, and the hazard ratios with 95% CI were 1.09 (0.79-1.49) and 1.54 (1.01-2.34) for patients with three or more resistant subclones. Coupled with several patient characteristics, evolutionary parameters strongly predict patients' PFS and OS. A stochastic computational framework was successfully developed to recapitulate individual patient evolutionary dynamics, which could predict clinical survival outcomes in mCRC patients. SIGNIFICANCE: A data analysis framework depicts the individual evolutionary dynamics of mCRC patients and can be generalized to project patient survival outcomes.
Collapse
Affiliation(s)
- Jiawei Zhou
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Yutong Liu
- School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Yubo Zhang
- Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Quefeng Li
- School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
15
|
Abstract
The tumor microenvironment (TME) is a complex ecosystem, including blood vessels,
immune cells, fibroblasts, extracellular matrix, cytokines, hormones, and so on.
The TME differs from the normal tissue environment (NTE) in many aspects, such
as tissue architecture, chronic inflammation, level of oxygen and pH,
nutritional state of the cells, as well as tissue firmness. The NTE can inhibit
the growth of cancer at the early tumorigenesis phase, whereas the TME promotes
the growth of cancer in general, although it may have some anticancer effects.
In particular, the TME plays a crucial role in the generation and maintenance of
cancer stem cells, which lie at the root of cancer growth. Therefore,
normalization of the TME to the NTE may inhibit cancer growth or improve cancer
therapeutic efficiency. This review focuses on the recent emerging approaches
for this normalization and the action mechanisms.
Collapse
Affiliation(s)
- Jie Zheng
- 1 Southeast University, Nanjing, China
| | - Peng Gao
- 2 Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
16
|
Finn KJ, Martin SE, Settleman J. A Single-Step, High-Dose Selection Scheme Reveals Distinct Mechanisms of Acquired Resistance to Oncogenic Kinase Inhibition in Cancer Cells. Cancer Res 2020; 80:79-90. [PMID: 31641034 DOI: 10.1158/0008-5472.can-19-0729] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 09/11/2019] [Accepted: 10/11/2019] [Indexed: 11/16/2022]
Abstract
Despite the remarkable clinical efficacy demonstrated by molecularly targeted cancer therapeutics, the benefits are typically temporary due to the emergence of acquired drug resistance. This has spurred a massive effort by the cancer research community to identify mechanisms used by cancer cells to evade treatment. Among the various methodologies developed and employed to identify such mechanisms, the most commonly used approach has been to model acquired resistance by exposing cancer cells in culture to gradually increasing concentrations of drug over an extended period of time. Here, we employed a less commonly used variation on this approach, wherein resistant cells are selected by immediately exposing cancer cells to a continuous, high concentration of drug. Using this approach, we isolated clones representing three distinct mechanisms of resistance to inhibition of MET kinase activity from a single clonally derived cancer cell line. The emergent clones had acquired resistance through engagement of alternative receptor tyrosine kinases either through upregulation of FGF3 or HBEGF or increased MAPK signaling through an activating V600E mutation in BRAF. Importantly, these mechanisms were not identified using the conventional "ramp-up" approach in previous studies that employed the same cell line. These results suggest that the particular nature of the selection scheme employed in cell culture modeling studies can determine which potential resistance mechanisms are identified and which ones may be missed, highlighting the need for careful consideration of the specific approach used to model resistance in cultured cells. SIGNIFICANCE: Through modeling resistance to MET kinase inhibition in cultured cancer cells using single-step, high-dose selection, these findings highlight that the specific nature of the selection protocol impacts which resistance mechanisms are identified.See related commentary by Floros et al., p. 25.
Collapse
Affiliation(s)
- Kenneth J Finn
- Calico Life Sciences LLC, South San Francisco, California
| | - Scott E Martin
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, California
| | - Jeff Settleman
- Calico Life Sciences LLC, South San Francisco, California.
| |
Collapse
|
17
|
Malone T, Schäfer L, Simon N, Heavey S, Cuffe S, Finn S, Moore G, Gately K. Current perspectives on targeting PIM kinases to overcome mechanisms of drug resistance and immune evasion in cancer. Pharmacol Ther 2019; 207:107454. [PMID: 31836451 DOI: 10.1016/j.pharmthera.2019.107454] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/03/2019] [Indexed: 12/22/2022]
Abstract
PIM kinases are a class of serine/threonine kinases that play a role in several of the hallmarks of cancer including cell cycle progression, metabolism, inflammation and immune evasion. Their constitutively active nature and unique catalytic structure has led them to be an attractive anticancer target through the use of small molecule inhibitors. This review highlights the enhanced activity of PIM kinases in cancer that can be driven by hypoxia in the tumour microenvironment and the important role that aberrant PIM kinase activity plays in resistance mechanisms to chemotherapy, radiotherapy, anti-angiogenic therapies and targeted therapies. We highlight an interaction of PIM kinases with numerous major oncogenic players, including but not limited to, stabilisation of p53, synergism with c-Myc, and notable parallel signalling with PI3K/Akt. We provide a comprehensive overview of PIM kinase's role as an escape mechanism to targeted therapies including PI3K/mTOR inhibitors, MET inhibitors, anti-HER2/EGFR treatments and the immunosuppressant rapamycin, providing a rationale for co-targeting treatment strategies for a more durable patient response. The current status of PIM kinase inhibitors and their use as a combination therapy with other targeted agents, in addition to the development of novel multi-molecularly targeted single therapeutic agents containing a PIM kinase targeting moiety are discussed.
Collapse
Affiliation(s)
- Tom Malone
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Lea Schäfer
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Nathalie Simon
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Susan Heavey
- Molecular Diagnostics and Therapeutics Group, University College London, London, UK
| | - Sinead Cuffe
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Stephen Finn
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Gillian Moore
- School of Pharmacy and Biomolecular Sciences, RCSI, Dublin, Ireland
| | - Kathy Gately
- Dept. of Clinical Medicine, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland.
| |
Collapse
|
18
|
Hatlen MA, Schmidt-Kittler O, Sherwin CA, Rozsahegyi E, Rubin N, Sheets MP, Kim JL, Miduturu C, Bifulco N, Brooijmans N, Shi H, Guzi T, Boral A, Lengauer C, Dorsch M, Kim RD, Kang YK, Wolf BB, Hoeflich KP. Acquired On-Target Clinical Resistance Validates FGFR4 as a Driver of Hepatocellular Carcinoma. Cancer Discov 2019; 9:1686-1695. [PMID: 31575540 DOI: 10.1158/2159-8290.cd-19-0367] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/26/2019] [Accepted: 09/26/2019] [Indexed: 11/16/2022]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer mortality worldwide with no clinically confirmed oncogenic driver. Although preclinical studies implicate the FGF19 receptor FGFR4 in hepatocarcinogenesis, the dependence of human cancer on FGFR4 has not been demonstrated. Fisogatinib (BLU-554) is a potent and selective inhibitor of FGFR4 and demonstrates clinical benefit and tumor regression in patients with HCC with aberrant FGF19 expression. Mutations were identified in the gatekeeper and hinge-1 residues in the kinase domain of FGFR4 upon disease progression in 2 patients treated with fisogatinib, which were confirmed to mediate resistance in vitro and in vivo. A gatekeeper-agnostic, pan-FGFR inhibitor decreased HCC xenograft growth in the presence of these mutations, demonstrating continued FGF19-FGFR4 pathway dependence. These results validate FGFR4 as an oncogenic driver and warrant further therapeutic targeting of this kinase in the clinic. SIGNIFICANCE: Our study is the first to demonstrate on-target FGFR4 kinase domain mutations as a mechanism of acquired clinical resistance to targeted therapy. This further establishes FGF19-FGFR4 pathway activation as an oncogenic driver. These findings support further investigation of fisogatinib in HCC and inform the profile of potential next-generation inhibitors.See related commentary by Subbiah and Pal, p. 1646.This article is highlighted in the In This Issue feature, p. 1631.
