251
|
Fairhurst RA, Furet P, Imbach-Weese P, Stauffer F, Rueeger H, McCarthy C, Ripoche S, Oswald S, Arnaud B, Jary A, Maira M, Schnell C, Guthy DA, Wartmann M, Kiffe M, Desrayaud S, Blasco F, Widmer T, Seiler F, Gutmann S, Knapp M, Caravatti G. Identification of NVP-CLR457 as an Orally Bioavailable Non-CNS-Penetrant pan-Class IA Phosphoinositol-3-Kinase Inhibitor. J Med Chem 2022; 65:8345-8379. [PMID: 35500094 DOI: 10.1021/acs.jmedchem.2c00267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Balanced pan-class I phosphoinositide 3-kinase inhibition as an approach to cancer treatment offers the prospect of treating a broad range of tumor types and/or a way to achieve greater efficacy with a single inhibitor. Taking buparlisib as the starting point, the balanced pan-class I PI3K inhibitor 40 (NVP-CLR457) was identified with what was considered to be a best-in-class profile. Key to the optimization to achieve this profile was eliminating a microtubule stabilizing off-target activity, balancing the pan-class I PI3K inhibition profile, minimizing CNS penetration, and developing an amorphous solid dispersion formulation. A rationale for the poor tolerability profile of 40 in a clinical study is discussed.
Collapse
Affiliation(s)
- Robin A Fairhurst
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Pascal Furet
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | | | - Frédéric Stauffer
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Heinrich Rueeger
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Clive McCarthy
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Sebastien Ripoche
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Susanne Oswald
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Bertrand Arnaud
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Aline Jary
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Michel Maira
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Christian Schnell
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Daniel A Guthy
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Markus Wartmann
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Michael Kiffe
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | | | - Francesca Blasco
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Toni Widmer
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Frank Seiler
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Sascha Gutmann
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| | - Mark Knapp
- Novartis Institutes for BioMedical Research, Emeryville, California 94608, United States
| | - Giorgio Caravatti
- Novartis Institutes for BioMedical Research, Basel CH-4002, Switzerland
| |
Collapse
|
252
|
Chandran SS, Ma J, Klatt MG, Dündar F, Bandlamudi C, Razavi P, Wen HY, Weigelt B, Zumbo P, Fu SN, Banks LB, Yi F, Vercher E, Etxeberria I, Bestman WD, Da Cruz Paula A, Aricescu IS, Drilon A, Betel D, Scheinberg DA, Baker BM, Klebanoff CA. Immunogenicity and therapeutic targeting of a public neoantigen derived from mutated PIK3CA. Nat Med 2022; 28:946-957. [PMID: 35484264 PMCID: PMC9117146 DOI: 10.1038/s41591-022-01786-3] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 03/16/2022] [Indexed: 01/05/2023]
Abstract
Public neoantigens (NeoAgs) represent an elite class of shared cancer-specific epitopes derived from recurrently mutated driver genes. Here we describe a high-throughput platform combining single-cell transcriptomic and T cell receptor (TCR) sequencing to establish whether mutant PIK3CA, among the most frequently genomically altered driver oncogenes, generates an immunogenic public NeoAg. Using this strategy, we developed a panel of TCRs that recognize an endogenously processed neopeptide encompassing a common PIK3CA hotspot mutation restricted by the prevalent human leukocyte antigen (HLA)-A*03:01 allele. Mechanistically, immunogenicity to this public NeoAg arises from enhanced neopeptide/HLA complex stability caused by a preferred HLA anchor substitution. Structural studies indicated that the HLA-bound neopeptide presents a comparatively 'featureless' surface dominated by the peptide's backbone. To bind this epitope with high specificity and affinity, we discovered that a lead TCR clinical candidate engages the neopeptide through an extended interface facilitated by an unusually long CDR3β loop. In patients with diverse malignancies, we observed NeoAg clonal conservation and spontaneous immunogenicity to the neoepitope. Finally, adoptive transfer of TCR-engineered T cells led to tumor regression in vivo in mice bearing PIK3CA-mutant tumors but not wild-type PIK3CA tumors. Together, these findings establish the immunogenicity and therapeutic potential of a mutant PIK3CA-derived public NeoAg.
Collapse
Affiliation(s)
- Smita S Chandran
- Human Oncology and Pathogenesis Program (HOPP), Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Parker Institute for Cancer Immunotherapy, New York, NY, USA.
| | - Jiaqi Ma
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| | - Martin G Klatt
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Friederike Dündar
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - Chaitanya Bandlamudi
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pedram Razavi
- Human Oncology and Pathogenesis Program (HOPP), Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Hannah Y Wen
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Paul Zumbo
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
| | - Si Ning Fu
- Human Oncology and Pathogenesis Program (HOPP), Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lauren B Banks
- Human Oncology and Pathogenesis Program (HOPP), Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fei Yi
- Human Oncology and Pathogenesis Program (HOPP), Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Enric Vercher
- Human Oncology and Pathogenesis Program (HOPP), Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Inaki Etxeberria
- Human Oncology and Pathogenesis Program (HOPP), Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Watchain D Bestman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Arnaud Da Cruz Paula
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ilinca S Aricescu
- Human Oncology and Pathogenesis Program (HOPP), Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexander Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
- Early Drug Development Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Doron Betel
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, USA
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - David A Scheinberg
- Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Brian M Baker
- Department of Chemistry and Biochemistry, University of Notre Dame, South Bend, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| | - Christopher A Klebanoff
- Human Oncology and Pathogenesis Program (HOPP), Immuno-Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Parker Institute for Cancer Immunotherapy, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Division of Hematology and Medical Oncology, Weill Cornell Medicine, New York, NY, USA.
- Early Drug Development Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Cell Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
253
|
Zhang C, Li D, Xiao B, Zhou C, Jiang W, Tang J, Li Y, Zhang R, Han K, Hou Z, Zhang L, Sui Q, Liao L, Pan Z, Zhang X, Ding P. B2M and JAK1/2-mutated MSI-H Colorectal Carcinomas Can Benefit From Anti-PD-1 Therapy. J Immunother 2022; 45:187-193. [PMID: 35343934 PMCID: PMC8986629 DOI: 10.1097/cji.0000000000000417] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/20/2022] [Indexed: 11/25/2022]
Abstract
β2-microglobulin (B2M) and Janus kinases 1 and 2 (JAK1/2) mutations have been suggested as genetic mechanisms of immune evasion for anti-programmed cell death protein 1 (PD-1) therapy. Whether B2M and JAK1/2 lose-of-function mutation can cause primary resistance to anti-PD-1 therapy in colorectal carcinoma (CRC) patients remains controversial. Here, we sought to compare the efficacy of anti-PD-1 therapy in DNA mismatch repair deficient/microsatellite instability-high CRC patients with or without B2M or JAK1/2 mutations. Thirty-Five CRC patients who received anti-PD-1 therapy were enrolled in this study. All tumor samples underwent next-generation sequencing. The clinical and molecular data from 110 CRC patients sequenced with the Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT) assay and accessed through cBioportal were also analyzed in this study. Of the 35 CRC patients from our center, 10 (28.6%) had a B2M loss-of-function mutation, and 8 (22.9%) had a JAK1/2 loss-of-function mutation. Compared with B2M wild-type CRCs, B2M-mutated CRCs did not show a higher frequency of resistance to anti-PD-1 therapy (P=0.71). There was even better response to anti-PD-1 therapy in patients with JAK1/2 mutation than in those without (P=0.015). Of the 110 CRC patients in the MSK-IMPACT datasets, 13 (11.8%) had a B2M mutation, and 15 (13.6%) had a JAK1/2 mutation. After analyzing the response to anti-PD-1 therapy in these 110 patients, we found similar results (P=0.438 and 0.071, respectively). Moreover, patients with B2M or JAK1/2 mutation had a lower tumor mutational burden score compared with those without. B2M and JAK1/2 loss-of-function mutations occur frequently in microsatellite instability-high CRC. Our study demonstrated that patients with CRC harboring B2M or JAK1/2 mutations should not be excluded from anti-PD-1 therapy.
Collapse
Affiliation(s)
- Chenzhi Zhang
- Departments of Colorectal Surgery
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, P.R. China
| | - Dandan Li
- Biological Therapy Center
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, P.R. China
| | - Binyi Xiao
- Departments of Colorectal Surgery
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, P.R. China
| | - Chi Zhou
- Departments of Colorectal Surgery
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, P.R. China
| | - Wu Jiang
- Departments of Colorectal Surgery
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, P.R. China
| | - Jinghua Tang
- Departments of Colorectal Surgery
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, P.R. China
| | - Yuan Li
- Departments of Colorectal Surgery
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, P.R. China
| | - Rongxin Zhang
- Departments of Colorectal Surgery
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, P.R. China
| | - Kai Han
- Departments of Colorectal Surgery
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, P.R. China
| | - Zhenlin Hou
- Departments of Colorectal Surgery
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, P.R. China
| | - Linjie Zhang
- Departments of Colorectal Surgery
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, P.R. China
| | - Qiaoqi Sui
- Departments of Colorectal Surgery
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, P.R. China
| | - Leen Liao
- Departments of Colorectal Surgery
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, P.R. China
| | - Zhizhong Pan
- Departments of Colorectal Surgery
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, P.R. China
| | - Xiaoshi Zhang
- Biological Therapy Center
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, P.R. China
| | - Peirong Ding
- Departments of Colorectal Surgery
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong Province, P.R. China
| |
Collapse
|
254
|
Luo ML, Huang W, Zhu HP, Peng C, Zhao Q, Han B. Advances in indole-containing alkaloids as potential anticancer agents by regulating autophagy. Biomed Pharmacother 2022; 149:112827. [PMID: 35316753 DOI: 10.1016/j.biopha.2022.112827] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/03/2022] [Accepted: 03/14/2022] [Indexed: 11/02/2022] Open
Abstract
Cancer is a leading cause of death worldwide, and cancer development is often associated with disturbances in the autophagy process. Autophagy is a catabolic process involved in many physiological processes, crucial for cell growth and survival. It is an intracellular lysosomal/vacuolar degradation system. In this system, inner cytoplasmic cell membrane is degraded by lysosomal hydrolases, and the products are released back into the cytoplasm. Indole alkaloids are natural products extensively found in nature and have been proven to possess various pharmacological activities. In recent years, pharmacological studies have demonstrated another potential of indole alkaloids, autophagy regulation. The regulation may contribute to the efficacy of indole alkaloids in preventing and treating cancer. This review summarizes the current understanding of indole alkaloids' effect on tumor cells and autophagy. Then, we focus on mechanisms by which indole alkaloids can target the autophagy process associated with cancer, including the PI3K/Akt/mTOR signaling pathway, MAPK signaling pathway, ROS signaling pathway, Beclin-1, and so on. Literature has been surveyed primarily from 2009 to Nov. 2021, and some semisynthetic or fully synthetic indole derivatives are also discussed.
