51
|
Chang J, Wang R, Yu K, Zhang T, Chen X, Liu Y, Shi R, Wang X, Xia Q, Ma S. Genome-wide CRISPR screening reveals genes essential for cell viability and resistance to abiotic and biotic stresses in Bombyx mori. Genome Res 2020; 30:757-767. [PMID: 32424075 PMCID: PMC7263191 DOI: 10.1101/gr.249045.119] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/30/2020] [Indexed: 12/19/2022]
Abstract
High-throughput genetic screens are powerful methods to interrogate gene function on a genome-wide scale and identify genes responsible to certain stresses. Here, we developed a piggyBac strategy to deliver pooled sgRNA libraries stably into cell lines. We used this strategy to conduct a screen based on genome-wide clustered regularly interspaced short palindromic repeat technology (CRISPR)-Cas9 in Bombyx mori cells. We first constructed a single guide RNA (sgRNA) library containing 94,000 sgRNAs, which targeted 16,571 protein-coding genes. We then generated knockout collections in BmE cells using the piggyBac transposon. We identified 1006 genes that are essential for cell viability under normal growth conditions. Of the identified genes, 82.4% (829 genes) were homologous to essential genes in seven animal species. We also identified 838 genes whose loss facilitated cell growth. Next, we performed context-specific positive screens for resistance to biotic or nonbiotic stresses using temperature and baculovirus separately, which identified several key genes and pathways from each screen. Collectively, our results provide a novel and versatile platform for functional annotations of B. mori genomes and deciphering key genes responsible for various conditions. This study also shows the effectiveness, practicality, and convenience of genome-wide CRISPR screens in nonmodel organisms.
Collapse
Affiliation(s)
- Jiasong Chang
- Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Ruolin Wang
- Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Kai Yu
- Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Tong Zhang
- Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Xiaoxu Chen
- Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Yue Liu
- Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Run Shi
- Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Xiaogang Wang
- Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Qingyou Xia
- Biological Science Research Center, Southwest University, Chongqing 400716, China
- Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing 400716, China
- Chongqing Key Laboratory of Sericulture, Southwest University, Chongqing 400716, China
| | - Sanyuan Ma
- Biological Science Research Center, Southwest University, Chongqing 400716, China
- Chongqing Engineering and Technology Research Center for Novel Silk Materials, Southwest University, Chongqing 400716, China
- Chongqing Key Laboratory of Sericulture, Southwest University, Chongqing 400716, China
| |
Collapse
|
52
|
Sato M. Phenotypic screening using large-scale genomic libraries to identify drug targets for the treatment of cancer. Oncol Lett 2020; 19:3617-3626. [PMID: 32391087 PMCID: PMC7204489 DOI: 10.3892/ol.2020.11512] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 02/04/2020] [Indexed: 02/06/2023] Open
Abstract
During malignant progression to overt cancer cells, normal cells accumulate multiple genetic and non-genetic changes, which result in the acquisition of various oncogenic properties, such as uncontrolled proliferation, drug resistance, invasiveness, anoikis-resistance, the ability to bypass oncogene-induced senescence and cancer stemness. To identify potential novel drug targets contributing to these malignant phenotypes, researchers have performed large-scale genomic screening using various in vitro and in vivo screening models and identified numerous promising cancer drug target genes. However, there are issues with these identified genes, such as low reproducibility between different datasets. In the present study, the recent advances in the functional screening for identification of cancer drug target genes are summarized, and current issues and future perspectives are discussed.