Collapse
MESH Headings
- Aged, 80 and over
- Animals
- Carcinoma, Hepatocellular/diagnostic imaging
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Cell Line, Tumor
- Drug Resistance, Neoplasm
- Female
- Fibroblast Growth Factors/metabolism
- Gene Expression Regulation, Neoplastic
- Humans
- Liver Neoplasms/diagnostic imaging
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Male
- Mice
- Middle Aged
- Models, Molecular
- Mutation
- Neoplasm Transplantation
- Protein Domains
- Pyrans/pharmacology
- Quinazolines/pharmacology
- Receptor, Fibroblast Growth Factor, Type 4/chemistry
- Receptor, Fibroblast Growth Factor, Type 4/genetics
- Receptor, Fibroblast Growth Factor, Type 4/metabolism
Collapse
Affiliation(s)
- Megan A Hatlen
- Blueprint Medicines Corporation, Cambridge, Massachusetts
| | | | | | | | - Nooreen Rubin
- Blueprint Medicines Corporation, Cambridge, Massachusetts
| | | | - Joseph L Kim
- Blueprint Medicines Corporation, Cambridge, Massachusetts
| | | | - Neil Bifulco
- Blueprint Medicines Corporation, Cambridge, Massachusetts
| | | | - Hongliang Shi
- Blueprint Medicines Corporation, Cambridge, Massachusetts
| | - Timothy Guzi
- Blueprint Medicines Corporation, Cambridge, Massachusetts
| | - Andy Boral
- Blueprint Medicines Corporation, Cambridge, Massachusetts
| | | | - Marion Dorsch
- Blueprint Medicines Corporation, Cambridge, Massachusetts
| | - Richard D Kim
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Yoon-Koo Kang
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Beni B Wolf
- Blueprint Medicines Corporation, Cambridge, Massachusetts
| | | |
Collapse
|
19
|
Kesireddy M, Mendiola VL, Jana B, Patel S. Long-term Response to Vismodegib in a Patient with Gorlin-Goltz Syndrome: A Case Report and Review of Pathological Mechanisms Involved. Cureus 2019; 11:e5383. [PMID: 31616614 PMCID: PMC6786843 DOI: 10.7759/cureus.5383] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Nevoid basal cell carcinoma syndrome (NBCCS), also known as Gorlin-Goltz syndrome or Gorlin syndrome, is a rare multisystem disorder with an estimated prevalence of around 1 in 100,000 on average. Vismodegib, an oral smoothened (SMO) inhibitor that blocks the activation of the sonic hedgehog (SHH) pathway, is used in patients with NBCCS. We present an interesting case of a 38-year-old female with Gorlin-Goltz syndrome and her response to vismodegib therapy over two and a half years. She had an excellent initial response to vismodegib for a year during which all her skin basal cell carcinoma (BCC) lesions decreased in size considerably; her dentigerous cysts remained the same size. Though she continued therapy despite several side effects, this was only followed by tumor regrowth and the emergence of new BCC lesions in a more aggressive manner. We discussed the proposed mechanism of resistance to vismodegib (based on our case and literature review) along with its clinical implications. Our case highlights that vismodegib resistance might lead to progression of Gorlin syndrome to a more aggressive version, and points out the need to determine the optimal regimen (combining vismodegib with other agents) and optimal therapy duration (continuous treatment vs. discontinuation after best response) for treatment of NBCCS.
Collapse
Affiliation(s)
- Meghana Kesireddy
- Internal Medicine, The University of Texas Medical Branch, Galveston, USA
| | - Vincent L Mendiola
- Internal Medicine, The University of Texas Medical Branch, Galveston, USA
| | - Bagi Jana
- Hematology Oncology, The University of Texas Medical Branch, Galveston, USA
| | - Shrestha Patel
- Oncology, The University of Texas Medical Branch, Galveston, USA
| |
Collapse
|
20
|
Tung SY, Lin CT, Chen CN, Huang WS. Effect of mitomycin C on X-ray repair cross complementing group 1 expression and consequent cytotoxicity regulation in human gastric cancer cells. J Cell Biochem 2019; 120:8333-8342. [PMID: 30614038 DOI: 10.1002/jcb.28116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/31/2018] [Indexed: 02/07/2023]
Abstract
Gastric cancer is the fourth most common cancer and ranks as the second leading cause of cancer-related deaths across the world. The combination therapy of surgery with chemotherapeutic drugs, that is, mitomycin C (MMC), is becoming a major strategy for patients with advanced gastric cancer. However, drug resistance is a major factor that limits the effectiveness of chemotherapy, which ultimately leads to the failure of cancer chemotherapy. X-ray repair cross complementing group 1 (XRCC1), a scaffold protein of the base excision repair process, has been implicated in the development of tumor chemoresistance. Thus, this study aimed to explore whether XRCC1 expression could be regulated, its role in gastric AGS cancer cells treated with MMC, and the underlying mechanism. The results of this study demonstrate that XRCC1 expression could be upregulated in AGS cells treated with MMC, and this upregulation could subsequently reduce the cytotoxicity of MMC in AGS cells. Furthermore, MMC-upregulated XRCC1 expression was regulated by MAPK signaling through activating the transcription factor Sp1. These results indicate the role of XRCC1 in the development of drug resistance to MMC in gastric AGS cells. Elucidating the mechanism concerning the MAPKs and transcription factor Sp1 may provide another notion for the development of a clinical chemotherapy strategy for gastric cancers in the future.
Collapse
Affiliation(s)
- Shui-Yi Tung
- Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital Chiayi Branch, Chiayi, Taiwan, ROC.,Chang Gung University College of Medicine, Taoyuan, Taiwan, ROC
| | - Chien-Tsong Lin
- Center for General Education, National Formosa University, Yunlin, Taiwan, ROC.,Department of Wood Based Materials and Design, National Chiayi University, Chiayi, Taiwan, ROC
| | - Cheng-Nan Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, Taiwan, ROC
| | - Wen-Shih Huang
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan, ROC
| |
Collapse
|
21
|
Lima ZS, Ghadamzadeh M, Arashloo FT, Amjad G, Ebadi MR, Younesi L. Recent advances of therapeutic targets based on the molecular signature in breast cancer: genetic mutations and implications for current treatment paradigms. J Hematol Oncol 2019; 12:38. [PMID: 30975222 PMCID: PMC6460547 DOI: 10.1186/s13045-019-0725-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 03/27/2019] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most common malignancy in women all over the world. Genetic background of women contributes to her risk of having breast cancer. Certain inherited DNA mutations can dramatically increase the risk of developing certain cancers and are responsible for many of the cancers that run in some families. Regarding the widespread multigene panels, whole exome sequencing is capable of providing the evaluation of genetic function mutations for development novel strategy in clinical trials. Targeting the mutant proteins involved in breast cancer can be an effective therapeutic approach for developing novel drugs. This systematic review discusses gene mutations linked to breast cancer, focusing on signaling pathways that are being targeted with investigational therapeutic strategies, where clinical trials could be potentially initiated in the future are being highlighted.
Collapse
Affiliation(s)
- Zeinab Safarpour Lima
- Shahid Akbar Abadi Clinical Research Development Unit (ShCRDU), Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mostafa Ghadamzadeh
- Departement of Radiology, Hasheminejad Kidney Centre (HKC), Iran University of Medical Sciences, Tehran, Iran
| | | | - Ghazaleh Amjad
- Shahid Akbar Abadi Clinical Research Development Unit (ShCRDU), Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Mohammad Reza Ebadi
- Shohadaye Haft-e-tir Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Ladan Younesi
- Shahid Akbar Abadi Clinical Research Development Unit (ShCRDU), Iran University of Medical Sciences (IUMS), Tehran, Iran
| |
Collapse
|
22
|
Dzobo K, Senthebane DA, Thomford NE, Rowe A, Dandara C, Parker MI. Not Everyone Fits the Mold: Intratumor and Intertumor Heterogeneity and Innovative Cancer Drug Design and Development. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2019; 22:17-34. [PMID: 29356626 DOI: 10.1089/omi.2017.0174] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Disruptive innovations in medicine are game-changing in nature and bring about radical shifts in the way we understand human diseases, their treatment, and/or prevention. Yet, disruptive innovations in cancer drug design and development are still limited. Therapies that cure all cancer patients are in short supply or do not exist at all. Chief among the causes of this predicament is drug resistance, a mechanism that is much more dynamic than previously understood. Drug resistance has limited the initial success experienced with biomarker-guided targeted therapies as well. A major contributor to drug resistance is intratumor heterogeneity. For example, within solid tumors, there are distinct subclones of cancer cells, presenting profound complexity to cancer treatment. Well-known contributors to intratumor heterogeneity are genomic instability, the microenvironment, cellular genotype, cell plasticity, and stochastic processes. This expert review explains that for oncology drug design and development to be more innovative, we need to take into account intratumor heterogeneity. Initially thought to be the preserve of cancer cells, recent evidence points to the highly heterogeneous nature and diverse locations of stromal cells, such as cancer-associated fibroblasts (CAFs) and cancer-associated macrophages (CAMs). Distinct subpopulations of CAFs and CAMs are now known to be located immediately adjacent and distant from cancer cells, with different subpopulations exerting different effects on cancer cells. Disruptive innovation and precision medicine in clinical oncology do not have to be a distant reality, but can potentially be achieved by targeting these spatially separated and exclusive cancer cell subclones and CAF subtypes. Finally, we emphasize that disruptive innovations in drug discovery and development will likely come from drugs whose effect is not necessarily tumor shrinkage.
Collapse
Affiliation(s)
- Kevin Dzobo
- 1 International Centre for Genetic Engineering and Biotechnology (ICGEB) , Cape Town, South Africa .,2 Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - Dimakatso Alice Senthebane
- 1 International Centre for Genetic Engineering and Biotechnology (ICGEB) , Cape Town, South Africa .,2 Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - Nicholas Ekow Thomford
- 3 Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - Arielle Rowe
- 1 International Centre for Genetic Engineering and Biotechnology (ICGEB) , Cape Town, South Africa
| | - Collet Dandara
- 3 Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology, Faculty of Health Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| | - M Iqbal Parker
- 2 Division of Medical Biochemistry, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town , Cape Town, South Africa
| |
Collapse
|
23
|
Wang W, Niu S, Qiao L, Wei F, Yin J, Wang S, Ouyang Y, Chen D. Usnea Acid as Multidrug Resistance (MDR) Reversing Agent against Human Chronic Myelogenous Leukemia K562/ADR Cells via an ROS Dependent Apoptosis. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8727935. [PMID: 30886864 PMCID: PMC6388510 DOI: 10.1155/2019/8727935] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/08/2018] [Accepted: 12/23/2018] [Indexed: 01/09/2023]
Abstract
PURPOSE Multidrug resistance (MDR) is a major obstacle in chemotherapy of leukemia treatments. In this paper, we investigated Usnea Acid (UA) as MDR reversal agent on hematologic K562/ADR cells via ROS dependent apoptosis. METHODS CCK8 assay was used to measure cell viability rate of K562/ADR. Intracellular reactive oxygen species (ROS) generation, cell cycle distribution, cell apoptosis were measured with flow cytometry, respectively. Proteins related to apoptosis were measured by Western blot. Intracellular Adriamycin accumulation was observed by confocal microscopy and measured by flow cytometry. RESULTS In vitro study showed intracellular Adriamycin accumulation was remarkably increased by UA. Cell viability treated with Adr (4 μM) was decreased from 89.8% ± 4.7 to 32% ± 8.9 by combined with UA (4 μM). Adr-induced apoptosis and G1/G0 phase cell cycle arrest were remarkably increased by UA, as well as, intracellular ROS level. However, MDR reversing activity of UA was inhibited by N-acetyl cysteine (NAC), a ROS scavenger. CONCLUSION These data provide compelling evidence that UA is a promising agent against MDR in leukemia cell line and suggest a promising therapeutic approach for leukemia.