Collapse
Affiliation(s)
- Meng-Lan Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong-Ping Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qian Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
255
|
Thorwarth A, Haase K, Röefzaad C, Pajtler KW, Schramm K, Hauptmann K, Behnke A, Vokuhl C, Elgeti T, Gratopp A, Schulte JH, Scheer M, Hernáiz Driever P, Nysom K, Eggert A, Henssen AG, Deubzer HE. Genomic Evolution and Personalized Therapy of an Infantile Fibrosarcoma Harboring an NTRK Oncogenic Fusion. JCO Precis Oncol 2022; 6:e2100283. [PMID: 35613412 PMCID: PMC9200398 DOI: 10.1200/po.21.00283] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Precision medicine for infantile fibrosarcoma by monitoring of spatial and temporal clonal evolution (requested from authors: Would you be so kind to let us know when the article is announced via Twitter?).![]()
Collapse
Affiliation(s)
- Anne Thorwarth
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Kerstin Haase
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany.,Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Partner Sites Berlin and Heidelberg, Germany
| | - Claudia Röefzaad
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany.,Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Kristian W Pajtler
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Partner Sites Berlin and Heidelberg, Germany.,Department of Pediatric Hematology and Oncology, University Hospital Heidelberg, Heidelberg, Germany.,Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
| | - Kathrin Schramm
- German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Partner Sites Berlin and Heidelberg, Germany.,Department of Pediatric Hematology and Oncology, University Hospital Heidelberg, Heidelberg, Germany.,Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
| | - Kathrin Hauptmann
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Anke Behnke
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Christian Vokuhl
- Section of Pediatric Pathology, Institute of Pathology, University Hospital Bonn, Bonn, Germany
| | - Thomas Elgeti
- Department of Radiology (including Pediatric Radiology), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Alexander Gratopp
- Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité-Universitäts-Medizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Johannes H Schulte
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Partner Sites Berlin and Heidelberg, Germany
| | - Monika Scheer
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Pablo Hernáiz Driever
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Karsten Nysom
- Department of Pediatrics and Adolescent Medicine, Juliane Marie Centre, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Angelika Eggert
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Partner Sites Berlin and Heidelberg, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Berlin, Germany
| | - Anton G Henssen
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany.,Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Partner Sites Berlin and Heidelberg, Germany
| | - Hedwig E Deubzer
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany.,Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), Partner Sites Berlin and Heidelberg, Germany.,Berliner Institut für Gesundheitsforschung (BIH), Berlin, Germany
| |
Collapse
|
256
|
Huyghe N, Benidovskaya E, Stevens P, Van den Eynde M. Biomarkers of Response and Resistance to Immunotherapy in Microsatellite Stable Colorectal Cancer: Toward a New Personalized Medicine. Cancers (Basel) 2022; 14:2241. [PMID: 35565369 PMCID: PMC9105843 DOI: 10.3390/cancers14092241] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 11/28/2022] Open
Abstract
Immune Checkpoint Inhibitors (ICIs) are well recognized as a major immune treatment modality for multiple types of solid cancers. However, for colorectal cancer (CRC), ICIs are only approved for the treatment of Mismatch-Repair-Deficient and Microsatellite Instability-High (dMMR/MSI-H) tumors. For the vast majority of CRC, that are not dMMR/MSI-H, ICIs alone provide limited to no clinical benefit. This discrepancy of response between CRC and other solid cancers suggests that CRC may be inherently resistant to ICIs alone. In translational research, efforts are underway to thoroughly characterize the immune microenvironment of CRC to better understand the mechanisms behind this resistance and to find new biomarkers of response. In the clinic, trials are being set up to study biomarkers along with treatments targeting newly discovered immune checkpoint molecules or treatments combining ICIs with other existing therapies to improve response in MSS CRC. In this review, we will focus on the characteristics of response and resistance to ICIs in CRC, and discuss promising biomarkers studied in recent clinical trials combining ICIs with other therapies.
Collapse
Affiliation(s)
- Nicolas Huyghe
- Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, 1200 Brussels, Belgium; (N.H.); (E.B.); (P.S.)
| | - Elena Benidovskaya
- Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, 1200 Brussels, Belgium; (N.H.); (E.B.); (P.S.)
| | - Philippe Stevens
- Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, 1200 Brussels, Belgium; (N.H.); (E.B.); (P.S.)
| | - Marc Van den Eynde
- Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, 1200 Brussels, Belgium; (N.H.); (E.B.); (P.S.)
- Institut Roi Albert II, Department of Medical Oncology and Gastroenterology, Cliniques Universitaires St-Luc, 1200 Brussels, Belgium
| |
Collapse
|
257
|
Phosphoinositide 3-Kinases as Potential Targets for Thrombosis Prevention. Int J Mol Sci 2022; 23:ijms23094840. [PMID: 35563228 PMCID: PMC9105564 DOI: 10.3390/ijms23094840] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 02/01/2023] Open
Abstract
As integral parts of pathological arterial thrombi, platelets are the targets of pharmacological regimens designed to treat and prevent thrombosis. A detailed understanding of platelet biology and function is thus key to design treatments that prevent thrombotic cardiovascular disease without significant disruption of the haemostatic balance. Phosphoinositide 3-kinases (PI3Ks) are a group of lipid kinases critical to various aspects of platelet biology. There are eight PI3K isoforms, grouped into three classes. Our understanding of PI3K biology has recently progressed with the targeting of specific isoforms emerging as an attractive therapeutic strategy in various human diseases, including for thrombosis. This review will focus on the role of PI3K subtypes in platelet function and subsequent thrombus formation. Understanding the mechanisms by which platelet function is regulated by the various PI3Ks edges us closer toward targeting specific PI3K isoforms for anti-thrombotic therapy.
Collapse
|
258
|
Skolariki A, D’Costa J, Little M, Lord S. Role of PI3K/Akt/mTOR pathway in mediating endocrine resistance: concept to clinic. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:172-199. [PMID: 36046843 PMCID: PMC9400772 DOI: 10.37349/etat.2022.00078] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/11/2022] [Indexed: 01/06/2023] Open
Abstract
The majority of breast cancers express the estrogen receptor (ER) and for this group of patients, endocrine therapy is the cornerstone of systemic treatment. However, drug resistance is common and a focus for breast cancer preclinical and clinical research. Over the past 2 decades, the PI3K/Akt/mTOR axis has emerged as an important driver of treatment failure, and inhibitors of mTOR and PI3K are now licensed for the treatment of women with advanced ER-positive breast cancer who have relapsed on first-line hormonal therapy. This review presents the preclinical and clinical data that led to this new treatment paradigm and discusses future directions.
Collapse
Affiliation(s)
- Aglaia Skolariki
- Department of Oncology, University of Oxford, Churchill Hospital, OX3 7LE Oxford, UK
| | - Jamie D’Costa
- Department of Oncology, University of Oxford, Churchill Hospital, OX3 7LE Oxford, UK
| | - Martin Little
- Department of Oncology, Churchill Hospital, OX3 7LE Oxford, UK
| | - Simon Lord
- Department of Oncology, University of Oxford, Churchill Hospital, OX3 7LE Oxford, UK
| |
Collapse
|
259
|
Nuclear translocation of p85β promotes tumorigenesis of PIK3CA helical domain mutant cancer. Nat Commun 2022; 13:1974. [PMID: 35418124 PMCID: PMC9007954 DOI: 10.1038/s41467-022-29585-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 03/14/2022] [Indexed: 12/11/2022] Open
Abstract
PI3Ks consist of p110 catalytic subunits and p85 regulatory subunits. PIK3CA, encoding p110α, is frequently mutated in human cancers. Most PIK3CA mutations are clustered in the helical domain or the kinase domain. Here, we report that p85β disassociates from p110α helical domain mutant protein and translocates into the nucleus through a nuclear localization sequence (NLS). Nuclear p85β recruits deubiquitinase USP7 to stabilize EZH1 and EZH2 and enhances H3K27 trimethylation. Knockout of p85β or p85β NLS mutant reduces the growth of tumors harboring a PIK3CA helical domain mutation. Our studies illuminate a novel mechanism by which PIK3CA helical domain mutations exert their oncogenic function. Finally, a combination of Alpelisib, a p110α-specific inhibitor, and an EZH inhibitor, Tazemetostat, induces regression of xenograft tumors harboring a PIK3CA helical domain mutation, but not tumors with either a WT PIK3CA or a PIK3CA kinase domain mutation, suggesting that the drug combination could be an effective therapeutic approach for PIK3CA helical domain mutant tumors. The mechanisms behind the oncogenic role of the PIK3CA helical domain mutant is poorly understood. Here, the authors show that its oncogenic function depends on the release of p85β from mutated p110α, its translocation to the nucleus and the consequent increased activity of EZH proteins.
Collapse
|
260
|
Jenkins ML, Ranga-prasad H, Parson MA, Harris NJ, Rathinaswamy MK, Burke JE. Oncogenic mutations of PIK3CAlead to increased membrane recruitment driven by reorientation of the ABD, p85 and C-terminus.. [DOI: 10.1101/2022.04.05.487205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
AbstractPIK3CAencoding the phosphoinositide 3-kinase (PI3K) p110α catalytic subunit is frequently mutated in cancer, with mutations occurring widely throughout the primary sequence. The full set of mechanisms underlying how PI3Ks are activated by all oncogenic mutations on membranes are unclear. Using a synergy of biochemical assays and hydrogen deuterium exchange mass spectrometry (HDX-MS), we reveal unique regulatory mechanisms underlying PI3K activation. Engagement of p110α on membranes leads to disengagement of the ABD of p110α from the catalytic core, and the C2 domain from the iSH2 domain of the p85 regulatory subunit. PI3K activation also requires reorientation of the p110α C-terminus, with mutations that alter the inhibited conformation of the C-terminus increasing membrane binding. Mutations at the C-terminus (M1043I/L, H1047R, G1049R, and N1068KLKR) activate p110α through distinct mechanisms, with this having important implications for mutant selective inhibitor development. This work reveals unique mechanisms underlying how PI3K is activated by oncogenic mutations, and explains how double mutants can synergistically increase PI3K activity.