Collapse
Affiliation(s)
- Mitsuo Sato
- Department of Pathophysiological Laboratory Sciences, Nagoya University Graduate School of Medicine, Nagoya, Aichi 461-8673, Japan
| |
Collapse
|
53
|
Li Z, Wang B, Gu S, Jiang P, Sahu A, Chen CH, Han T, Shi S, Wang X, Traugh N, Liu H, Liu Y, Wu Q, Brown M, Xiao T, Boland GM, Shirley Liu X. CRISPR Screens Identify Essential Cell Growth Mediators in BRAF Inhibitor-resistant Melanoma. GENOMICS, PROTEOMICS & BIOINFORMATICS 2020; 18:26-40. [PMID: 32413516 PMCID: PMC7393575 DOI: 10.1016/j.gpb.2020.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/20/2020] [Accepted: 02/26/2020] [Indexed: 12/21/2022]
Abstract
BRAF is a serine/threonine kinase that harbors activating mutations in ∼7% of human malignancies and ∼60% of melanomas. Despite initial clinical responses to BRAF inhibitors, patients frequently develop drug resistance. To identify candidate therapeutic targets for BRAF inhibitor resistant melanoma, we conduct CRISPR screens in melanoma cells harboring an activating BRAF mutation that had also acquired resistance to BRAF inhibitors. To investigate the mechanisms and pathways enabling resistance to BRAF inhibitors in melanomas, we integrate expression, ATAC-seq, and CRISPR screen data. We identify the JUN family transcription factors and the ETS family transcription factor ETV5 as key regulators of CDK6, which together enable resistance to BRAF inhibitors in melanoma cells. Our findings reveal genes contributing to resistance to a selective BRAF inhibitor PLX4720, providing new insights into gene regulation in BRAF inhibitor resistant melanoma cells.
Collapse
Affiliation(s)
- Ziyi Li
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA
| | - Binbin Wang
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Department of Data Sciences, Dana-Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Shengqing Gu
- Department of Data Sciences, Dana-Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Peng Jiang
- Department of Data Sciences, Dana-Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Avinash Sahu
- Department of Data Sciences, Dana-Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Chen-Hao Chen
- Department of Data Sciences, Dana-Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Tong Han
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Sailing Shi
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Xiaoqing Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA
| | - Nicole Traugh
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA
| | - Hailing Liu
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yin Liu
- Department of Clinical Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Qiu Wu
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Myles Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA; Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Tengfei Xiao
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02215, USA.
| | - Genevieve M Boland
- Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - X Shirley Liu
- Department of Data Sciences, Dana-Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| |
Collapse
|
54
|
Fletcher L, Joshi SK, Traer E. Profile of Quizartinib for the Treatment of Adult Patients with Relapsed/Refractory FLT3-ITD-Positive Acute Myeloid Leukemia: Evidence to Date. Cancer Manag Res 2020; 12:151-163. [PMID: 32021432 PMCID: PMC6955578 DOI: 10.2147/cmar.s196568] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 12/05/2019] [Indexed: 01/16/2023] Open
Abstract
Acute myeloid leukemia (AML) is a clonal hematologic neoplasm characterized by rapid, uncontrolled cell growth of immature myeloid cells (blasts). There are numerous genetic abnormalities in AML, many of which are prognostic, but an increasing number are targets for drug therapy. One of the most common genetic abnormalities in AML are activating mutations in the FMS-like tyrosine kinase 3 receptor (FLT3). As a receptor tyrosine kinase, FLT3 was the first targetable genetic abnormality in AML. The first generation of FLT3 inhibitors were broad-spectrum kinase inhibitors that inhibited FLT3 among other proteins. Although clinically active, first-generation FLT3 inhibitors had limited success as single agents. This led to the development of a second generation of more selective FLT3 inhibitors. This review focuses on quizartinib, a potent second-generation FLT3 inhibitor. We discuss the clinical trial development, mechanisms of resistance, and the recent FDA decision to deny approval for quizartinib as a single agent in relapsed/refractory AML.