Collapse
MESH Headings
- Acetylcysteine/pharmacology
- Apoptosis/drug effects
- Benzofurans/antagonists & inhibitors
- Benzofurans/pharmacology
- Cell Cycle Checkpoints/drug effects
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Doxorubicin/pharmacology
- Drug Resistance, Multiple/drug effects
- Drug Resistance, Neoplasm/drug effects
- Flow Cytometry
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Reactive Oxygen Species/metabolism
Collapse
Affiliation(s)
- Wenjing Wang
- Capital Medical University Affiliated Beijing You An Hospital, Beijing, 100069, China
- Beijing Institute of Hepatology, Beijing, 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, 100069, China
| | - Shubin Niu
- School of biomedicine, Beijing City University, No. 6 Huanghoudian Road Haidian District, Beijing, 100094, China
| | - Luxin Qiao
- Capital Medical University Affiliated Beijing You An Hospital, Beijing, 100069, China
- Beijing Institute of Hepatology, Beijing, 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, 100069, China
| | - Feili Wei
- Capital Medical University Affiliated Beijing You An Hospital, Beijing, 100069, China
- Beijing Institute of Hepatology, Beijing, 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, 100069, China
| | - Jiming Yin
- Capital Medical University Affiliated Beijing You An Hospital, Beijing, 100069, China
- Beijing Institute of Hepatology, Beijing, 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, 100069, China
| | - Shanshan Wang
- Capital Medical University Affiliated Beijing You An Hospital, Beijing, 100069, China
- Beijing Institute of Hepatology, Beijing, 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, 100069, China
| | - Yabo Ouyang
- Capital Medical University Affiliated Beijing You An Hospital, Beijing, 100069, China
- Beijing Institute of Hepatology, Beijing, 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, 100069, China
| | - Dexi Chen
- Capital Medical University Affiliated Beijing You An Hospital, Beijing, 100069, China
- Beijing Institute of Hepatology, Beijing, 100069, China
- Beijing Precision Medicine and Transformation Engineering Technology Research Center of Hepatitis and Liver Cancer, 100069, China
| |
Collapse
|
24
|
Singer F, Irmisch A, Toussaint NC, Grob L, Singer J, Thurnherr T, Beerenwinkel N, Levesque MP, Dummer R, Quagliata L, Rothschild SI, Wicki A, Beisel C, Stekhoven DJ. SwissMTB: establishing comprehensive molecular cancer diagnostics in Swiss clinics. BMC Med Inform Decis Mak 2018; 18:89. [PMID: 30373609 PMCID: PMC6206832 DOI: 10.1186/s12911-018-0680-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 10/18/2018] [Indexed: 12/18/2022] Open
Abstract
Background Molecular precision oncology is an emerging practice to improve cancer therapy by decreasing the risk of choosing treatments that lack efficacy or cause adverse events. However, the challenges of integrating molecular profiling into routine clinical care are manifold. From a computational perspective these include the importance of a short analysis turnaround time, the interpretation of complex drug-gene and gene-gene interactions, and the necessity of standardized high-quality workflows. In addition, difficulties faced when integrating molecular diagnostics into clinical practice are ethical concerns, legal requirements, and limited availability of treatment options beyond standard of care as well as the overall lack of awareness of their existence. Methods To the best of our knowledge, we are the first group in Switzerland that established a workflow for personalized diagnostics based on comprehensive high-throughput sequencing of tumors at the clinic. Our workflow, named SwissMTB (Swiss Molecular Tumor Board), links genetic tumor alterations and gene expression to therapeutic options and clinical trial opportunities. The resulting treatment recommendations are summarized in a clinical report and discussed in a molecular tumor board at the clinic to support therapy decisions. Results Here we present results from an observational pilot study including 22 late-stage cancer patients. In this study we were able to identify actionable variants and corresponding therapies for 19 patients. Half of the patients were analyzed retrospectively. In two patients we identified resistance-associated variants explaining lack of therapy response. For five out of eleven patients analyzed before treatment the SwissMTB diagnostic influenced treatment decision. Conclusions SwissMTB enables the analysis and clinical interpretation of large numbers of potentially actionable molecular targets. Thus, our workflow paves the way towards a more frequent use of comprehensive molecular diagnostics in Swiss hospitals. Electronic supplementary material The online version of this article (10.1186/s12911-018-0680-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Franziska Singer
- NEXUS Personalized Health Technologies, ETH Zurich, Otto-Stern-Weg 7, 8093, Zurich, Switzerland.,SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland
| | - Anja Irmisch
- Department of Dermatology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Nora C Toussaint
- NEXUS Personalized Health Technologies, ETH Zurich, Otto-Stern-Weg 7, 8093, Zurich, Switzerland.,SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland
| | - Linda Grob
- NEXUS Personalized Health Technologies, ETH Zurich, Otto-Stern-Weg 7, 8093, Zurich, Switzerland.,SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland
| | - Jochen Singer
- SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland.,Department of Biosystems Science and Engineering, ETH Zurich, 4058, Basel, Switzerland
| | - Thomas Thurnherr
- SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland.,Department of Biosystems Science and Engineering, ETH Zurich, 4058, Basel, Switzerland
| | - Niko Beerenwinkel
- SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland.,Department of Biosystems Science and Engineering, ETH Zurich, 4058, Basel, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Luca Quagliata
- Department of Pathology, University Hospital Basel, Schönbeinstrasse 40, 4056, Basel, Switzerland
| | - Sacha I Rothschild
- Division of Oncology, Department of Biomedicine, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Andreas Wicki
- Division of Oncology, Department of Biomedicine, University Hospital Basel, Petersgraben 4, 4031, Basel, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zurich, 4058, Basel, Switzerland
| | - Daniel J Stekhoven
- NEXUS Personalized Health Technologies, ETH Zurich, Otto-Stern-Weg 7, 8093, Zurich, Switzerland. .,SIB Swiss Institute of Bioinformatics, 4058, Basel, Switzerland.
| |
Collapse
|
25
|
Dai J, Huang Q, Niu K, Wang B, Li Y, Dai C, Chen Z, Tao K, Dai J. Sestrin 2 confers primary resistance to sorafenib by simultaneously activating AKT and AMPK in hepatocellular carcinoma. Cancer Med 2018; 7:5691-5703. [PMID: 30311444 PMCID: PMC6247041 DOI: 10.1002/cam4.1826] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 09/04/2018] [Accepted: 09/10/2018] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the malignancy derived from normal hepatocytes with increasing incidence and extremely poor prognosis worldwide. The only approved first‐line systematic treatment agent for HCC, sorafenib, is capable to effectively improve advanced HCC patients’ survival. However, it is gradually recognized that the therapeutic response to sorafenib could be drastically diminished after short‐term treatment, defined as primary resistance. The present study is aimed to explore the role of stress‐inducible protein Sestrin2 (SESN2), one of the most important sestrins family members, in sorafenib primary resistance. Herein, we initially found that SESN2 expression was significantly up‐regulated in both HCC cell lines and tissues compared to normal human hepatocytes and corresponding adjacent liver tissues, respectively. In addition, SESN2 expression was highly correlated with sorafenib IC50 of HCC cell lines. Thereafter, we showed that sorafenib treatment resulted in an increase of SESN2 expression and the knockdown of SESN2 exacerbated sorafenib‐induced proliferation inhibition and cell apoptosis. Further mechanistic study uncovered that SESN2 deficiency impaired both AKT and AMPK phosphorylation and activation after sorafenib treatment. Moreover, the correlations between SESN2 expression and both phosphor‐AKT and phosphor‐AMPK expression were illustrated in HCC tissues. Taken together, our study demonstrates that SESN2 activates AKT and AMPK signaling as a novel mechanism to induce sorafenib primary resistance in HCC.