Collapse
|
261
|
Alharbey R, Kim JI, Daud A, Song M, Alshdadi AA, Hayat MK. Indexing important drugs from medical literature. Scientometrics 2022. [DOI: 10.1007/s11192-022-04340-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
262
|
Chen X, Tan F, Zhang H, Zhang X, Xu F, Yuan J, Sun C, Huang L, Guan H, Luo C, Huang RP, Yang Z. Serum Cytokine Profiles in Phlegm-dampness Constitution and Damp-heat Constitution Using Proteomic Antibody Microarray. Eur J Integr Med 2022. [DOI: 10.1016/j.eujim.2022.102126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
263
|
Bamberger C, Diedrich J, Martìnez-Bartholomé S, Yates JR. Cancer Conformational Landscape Shapes Tumorigenesis. J Proteome Res 2022; 21:1017-1028. [PMID: 35271278 PMCID: PMC9653087 DOI: 10.1021/acs.jproteome.1c00906] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
During tumorigenesis, DNA mutations in protein coding sequences can alter amino acid sequences which can change the structures of proteins. While the 3D structure of mutated proteins has been studied with atomic resolution, the precise impact of somatic mutations on the 3D proteome during malignant transformation remains unknown because methods to reveal in vivo protein structures in high throughput are limited. Here, we measured the accessibility of the lysine ε-amine for chemical modification across proteomes using covalent protein painting (CPP) to indirectly determine alterations in the 3D proteome. CPP is a novel, high-throughput quantitative mass spectrometric method that surveyed a total of 8052 lysine sites across the 60 cell lines of the well-studied anticancer cell line panel (NCI60). Overall, 5.2 structural alterations differentiated any cancer cell line from the other 59. Structural aberrations in 98 effector proteins correlated with the selected presence of 90 commonly mutated proteins in the NCI60 cell line panel, suggesting that different tumor genotypes reshape a limited set of effector proteins. We searched our dataset for druggable conformational aberrations and identified 49 changes in the cancer conformational landscape that correlated with the growth inhibition profiles of 300 drug candidates out of 50,000 small molecules. We found that alterations in heat shock proteins are key predictors of anticancer drug efficacy, which implies that the proteostasis network may have a general but hitherto unrecognized role in maintaining malignancy. Individual lysine sites may serve as biomarkers to guide drug selection or may be directly targeted for anticancer drug development.
Collapse
Affiliation(s)
- Casimir Bamberger
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Jolene Diedrich
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Salvador Martìnez-Bartholomé
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - John R Yates
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
264
|
Jiao S, Zhang X, Wang D, Fu H, Xia Q. Genetic Alteration and Their Significance on Clinical Events in Small Cell Lung Cancer. Cancer Manag Res 2022; 14:1493-1505. [PMID: 35469134 PMCID: PMC9034895 DOI: 10.2147/cmar.s356037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/07/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Small cell lung cancer (SCLC), an aggressive subtype of lung cancer characterized by the development of neuroendocrine tumors, is prone to distant metastasis, resistant to platinum-based drugs and has a poor prognosis. The development of next-generation sequencing technology (NGS) has led to the identification of many genetic alterations in SCLC. Few druggable targeted molecules can be used in clinical practice. Currently, NGS is widely employed in routine clinical practice of non-small cell lung cancer to assist in therapeutic options and prognosis evaluation. This study aims to investigate genes involved in small cell lung cancer (SCLC), their occurrence and their significance in clinical events. Methods Tumor tissue specimens from 18 Chinese SCLC patients were collected through a 520 cancer‐related genes panel for next-generation sequencing. First, the association between sequence results and clinical outcomes was examined. Subsequently, data on clinical pathology and sequencing results were analyzed. Results The Kaplan–Meier curve displayed a significant reduction in PFS for SCLC patients with LRP1B or MAP3K13 mutations. Overall survival (OS) of SCLC patients with MSH6 mutation was significantly higher than those with SPEN mutation. Conclusion Next-generation sequencing demonstrates that the genetic landscape of SCLC. Mutation status of LRP1B, MAP3K13, MSH6 and SPEN has prognostic significance, which might be potential therapeutic targets. We found possible genes and related signaling pathways that affect metastasis. These results can improve our understanding of the mutation characteristics of SCLC and identify potential biomarkers to guide targeted therapies.
Collapse
Affiliation(s)
- Shuyue Jiao
- Department of Oncology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Xin Zhang
- Department of Pathology, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, People’s Republic of China
| | - Dapeng Wang
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University; Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou, People’s Republic of China
| | - Hongyong Fu
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University; Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou, People’s Republic of China
| | - Qingxin Xia
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University; Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou, People’s Republic of China
- Correspondence: Qingxin Xia; Hongyong Fu, Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University; Henan Medical Key Laboratory of Tumor Pathology and Artificial Intelligence Diagnosis, Zhengzhou Key Laboratory of Accurate Pathological Diagnosis of Intractable Tumors, Zhengzhou, 450000, People’s Republic of China, Email ;
| |
Collapse
|
265
|
Abstract
Colorectal cancer is the second leading cause of cancer-associated mortality, with a lifetime risk of approximately 4% to 5%. Colorectal cancer develops from the sequential acquisition of defined genetic mutations in the colonic epithelium. Tumorigenesis from normal tissue to cancer occurs largely through 3 pathways: the chromosomal instability pathway, the microsatellite instability pathway, and the sessile serrated pathway. Colorectal cancer incidence and mortality have decreased by approximately 35% since the beginning of screening programs in the 1990s, although other factors such as use of aspirin for coronary disease prevention and decreased smoking rates may also be important. In this review, we discuss the etiology, epidemiology, and histology of colorectal polyps and cancer.
Collapse
|
266
|
Borsari C, Keles E, McPhail JA, Schaefer A, Sriramaratnam R, Goch W, Schaefer T, De Pascale M, Bal W, Gstaiger M, Burke JE, Wymann MP. Covalent Proximity Scanning of a Distal Cysteine to Target PI3Kα. J Am Chem Soc 2022; 144:6326-6342. [PMID: 35353516 PMCID: PMC9011356 DOI: 10.1021/jacs.1c13568] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
![]()
Covalent protein
kinase inhibitors exploit currently noncatalytic
cysteines in the adenosine 5′-triphosphate (ATP)-binding site
via electrophiles directly appended to a reversible-inhibitor scaffold.
Here, we delineate a path to target solvent-exposed cysteines at a
distance >10 Å from an ATP-site-directed core module and produce
potent covalent phosphoinositide 3-kinase α (PI3Kα) inhibitors.
First, reactive warheads are used to reach out to Cys862 on PI3Kα,
and second, enones are replaced with druglike warheads while linkers
are optimized. The systematic investigation of intrinsic warhead reactivity
(kchem), rate of covalent bond formation
and proximity (kinact and reaction space
volume Vr), and integration of structure
data, kinetic and structural modeling, led to the guided identification
of high-quality, covalent chemical probes. A novel stochastic approach
provided direct access to the calculation of overall reaction rates
as a function of kchem, kinact, Ki, and Vr, which was validated with compounds with varied linker
lengths. X-ray crystallography, protein mass spectrometry (MS), and
NanoBRET assays confirmed covalent bond formation of the acrylamide
warhead and Cys862. In rat liver microsomes, compounds 19 and 22 outperformed the rapidly metabolized CNX-1351,
the only known PI3Kα irreversible inhibitor. Washout experiments
in cancer cell lines with mutated, constitutively activated PI3Kα
showed a long-lasting inhibition of PI3Kα. In SKOV3 cells, compounds 19 and 22 revealed PI3Kβ-dependent signaling,
which was sensitive to TGX221. Compounds 19 and 22 thus qualify as specific chemical probes to explore PI3Kα-selective
signaling branches. The proposed approach is generally suited to develop
covalent tools targeting distal, unexplored Cys residues in biologically
active enzymes.
Collapse
Affiliation(s)
- Chiara Borsari
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Erhan Keles
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Jacob A McPhail
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada
| | - Alexander Schaefer
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Otto-Stern-Weg 3, 8093 Zürich, Switzerland
| | - Rohitha Sriramaratnam
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Wojciech Goch
- Department of Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Thorsten Schaefer
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Martina De Pascale
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| | - Wojciech Bal
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | - Matthias Gstaiger
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Otto-Stern-Weg 3, 8093 Zürich, Switzerland
| | - John E Burke
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8W 2Y2, Canada
| | - Matthias P Wymann
- Department of Biomedicine, University of Basel, Mattenstrasse 28, 4058 Basel, Switzerland
| |
Collapse
|
267
|
Small-Molecule RAS Inhibitors as Anticancer Agents: Discovery, Development, and Mechanistic Studies. Int J Mol Sci 2022; 23:ijms23073706. [PMID: 35409064 PMCID: PMC8999084 DOI: 10.3390/ijms23073706] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/21/2022] [Accepted: 03/24/2022] [Indexed: 12/11/2022] Open
Abstract
Mutations of RAS oncogenes are responsible for about 30% of all human cancer types, including pancreatic, lung, and colorectal cancers. While KRAS1 is a pseudogene, mutation of KRAS2 (commonly known as KRAS oncogene) is directly or indirectly associated with human cancers. Among the RAS family, KRAS is the most abundant oncogene related to uncontrolled cellular proliferation to generate solid tumors in many types of cancer such as pancreatic carcinoma (over 80%), colon carcinoma (40-50%), lung carcinoma (30-50%), and other types of cancer. Once described as 'undruggable', RAS proteins have become 'druggable', at least to a certain extent, due to the continuous efforts made during the past four decades. In this account, we discuss the chemistry and biology (wherever available) of the small-molecule inhibitors (synthetic, semi-synthetic, and natural) of KRAS proteins that were published in the past decades. Commercial drugs, as well as investigational molecules from preliminary stages to clinical trials, are categorized and discussed in this study. In summary, this study presents an in-depth discussion of RAS proteins, classifies the RAS superfamily, and describes the molecular mechanism of small-molecule RAS inhibitors.
Collapse
|
268
|
Deng S, Leong HC, Datta A, Gopal V, Kumar AP, Yap CT. PI3K/AKT Signaling Tips the Balance of Cytoskeletal Forces for Cancer Progression. Cancers (Basel) 2022; 14:1652. [PMID: 35406424 PMCID: PMC8997157 DOI: 10.3390/cancers14071652] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/13/2022] [Accepted: 03/21/2022] [Indexed: 02/01/2023] Open
Abstract
The PI3K/AKT signaling pathway plays essential roles in multiple cellular processes, which include cell growth, survival, metabolism, and motility. In response to internal and external stimuli, the PI3K/AKT signaling pathway co-opts other signaling pathways, cellular components, and cytoskeletal proteins to reshape individual cells. The cytoskeletal network comprises three main components, which are namely the microfilaments, microtubules, and intermediate filaments. Collectively, they are essential for many fundamental structures and cellular processes. In cancer, aberrant activation of the PI3K/AKT signaling cascade and alteration of cytoskeletal structures have been observed to be highly prevalent, and eventually contribute to many cancer hallmarks. Due to their critical roles in tumor progression, pharmacological agents targeting PI3K/AKT, along with cytoskeletal components, have been developed for better intervention strategies against cancer. In our review, we first discuss existing evidence in-depth and then build on recent advances to propose new directions for therapeutic intervention.
Collapse
Affiliation(s)
- Shuo Deng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (S.D.); (V.G.)
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
| | - Hin Chong Leong
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
- Departments of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Arpita Datta
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
| | - Vennila Gopal
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (S.D.); (V.G.)
| | - Alan Prem Kumar
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
- Departments of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| | - Celestial T. Yap
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (S.D.); (V.G.)