Collapse
Affiliation(s)
- Luke Fletcher
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Sunil K Joshi
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA.,School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Elie Traer
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR 97239, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
55
|
Morales ML, Arenas A, Ortiz-Ruiz A, Leivas A, Rapado I, Rodríguez-García A, Castro N, Zagorac I, Quintela-Fandino M, Gómez-López G, Gallardo M, Ayala R, Linares M, Martínez-López J. MEK inhibition enhances the response to tyrosine kinase inhibitors in acute myeloid leukemia. Sci Rep 2019; 9:18630. [PMID: 31819100 PMCID: PMC6901485 DOI: 10.1038/s41598-019-54901-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 11/19/2019] [Indexed: 12/28/2022] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3) is a key driver of acute myeloid leukemia (AML). Several tyrosine kinase inhibitors (TKIs) targeting FLT3 have been evaluated clinically, but their effects are limited when used in monotherapy due to the emergence of drug-resistance. Thus, a better understanding of drug-resistance pathways could be a good strategy to explore and evaluate new combinational therapies for AML. Here, we used phosphoproteomics to identify differentially-phosphorylated proteins in patients with AML and TKI resistance. We then studied resistance mechanisms in vitro and evaluated the efficacy and safety of rational combinational therapy in vitro, ex vivo and in vivo in mice. Proteomic and immunohistochemical studies showed the sustained activation of ERK1/2 in bone marrow samples of patients with AML after developing resistance to FLT3 inhibitors, which was identified as a common resistance pathway. We examined the concomitant inhibition of MEK-ERK1/2 and FLT3 as a strategy to overcome drug-resistance, finding that the MEK inhibitor trametinib remained potent in TKI-resistant cells and exerted strong synergy when combined with the TKI midostaurin in cells with mutated and wild-type FLT3. Importantly, this combination was not toxic to CD34+ cells from healthy donors, but produced survival improvements in vivo when compared with single therapy groups. Thus, our data point to trametinib plus midostaurin as a potentially beneficial therapy in patients with AML.
Collapse
Affiliation(s)
- María Luz Morales
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Alicia Arenas
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Alejandra Ortiz-Ruiz
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Alejandra Leivas
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Inmaculada Rapado
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
- Servicio de Hematología, Hospital 12 de Octubre, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), ISCIII, Madrid, Spain
| | - Alba Rodríguez-García
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Nerea Castro
- Servicio de Hematología, Hospital 12 de Octubre, Madrid, Spain
| | - Ivana Zagorac
- Breast Cancer Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Miguel Quintela-Fandino
- Breast Cancer Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Gonzalo Gómez-López
- Bioinformatics Unit, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Miguel Gallardo
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Rosa Ayala
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
- Servicio de Hematología, Hospital 12 de Octubre, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), ISCIII, Madrid, Spain
- Universidad Complutense de Madrid, Madrid, Spain
| | - María Linares
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain.
- Universidad Complutense de Madrid, Madrid, Spain.
| | - Joaquín Martínez-López
- H12O-CNIO Haematological Malignancies Clinical Research Unit, Hospital 12 de Octubre - Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
- Servicio de Hematología, Hospital 12 de Octubre, Madrid, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), ISCIII, Madrid, Spain
- Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
56
|
Mudgapalli N, Nallasamy P, Chava H, Chava S, Pathania AS, Gunda V, Gorantla S, Pandey MK, Gupta SC, Challagundla KB. The role of exosomes and MYC in therapy resistance of acute myeloid leukemia: Challenges and opportunities. Mol Aspects Med 2019; 70:21-32. [PMID: 31623866 PMCID: PMC7775410 DOI: 10.1016/j.mam.2019.10.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/27/2019] [Accepted: 10/06/2019] [Indexed: 12/12/2022]
Abstract
Acute myeloid leukemia (AML) is caused by abnormal production of white blood cells, red blood cells or platelets. The leukemia cells communicate with their microenvironment through nano-vesicle exosomes that are 30-100 nm in diameter. These nano-vesicles are released from body fluids upon fusion of an endocytic compartment with the cell membrane. Exosomes function as cargo to deliver signaling molecules to distant cells. This allows cross-talk between hematopoietic cells and other distant target cell environments. Exosomes support leukemia growth by acting as messengers between tumor cells and the microenvironment as well as inducing oncogenic factors such as c-Myc. Exosomes have also been used as biomarkers in the clinical diagnosis of leukemia. Glycogen synthase kinase-3 (GSK-3) and protein phosphatase 2A (PP2A) are two crucial signaling molecules involved in the AML pathogenesis and MYC stability. GSK-3 is a serine/threonine protein kinase that coordinates with over 40 different proteins during physiological/pathological conditions in blood cells. The dysregulation in GSK-3 has been reported during hematological malignancies. GSK-3 acts as a tumor suppressor by targeting c-MYC, MCL-1 and β-catenin. Conversely, GSK-3 can also act as tumor promoter in some instances. The pharmacological modulators of GSK-3 such as ABT-869, 6-Bromoindirubin-3'-oxime (BIO), GS-87 and LY2090314 have shown promise in the treatment of hematological malignancy. PP2A is a heterotrimeric serine/threonine phosphatase involved in the regulation of hematological malignancy. PP2A-activating drugs (PADs) can effectively antagonize leukemogenesis. The discovery of exosomes, kinase inhibitors and phosphatase activators have provided new hope to the leukemia patients. This review discusses the role of exosomes, GSK-3 and PP2A in the pathogenesis of leukemia. We provide evidence from both preclinical and clinical studies.