Collapse
Affiliation(s)
- Jimin Dai
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Air Force Medical University, Xi'an, China.,The Cadet Team 6 (Regiment 6) of School of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Qichao Huang
- State Key Laboratory of Cancer Biology and Experimental Teaching Center of Basic Medicine, Air Force Medical University, Xi'an, China
| | - Kunwei Niu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Bo Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Yijie Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Chen Dai
- Department of Orthopedics, The First Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Zhinan Chen
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Air Force Medical University, Xi'an, China
| | - Kaishan Tao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Air Force Medical University, Xi'an, China
| | - Jingyao Dai
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Air Force Medical University, Xi'an, China.,Department of Cell Biology, National Translational Science Center for Molecular Medicine, Air Force Medical University, Xi'an, China
| |
Collapse
|
26
|
Aronchik I, Dai Y, Labenski M, Barnes C, Jones T, Qiao L, Beebe L, Malek M, Elis W, Shi T, Mavrommatis K, Bray GL, Filvaroff EH. Efficacy of a Covalent ERK1/2 Inhibitor, CC-90003, in KRAS-Mutant Cancer Models Reveals Novel Mechanisms of Response and Resistance. Mol Cancer Res 2018; 17:642-654. [DOI: 10.1158/1541-7786.mcr-17-0554] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 05/31/2018] [Accepted: 09/21/2018] [Indexed: 11/16/2022]
|
27
|
Migliore C, Morando E, Ghiso E, Anastasi S, Leoni VP, Apicella M, Cora' D, Sapino A, Pietrantonio F, De Braud F, Columbano A, Segatto O, Giordano S. miR-205 mediates adaptive resistance to MET inhibition via ERRFI1 targeting and raised EGFR signaling. EMBO Mol Med 2018; 10:e8746. [PMID: 30021798 PMCID: PMC6127885 DOI: 10.15252/emmm.201708746] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 06/25/2018] [Accepted: 06/27/2018] [Indexed: 12/24/2022] Open
Abstract
The onset of secondary resistance represents a major limitation to long-term efficacy of target therapies in cancer patients. Thus, the identification of mechanisms mediating secondary resistance is the key to the rational design of therapeutic strategies for resistant patients. MiRNA profiling combined with RNA-Seq in MET-addicted cancer cell lines led us to identify the miR-205/ERRFI1 (ERBB receptor feedback inhibitor-1) axis as a novel mediator of resistance to MET tyrosine kinase inhibitors (TKIs). In cells resistant to MET-TKIs, epigenetically induced miR-205 expression determined the downregulation of ERRFI1 which, in turn, caused EGFR activation, sustaining resistance to MET-TKIs. Anti-miR-205 transduction reverted crizotinib resistance in vivo, while miR-205 over-expression rendered wt cells refractory to TKI treatment. Importantly, in the absence of EGFR genetic alterations, miR-205/ERRFI1-driven EGFR activation rendered MET-TKI-resistant cells sensitive to combined MET/EGFR inhibition. As a proof of concept of the clinical relevance of this new mechanism of adaptive resistance, we report that a patient with a MET-amplified lung adenocarcinoma displayed deregulation of the miR-205/ERRFI1 axis in concomitance with onset of clinical resistance to anti-MET therapy.
Collapse
Affiliation(s)
- Cristina Migliore
- Department of Oncology, University of Torino, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Elena Morando
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Elena Ghiso
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Sergio Anastasi
- Unit of Oncogenomics and Epigenetics, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Vera P Leoni
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
| | | | - Davide Cora'
- Department of Oncology, University of Torino, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
- Department of Translational Medicine, Piemonte Orientale University "Amedeo Avogadro", Novara, Italy
| | - Anna Sapino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
- Department of Medical Science, University of Torino, Torino, Italy
| | - Filippo Pietrantonio
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milano, Milan, Italy
| | - Filippo De Braud
- Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- Department of Oncology and Hemato-oncology, University of Milano, Milan, Italy
| | - Amedeo Columbano
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
| | - Oreste Segatto
- Unit of Oncogenomics and Epigenetics, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Giordano
- Department of Oncology, University of Torino, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| |
Collapse
|
28
|
Świderska KW, Szlachcic A, Opaliński Ł, Zakrzewska M, Otlewski J. FGF2 Dual Warhead Conjugate with Monomethyl Auristatin E and α-Amanitin Displays a Cytotoxic Effect towards Cancer Cells Overproducing FGF Receptor 1. Int J Mol Sci 2018; 19:ijms19072098. [PMID: 30029518 PMCID: PMC6073801 DOI: 10.3390/ijms19072098] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/12/2018] [Accepted: 07/18/2018] [Indexed: 02/03/2023] Open
Abstract
In the rapidly developing field of targeted cancer therapy there is growing interest towards therapeutics combining two or more compounds to achieve synergistic action and minimize the chance of cancer resistance to treatment. We developed a fibroblast growth factor 2 (FGF2)-conjugate bearing two cytotoxic drugs with independent mode of action: α-amanitin and monomethyl auristatin E. Drugs are covalently attached to the targeting protein in a site-specific manner via maleimide-thiol conjugation and Cu(I)-catalyzed alkyne-azide cycloaddition. The dual warhead conjugate binds to FGF receptor 1 (FGFR1) and utilizes receptor-mediated endocytosis for selective internalization into cancer cells with FGFR1. The developed conjugate displays high cytotoxicity towards all tested FGFR1-positive cell lines. Most importantly, the improved cytotoxic effect of both drugs is observed for lung cancer cell line NCI-H446. The single drug-FGF2 conjugates have no impact on the viability of NCI-H446 cells, whereas the dual warhead-FGF2 conjugate selectively and efficiently kills these FGFR1 positive cancer cells. Due to the diversified mode of action the dual warhead-FGF2 conjugate may overcome the potential acquired resistance of FGFR1-overproducing cancer cells towards single cytotoxic drugs.
Collapse
Affiliation(s)
- Karolina Weronika Świderska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Anna Szlachcic
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Łukasz Opaliński
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Małgorzata Zakrzewska
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| | - Jacek Otlewski
- Faculty of Biotechnology, Department of Protein Engineering, University of Wroclaw, Joliot-Curie 14a, 50-383 Wroclaw, Poland.
| |
Collapse
|
29
|
Nikolaou M, Pavlopoulou A, Georgakilas AG, Kyrodimos E. The challenge of drug resistance in cancer treatment: a current overview. Clin Exp Metastasis 2018; 35:309-318. [DOI: 10.1007/s10585-018-9903-0] [Citation(s) in RCA: 248] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 05/16/2018] [Indexed: 12/14/2022]
|
30
|
Basu R, Qian Y, Kopchick JJ. MECHANISMS IN ENDOCRINOLOGY: Lessons from growth hormone receptor gene-disrupted mice: are there benefits of endocrine defects? Eur J Endocrinol 2018; 178:R155-R181. [PMID: 29459441 DOI: 10.1530/eje-18-0018] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 02/19/2018] [Indexed: 12/12/2022]
Abstract
Growth hormone (GH) is produced primarily by anterior pituitary somatotroph cells. Numerous acute human (h) GH treatment and long-term follow-up studies and extensive use of animal models of GH action have shaped the body of GH research over the past 70 years. Work on the GH receptor (R)-knockout (GHRKO) mice and results of studies on GH-resistant Laron Syndrome (LS) patients have helped define many physiological actions of GH including those dealing with metabolism, obesity, cancer, diabetes, cognition and aging/longevity. In this review, we have discussed several issues dealing with these biological effects of GH and attempt to answer the question of whether decreased GH action may be beneficial.
Collapse
Affiliation(s)
- Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA
| | - Yanrong Qian
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA
| | - John J Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, Ohio, USA
- Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio, USA
| |
Collapse
|
31
|
Gallaher JA, Enriquez-Navas PM, Luddy KA, Gatenby RA, Anderson ARA. Spatial Heterogeneity and Evolutionary Dynamics Modulate Time to Recurrence in Continuous and Adaptive Cancer Therapies. Cancer Res 2018; 78:2127-2139. [PMID: 29382708 PMCID: PMC5899666 DOI: 10.1158/0008-5472.can-17-2649] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 12/05/2017] [Accepted: 01/24/2018] [Indexed: 12/31/2022]
Abstract
Treatment of advanced cancers has benefited from new agents that supplement or bypass conventional therapies. However, even effective therapies fail as cancer cells deploy a wide range of resistance strategies. We propose that evolutionary dynamics ultimately determine survival and proliferation of resistant cells. Therefore, evolutionary strategies should be used with conventional therapies to delay or prevent resistance. Using an agent-based framework to model spatial competition among sensitive and resistant populations, we applied antiproliferative drug treatments to varying ratios of sensitive and resistant cells. We compared a continuous maximum-tolerated dose schedule with an adaptive schedule aimed at tumor control via competition between sensitive and resistant cells. Continuous treatment cured mostly sensitive tumors, but with any resistant cells, recurrence was inevitable. We identified two adaptive strategies that control heterogeneous tumors: dose modulation controls most tumors with less drug, while a more vacation-oriented schedule can control more invasive tumors. These findings offer potential modifications to treatment regimens that may improve outcomes and reduce resistance and recurrence.Significance: By using drug dose modulation or treatment vacations, adaptive therapy strategies control the emergence of tumor drug resistance by spatially suppressing less fit resistant populations in favor of treatment sensitive ones. Cancer Res; 78(8); 2127-39. ©2018 AACR.