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| |
Collapse
|
269
|
Youssef ASED, Abdel-Fattah MA, Lotfy MM, Nassar A, Abouelhoda M, Touny AO, Hassan ZK, Mohey Eldin M, Bahnassy AA, Khaled H, Zekri ARN. Multigene Panel Sequencing Reveals Cancer-Specific and Common Somatic Mutations in Colorectal Cancer Patients: An Egyptian Experience. Curr Issues Mol Biol 2022; 44:1332-1352. [PMID: 35723313 PMCID: PMC8947625 DOI: 10.3390/cimb44030090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 12/11/2022] Open
Abstract
This study aims at identifying common pathogenic somatic mutations at different stages of colorectal carcinogenesis in Egyptian patients. Our cohort included colonoscopic biopsies collected from 120 patients: 20 biopsies from patients with inflammatory bowel disease, 38 from colonic polyp patients, and 62 from patients with colorectal cancer. On top of this, the cohort included 20 biopsies from patients with non-specific mild to moderated colitis. Targeted DNA sequencing using a customized gene panel of 96 colorectal related genes running on the Ion Torrent NGS technology was used to process the samples. Our results revealed that 69% of all cases harbored at least one somatic mutation. Fifty-seven genes were found to carry 232 somatic non-synonymous variants. The most frequently pathogenic somatic mutations were localized in TP53, APC, KRAS, and PIK3CA. In total, 16 somatic mutations were detected in the CRC group and in either the IBD or CP group. In addition, our data showed that 51% of total somatic variants were CRC-specific variants. The average number of CRC-specific variants per sample is 2.4. The top genes carrying CRC-specific mutations are APC, TP53, PIK3CA, FBXW7, ATM, and SMAD4. It seems obvious that TP53 and APC genes were the most affected genes with somatic mutations in all groups. Of interest, 85% and 28% of the APC and TP53 deleterious somatic mutations were located in Exon 14 and Exon 3, respectively. Besides, 37% and 28% of the total somatic mutations identified in APC and TP53 were CRC-specific variants, respectively. Moreover, we identified that, in 29 somatic mutations in 21 genes, their association with CRC patients was unprecedented. Ten detected variants were likely to be novel: six in PIK3CA and four variants in FBXW7. The detected P53, Wnt/βcatenin, Angiogenesis, EGFR, TGF-β and Interleukin signaling pathways were the most altered pathways in 22%, 16%, 12%, 10%, 9% and 9% of the CRC patients, respectively. These results would contribute to a better understanding of the colorectal cancer and in introducing personalized therapies for Egyptian CRC patients.
Collapse
Affiliation(s)
- Amira Salah El-Din Youssef
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt; (M.M.L.); (A.N.); (Z.K.H.)
| | | | - Mai M. Lotfy
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt; (M.M.L.); (A.N.); (Z.K.H.)
| | - Auhood Nassar
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt; (M.M.L.); (A.N.); (Z.K.H.)
| | | | - Ahmed O. Touny
- Surgical Oncology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt;
| | - Zeinab K. Hassan
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt; (M.M.L.); (A.N.); (Z.K.H.)
| | - Mohammed Mohey Eldin
- Tropical Medicine Department, El Kasr Al-Aini, Cairo University, Cairo 11562, Egypt;
| | - Abeer A. Bahnassy
- Molecular Pathology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt;
| | - Hussein Khaled
- Medical Oncology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt;
| | - Abdel Rahman N. Zekri
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo 11796, Egypt; (M.M.L.); (A.N.); (Z.K.H.)
| |
Collapse
|
270
|
Singhal S, Maheshwari P, Krishnamurthy PT, Patil VM. Drug Repurposing Strategies for Non-Cancer to Cancer Therapeutics. Anticancer Agents Med Chem 2022; 22:2726-2756. [PMID: 35301945 DOI: 10.2174/1871520622666220317140557] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/15/2021] [Accepted: 11/27/2021] [Indexed: 11/22/2022]
Abstract
Global efforts invested for the prevention and treatment of cancer need to be repositioned to develop safe, effective, and economic anticancer therapeutics by adopting rational approaches of drug discovery. Drug repurposing is one of the established approaches to reposition old, clinically approved off patent noncancer drugs with known targets into newer indications. The literature review suggests key role of drug repurposing in the development of drugs intended for cancer as well as noncancer therapeutics. A wide category of noncancer drugs namely, drugs acting on CNS, anthelmintics, cardiovascular drugs, antimalarial drugs, anti-inflammatory drugs have come out with interesting outcomes during preclinical and clinical phases. In the present article a comprehensive overview of the current scenario of drug repurposing for the treatment of cancer has been focused. The details of some successful studies along with examples have been included followed by associated challenges.
Collapse
Affiliation(s)
- Shipra Singhal
- Department of Pharmaceutical Chemistry KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, India
| | - Priyal Maheshwari
- Department of Pharmaceutical Chemistry KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, India
| | | | - Vaishali M Patil
- Department of Pharmaceutical Chemistry KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, India
| |
Collapse
|
271
|
Wang T, Long K, Zhou Y, Jiang X, Liu J, Fong JH, Wong AS, Ng WL, Wang W. Optochemical Control of mTOR Signaling and mTOR-Dependent Autophagy. ACS Pharmacol Transl Sci 2022; 5:149-155. [PMID: 35311017 PMCID: PMC8922298 DOI: 10.1021/acsptsci.1c00230] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Indexed: 11/29/2022]
Abstract
As an important regulator of cell metabolism, proliferation, and survival, mTOR (mammalian target of rapamycin) signaling provides both a potential target for cancer treatment and a research tool for investigation of cell metabolism. One inhibitor for both mTORC1 and mTORC2 pathways, OSI-027, exhibited robust anticancer efficacy but induced side effects. Herein, we designed a photoactivatable OSI-027 prodrug, which allowed the release of OSI-027 after light irradiation to inhibit the mTOR signaling pathway, triggering autophagy and leading to cell death. This photoactivatable prodrug can provide novel strategies for mTOR-targeting cancer therapy and act as a new tool for investigating mTOR signaling and its related biological processes.
Collapse
Affiliation(s)
- Tianyi Wang
- State
Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department
of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Dr.
Li Dak-Sum Research Centre, The University
of Hong Kong, Hong Kong 0000, China
| | - Kaiqi Long
- State
Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department
of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Dr.
Li Dak-Sum Research Centre, The University
of Hong Kong, Hong Kong 0000, China
| | - Yang Zhou
- State
Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department
of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Dr.
Li Dak-Sum Research Centre, The University
of Hong Kong, Hong Kong 0000, China
| | - Xiaoding Jiang
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Hong Kong, China
| | - Jinzhao Liu
- Dr.
Li Dak-Sum Research Centre, The University
of Hong Kong, Hong Kong 0000, China
| | - John H.C. Fong
- Laboratory
of Combinatorial Genetics and Synthetic Biology, School of Biomedical
Sciences, The University of Hong Kong, Hong Kong China
| | - Alan S.L. Wong
- Laboratory
of Combinatorial Genetics and Synthetic Biology, School of Biomedical
Sciences, The University of Hong Kong, Hong Kong China
- Department
of Electrical and Electronic Engineering, The University of Hong Kong, Hong
Kong, China
| | - Wai-Lung Ng
- School
of Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Hong Kong, China
| | - Weiping Wang
- State
Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department
of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Dr.
Li Dak-Sum Research Centre, The University
of Hong Kong, Hong Kong 0000, China
| |
Collapse
|
272
|
Collins NB, Al Abosy R, Miller BC, Bi K, Zhao Q, Quigley M, Ishizuka JJ, Yates KB, Pope HW, Manguso RT, Shrestha Y, Wadsworth M, Hughes T, Shalek AK, Boehm JS, Hahn WC, Doench JG, Haining WN. PI3K activation allows immune evasion by promoting an inhibitory myeloid tumor microenvironment. J Immunother Cancer 2022; 10:jitc-2021-003402. [PMID: 35264433 PMCID: PMC8915320 DOI: 10.1136/jitc-2021-003402] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Oncogenes act in a cell-intrinsic way to promote tumorigenesis. Whether oncogenes also have a cell-extrinsic effect on suppressing the immune response to cancer is less well understood. METHODS We use an in vivo expression screen of known cancer-associated somatic mutations in mouse syngeneic tumor models treated with checkpoint blockade to identify oncogenes that promote immune evasion. We then validated candidates from this screen in vivo and analyzed the tumor immune microenvironment of tumors expressing mutant protein to identify mechanisms of immune evasion. RESULTS We found that expression of a catalytically active mutation in phospho-inositol 3 kinase (PI3K), PIK3CA c.3140A>G (H1047R) confers a selective growth advantage to tumors treated with immunotherapy that is reversed by pharmacological PI3K inhibition. PIK3CA H1047R-expression in tumors decreased the number of CD8+ T cells but increased the number of inhibitory myeloid cells following immunotherapy. Inhibition of myeloid infiltration by pharmacological or genetic modulation of Ccl2 in PIK3CA H1047R tumors restored sensitivity to programmed cell death protein 1 (PD-1) checkpoint blockade. CONCLUSIONS PI3K activation enables tumor immune evasion by promoting an inhibitory myeloid microenvironment. Activating mutations in PI3K may be useful as a biomarker of poor response to immunotherapy. Our data suggest that some oncogenes promote tumorigenesis by enabling tumor cells to avoid clearance by the immune system. Identification of those mechanisms can advance rational combination strategies to increase the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Natalie B Collins
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA,Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Rose Al Abosy
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Brian C Miller
- Lineberger Comprehensive Cancer Center, Department of Medicine, Division of Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kevin Bi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Qihong Zhao
- Oncology Discovery Biology, Bristol Myers Squibb, Lawrenceville, New Jersey, USA
| | - Michael Quigley
- Research Biology, Gilead Sciences Inc, Foster City, California, USA
| | - Jeffrey J Ishizuka
- Department of Internal Medicine (Oncology), Yale Cancer Center and Yale School of Medicine, New Haven, New Jersey, USA
| | - Kathleen B Yates
- Broad Institute, Cambridge, Massachusetts, USA,Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Hans W Pope
- Arsenal Biosciences, San Francisco, California, USA
| | - Robert T Manguso
- Broad Institute, Cambridge, Massachusetts, USA,Center for Cancer Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
| | | | - Marc Wadsworth
- Broad Institute, Cambridge, Massachusetts, USA,Institute for Medical Engineering & Science (IMES), Department of Chemistry and Koch Institute for Integrative Cancer Research, Ragon Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA,Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, USA
| | - Travis Hughes
- Broad Institute, Cambridge, Massachusetts, USA,Institute for Medical Engineering & Science (IMES), Department of Chemistry and Koch Institute for Integrative Cancer Research, Ragon Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA,Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, USA
| | - Alex K Shalek
- Broad Institute, Cambridge, Massachusetts, USA,Institute for Medical Engineering & Science (IMES), Department of Chemistry and Koch Institute for Integrative Cancer Research, Ragon Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA,Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts, USA
| | | | - William C Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA,Broad Institute, Cambridge, Massachusetts, USA,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
273
|
PIK3CA Gene Mutations in HNSCC: Systematic Review and Correlations with HPV Status and Patient Survival. Cancers (Basel) 2022; 14:cancers14051286. [PMID: 35267596 PMCID: PMC8909011 DOI: 10.3390/cancers14051286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 12/24/2022] Open
Abstract
PIK3CA mutations are believed to contribute to the pathogenesis of human papillomavirus (HPV)-associated head and neck squamous cell carcinomas (HNSCC). This study aims to establish the frequency of PIK3CA mutations in a Portuguese HNSCC cohort and to determine their association with the HPV status and patient survival. A meta-analysis of scientific literature also revealed widely different mutation rates in cohorts from different world regions and a trend towards improved prognosis among patients with PIK3CA mutations. DNA samples were available from 95 patients diagnosed with HNSCC at the Portuguese Institute of Oncology in Lisbon between 2010 and 2019. HPV status was established based on viral DNA detected using real-time PCR. The evaluation of PIK3CA gene mutations was performed by real-time PCR for four mutations (H1047L; E542K, E545K, and E545D). Thirty-seven cases were found to harbour PIK3CA mutations (39%), with the E545D mutation (73%) more frequently detected. There were no significant associations between the mutational status and HPV status (74% WT and 68% MUT were HPV (+); p = 0.489) or overall survival (OS) (3-year OS: WT 54% and MUT 65%; p = 0.090). HPV status was the only factor significantly associated with both OS and disease-free survival (DFS), with HPV (+) patients having consistently better outcomes (3-year OS: HPV (+) 65% and HPV (-) 36%; p = 0.007; DFS HPV (+) 83% and HPV (-) 43%; p = 0.001). There was a statistically significant interaction effect between HPV status and PIK3CA mutation regarding DFS (Interaction test: p = 0.026). In HPV (+) patients, PIK3CA wild-type is associated with a significant 4.64 times increase in the hazard of recurrence or death (HR = 4.64; 95% CI 1.02-20.99; p = 0.047). Overall, PIK3CA gene mutations are present in a large number of patients and may help define patient subsets who can benefit from therapies targeting the PI3K pathway. The systematic assessment of PIK3CA gene mutations in HNSCC patients will require further methodological standardisation.