Collapse
Affiliation(s)
- Nithya Mudgapalli
- Department of Biochemistry and Molecular Biology, The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
; UNMC Summer Undergraduate Research Program, University of Nebraska Medical Center, Omaha, NE, USA
| | - Palanisamy Nallasamy
- Department of Biochemistry and Molecular Biology, The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Haritha Chava
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Srinivas Chava
- Department of Biochemistry and Molecular Biology, The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anup S Pathania
- Department of Biochemistry and Molecular Biology, The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Venugopal Gunda
- Pediatric Oncology Laboratory, Child Health Research Institute, University of Nebraska Medical Center, Omaha, NE, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Manoj K Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | - Subash C Gupta
- Laboratory for Translational Cancer Research, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221 005, India
| | - Kishore B Challagundla
- Department of Biochemistry and Molecular Biology, The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
.
| |
Collapse
|
57
|
Paul S, DiPippo AJ, Ravandi F, Kadia TM. Quizartinib in the treatment of FLT3-internal-tandem duplication-positive acute myeloid leukemia. Future Oncol 2019; 15:3885-3894. [PMID: 31559849 DOI: 10.2217/fon-2019-0353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
FLT3 mutations, characterized by an internal-tandem duplication or missense mutations in the tyrosine kinase domain, are observed in a third of patients with newly diagnosed acute myeloid leukemia. FLT3-ITD mutations are associated with high relapse rates and short overall survival with conventional chemotherapy. Several tyrosine kinase inhibitors targeting FLT3 have been developed in an effort to improve survival and therapeutic options. This review focuses on quizartinib, a second-generation FLT3 inhibitor that has demonstrated efficacy and safety as a single agent and in combination with chemotherapy. We discuss its clinical development as well as its place in the treatment of FLT3-mutated acute myeloid leukemia among the other FLT3 inhibtors currently available and its mechanisms of resistance.
Collapse
Affiliation(s)
- Shilpa Paul
- Department of Clinical Pharmacy, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0377, Houston, TX 77030, USA
| | - Adam J DiPippo
- Department of Clinical Pharmacy, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0377, Houston, TX 77030, USA
| | - Farhad Ravandi
- Department of Leukemia, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0428 Houston, TX 77030, USA
| | - Tapan M Kadia
- Department of Leukemia, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0428 Houston, TX 77030, USA
| |
Collapse
|
58
|
CRISPR Loss-of-Function Screen Identifies the Hippo Signaling Pathway as the Mediator of Regorafenib Efficacy in Hepatocellular Carcinoma. Cancers (Basel) 2019; 11:cancers11091362. [PMID: 31540262 PMCID: PMC6770429 DOI: 10.3390/cancers11091362] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/03/2019] [Accepted: 09/10/2019] [Indexed: 12/26/2022] Open
Abstract
Regorafenib is used for hepatocellular carcinoma (HCC), but its response does not last long, partly due to chemoresistance acquisition. We performed a clustered regularly interspaced short palindromic repeats (CRISPR)-based loss-of-function genetic screen and aimed to discover molecules involved in regorafenib resistance in HCC. Xenograft tumors established from Cas9-expressing HCC cells with pooled CRISPR kinome libraries were treated with regorafenib or a vehicle. Sequencing analysis identified 31 genes, with the abundance of multiple guide RNAs increased in regorafenib-treated tumors compared to that in vehicle-treated tumors, including 2 paralogues, LATS2 and LATS1, core components of the Hippo signaling pathway. Notably, all eight designed guide RNAs targeting LATS2 increased in regorafenib-treated tumors, suggesting that LATS2 deletion confers regorafenib resistance in HCC cells. LATS2 knockdown significantly mitigated the cytotoxic and proapoptotic effects of regorafenib on HCC cells. LATS2 inhibition stabilized the Hippo signaling mediator YAP, leading to the upregulation of antiapoptotic Bcl-xL and the multidrug resistance transporter ABCB1. Among 12 hepatoma cell lines, the half maximal inhibitory concentration (IC50) values of regorafenib were positively correlated with any of YAP, Bcl-xL and ABCB1 levels. The inhibition of YAP or Bcl-xL in regorafenib-insensitive HCC cells restored their susceptibility to regorafenib. In conclusion, our screen identified the Hippo signaling pathway as the mediator of regorafenib efficacy in HCC.