Collapse
Affiliation(s)
- Jill A Gallaher
- H. Lee Moffitt Cancer Center, Integrated Mathematical Oncology, Tampa, Florida
| | | | - Kimberly A Luddy
- H. Lee Moffitt Cancer Center, Cancer Imaging and Metabolism, Tampa, Florida
| | - Robert A Gatenby
- H. Lee Moffitt Cancer Center, Integrated Mathematical Oncology, Tampa, Florida
| | | |
Collapse
|
32
|
Vlahovic G, Meadows KL, Hatch AJ, Jia J, Nixon AB, Uronis HE, Morse MA, Selim MA, Crawford J, Riedel RF, Zafar SY, Howard LA, O'Neill M, Meadows JJ, Haley ST, Arrowood CC, Rushing C, Pang H, Hurwitz HI. A Phase I Trial of the IGF-1R Antibody Ganitumab (AMG 479) in Combination with Everolimus (RAD001) and Panitumumab in Patients with Advanced Cancer. Oncologist 2018; 23:782-790. [PMID: 29572245 DOI: 10.1634/theoncologist.2016-0377] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 08/17/2017] [Indexed: 11/17/2022] Open
Abstract
PURPOSE This study evaluated the maximum tolerated dose or recommended phase II dose (RPTD) and safety and tolerability of the ganitumab and everolimus doublet regimen followed by the ganitumab, everolimus, and panitumumab triplet regimen. MATERIALS AND METHODS This was a standard 3 + 3 dose escalation trial. Doublet therapy consisted of ganitumab at 12 mg/kg every 2 weeks; doses of everolimus were adjusted according to dose-limiting toxicities (DLTs). Panitumumab at 4.8 mg/kg every 2 weeks was added to the RPTD of ganitumab and everolimus. DLTs were assessed in cycle 1; toxicity evaluation was closely monitored throughout treatment. Treatment continued until disease progression or undesirable toxicity. Pretreatment and on-treatment skin biopsies were collected to assess insulin-like growth factor 1 receptor and mammalian target of rapamycin (mTOR) target modulation. RESULTS Forty-three subjects were enrolled. In the doublet regimen, two DLTs were observed in cohort 1, no DLTs in cohort -1, and one in cohort -1B. The triplet combination was discontinued because of unacceptable toxicity. Common adverse events were thrombocytopenia/neutropenia, skin rash, mucositis, fatigue, and hyperglycemia. In the doublet regimen, two patients with refractory non-small cell lung cancer (NSCLC) achieved prolonged complete responses ranging from 18 to >60 months; one treatment-naïve patient with chondrosarcoma achieved prolonged stable disease >24 months. In dermal granulation tissue, the insulin-like growth factor receptor and mTOR pathways were potently and specifically inhibited by ganitumab and everolimus, respectively. CONCLUSION The triplet regimen of ganitumab, everolimus, and panitumumab was associated with unacceptable toxicity. However, the doublet of ganitumab at 12 mg/kg every 2 weeks and everolimus five times weekly had an acceptable safety profile and demonstrated notable clinical activity in patients with refractory NSCLC and sarcoma. IMPLICATIONS FOR PRACTICE This trial evaluated the maximum tolerated dose or recommended phase II dose and safety and tolerability of the ganitumab and everolimus doublet regimen followed by the ganitumab, everolimus, and panitumumab triplet regimen. Although the triplet regimen of ganitumab, everolimus, and panitumumab was associated with unacceptable toxicity, the doublet of ganitumab at 12 mg/kg every 2 weeks and everolimus at five times weekly had an acceptable safety profile and demonstrated notable clinical activity in patients with refractory non-small cell lung cancer and sarcoma.
Collapse
Affiliation(s)
| | | | - Ace J Hatch
- Duke Cancer Institute, Durham, North Carolina, USA
| | - Jingquan Jia
- Duke Cancer Institute, Durham, North Carolina, USA
| | | | | | | | - M Angelica Selim
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | | | | | | | | | | | | | | | - Herbert Pang
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina, USA
| | | |
Collapse
|
33
|
Belfiore L, Saunders DN, Ranson M, Thurecht KJ, Storm G, Vine KL. Towards clinical translation of ligand-functionalized liposomes in targeted cancer therapy: Challenges and opportunities. J Control Release 2018; 277:1-13. [PMID: 29501721 DOI: 10.1016/j.jconrel.2018.02.040] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/26/2018] [Accepted: 02/27/2018] [Indexed: 01/03/2023]
Abstract
The development of therapeutic resistance to targeted anticancer therapies remains a significant clinical problem, with intratumoral heterogeneity playing a key role. In this context, improving the therapeutic outcome through simultaneous targeting of multiple tumor cell subtypes within a heterogeneous tumor is a promising approach. Liposomes have emerged as useful drug carriers that can reduce systemic toxicity and increase drug delivery to the tumor site. While clinically used liposomal drug formulations show marked therapeutic advantages over free drug formulations, ligand-functionalized liposomes that can target multiple tumor cell subtypes may further improve the therapeutic efficacy by facilitating drug delivery to a broader population of tumor cells making up the heterogeneous tumor tissue. Ligand-directed liposomes enable the so-called active targeting of cell receptors via surface-attached ligands that direct drug uptake into tumor cells or tumor-associated stromal cells, and so can increase the selectivity of drug delivery. Despite promising preclinical results demonstrating improved targeting and anti-tumor effects of ligand-directed liposomes, there has been limited translation of this approach to the clinic. Key challenges for translation include the lack of established methods to scale up production and comprehensively characterize ligand-functionalized liposome formulations, as well as the inadequate recapitulation of in vivo tumors in the preclinical models currently used to evaluate their performance. Herein, we discuss the utility of recent ligand-directed liposome approaches, with a focus on dual-ligand liposomes, for the treatment of solid tumors and examine the drawbacks limiting their progression to clinical adoption.
Collapse
Affiliation(s)
- Lisa Belfiore
- Illawarra Health and Medical Research Institute, Centre for Medical and Molecular Bioscience, School of Biological Sciences, University of Wollongong, Wollongong, Australia
| | - Darren N Saunders
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Marie Ranson
- Illawarra Health and Medical Research Institute, Centre for Medical and Molecular Bioscience, School of Biological Sciences, University of Wollongong, Wollongong, Australia
| | - Kristofer J Thurecht
- Australian Institute for Bioengineering and Nanotechnology (AIBN), Centre for Advanced Imaging (CAI), Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, Brisbane, Australia
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, CG, The Netherlands
| | - Kara L Vine
- Illawarra Health and Medical Research Institute, Centre for Medical and Molecular Bioscience, School of Biological Sciences, University of Wollongong, Wollongong, Australia.
| |
Collapse
|
34
|
Wang W, Xu B, Li Q, Jiang D, Yan S. Anti‑cancer effects of a novel Pan‑RAF inhibitor in a hepatocellular carcinoma cell line. Mol Med Rep 2018; 17:6185-6193. [PMID: 29484394 DOI: 10.3892/mmr.2018.8615] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 02/01/2018] [Indexed: 11/06/2022] Open
Abstract
The RAF/mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) kinase (MEK)/ERK (RAF/MEK/ERK) signaling cascade serves a prominent role in hepatocellular carcinoma (HCC) proliferation. Sorafenib (BAY 43‑9006) is a potent multikinase inhibitor of RAF kinases and a few receptor tyrosine kinases. Additionally, sorafenib causes apoptosis in a number of human tumor cell lines such as leukemia cell lines. Sorafenib is the first targeted drug to prolong the overall survival of patients with advanced HCC. However, sorafenib activity is less favorable in certain cancers, including sarcomas and melanomas, due to patient insensitivity and drug resistance. In the present study, a novel bi‑aryl urea, N‑(3‑trifluoromethylphenyl)‑N'-(2-methyl-4-(6‑cyclopropanecarboxamido-pyrimidin-4-yl) oxyphenyl) urea (CBI‑5725), is shown to be a potential candidate for the treatment of liver cancer. In the present study, the in vitro activities of CBI‑5725 and sorafenib in PLC/PRF/5 HCC cells were examined and the corresponding in vivo antitumor activities in PLC/PRF/5 human tumor xenografts. An alamar blue assay confirmed that CBI‑5725 was more cytotoxic than sorafenib to PLC/PRF/5 cells, suggesting that CBI‑5725 inhibited tumor cell proliferation more potently than sorafenib. CBI‑5725 inhibited the RAF/MEK/ERK signaling pathway to the same extent as sorafenib. In addition, CBI‑5725 elicited cell cycle arrest in the G2/M phase, while sorafenib did not markedly alter the cell cycle. Furthermore, CBI‑5725 induced apoptosis more strongly than sorafenib in a dose‑dependent manner, which may be attributed to greater caspase‑3 and poly(adenosine 5'‑diphosphate‑ribose) polymerase activation by CBI‑5725. In the PLC/PRF/5 xenograft model, 2 mg/kg CBI‑5725 inhibited tumor growth by 73%. At doses ranging from 6 to 18 mg/kg, CBI‑5725 nearly completely prevented tumor growth. These results imply that the antitumor efficacy of CBI‑5725 in HCC models may result from the suppression of the RAF/MEK/ERK signaling pathway, the induction of cell cycle arrest in the G2/M phase, and the initiation of caspase‑3‑dependent apoptosis. These observations suggested that CBI‑5725 may be a potent novel compound for the treatment of HCC.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Bo Xu
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University Health Science Center, Beijing 100191, P.R. China
| | - Qixiang Li
- Biomarker and Diagnostic Technology, Crown Bioscience Inc., Beijing 102200, P.R. China
| | - Dechun Jiang
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Suying Yan
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| |
Collapse
|
35
|
Frequent Coamplification of Receptor Tyrosine Kinase and Downstream Signaling Genes in Japanese Primary Gastric Cancer and Conversion in Matched Lymph Node Metastasis. Ann Surg 2018; 267:114-121. [DOI: 10.1097/sla.0000000000002042] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
36
|
Anderson GR, Wardell SE, Cakir M, Crawford L, Leeds JC, Nussbaum DP, Shankar PS, Soderquist RS, Stein EM, Tingley JP, Winter PS, Zieser-Misenheimer EK, Alley HM, Yllanes A, Haney V, Blackwell KL, McCall SJ, McDonnell DP, Wood KC. PIK3CA mutations enable targeting of a breast tumor dependency through mTOR-mediated MCL-1 translation. Sci Transl Med 2017; 8:369ra175. [PMID: 27974663 DOI: 10.1126/scitranslmed.aae0348] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/06/2016] [Accepted: 10/05/2016] [Indexed: 12/23/2022]
Abstract
Therapies that efficiently induce apoptosis are likely to be required for durable clinical responses in patients with solid tumors. Using a pharmacological screening approach, we discovered that combined inhibition of B cell lymphoma-extra large (BCL-XL) and the mammalian target of rapamycin (mTOR)/4E-BP axis results in selective and synergistic induction of apoptosis in cellular and animal models of PIK3CA mutant breast cancers, including triple-negative tumors. Mechanistically, inhibition of mTOR/4E-BP suppresses myeloid cell leukemia-1 (MCL-1) protein translation only in PIK3CA mutant tumors, creating a synthetic dependence on BCL-XL This dual dependence on BCL-XL and MCL-1, but not on BCL-2, appears to be a fundamental property of diverse breast cancer cell lines, xenografts, and patient-derived tumors that is independent of the molecular subtype or PIK3CA mutational status. Furthermore, this dependence distinguishes breast cancers from normal breast epithelial cells, which are neither primed for apoptosis nor dependent on BCL-XL/MCL-1, suggesting a potential therapeutic window. By tilting the balance of pro- to antiapoptotic signals in the mitochondria, dual inhibition of MCL-1 and BCL-XL also sensitizes breast cancer cells to standard-of-care cytotoxic and targeted chemotherapies. Together, these results suggest that patients with PIK3CA mutant breast cancers may benefit from combined treatment with inhibitors of BCL-XL and the mTOR/4E-BP axis, whereas alternative methods of inhibiting MCL-1 and BCL-XL may be effective in tumors lacking PIK3CA mutations.