Collapse
|
274
|
Nussinov R, Zhang M, Maloney R, Tsai C, Yavuz BR, Tuncbag N, Jang H. Mechanism of activation and the rewired network: New drug design concepts. Med Res Rev 2022; 42:770-799. [PMID: 34693559 PMCID: PMC8837674 DOI: 10.1002/med.21863] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/06/2021] [Accepted: 10/07/2021] [Indexed: 12/13/2022]
Abstract
Precision oncology benefits from effective early phase drug discovery decisions. Recently, drugging inactive protein conformations has shown impressive successes, raising the cardinal questions of which targets can profit and what are the principles of the active/inactive protein pharmacology. Cancer driver mutations have been established to mimic the protein activation mechanism. We suggest that the decision whether to target an inactive (or active) conformation should largely rest on the protein mechanism of activation. We next discuss the recent identification of double (multiple) same-allele driver mutations and their impact on cell proliferation and suggest that like single driver mutations, double drivers also mimic the mechanism of activation. We further suggest that the structural perturbations of double (multiple) in cis mutations may reveal new surfaces/pockets for drug design. Finally, we underscore the preeminent role of the cellular network which is deregulated in cancer. Our structure-based review and outlook updates the traditional Mechanism of Action, informs decisions, and calls attention to the intrinsic activation mechanism of the target protein and the rewired tumor-specific network, ushering innovative considerations in precision medicine.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer ImmunometabolismNational Cancer InstituteFrederickMarylandUSA
- Department of Human Molecular Genetics and Biochemistry, Sackler School of MedicineTel Aviv UniversityTel AvivIsrael
| | - Mingzhen Zhang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer ImmunometabolismNational Cancer InstituteFrederickMarylandUSA
| | - Ryan Maloney
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer ImmunometabolismNational Cancer InstituteFrederickMarylandUSA
| | - Chung‐Jung Tsai
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer ImmunometabolismNational Cancer InstituteFrederickMarylandUSA
| | - Bengi Ruken Yavuz
- Department of Health Informatics, Graduate School of InformaticsMiddle East Technical UniversityAnkaraTurkey
| | - Nurcan Tuncbag
- Department of Health Informatics, Graduate School of InformaticsMiddle East Technical UniversityAnkaraTurkey
- Department of Chemical and Biological Engineering, College of EngineeringKoc UniversityIstanbulTurkey
- Koc University Research Center for Translational Medicine, School of MedicineKoc UniversityIstanbulTurkey
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Laboratory of Cancer ImmunometabolismNational Cancer InstituteFrederickMarylandUSA
| |
Collapse
|
275
|
The potential of PIK3CA, KRAS, BRAF, and APC hotspot mutations as a non-invasive detection method for colorectal cancer. Mol Cell Probes 2022; 63:101807. [DOI: 10.1016/j.mcp.2022.101807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 03/09/2022] [Accepted: 03/09/2022] [Indexed: 02/07/2023]
|
276
|
Michmerhuizen NL, Heenan C, Wang J, Leonard E, Bellile E, Loganathan SK, Wong SY, Lei YL, Brenner JC. Combined Pik3ca-H1047R and loss-of-function Notch1 alleles decrease survival time in a 4-nitroquinoline N-oxide-driven head and neck squamous cell carcinoma model. Oral Oncol 2022; 126:105770. [DOI: 10.1016/j.oraloncology.2022.105770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 11/27/2022]
|
277
|
Dechaphunkul A, Thongwatchara P, Thongsuksai P, Dechaphunkul T, Geater SL. Frequency of PIK3CA mutations in different subsites of head and neck squamous cell carcinoma in southern Thailand. J Pathol Transl Med 2022; 56:126-133. [PMID: 35209701 PMCID: PMC9119806 DOI: 10.4132/jptm.2022.01.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/04/2022] [Indexed: 12/02/2022] Open
Abstract
Background Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) mutations have been reported in many cancers, including head and neck squamous cell carcinoma (HNSCC). The frequency of these mutations varies among tumor locations and might be relevant to treatment outcomes among HNSCC. In this study, we examined the frequency of PIK3CA mutations in the different subsites of HNSCC. Methods Ninety-six fresh biopsy specimens were investigated for mutations in PIK3CA exons 4, 9, and 20 using allele-specific real-time polymerase chain reaction. Patient characteristics and survival were analyzed and compared between specimens with or without PIK3CA mutations. Results The study included primary tumors originating from the oral cavity (n = 63), hypopharynx (n = 23), and oropharynx (n = 10). We identified mutations in 10.4% of patients (10 of 96 specimens). The overall mutational frequency was 17.4% (4/23) and 9.5% (6/63) in the hypopharynx and oral cavity, respectively. No patients with oropharyngeal carcinoma had mutations. Among the 10 mutant specimens, five were missense mutations (exon 9 [E545K] in two samples and exon 20 [H1047R] in three samples) and five were silent mutations in exon 20 (T1025T). Mutations were not found in exon 4. Among 84 patients with available clinical data, we found no significant differences in clinical characteristics and survival based on the presence or absence of PIK-3CA mutations. Conclusions The results indicate that PIK3CA mutations are involved in HNSCC carcinogenesis, and the hypopharynx should be considered a primary site of interest for future studies, particularly in Southeast Asian populations.
Collapse
Affiliation(s)
- Arunee Dechaphunkul
- Holistic Center for Cancer Study and Care (HOCC-PSU), Medical Oncology Unit, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Phatcharaporn Thongwatchara
- Holistic Center for Cancer Study and Care (HOCC-PSU), Medical Oncology Unit, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Paramee Thongsuksai
- Department of Pathology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Tanadech Dechaphunkul
- Department of Otorhinolaryngology Head and Neck Surgery, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Sarayut Lucien Geater
- Division of Respiratory and Respiratory Critical Care Medicine, Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| |
Collapse
|
278
|
Pinto N, Ruicci KM, Khan MI, Shaikh MH, Zeng YFP, Yoo J, Fung K, MacNeil SD, Mendez A, Mymryk JS, Barrett JW, Boutros PC, Nichols AC. Introduction and expression of PIK3CAE545K in a papillary thyroid cancer BRAFV600E cell line leads to a dedifferentiated aggressive phenotype. JOURNAL OF OTOLARYNGOLOGY - HEAD & NECK SURGERY 2022; 51:7. [PMID: 35193694 PMCID: PMC8862267 DOI: 10.1186/s40463-022-00558-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/02/2022] [Indexed: 01/03/2023]
Abstract
Anaplastic thyroid cancer (ATC) is a rare, aggressive form of undifferentiated thyroid cancer, which exhibits rapid progression and is almost universally fatal. At least a subset of ATC is thought to arise from pre-existing well-differentiated thyroid cancer, most frequently papillary thyroid cancer (PTC). While PIK3CA mutations are rare in PTC, they are common in ATC and tend to co-occur with BRAF mutations. This provided the rationale for our study to identify the potential role of PIK3CA mutations in the progression from well-differentiated to undifferentiated thyroid cancer. We introduced PIK3CAE545K into the LAM1 PTC cell line, which carries a BRAFV600E mutation. In culture, the engineered cell line (LAM1:PIK3CAE545K) proliferated faster and demonstrated increased clonogenic potential relative to the parental line carrying an empty vector (LAM1EV). Both the LAM1EV and LAM1:PIK3CAE545K edited lines were implanted into hind flanks of athymic nude mice for in vivo determination of disease progression. While tumour weights and volumes were not significantly higher in LAM1:PIK3CAE545K mice, there was a decrease in expression of thyroid differentiation markers TTF-1, thyroglobulin, PAX8 and B-catenin, suggesting that introduction of PIK3CAE545K led to dedifferentiation in vivo. Collectively, this study provides evidence of a role for PIK3CAE545K in driving disease progression from a well-differentiated to an undifferentiated thyroid cancer; however, over-expression was not a determinant of an accelerated growth phenotype in ATC.
Collapse
|
279
|
Immunotherapy for Colorectal Cancer: Mechanisms and Predictive Biomarkers. Cancers (Basel) 2022; 14:cancers14041028. [PMID: 35205776 PMCID: PMC8869923 DOI: 10.3390/cancers14041028] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Late-stage colorectal cancer treatment often involves chemotherapy and radiation that can cause dose-limiting toxicity, and therefore there is great interest in developing targeted therapies for this disease. Immunotherapy is a targeted therapy that uses peptides, cells, antibodies, viruses, or small molecules to engage or train the immune system to kill cancer. Here, we discuss the preclinical and clinical development of immunotherapy for treatment of colorectal cancer and provide an overview of predictive biomarkers for such treatments. We also consider open questions including optimal combination treatments and sensitization of colorectal cancer patients with proficient mismatch repair enzymes. Abstract Though early-stage colorectal cancer has a high 5 year survival rate of 65–92% depending on the specific stage, this probability drops to 13% after the cancer metastasizes. Frontline treatments for colorectal cancer such as chemotherapy and radiation often produce dose-limiting toxicities in patients and acquired resistance in cancer cells. Additional targeted treatments are needed to improve patient outcomes and quality of life. Immunotherapy involves treatment with peptides, cells, antibodies, viruses, or small molecules to engage or train the immune system to kill cancer cells. Preclinical and clinical investigations of immunotherapy for treatment of colorectal cancer including immune checkpoint blockade, adoptive cell therapy, monoclonal antibodies, oncolytic viruses, anti-cancer vaccines, and immune system modulators have been promising, but demonstrate limitations for patients with proficient mismatch repair enzymes. In this review, we discuss preclinical and clinical studies investigating immunotherapy for treatment of colorectal cancer and predictive biomarkers for response to these treatments. We also consider open questions including optimal combination treatments to maximize efficacy, minimize toxicity, and prevent acquired resistance and approaches to sensitize mismatch repair-proficient patients to immunotherapy.