Collapse
|
59
|
Current and Future Horizons of Patient-Derived Xenograft Models in Colorectal Cancer Translational Research. Cancers (Basel) 2019; 11:cancers11091321. [PMID: 31500168 PMCID: PMC6770280 DOI: 10.3390/cancers11091321] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/27/2019] [Accepted: 09/02/2019] [Indexed: 12/18/2022] Open
Abstract
Our poor understanding of the intricate biology of cancer and the limited availability of preclinical models that faithfully recapitulate the complexity of tumors are primary contributors to the high failure rate of novel therapeutics in oncology clinical studies. To address this need, patient-derived xenograft (PDX) platforms have been widely deployed and have reached a point of development where we can critically review their utility to model and interrogate relevant clinical scenarios, including tumor heterogeneity and clonal evolution, contributions of the tumor microenvironment, identification of novel drugs and biomarkers, and mechanisms of drug resistance. Colorectal cancer (CRC) constitutes a unique case to illustrate clinical perspectives revealed by PDX studies, as they overcome limitations intrinsic to conventional ex vivo models. Furthermore, the success of molecularly annotated "Avatar" models for co-clinical trials in other diseases suggests that this approach may provide an additional opportunity to improve clinical decisions, including opportunities for precision targeted therapeutics, for patients with CRC in real time. Although critical weaknesses have been identified with regard to the ability of PDX models to predict clinical outcomes, for now, they are certainly the model of choice for preclinical studies in CRC. Ongoing multi-institutional efforts to develop and share large-scale, well-annotated PDX resources aim to maximize their translational potential. This review comprehensively surveys the current status of PDX models in translational CRC research and discusses the opportunities and considerations for future PDX development.
Collapse
|
60
|
Larrimore KE, Rancati G. The conditional nature of gene essentiality. Curr Opin Genet Dev 2019; 58-59:55-61. [PMID: 31470233 DOI: 10.1016/j.gde.2019.07.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 07/17/2019] [Accepted: 07/21/2019] [Indexed: 11/18/2022]
Abstract
Essential genes are classically defined as required for cellular viability and reproductive success. Despite this deceptively simple definition, several lines of evidence suggest that gene essentiality is instead a conditional trait. Indeed, gene essentiality has been shown to depend on the environmental and genetic context as well as the variable ability of cells to acquire adaptive mutations to survive inactivation of seemingly essential genes. Here, we will discuss these findings and highlight the mechanisms underlying the ability of cells to survive an essential gene deletion. Also, since essential genes are prioritized as targets for anticancer therapy, we discuss emergence of bypass resistance mechanisms toward targeted therapies as the result of the conditional nature of gene essentiality. To identify targets associated to a lower risk of relapse (i.e. the return of cancer following remission), we finally call for a coordinated effort to quantify the variable nature of gene essentiality across species, cell types, and growth conditions.