Collapse
Affiliation(s)
- Grace R Anderson
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Suzanne E Wardell
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Merve Cakir
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA.,Program in Computational Biology and Bioinformatics, Duke University, Durham, NC 27708, USA
| | - Lorin Crawford
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA.,Department of Statistical Science, Duke University, Durham, NC 27708, USA
| | - Jim C Leeds
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Daniel P Nussbaum
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA.,Department of Surgery, Duke University, Durham, NC 27710, USA
| | - Pallavi S Shankar
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Ryan S Soderquist
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Elizabeth M Stein
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Jennifer P Tingley
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Peter S Winter
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA.,Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
| | | | - Holly M Alley
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Alexander Yllanes
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Victoria Haney
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | | | | | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
| | - Kris C Wood
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
37
|
Identification of cancer genes that are independent of dominant proliferation and lineage programs. Proc Natl Acad Sci U S A 2017; 114:E11276-E11284. [PMID: 29229826 PMCID: PMC5748209 DOI: 10.1073/pnas.1714877115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Large, multidimensional “landscaping” projects have provided datasets that can be mined to identify potential targets for subgroups of tumors. Here, we analyzed genomic and transcriptomic data from human breast tumors to identify genes whose expression is enriched in tumors harboring specific genetic alterations. However, this analysis revealed that two other factors, proliferation rate and tumor lineage, are more dominant factors in shaping tumor transcriptional programs than genetic alterations. This discovery shifted our attention to identifying genes that are independent of the dominant proliferation and lineage programs. A small subset of these genes represents candidate targets for combination cancer therapies because they are druggable, maintained after treatment with chemotherapy, essential for cell line survival, and elevated in drug-resistant stem-like cancer cells. Large, multidimensional cancer datasets provide a resource that can be mined to identify candidate therapeutic targets for specific subgroups of tumors. Here, we analyzed human breast cancer data to identify transcriptional programs associated with tumors bearing specific genetic driver alterations. Using an unbiased approach, we identified thousands of genes whose expression was enriched in tumors with specific genetic alterations. However, expression of the vast majority of these genes was not enriched if associations were analyzed within individual breast tumor molecular subtypes, across multiple tumor types, or after gene expression was normalized to account for differences in proliferation or tumor lineage. Together with linear modeling results, these findings suggest that most transcriptional programs associated with specific genetic alterations in oncogenes and tumor suppressors are highly context-dependent and are predominantly linked to differences in proliferation programs between distinct breast cancer subtypes. We demonstrate that such proliferation-dependent gene expression dominates tumor transcriptional programs relative to matched normal tissues. However, we also identified a relatively small group of cancer-associated genes that are both proliferation- and lineage-independent. A subset of these genes are attractive candidate targets for combination therapy because they are essential in breast cancer cell lines, druggable, enriched in stem-like breast cancer cells, and resistant to chemotherapy-induced down-regulation.
Collapse
|
38
|
Wu Q, Sharma S, Cui H, LeBlanc SE, Zhang H, Muthuswami R, Nickerson JA, Imbalzano AN. Targeting the chromatin remodeling enzyme BRG1 increases the efficacy of chemotherapy drugs in breast cancer cells. Oncotarget 2017; 7:27158-75. [PMID: 27029062 PMCID: PMC5053639 DOI: 10.18632/oncotarget.8384] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 03/16/2016] [Indexed: 12/31/2022] Open
Abstract
Brahma related gene product 1 (BRG1) is an ATPase that drives the catalytic activity of a subset of the mammalian SWI/SNF chromatin remodeling enzymes. BRG1 is overexpressed in most human breast cancer tumors without evidence of mutation and is required for breast cancer cell proliferation. We demonstrate that knockdown of BRG1 sensitized triple negative breast cancer cells to chemotherapeutic drugs used to treat breast cancer. An inhibitor of the BRG1 bromodomain had no effect on breast cancer cell viability, but an inhibitory molecule that targets the BRG1 ATPase activity recapitulated the increased drug efficacy observed in the presence of BRG1 knockdown. We further demonstrate that inhibition of BRG1 ATPase activity blocks the induction of ABC transporter genes by these chemotherapeutic drugs and that BRG1 binds to ABC transporter gene promoters. This inhibition increased intracellular concentrations of the drugs, providing a likely mechanism for the increased chemosensitivity. Since ABC transporters and their induction by chemotherapy drugs are a major cause of chemoresistance and treatment failure, these results support the idea that targeting the enzymatic activity of BRG1 would be an effective adjuvant therapy for breast cancer.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Soni Sharma
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India
| | - Hang Cui
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, USA.,Abace Biotech Co Ltd., Yi Zhuang Biomedical Park, BDA, Beijing, China
| | - Scott E LeBlanc
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Hong Zhang
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Rohini Muthuswami
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India
| | - Jeffrey A Nickerson
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Anthony N Imbalzano
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
39
|
Swamy SG, Kameshwar VH, Shubha PB, Looi CY, Shanmugam MK, Arfuso F, Dharmarajan A, Sethi G, Shivananju NS, Bishayee A. Targeting multiple oncogenic pathways for the treatment of hepatocellular carcinoma. Target Oncol 2017; 12:1-10. [PMID: 27510230 DOI: 10.1007/s11523-016-0452-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common forms of liver cancer diagnosed worldwide. HCC occurs due to chronic liver disease and is often diagnosed at advanced stages. Chemotherapeutic agents such as doxorubicin are currently used as first-line agents for HCC therapy, but these are non-selective cytotoxic molecules with significant side effects. Sorafenib, a multi-targeted tyrosine kinase inhibitor, is the only approved targeted drug for HCC patients. However, due to adverse side effects and limited efficacy, there is a need for the identification of novel pharmacological drugs beyond sorafenib. Several agents that target and inhibit various signaling pathways involved in HCC are currently being assessed for HCC treatment. In the present review article, we summarize the diverse signal transduction pathways responsible for initiation as well as progression of HCC and also the potential anticancer effects of selected targeted therapies that can be employed for HCC therapy.
Collapse
Affiliation(s)
- Supritha G Swamy
- Department of Biotechnology, JSS Science and Technology University, JSS Technical Institutions Campus, Mysore, Karnataka, 570006, India
| | - Vivek H Kameshwar
- Department of Biotechnology, JSS Science and Technology University, JSS Technical Institutions Campus, Mysore, Karnataka, 570006, India
| | - Priya B Shubha
- Department of Studies in Chemistry, University of Mysore, Manasagangotri, Mysore, 570 006, Karnataka, India
| | - Chung Yeng Looi
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Frank Arfuso
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Biosciences Research Precinct, Curtin University, Bentley, Western Australia, 6009, Australia
| | - Arunasalam Dharmarajan
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Biosciences Research Precinct, Curtin University, Bentley, Western Australia, 6009, Australia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Biosciences Research Precinct, Curtin University, Bentley, Western Australia, 6009, Australia
| | - Nanjunda Swamy Shivananju
- Department of Biotechnology, JSS Science and Technology University, JSS Technical Institutions Campus, Mysore, Karnataka, 570006, India.