Collapse
|
280
|
Genetic and Molecular Determinants of Lymphatic Malformations: Potential Targets for Therapy. J Dev Biol 2022; 10:jdb10010011. [PMID: 35225964 PMCID: PMC8883961 DOI: 10.3390/jdb10010011] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/28/2022] [Accepted: 02/01/2022] [Indexed: 12/15/2022] Open
Abstract
Lymphatic malformations are fluid-filled congenital defects of lymphatic channels occurring in 1 in 6000 to 16,000 patients. There are various types, and they often exist in conjunction with other congenital anomalies and vascular malformations. Great strides have been made in understanding these malformations in recent years. This review summarize known molecular and embryological precursors for lymphangiogenesis. Gene mutations and dysregulations implicated in pathogenesis of lymphatic malformations are discussed. Finally, we touch on current and developing therapies with special attention on targeted biotherapeutics.
Collapse
|
281
|
Huang Y, He S, Chen Y, Sheng J, Fu Y, Du X, Yang Y, Liu H, Han Z, Huang Y, Wen Q, Zhou C, Zhou X, Hu S, Ma L. UCHL1 Promoted Polarization of M1 Macrophages by Regulating the PI3K/AKT Signaling Pathway. J Inflamm Res 2022; 15:735-746. [PMID: 35153498 PMCID: PMC8824699 DOI: 10.2147/jir.s343487] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Affiliation(s)
- Yulan Huang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Shitong He
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Yitian Chen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Junli Sheng
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Yuling Fu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Xialin Du
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Yalong Yang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Honglin Liu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Zhenyu Han
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Yingqi Huang
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Qian Wen
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Chaoying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Xinying Zhou
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Shengfeng Hu
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
| | - Li Ma
- Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China
- Correspondence: Li Ma; Shengfeng Hu, Institute of Molecular Immunology, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, People’s Republic of China, Email ;
| |
Collapse
|
282
|
He XS, Ye WL, Zhang YJ, Yang XQ, Liu F, Wang JR, Ding XL, Yang Y, Zhang RN, Zhao YY, Bi HX, Guo LC, Gan WJ, Wu H. Oncogenic potential of BEST4 in colorectal cancer via activation of PI3K/Akt signaling. Oncogene 2022; 41:1166-1177. [PMID: 35058597 DOI: 10.1038/s41388-021-02160-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 12/02/2021] [Accepted: 12/14/2021] [Indexed: 12/11/2022]
Abstract
BEST4 is a member of the bestrophin protein family that plays a critical role in human intestinal epithelial cells. However, its role and mechanism in colorectal cancer (CRC) remain largely elusive. Here, we investigated the role and clinical significance of BEST4 in CRC. Our results demonstrate that BEST4 expression is upregulated in clinical CRC samples and its high-level expression correlates with advanced TNM (tumor, lymph nodes, distant metastasis) stage, LNM (lymph node metastasis), and poor survival. Functional studies revealed that ectopic expression of BEST4 promoted CRC cell proliferation and metastasis, whereas the depletion of BEST4 had the opposite effect both in vitro and in vivo. Mechanistically, BEST4 binds to the p85α regulatory subunit of phosphatidylinositol-3-kinase (PI3K) and promotes p110 kinase activity; this leads to activation of Akt signaling and expression of MYC and CCND1, which are critical regulators of cell proliferation and metastasis. In clinical samples, the expression of BEST4 is positively associated with the expression of phosphorylated Akt, MYC and CCND1. Pharmacological inhibition of Akt activity markedly repressed BEST4-mediated Akt signaling and proliferation and metastasis of CRC cells. Importantly, the interaction between BEST4 and p85α was also enhanced by epidermal growth factor (EGF) in CRC cells. Therapeutically, BEST4 suppression effectively sensitized CRC cells to gefitinib treatment in vivo. Taken together, our findings indicate the oncogenic potential of BEST4 in colorectal carcinogenesis and metastasis by modulating BEST4/PI3K/Akt signaling, highlighting a potential strategy for CRC therapy.
Collapse
Affiliation(s)
- Xiao-Shun He
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Wen-Long Ye
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Yu-Juan Zhang
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Xiao-Qin Yang
- Department of Bioinformatics, Medical College of Soochow University, Soochow University, Suzhou, 215123, China
| | - Feng Liu
- Department of General Surgery, Canglang Hospital of Suzhou, Suzhou, 215009, China
| | - Jing-Ru Wang
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Xiao-Lu Ding
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Yun Yang
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Ruo-Nan Zhang
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Yuan-Yuan Zhao
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Hai-Xia Bi
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China
| | - Ling-Chuan Guo
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China.
| | - Wen-Juan Gan
- Department of Pathology, Dushu Lake Hospital Affiliated to Soochow University, Soochow University, Suzhou, 215124, China.
- Department of Pathology, Medical Center of Soochow University, Soochow University, Suzhou, 215124, China.
| | - Hua Wu
- Department of Pathology, Medical College of Soochow University & The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215123, China.
- Department of Pathology, Dushu Lake Hospital Affiliated to Soochow University, Soochow University, Suzhou, 215124, China.
| |
Collapse
|
283
|
Zhou XR, Li X, Liao LP, Han J, Huang J, Li JC, Tao HR, Fan SJ, Chen ZF, Li Q, Chen SJ, Ding H, Yang YX, Zhou B, Jiang HL, Chen KX, Zhang YY, Huang CX, Luo C. P300/CBP inhibition sensitizes mantle cell lymphoma to PI3Kδ inhibitor idelalisib. Acta Pharmacol Sin 2022; 43:457-469. [PMID: 33850273 PMCID: PMC8791947 DOI: 10.1038/s41401-021-00643-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/08/2021] [Indexed: 02/03/2023]
Abstract
Mantle cell lymphoma (MCL) is a lymphoproliferative disorder lacking reliable therapies. PI3K pathway contributes to the pathogenesis of MCL, serving as a potential target. However, idelalisib, an FDA-approved drug targeting PI3Kδ, has shown intrinsic resistance in MCL treatment. Here we report that a p300/CBP inhibitor, A-485, could overcome resistance to idelalisib in MCL cells in vitro and in vivo. A-485 was discovered in a combinational drug screening from an epigenetic compound library containing 45 small molecule modulators. We found that A-485, the highly selective catalytic inhibitor of p300 and CBP, was the most potent compound that enhanced the sensitivity of MCL cell line Z-138 to idelalisib. Combination of A-485 and idelalisib remarkably decreased the viability of three MCL cell lines tested. Co-treatment with A-485 and idelalisib in Maver-1 and Z-138 MCL cell xenograft mice for 3 weeks dramatically suppressed the tumor growth by reversing the unsustained inhibition in PI3K downstream signaling. We further demonstrated that p300/CBP inhibition decreased histone acetylation at RTKs gene promoters and reduced transcriptional upregulation of RTKs, thereby inhibiting the downstream persistent activation of MAPK/ERK signaling, which also contributed to the pathogenesis of MCL. Therefore, additional inhibition of p300/CBP blocked MAPK/ERK signaling, which rendered maintaining activation to PI3K-mTOR downstream signals p-S6 and p-4E-BP1, thus leading to suppression of cell growth and tumor progression and eliminating the intrinsic resistance to idelalisib ultimately. Our results provide a promising combination therapy for MCL and highlight the potential use of epigenetic inhibitors targeting p300/CBP to reverse drug resistance in tumor.
Collapse
Affiliation(s)
- Xiao-ru Zhou
- grid.9227.e0000000119573309Drug Discovery and Design Center, The Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China ,grid.440637.20000 0004 4657 8879School of Life Science and Technology, ShanghaiTech University, Shanghai, 200031 China
| | - Xiao Li
- grid.9227.e0000000119573309Drug Discovery and Design Center, The Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Li-ping Liao
- grid.9227.e0000000119573309Drug Discovery and Design Center, The Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Jie Han
- grid.9227.e0000000119573309Drug Discovery and Design Center, The Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Jing Huang
- grid.9227.e0000000119573309Drug Discovery and Design Center, The Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Jia-cheng Li
- grid.9227.e0000000119573309Drug Discovery and Design Center, The Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Hong-ru Tao
- grid.9227.e0000000119573309Drug Discovery and Design Center, The Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Shi-jie Fan
- grid.9227.e0000000119573309Drug Discovery and Design Center, The Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Zhi-feng Chen
- grid.9227.e0000000119573309Drug Discovery and Design Center, The Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Qi Li
- grid.9227.e0000000119573309Drug Discovery and Design Center, The Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Shi-jie Chen
- grid.9227.e0000000119573309Drug Discovery and Design Center, The Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Hong Ding
- grid.9227.e0000000119573309Drug Discovery and Design Center, The Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Ya-xi Yang
- grid.9227.e0000000119573309Department of Medicinal Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Bing Zhou
- grid.9227.e0000000119573309Department of Medicinal Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Hua-liang Jiang
- grid.9227.e0000000119573309Drug Discovery and Design Center, The Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Kai-xian Chen
- grid.9227.e0000000119573309Drug Discovery and Design Center, The Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China ,grid.440637.20000 0004 4657 8879School of Life Science and Technology, ShanghaiTech University, Shanghai, 200031 China
| | - Yuan-yuan Zhang
- grid.9227.e0000000119573309Drug Discovery and Design Center, The Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Chuan-xin Huang
- grid.16821.3c0000 0004 0368 8293Shanghai Institute of Immunology and Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Cheng Luo
- grid.9227.e0000000119573309Drug Discovery and Design Center, The Center for Chemical Biology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,grid.410726.60000 0004 1797 8419University of Chinese Academy of Sciences, Beijing, 100049 China ,grid.440637.20000 0004 4657 8879School of Life Science and Technology, ShanghaiTech University, Shanghai, 200031 China
| |
Collapse
|
284
|
Tsygankova S, Komova D, Boulygina E, Slobodova N, Sharko F, Rastorguev S, Gladysheva-Azgari M, Koroleva D, Smol’yaninova A, Tatarnikova S, Obuchova T, Nedoluzhko A, Gabeeva N, Zvonkov E. Non-GCB Diffuse Large B-Cell Lymphoma With an Atypical Disease Course: A Case Report and Clinical Exome Analysis. World J Oncol 2022; 13:38-47. [PMID: 35317330 PMCID: PMC8913013 DOI: 10.14740/wjon1436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/31/2021] [Indexed: 11/27/2022] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoid tumor among other non-Hodgkin lymphomas (30-40% of all cases). This type of lymphoma is characterized by significant differences in treatment response and the heterogeneity of clinical traits. Approximately 60% of patients are cured using standard chemotherapy (CT), while in 10-15% of cases, the tumor is characterized by an extremely aggressive course and resistance to even the most high-dose programs with autologous stem cell transplantation (auto-SCT). The activated B-cell (ABC) subtype of DLBCL is characterized by poor prognosis. Here, we describe a clinical case of diffuse ABC-DLBCL with an atypical disease course. Complete remission was achieved after four courses of CT, followed by autologous hematopoietic stem cell transplantation (auto-HSCT). However, early relapse occurred 2 months after the completion of treatment. According to the results of cytogenetic studies, significant chromosome breakdowns were observed. Exome sequencing allowed for the detection of several novel mutations that affect components of the NOTCH2 and NF-κB signaling pathways, a number of epigenetic regulators (KMT2D, CREBBP, EP300, ARID1A, MEF2B), as well as members of the immunoglobulin superfamily (CD58 and CD70). Whether these mutations were the result of therapy or were originally present in the lymphoid tumor remains unclear. Nevertheless, the introduction of genomic technologies into clinical practice is important for making a diagnosis and developing a DLBCL treatment regimen with the use of targeted drugs.