Collapse
Affiliation(s)
- Katherine E Larrimore
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A⁎STAR), 8A Biomedical Grove, Immunos #05, Singapore 138648, Singapore
| | - Giulia Rancati
- Institute of Medical Biology (IMB), Agency for Science, Technology and Research (A⁎STAR), 8A Biomedical Grove, Immunos #05, Singapore 138648, Singapore.
| |
Collapse
|
61
|
Zhao WS, Yan WP, Chen DB, Dai L, Yang YB, Kang XZ, Fu H, Chen P, Deng KJ, Wang XY, Xie XW, Chen HS, Chen KN. Genome-scale CRISPR activation screening identifies a role of ELAVL2-CDKN1A axis in paclitaxel resistance in esophageal squamous cell carcinoma. Am J Cancer Res 2019; 9:1183-1200. [PMID: 31285951 PMCID: PMC6610048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 05/07/2019] [Indexed: 06/09/2023] Open
Abstract
Neoadjuvant chemotherapy (NAC) may provide survival benefits for patients with advanced esophageal squamous cell carcinoma. However, tumor cells can display primary or secondary resistance to paclitaxel (PTX), a primary component of induction chemotherapy regimen. To identify genes capable of conveying PTX resistance, we performed a genome-wide CRISPR transcriptional activation library in human KYSE-180 cells. High throughput next generation sequencing was further applied to establish the phenotype-to-genotype relationship. Our highest-ranking hits are CDKN1A, TSPAN4, ELAVL2, JUNB and PAAF1. We generated evidence that esophageal tumors with high CDKN1A, ELAVL2 and TSPAN4 levels, quantified using qRT-PCR and Western blot assays, showed poorer chemotherapy response. Higher expression levels of TSPAN4 and ELAVL2 protein are independent risk factors for poor chemotherapy response in ESCC patients. We then found that overexpression of CDKN1A, ELAVL2 or TSPAN4 in ESCC cell lines significantly promoted the resistance to PTX by inhibiting cell apoptosis. Interestingly, ESCC cells overexpressed CDKN1A, ELAVL2 or TSPAN4 also acquired resistance to cisplatin (DDP). This phenomenon may be explained by cross-resistance of chemotherapy. We additionally found an association between ELAVL2 and CDKN1A, which may be regarded as the upstream and downstream factors that synergistically involved in the regulation of chemo-resistance in ESCC. Therefore, our study demonstrated that the genome-wide CRISPR activation library is a powerful strategy for the discovery of chemo-resistant genes critical for ESCC and we reported the first evidence that the ELAVL2-CDKN1A axis may be an important mechanism involved in chemo-resistance in ESCC.
Collapse
Affiliation(s)
- Wen-Si Zhao
- Department of Thoracic Surgery I, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Wan-Pu Yan
- Department of Thoracic Surgery I, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Dong-Bo Chen
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver DiseaseBeijing 100044, China
| | - Liang Dai
- Department of Thoracic Surgery I, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Yong-Bo Yang
- Department of Thoracic Surgery I, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Xiao-Zheng Kang
- Department of Thoracic Surgery I, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Hao Fu
- Department of Thoracic Surgery I, Peking University Cancer Hospital and InstituteBeijing 100142, China
| | - Pu Chen
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver DiseaseBeijing 100044, China
| | - Kang-Jian Deng
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver DiseaseBeijing 100044, China
| | - Xue-Yan Wang
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver DiseaseBeijing 100044, China
| | - Xing-Wang Xie
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver DiseaseBeijing 100044, China
| | - Hong-Song Chen
- Peking University People’s Hospital, Peking University Hepatology Institute, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver DiseaseBeijing 100044, China
| | - Ke-Neng Chen
- Department of Thoracic Surgery I, Peking University Cancer Hospital and InstituteBeijing 100142, China
| |
Collapse
|
62
|