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin Health Sciences Institute, 18301 N. Miami Avenue, Miami, FL, 33169, USA.
| |
Collapse
|
40
|
Bosch-Barrera J, Queralt B, Menendez JA. Targeting STAT3 with silibinin to improve cancer therapeutics. Cancer Treat Rev 2017; 58:61-69. [DOI: 10.1016/j.ctrv.2017.06.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 06/15/2017] [Indexed: 02/08/2023]
|
41
|
Synergistic effects of various Her inhibitors in combination with IGF-1R, C-MET and Src targeting agents in breast cancer cell lines. Sci Rep 2017. [PMID: 28638122 PMCID: PMC5479850 DOI: 10.1038/s41598-017-04301-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Overexpression of HER2 has been reported in around 25% of human breast cancers. Despite recent advances in HER2 targeted therapy, many patients still experience primary and secondary resistance to such treatments, the mechanisms for which are poorly understood. Here, we investigated the sensitivity of a panel of breast cancer cell lines to treatment with various types of HER-family inhibitors alone or in combination with other tyrosine kinase inhibitors or chemotherapeutic agents. We found that treatment with the second-generation irreversible HER-family inhibitors, particularly afatinib and neratinib, were more effective than treatment with the first-generation reversible inhibitors in inhibiting growth, migration and downstream cell signalling in breast cancer cells. Of the three HER2 overexpressing cell lines in this panel, SKBr3 and BT474 were highly sensitive to treatment with HER-family inhibitors, while MDA-MB-453 was comparatively resistant. Combinations of HER-family inhibitors with NVP-AEW541, dasatinib or crizotinib (inhibitors of IGF-1R, Src and c-Met/ALK, respectively) led to synergistic effects in some of the cell lines examined. In particular, treatment with a combination of Src and HER-family member inhibitors resulted in synergistic growth inhibition of MDA-MB453 cells, implicating Src as a mediator of resistance to HER2-targeting agents. Our results suggest that combining HER-family inhibitors with other TKIs such as dasatinib may have therapeutic advantages in certain breast cancer subtypes and warrants further investigation.
Collapse
|
42
|
Basu R, Baumgaertel N, Wu S, Kopchick JJ. Growth Hormone Receptor Knockdown Sensitizes Human Melanoma Cells to Chemotherapy by Attenuating Expression of ABC Drug Efflux Pumps. HORMONES & CANCER 2017; 8:143-156. [PMID: 28293855 PMCID: PMC10355985 DOI: 10.1007/s12672-017-0292-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 03/02/2017] [Indexed: 12/16/2022]
Abstract
Melanoma remains one of the most therapy-resistant forms of human cancer despite recent introductions of highly efficacious targeted therapies. The intrinsic therapy resistance of human melanoma is largely due to abundant expression of a repertoire of xenobiotic efflux pumps of the ATP-binding cassette (ABC) transporter family. Here, we report that GH action is a key mediator of chemotherapeutic resistance in human melanoma cells. We investigated multiple ABC efflux pumps (ABCB1, ABCB5, ABCB8, ABCC1, ABCC2, ABCG1, and ABCG2) reportedly associated with melanoma drug resistance in different human melanoma cells and tested the efficacy of five different anti-cancer compounds (cisplatin, doxorubicin, oridonin, paclitaxel, vemurafenib) with decreased GH action. We found that GH treatment of human melanoma cells upregulates expression of multiple ABC transporters and increases the EC50 of melanoma drug vemurafenib. Also, vemurafenib-resistant melanoma cells had upregulated levels of GH receptor (GHR) expression as well as ABC efflux pumps. GHR knockdown (KD) using siRNA in human melanoma cells treated with sub-EC50 doses of anti-tumor compounds resulted in significantly increased drug retention, decreased cell proliferation and increased drug efficacy, compared to mock-transfected controls. Our set of findings identify an unknown mechanism of GH regulation in mediating melanoma drug resistance and validates GHR as a unique therapeutic target for sensitizing highly therapy-resistant human melanoma cells to lower doses of anti-cancer drugs.
Collapse
Affiliation(s)
- Reetobrata Basu
- Edison Biotechnology Institute, Konneker Research Laboratory 206, Ohio University, Athens, OH, 45701, USA
- Molecular and Cell Biology Program, Ohio University, Athens, OH, USA
| | - Nicholas Baumgaertel
- Edison Biotechnology Institute, Konneker Research Laboratory 206, Ohio University, Athens, OH, 45701, USA
- Department of Biological Sciences, Ohio University, Athens, OH, USA
| | - Shiyong Wu
- Edison Biotechnology Institute, Konneker Research Laboratory 206, Ohio University, Athens, OH, 45701, USA
- Molecular and Cell Biology Program, Ohio University, Athens, OH, USA
| | - John J Kopchick
- Edison Biotechnology Institute, Konneker Research Laboratory 206, Ohio University, Athens, OH, 45701, USA.
- Molecular and Cell Biology Program, Ohio University, Athens, OH, USA.
- Heritage College of Osteopathic Medicine, Athens, OH, USA.
| |
Collapse
|
43
|
Koetz-Ploch L, Hanniford D, Dolgalev I, Sokolova E, Zhong J, Díaz-Martínez M, Bernstein E, Darvishian F, Flaherty KT, Chapman PB, Tawbi H, Hernando E. MicroRNA-125a promotes resistance to BRAF inhibitors through suppression of the intrinsic apoptotic pathway. Pigment Cell Melanoma Res 2017; 30:328-338. [PMID: 28140520 DOI: 10.1111/pcmr.12578] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 01/16/2017] [Indexed: 12/20/2022]
Abstract
Melanoma patients with BRAFV600E -mutant tumors display striking responses to BRAF inhibitors (BRAFi); however, almost all invariably relapse with drug-resistant disease. Here, we report that microRNA-125a (miR-125a) expression is upregulated in human melanoma cells and patient tissues upon acquisition of BRAFi resistance. We show that miR-125a induction confers resistance to BRAFV600E melanoma cells to BRAFi by directly suppressing pro-apoptotic components of the intrinsic apoptosis pathway, including BAK1 and MLK3. Apoptotic suppression and prolonged survival favor reactivation of the MAPK and AKT pathways by drug-resistant melanoma cells. We demonstrate that miR-125a inhibition suppresses the emergence of resistance to BRAFi and, in a subset of resistant melanoma cell lines, leads to partial drug resensitization. Finally, we show that miR-125a upregulation is mediated by TGFβ signaling. In conclusion, the identification of this novel role for miR-125a in BRAFi resistance exposes clinically relevant mechanisms of melanoma cell survival that can be exploited therapeutically.
Collapse
Affiliation(s)
- Lisa Koetz-Ploch
- Department of Pathology, NYU School of Medicine, NYU Langone Medical Center, New York, NY, USA.,NYU Interdisciplinary Melanoma Cooperative Group, NYU School of Medicine, NYU Langone Medical Center, New York, NY, USA
| | - Douglas Hanniford
- Department of Pathology, NYU School of Medicine, NYU Langone Medical Center, New York, NY, USA.,NYU Interdisciplinary Melanoma Cooperative Group, NYU School of Medicine, NYU Langone Medical Center, New York, NY, USA
| | - Igor Dolgalev
- Genomics Technology Center, NYU School of Medicine, NYU Langone Medical Center, New York, NY, USA
| | - Elena Sokolova
- Department of Pathology, NYU School of Medicine, NYU Langone Medical Center, New York, NY, USA.,NYU Interdisciplinary Melanoma Cooperative Group, NYU School of Medicine, NYU Langone Medical Center, New York, NY, USA
| | - Judy Zhong
- NYU Interdisciplinary Melanoma Cooperative Group, NYU School of Medicine, NYU Langone Medical Center, New York, NY, USA.,Division of Biostatistics, Department of Environmental Medicine, NYU School of Medicine, NYU Langone Medical Center, New York, NY, USA
| | | | | | - Farbod Darvishian
- Department of Pathology, NYU School of Medicine, NYU Langone Medical Center, New York, NY, USA.,NYU Interdisciplinary Melanoma Cooperative Group, NYU School of Medicine, NYU Langone Medical Center, New York, NY, USA
| | - Keith T Flaherty
- Massachusetts General Hospital, Harvard University, Boston, MA, USA
| | - Paul B Chapman
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | | | - Eva Hernando
- Department of Pathology, NYU School of Medicine, NYU Langone Medical Center, New York, NY, USA.,NYU Interdisciplinary Melanoma Cooperative Group, NYU School of Medicine, NYU Langone Medical Center, New York, NY, USA
| |
Collapse
|
44
|
Masoud V, Pagès G. Targeted therapies in breast cancer: New challenges to fight against resistance. World J Clin Oncol 2017; 8:120-134. [PMID: 28439493 PMCID: PMC5385433 DOI: 10.5306/wjco.v8.i2.120] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 09/16/2016] [Accepted: 10/17/2016] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is the most common type of cancer found in women and today represents a significant challenge to public health. With the latest breakthroughs in molecular biology and immunotherapy, very specific targeted therapies have been tailored to the specific pathophysiology of different types of breast cancers. These recent developments have contributed to a more efficient and specific treatment protocol in breast cancer patients. However, the main challenge to be further investigated still remains the emergence of therapeutic resistance mechanisms, which develop soon after the onset of therapy and need urgent attention and further elucidation. What are the recent emerging molecular resistance mechanisms in breast cancer targeted therapy and what are the best strategies to apply in order to circumvent this important obstacle? The main scope of this review is to provide a thorough update of recent developments in the field and discuss future prospects for preventing resistance mechanisms in the quest to increase overall survival of patients suffering from the disease.