Collapse
Affiliation(s)
- Svetlana Tsygankova
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
- These authors contributed equally
| | - Daria Komova
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
- These authors contributed equally
| | - Eugenia Boulygina
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
| | - Natalia Slobodova
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
| | - Fedor Sharko
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
| | - Sergey Rastorguev
- National Research Center “Kurchatov Institute”, 123182 Moscow, Russia
| | | | - Daria Koroleva
- National Medical Hematology Research Center, 125167 Moscow, Russia
| | | | | | - Tatiana Obuchova
- National Medical Hematology Research Center, 125167 Moscow, Russia
| | - Artem Nedoluzhko
- Moscow Healthcare Department, Mental-Health Clinic No. 1 Named After N.A. Alexeev, 115191 Moscow, Russia
| | - Nelli Gabeeva
- National Medical Hematology Research Center, 125167 Moscow, Russia
| | - Eugene Zvonkov
- National Medical Hematology Research Center, 125167 Moscow, Russia
| |
Collapse
|
285
|
Murugan AK, Alzahrani AS. Isocitrate Dehydrogenase IDH1 and IDH2 Mutations in Human Cancer: Prognostic Implications for Gliomas. Br J Biomed Sci 2022; 79:10208. [PMID: 35996504 PMCID: PMC8915566 DOI: 10.3389/bjbs.2021.10208] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/02/2021] [Indexed: 12/15/2022]
Abstract
Background: There are isolated reports of mutations in genes for isocitrate dehydrogenases (IDH1 and IDH2), but few have been examined in a large number of different malignancies. We aimed to analyze mutational prevalence of these genes in a large series of cancers and determine their significance in most mutated phenotype. Methods: We analyzed the frequencies of IDH1 and IDH2 mutations in 14,726 malignancies of 37 cancers. Furthermore, we examined these mutations in the most frequent cancer (gliomas, 923 cases) from a single cohort, and determined their clinical significance. Results:IDH1 mutations were present in 3% (473/14,726) of cancers. The highest frequencies were in oligodendrogliomas (91/102, 89%), anaplastic oligodendrogliomas (40/46, 87%), and diffuse astrocytomas (89/116, 77%). IDH2 mutation was detected in <1% (83/14,726) of cancers, but were present in 13% (6/46) of anaplastic oligodendrogliomas, 9% (9/102) of oligodendrogliomas, and in 5% (2/39) of cutaneous squamous cell carcinomas. Further analyses of 923 gliomas revealed 34 and 1% of IDH1 and IDH2 mutations, respectively. In up to 342 months of follow-up, IDH1 and IDH2 mutations were significantly linked with better overall (OS) (both p = 0.01) and progression-free survival (PFS) (p = 0.01; p = 0.004), respectively. Conclusion:IDH1 and IDH2 are often mutated in a tissue-specific manner, most commonly in gliomas. Mutation in both genes is linked to OS and PFS. Our findings suggest that these genes are promising therapeutic targets and strong prognostic biomarkers in gliomas.
Collapse
Affiliation(s)
- A. K. Murugan
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- *Correspondence: A. K. Murugan,
| | - A. S. Alzahrani
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
- Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
286
|
Kim JC, Bodmer WF. Genomic landscape of colorectal carcinogenesis. J Cancer Res Clin Oncol 2022; 148:533-545. [PMID: 35048197 DOI: 10.1007/s00432-021-03888-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/11/2021] [Indexed: 12/19/2022]
|
287
|
Bernardini A, Dueñas M, Martín-Soberon MC, Rubio C, Suarez-Cabrera C, Ruiz-Palomares R, Munera-Maravilla E, Lázaro S, Lodewijk I, Rueda D, Gómez-Sánchez D, Alonso-Gordoa T, Puente J, Pinto Á, González-Peramato P, Aguado C, Herrera M, López F, Martinez VMG, Morales L, Castellano D, Paramio JM, de Velasco G. Genomic Landscape of Vinflunine Response in Metastatic Urothelial Cancer. Cancers (Basel) 2022; 14:cancers14020378. [PMID: 35053540 PMCID: PMC8773703 DOI: 10.3390/cancers14020378] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/28/2021] [Accepted: 12/29/2021] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Few metastatic urothelial cancer patients achieve durable clinical benefit with vinflunine. Predictive biomarkers to help to identify better treatment strategies are extremely needed. The objective of this study was to identify molecular differences between extreme responders to vinflunine in urothelial cancer. Genomic and immune markers are potentially useful identifying patients that may achieve greater benefit with vinflunine. Abstract Background and Aims: Metastatic urothelial carcinoma (mUC) remains an incurable disease with limited treatment options after platinum-based chemotherapy and immune checkpoint blockade (ICB). Vinflunine has shown a modest increase in overall survival and remains a therapeutic option for chemo- and immunotherapy refractory tumours. However, biomarkers that could identify responding patients to vinflunine and possible alternative therapies after failure to treatment are still missing. In this study, we aimed to identify potential genomic biomarkers of vinflunine response in mUC patient samples and potential management alternatives. Methods: Formalin-fixed paraffin-embedded samples of mUC patients (n = 23) from three university hospitals in Spain were used for genomic targeted-sequencing and transcriptome (using the Immune Profile panel by NanoString) analyses. Patients who received vinflunine after platinum-based chemotherapy failure were classified in non-responders (NR: progressive disease ≤ 3 months; n= 11) or responders (R: response ≥ 6 months; n = 12). Results: Genomic characterization revealed that the most common alteration, TP53 mutations, had comparable frequency in R (6/12; 50%) and NR (4/11; 36%). Non-synonymous mutations in KTM2C (4/12; 33.3%), PIK3CA (3/12; 25%) and ARID2 (3/12; 25%) were predominantly associated with response. No significant difference was observed in tumour mutational burden (TMB) between R and NR patients. The NR tumours showed increased expression of diverse immune-related genes and pathways, including various interferon gamma-related genes. We also identified increased MAGEA4 expression as a potential biomarker of non-responding tumours to vinflunine treatment. Conclusions: Our data may help to identify potential genomic biomarkers of response to vinflunine. Moreover, tumours refractory to vinflunine showed immune signatures potentially associated with response to ICB. Extensive validation studies, including longitudinal series, are needed to corroborate these findings.
Collapse
Affiliation(s)
- Alejandra Bernardini
- Instituto de Investigación i+12, Hospital University “12 de Octubre”, 28040 Madrid, Spain; (A.B.); (M.D.); (M.C.M.-S.); (C.R.); (C.S.-C.); (R.R.-P.); (E.M.-M.); (I.L.); (D.R.); (D.G.-S.); (V.M.G.M.); (L.M.); (D.C.)
- Unidad de Oncología Molecular, CIEMAT, 28045 Madrid, Spain;
- Centro de Investigacion en Red de Cáncer CIBERONC, 28029 Madrid, Spain
| | - Marta Dueñas
- Instituto de Investigación i+12, Hospital University “12 de Octubre”, 28040 Madrid, Spain; (A.B.); (M.D.); (M.C.M.-S.); (C.R.); (C.S.-C.); (R.R.-P.); (E.M.-M.); (I.L.); (D.R.); (D.G.-S.); (V.M.G.M.); (L.M.); (D.C.)
- Unidad de Oncología Molecular, CIEMAT, 28045 Madrid, Spain;
- Centro de Investigacion en Red de Cáncer CIBERONC, 28029 Madrid, Spain
| | - María Cruz Martín-Soberon
- Instituto de Investigación i+12, Hospital University “12 de Octubre”, 28040 Madrid, Spain; (A.B.); (M.D.); (M.C.M.-S.); (C.R.); (C.S.-C.); (R.R.-P.); (E.M.-M.); (I.L.); (D.R.); (D.G.-S.); (V.M.G.M.); (L.M.); (D.C.)
- Departamento de Oncología Médica, Hospital 12 de Octubre, 28040 Madrid, Spain; (M.H.); (F.L.)
| | - Carolina Rubio
- Instituto de Investigación i+12, Hospital University “12 de Octubre”, 28040 Madrid, Spain; (A.B.); (M.D.); (M.C.M.-S.); (C.R.); (C.S.-C.); (R.R.-P.); (E.M.-M.); (I.L.); (D.R.); (D.G.-S.); (V.M.G.M.); (L.M.); (D.C.)
- Unidad de Oncología Molecular, CIEMAT, 28045 Madrid, Spain;
- Centro de Investigacion en Red de Cáncer CIBERONC, 28029 Madrid, Spain
| | - Cristian Suarez-Cabrera
- Instituto de Investigación i+12, Hospital University “12 de Octubre”, 28040 Madrid, Spain; (A.B.); (M.D.); (M.C.M.-S.); (C.R.); (C.S.-C.); (R.R.-P.); (E.M.-M.); (I.L.); (D.R.); (D.G.-S.); (V.M.G.M.); (L.M.); (D.C.)
- Unidad de Oncología Molecular, CIEMAT, 28045 Madrid, Spain;
- Centro de Investigacion en Red de Cáncer CIBERONC, 28029 Madrid, Spain
| | - Raquel Ruiz-Palomares
- Instituto de Investigación i+12, Hospital University “12 de Octubre”, 28040 Madrid, Spain; (A.B.); (M.D.); (M.C.M.-S.); (C.R.); (C.S.-C.); (R.R.-P.); (E.M.-M.); (I.L.); (D.R.); (D.G.-S.); (V.M.G.M.); (L.M.); (D.C.)
- Unidad de Oncología Molecular, CIEMAT, 28045 Madrid, Spain;
| | - Ester Munera-Maravilla
- Instituto de Investigación i+12, Hospital University “12 de Octubre”, 28040 Madrid, Spain; (A.B.); (M.D.); (M.C.M.-S.); (C.R.); (C.S.-C.); (R.R.-P.); (E.M.-M.); (I.L.); (D.R.); (D.G.-S.); (V.M.G.M.); (L.M.); (D.C.)