Zhou F, Ge Z, Chen B. Quizartinib (AC220): a promising option for acute myeloid leukemia. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:1117-1125. [PMID: 31114157 PMCID: PMC6497874 DOI: 10.2147/dddt.s198950] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/04/2019] [Indexed: 12/27/2022]
Abstract
Quizartinib is an effective therapy for patients with FLT3-ITD acute myeloid leukemia (AML) by continuing to inhibit the activity of FLT3 gene, leading to apoptosis of tumor cells. Multiple clinical trials have proved that it is effective in relapsed or refractory AML with an FLT3-ITD mutation. In this review, we focus on the characteristics of FLT3/ITD mutations, the mechanism and pharmacokinetics of quizartinib, and the mechanisms of resistance to quizartinib. We also summarize clinical experiences and adverse effects with quizartinib and recommend crucial approaches of quizartinib in the therapy of patients with newly diagnosed AML and patients with relapsed/refractory AML, particularly those with FLT3-ITD mutation. Quizartinib presents its advantages as a very promising agent in the treatment of AML, especially in patients with FLT3-ITD mutations. FLT3/ITD mutation can lead to constitutive autophosphorylation of FLT3 and activation of its downstream effectors including RAS/RAF/MEK, MAPK/ERK, PI3K/AKT/mTOR and JAK/STAT5 signal pathways, while Quizartinib can inhibit these downstream pathways through specific FLT3 inhibition. Quizartinib has received US Food and Drug Administration breakthrough therapy designation in patients with relapsed/refractory FLT3-ITD AML based on clinical trials. A larger sample of clinical trials are needed to verify its safety and efficacy, and the efficacy of quizartinib combined with chemotherapy or allogeneic hematopoietic cell transplantation should also be estimated in clinical trials. Meanwhile, for the side effects of quizartinib, further studies are needed to find a way to reduce its toxicity.
Collapse
Affiliation(s)
- Fang Zhou
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu Province, People's Republic of China
| | - Zheng Ge
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu Province, People's Republic of China
| | - Baoan Chen
- Department of Hematology and Oncology (Key Department of Jiangsu Medicine), Zhongda Hospital, Medical School, Southeast University, Nanjing, Jiangsu Province, People's Republic of China
| |
Collapse
|
63
|
Bakke J, Wright WC, Zamora AE, Oladimeji P, Crawford JC, Brewer CT, Autry RJ, Evans WE, Thomas PG, Chen T. Genome-wide CRISPR screen reveals PSMA6 to be an essential gene in pancreatic cancer cells. BMC Cancer 2019; 19:253. [PMID: 30898113 PMCID: PMC6429770 DOI: 10.1186/s12885-019-5455-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 03/12/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Despite its relatively low incidence, pancreatic ductal adenocarcinoma (PDAC) is a leading cause of cancer deaths because of the aggressive growth/metastasis of the tumor, the lack of early symptoms, and the poor treatment options. Basic research to identify potential therapeutic targets for PDAC is greatly needed. METHODS We used a negative-selection genome-wide CRISPR screen to identify essential genes in the PANC-1 human pancreatic carcinoma cell line. We validated the top hits with follow-up siRNA screens, using the HPNE, HPAF-II, AsPC-1, and Mia PaCa-2 cell lines. RESULTS The PSMA6 gene was an identified candidate hit after the CRISPR screen, siRNA validation screen, and siRNA deconvolution screen. Spheroid formation assays and flow cytometry analysis showed that PSMA6 is critical for survival in many pancreatic ductal carcinoma cell models. Lastly, as PSMA6 protein is a proteosomal subunit of the 20S core complex, we showed that bortezomib, a proteasome inhibitor, was especially toxic in PANC-1 cells. CONCLUSIONS Further study of PSMA6 and the proteasome subunit that it encodes, along with other hits identified in our CRISPR screens, may provide valuable insights into potential therapeutic targets for PDAC.
Collapse
Affiliation(s)
- Jesse Bakke
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Department of Foundational Sciences, College of Medicine, Central Michigan University, Mount Pleasant, MI, USA.