Collapse
|
45
|
Kim MH, Kim J. Role of YAP/TAZ transcriptional regulators in resistance to anti-cancer therapies. Cell Mol Life Sci 2017; 74:1457-1474. [PMID: 27826640 PMCID: PMC11107740 DOI: 10.1007/s00018-016-2412-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 10/15/2016] [Accepted: 11/03/2016] [Indexed: 12/11/2022]
Abstract
A diverse range of drug resistance mechanisms in cancer cells and their microenvironment significantly reduces the effectiveness of anti-cancer therapies. Growing evidence suggests that transcriptional effectors of the Hippo pathway, YAP and TAZ, promote resistance to various anti-cancer therapies, including cytotoxic chemotherapy, molecular targeted therapy, and radiation therapy. Here, we overview the role of YAP and TAZ as drug resistance mediators, and also discuss potential upstream regulators and downstream targets of YAP/TAZ in cancer. The widespread involvement of YAP and TAZ in resistance mechanisms suggests that therapeutic targeting of YAP and TAZ may expedite the development of effective anti-resistance therapies.
Collapse
Affiliation(s)
- Min Hwan Kim
- Graduate School of Medical Science and Engineering, KAIST, 291 Daehak-ro, Taejon, 34141, Republic of Korea
| | - Joon Kim
- Graduate School of Medical Science and Engineering, KAIST, 291 Daehak-ro, Taejon, 34141, Republic of Korea.
| |
Collapse
|
46
|
Bergamo A, Pelillo C, Chambery A, Sava G. Influence of components of tumour microenvironment on the response of HCT-116 colorectal cancer to the ruthenium-based drug NAMI-A. J Inorg Biochem 2017; 168:90-97. [DOI: 10.1016/j.jinorgbio.2016.11.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 11/22/2016] [Accepted: 11/30/2016] [Indexed: 12/30/2022]
|
47
|
Sui J, Cui Y, Cai H, Bian S, Xu Z, Zhou L, Sun Y, Liang J, Fan Y, Zhang X. Synergistic chemotherapeutic effect of sorafenib-loaded pullulan-Dox conjugate nanoparticles against murine breast carcinoma. NANOSCALE 2017; 9:2755-2767. [PMID: 28155940 DOI: 10.1039/c6nr09639e] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
pH-Sensitive pullulan-doxorubicin conjugates encapsulating sorafenib (P-Dox/S) nanoparticles were developed as a synergistic combinatorial delivery system against murine breast carcinoma. The nanoparticles can encapsulate Dox and sorafenib with ultra-high loading capacity (65.34 wt%) through chemical conjugation and physical loading, whereas can remain stable under physiological conditions and gradually release Dox and sorafenib with the decreasing pH. These conjugates can be effectively internalized and clearly suppress 4T1 cell growth in vitro. Furthermore, research data of in vivo animal models revealed that the synergistic combinatorial P-Dox/S nanoparticles heavily accumulated in solid tumor tissue sites to maximize therapeutic efficacy; they also significantly inhibited solid tumor growth, even remarkably reduced solid tumor volume in comparison to the initial volume, and obviously diminished adverse effects. The anti-tumor therapeutic effect obviously outperformed the delivery of combinational chemotherapy of free drugs or single drug-loaded P-Dox nanoparticles at the same concentration. These promising results indicate the high-efficiency synergistic chemotherapeutic effects of these nanoparticles. Combinational chemotherapy using P-Dox/S nanoparticles has important potential in the clinical treatment of malignancy for overcoming drug resistance and heterogeneity.
Collapse
Affiliation(s)
- Junhui Sui
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.
| | - Yani Cui
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.
| | - Hanxu Cai
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.
| | - Shaoquan Bian
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.
| | - Zhiyi Xu
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.
| | - Ling Zhou
- Cancer Center, West China hospital, Sichuan University, 37 Guoxue Lane, Chengdu 610064, China
| | - Yong Sun
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.
| | - Jie Liang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu 610064, China.
| |
Collapse
|
48
|
Xie F, Li BX, Xiao X. Design, synthesis and biological evaluation of regioisomers of 666-15 as inhibitors of CREB-mediated gene transcription. Bioorg Med Chem Lett 2017; 27:994-998. [PMID: 28073675 PMCID: PMC5296214 DOI: 10.1016/j.bmcl.2016.12.078] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/28/2016] [Accepted: 12/28/2016] [Indexed: 11/19/2022]
Abstract
cAMP-response element binding protein (CREB) is a nuclear transcription factor that has been implicated in the pathogenesis and maintenance of various types of human cancers. Identification of small molecule inhibitors of CREB-mediated gene transcription has been pursued as a novel strategy for developing cancer therapeutics. We recently discovered a potent and cell-permeable CREB inhibitor called 666-15. 666-15 is a bisnaphthamide and has been shown to possess efficacious anti-breast cancer activity without toxicity in vivo. In this study, we designed and synthesized a series of analogs of 666-15 to probe the importance of regiochemistry in naphthalene ring B. Biological evaluations of these analogs demonstrated that the substitution pattern of the alkoxy and carboxamide in naphthalene ring B is very critical for maintaining potent CREB inhibition activity, suggesting that the unique bioactive conformation accessible in 666-15 is critically important.
Collapse
Affiliation(s)
- Fuchun Xie
- Program in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Bingbing X Li
- Program in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA
| | - Xiangshu Xiao
- Program in Chemical Biology, Department of Physiology and Pharmacology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA; Knight Cardiovascular Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239, USA.
| |
Collapse
|
49
|
Wilms D, Andrä J. Comparison of patient-derived high and low phosphatidylserine-exposing colorectal carcinoma cells in their interaction with anti-cancer peptides. J Pept Sci 2017; 23:56-67. [PMID: 28066958 DOI: 10.1002/psc.2963] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 12/01/2016] [Accepted: 12/07/2016] [Indexed: 01/10/2023]
Abstract
Current cancer treatment is frequently compromised by severe adverse effects on healthy cells and tissues as well as by the increasing burden of (multi-)drug resistances. Some representatives of small, amphipathic peptides known as host defense peptides possess the potential to overcome these limitations and to evolve as future anti-cancer therapeutics. Peptide NK-2, derived from porcine NK-lysin, was originally discovered due to its broad-spectrum antimicrobial activities. Today, also potent anti-cancer activity is proven and accompanied by low toxicity towards normal human cells. The molecular basis underlying this target selectivity remains rather elusive. Nevertheless, it is presumptive that preferential peptide interactions with surface factors non-abundant on healthy human cells play a key role. Here, we investigated the cytotoxicity of peptide NK-2 and structurally improved anti-cancer variants thereof against two patient-derived colorectal cancer cell lines, exposing high and low levels of phosphatidylserine on their cell surfaces, respectively. Concluding from a range of in vitro tests involving cellular as well as lipid vesicle-based methods, it is proposed that the magnitude of the accessible membrane surface charge is not a primarily decisive factor for selective peptide interactions. Instead, it is suggested that the level of membrane surface-exposed phosphatidylserine is of crucial importance for the activity of peptide NK-2 and enhanced variants thereof in terms of their cancer cell selectivity, the overall efficacy, as well as the underlying mode of action and kinetics. Copyright © 2017 European Peptide Society and John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Dominik Wilms
- Faculty of Life Sciences, Department of Biotechnology, Hamburg University of Applied Sciences, Ulmenliet 20, D-21033, Hamburg, Germany
| | - Jörg Andrä
- Faculty of Life Sciences, Department of Biotechnology, Hamburg University of Applied Sciences, Ulmenliet 20, D-21033, Hamburg, Germany
| |
Collapse
|
50
|
Gogoi B, Gogoi D, Silla Y, Kakoti BB, Bhau BS. Network pharmacology-based virtual screening of natural products from Clerodendrum species for identification of novel anti-cancer therapeutics. MOLECULAR BIOSYSTEMS 2017; 13:406-416. [DOI: 10.1039/c6mb00807k] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the present work, latest network pharmacological approach has been used for the screening of natural anticancer compounds from Clerodendrum species.
Collapse
Affiliation(s)
- Barbi Gogoi
- Plant Genomic Laboratory
- Medicinal Aromatic & Economic Plants (MAEP) Group
- Biological Sciences & Technology Division (BSTD)
- CSIR-North East Institute of Science and Technology
- Jorhat-785006
| | - Dhrubajyoti Gogoi
- DBT-BIF
- Centre for Biotechnology and Bioinformatics
- Dibrugarh University
- Dibrugarh
- India
| | - Yumnam Silla
- Biotechnology Group
- Biological Sciences & Technology Division (BSTD)
- CSIR-North East Institute of Science and Technology
- Jorhat-785006
- India
| | | | - Brijmohan Singh Bhau
- Plant Genomic Laboratory
- Medicinal Aromatic & Economic Plants (MAEP) Group
- Biological Sciences & Technology Division (BSTD)
- CSIR-North East Institute of Science and Technology
- Jorhat-785006
| |
Collapse
|