| | - William C Wright
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
- Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Anthony E Zamora
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Peter Oladimeji
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Christopher T Brewer
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA
- Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Robert J Autry
- Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - William E Evans
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Taosheng Chen
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
64
|
Ghosh D, Venkataramani P, Nandi S, Bhattacharjee S. CRISPR-Cas9 a boon or bane: the bumpy road ahead to cancer therapeutics. Cancer Cell Int 2019; 19:12. [PMID: 30636933 PMCID: PMC6325665 DOI: 10.1186/s12935-019-0726-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 01/02/2019] [Indexed: 12/13/2022] Open
Abstract
Genome editing allows for the precise manipulation of DNA sequences in a cell making this technology essential for understanding gene function. CRISPR/Cas9 is a targeted genome-editing platform derived from bacterial adaptive immune system and has been repurposed into a genome-editing tool. The RNA-guided DNA endonuclease, Cas9 can be easily programmed to target new sites by altering its guide RNA sequence, making this technology easier, more efficient, scalable and an indispensable tool in biological research. This technology has helped genetically engineer animal models to understand disease mechanisms and elucidate molecular details that can be exploited for improved therapeutic outcomes. In this review, we describe the CRISPR-Cas9 gene-editing mechanism, CRISPR-screening methods, therapeutic targeting of CRISPR in animal models and in cancer immunotherapy. We also discuss the ongoing clinical trials using this tool, limitations of this tool that might impede the clinical applicability of CRISPR-Cas9 and future directions for developing effective CRISPR-Cas9 delivery systems that may improve cancer therapeutics.
Collapse
Affiliation(s)
- Debarati Ghosh
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY USA
| | | | - Saikat Nandi
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY USA
| | | |
Collapse
|
65
|
Chow RD, Chen S. Cancer CRISPR Screens In Vivo. Trends Cancer 2018; 4:349-358. [PMID: 29709259 DOI: 10.1016/j.trecan.2018.03.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 03/04/2018] [Accepted: 03/05/2018] [Indexed: 02/08/2023]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR) screening is a powerful toolset for investigating diverse biological processes. Most CRISPR screens to date have been performed with in vitro cultures or cellular transplant models. To interrogate cancer in animal models that more closely recapitulate the human disease, autochthonous direct in vivo CRISPR screens have recently been developed that can identify causative drivers in the native tissue microenvironment. By empowering multiplexed mutagenesis in fully immunocompetent animals, direct in vivo CRISPR screens enable the rapid generation of patient-specific avatars that can guide precision medicine. This Opinion article discusses the current status of in vivo CRISPR screens in cancer and offers perspectives on future applications.
Collapse
Affiliation(s)
- Ryan D Chow
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA; Systems Biology Institute, Yale University School of Medicine, West Haven, CT, USA; Medical Scientist Training Program, Yale University School of Medicine, New Haven, CT, USA
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA; Systems Biology Institute, Yale University School of Medicine, West Haven, CT, USA; Medical Scientist Training Program, Yale University School of Medicine, New Haven, CT, USA; Biological and Biomedical Sciences Program, Yale University School of Medicine, New Haven, CT, USA; Immunobiology Program, Yale University School of Medicine, New Haven, CT, USA; Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA; Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
66
|
Sharma S, Petsalaki E. Application of CRISPR-Cas9 Based Genome-Wide Screening Approaches to Study Cellular Signalling Mechanisms. Int J Mol Sci 2018; 19:E933. [PMID: 29561791 PMCID: PMC5979383 DOI: 10.3390/ijms19040933] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 03/15/2018] [Accepted: 03/18/2018] [Indexed: 12/26/2022] Open
Abstract
The cellular signalling process is a highly complex mechanism, involving multiple players, which together orchestrate the cell's response to environmental changes and perturbations. Given the multitude of genes that participate in the process of cellular signalling, its study in a genome-wide manner has proven challenging. Recent advances in gene editing technologies, including clustered regularly-interspaced short palindromic repeats/Cas9 (CRISPR/Cas9) approaches, have opened new opportunities to investigate global regulatory signalling programs of cells in an unbiased manner. In this review, we focus on how the application of pooled genetic screening approaches using the CRISPR/Cas9 system has contributed to a systematic understanding of cellular signalling processes in normal and disease contexts.
Collapse
Affiliation(s)
- Sumana Sharma
- European Bioinformatics Institute, European Molecular Biology Laboratory, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK.
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK.
| | - Evangelia Petsalaki
- European Bioinformatics Institute, European Molecular Biology Laboratory, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK.
| |
Collapse
|