- Unidad de Oncología Molecular, CIEMAT, 28045 Madrid, Spain;
- Centro de Investigacion en Red de Cáncer CIBERONC, 28029 Madrid, Spain
| | - Sara Lázaro
- Unidad de Oncología Molecular, CIEMAT, 28045 Madrid, Spain;
| | - Iris Lodewijk
- Instituto de Investigación i+12, Hospital University “12 de Octubre”, 28040 Madrid, Spain; (A.B.); (M.D.); (M.C.M.-S.); (C.R.); (C.S.-C.); (R.R.-P.); (E.M.-M.); (I.L.); (D.R.); (D.G.-S.); (V.M.G.M.); (L.M.); (D.C.)
- Unidad de Oncología Molecular, CIEMAT, 28045 Madrid, Spain;
- Centro de Investigacion en Red de Cáncer CIBERONC, 28029 Madrid, Spain
| | - Daniel Rueda
- Instituto de Investigación i+12, Hospital University “12 de Octubre”, 28040 Madrid, Spain; (A.B.); (M.D.); (M.C.M.-S.); (C.R.); (C.S.-C.); (R.R.-P.); (E.M.-M.); (I.L.); (D.R.); (D.G.-S.); (V.M.G.M.); (L.M.); (D.C.)
| | - David Gómez-Sánchez
- Instituto de Investigación i+12, Hospital University “12 de Octubre”, 28040 Madrid, Spain; (A.B.); (M.D.); (M.C.M.-S.); (C.R.); (C.S.-C.); (R.R.-P.); (E.M.-M.); (I.L.); (D.R.); (D.G.-S.); (V.M.G.M.); (L.M.); (D.C.)
| | | | - Javier Puente
- Departamento de Oncología Médica, Hospital Clínico San Carlos, 28040 Madrid, Spain; (J.P.); (C.A.)
| | - Álvaro Pinto
- Departamento de Oncología Médica, Hospital La Paz, 28046 Madrid, Spain; (Á.P.); (P.G.-P.)
| | | | - Carlos Aguado
- Departamento de Oncología Médica, Hospital Clínico San Carlos, 28040 Madrid, Spain; (J.P.); (C.A.)
| | - Mercedes Herrera
- Departamento de Oncología Médica, Hospital 12 de Octubre, 28040 Madrid, Spain; (M.H.); (F.L.)
| | - Flora López
- Departamento de Oncología Médica, Hospital 12 de Octubre, 28040 Madrid, Spain; (M.H.); (F.L.)
| | - Victor M. G. Martinez
- Instituto de Investigación i+12, Hospital University “12 de Octubre”, 28040 Madrid, Spain; (A.B.); (M.D.); (M.C.M.-S.); (C.R.); (C.S.-C.); (R.R.-P.); (E.M.-M.); (I.L.); (D.R.); (D.G.-S.); (V.M.G.M.); (L.M.); (D.C.)
- Unidad de Oncología Molecular, CIEMAT, 28045 Madrid, Spain;
- Centro de Investigacion en Red de Cáncer CIBERONC, 28029 Madrid, Spain
| | - Lucía Morales
- Instituto de Investigación i+12, Hospital University “12 de Octubre”, 28040 Madrid, Spain; (A.B.); (M.D.); (M.C.M.-S.); (C.R.); (C.S.-C.); (R.R.-P.); (E.M.-M.); (I.L.); (D.R.); (D.G.-S.); (V.M.G.M.); (L.M.); (D.C.)
- Unidad de Oncología Molecular, CIEMAT, 28045 Madrid, Spain;
| | - Daniel Castellano
- Instituto de Investigación i+12, Hospital University “12 de Octubre”, 28040 Madrid, Spain; (A.B.); (M.D.); (M.C.M.-S.); (C.R.); (C.S.-C.); (R.R.-P.); (E.M.-M.); (I.L.); (D.R.); (D.G.-S.); (V.M.G.M.); (L.M.); (D.C.)
- Centro de Investigacion en Red de Cáncer CIBERONC, 28029 Madrid, Spain
- Departamento de Oncología Médica, Hospital 12 de Octubre, 28040 Madrid, Spain; (M.H.); (F.L.)
| | - Jesús M. Paramio
- Instituto de Investigación i+12, Hospital University “12 de Octubre”, 28040 Madrid, Spain; (A.B.); (M.D.); (M.C.M.-S.); (C.R.); (C.S.-C.); (R.R.-P.); (E.M.-M.); (I.L.); (D.R.); (D.G.-S.); (V.M.G.M.); (L.M.); (D.C.)
- Unidad de Oncología Molecular, CIEMAT, 28045 Madrid, Spain;
- Centro de Investigacion en Red de Cáncer CIBERONC, 28029 Madrid, Spain
- Correspondence: (J.M.P.); (G.d.V.)
| | - Guillermo de Velasco
- Instituto de Investigación i+12, Hospital University “12 de Octubre”, 28040 Madrid, Spain; (A.B.); (M.D.); (M.C.M.-S.); (C.R.); (C.S.-C.); (R.R.-P.); (E.M.-M.); (I.L.); (D.R.); (D.G.-S.); (V.M.G.M.); (L.M.); (D.C.)
- Departamento de Oncología Médica, Hospital 12 de Octubre, 28040 Madrid, Spain; (M.H.); (F.L.)
- Correspondence: (J.M.P.); (G.d.V.)
| |
Collapse
|
288
|
Exploring the selective mechanism of inhibitors towards different subtypes of class I PI3K. Chem Phys Lett 2022. [DOI: 10.1016/j.cplett.2021.139174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
289
|
Reinhardt K, Stückrath K, Hartung C, Kaufhold S, Uleer C, Hanf V, Lantzsch T, Peschel S, John J, Pöhler M, Bauer M, Bürrig FK, Weigert E, Buchmann J, Kantelhardt EJ, Thomssen C, Vetter M. PIK3CA-mutations in breast cancer. Breast Cancer Res Treat 2022; 196:483-493. [PMID: 36279023 PMCID: PMC9633529 DOI: 10.1007/s10549-022-06637-w] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/14/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE Phosphatidylinositide-3-kinase (PI3K) regulates proliferation and apoptosis; somatic PIK3CA-mutations may activate these processes. Aim of this study was to determine the prevalence of PIK3CA-mutations in a cohort of early stage breast cancer patients and the association to the course of disease. PATIENTS AND METHODS From an unselected cohort of 1270 breast cancer patients (PiA, Prognostic Assessment in routine application, NCT01592825) 1123 tumours were tested for the three PIK3CA hotspot-mutations H1047R, E545K, and E542K by qPCR. Primary objectives were the prevalence of somatic PIK3CA-mutations and their association to tumour characteristics. Secondary objective was the association of PIK3CA-mutations to recurrence-free interval (RFI) and overall survival. RESULTS PIK3CA-mutation rate was 26.7% (300 of 1123). PIK3CA-mutations were significantly more frequent in steroid hormone-receptor (SHR)-positive HER2-negative (31.4%), and G1 and G2 tumours (32.8%). Overall, we did not observe a significant association of PIK3CA-mutations to RFI. In SHR-positive BCs with PIK3CA-mutations, a strong trend for impaired RFI was observed (adjusted HR 1.64, 95% CI 0.958-2.807), whilst in SHR-negative BCs PIK3CA-mutations were insignificantly associated with improved RFI (adjusted HR 0.49; 95% CI 0.152-1.597). Of note, we observed a significantly detrimental prognostic impact of PIK3CA-mutations on RFI in SHR-positive, HER2-negative BCs if only aromatase inhibitors were administered as adjuvant therapy (adjusted HR 4.44, 95% CI 1.385-13.920), whilst no impact was observed in tamoxifen treated patients. CONCLUSION This cohort study speficies the overall mutation rate of PIK3CA in early breast cancer. The impact of PIK3CA-mutations on RFI and OS was heterogeneous. Our results suggest that estrogen deprivation failes to be active in case of PIK3CA-mutation.
Collapse
Affiliation(s)
- Kristin Reinhardt
- Department of Gynaecology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle (Saale), Germany
| | - Kathrin Stückrath
- Department of Gynaecology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle (Saale), Germany
| | - Carolin Hartung
- Department of Gynaecology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle (Saale), Germany
| | - Sandy Kaufhold
- Department of Gynaecology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle (Saale), Germany
| | | | - Volker Hanf
- Department of Gynaecology, Nathanstift, Hospital Fuerth, Fürth, Germany
| | - Tillmann Lantzsch
- Department of Gynaecology, Hospital St. Elisabeth and St. Barbara, Halle (Saale), Germany
| | - Susanne Peschel
- Department of Gynaecology, St. Bernward Hospital, Hildesheim, Germany
| | - Jutta John
- Department of Gynaecology, Helios Hospital Hildesheim, Hildesheim, Germany
| | - Marleen Pöhler
- Department of Gynaecology, Asklepios Hospital Goslar, Goslar, Germany ,Present Address: Department of Gynaecology and Obstretrics, Hospital Wolfenbüttel, Wolfenbüttel, Germany
| | - Marcus Bauer
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | | | - Edith Weigert
- Institute of Pathology, Hospital Fürth, Fürth, Germany ,Present Address: Gemeinschaftspraxis Pathologie Amberg, Amberg, Germany
| | - Jörg Buchmann
- Institute of Pathology, Hospital Martha-Maria, Halle (Saale), Germany
| | - Eva Johanna Kantelhardt
- Department of Gynaecology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle (Saale), Germany ,Institute of Epidemiology, Biometry and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Christoph Thomssen
- Department of Gynaecology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle (Saale), Germany
| | - Martina Vetter
- Department of Gynaecology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle (Saale), Germany
| |
Collapse
|
290
|
Brito C, Tomás A, Azevedo A, Esteves S, Mafra M, Roque L, Pojo M. PIK3CA Mutations in Diffuse Gliomas: An Update on Molecular Stratification, Prognosis, Recurrence, and Aggressiveness. CLINICAL MEDICINE INSIGHTS: ONCOLOGY 2022; 16:11795549211068804. [PMID: 35023985 PMCID: PMC8743979 DOI: 10.1177/11795549211068804] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/06/2021] [Indexed: 11/15/2022] Open
Abstract
Introduction: PIK3CA is one of the most mutated oncogenes in solid tumors. In breast cancer (ER-positive, HER2-negative), these events represent a predictive biomarker of response to alpelisib. In glioblastomas (GBM), PIK3CA mutations were described as early constitutive events. Here, we investigated PIK3CA mutational profile across glioma molecular subgroups and its relevance during glioma recurrence. Furthermore, PIK3CA mutations’ effect in PI3K pathway, prognosis, and response to therapy was also explored. Material and Methods: Exons 10 and 21 of PIK3CA mutations were evaluated in 394 gliomas and 19 glioma recurrences from Instituto Português de Oncologia Lisboa Francisco Gentil (IPOLFG) and compared with The Cancer Genome Atlas (TCGA) data. TIMER2.0 and NetMHCpan4.1 were used to assess the immune-microenvironment contribution. Results: PIK3CA mutations were identified among all glioma subgroups, although with no impact on their stratification or prognosis. In both cohorts (IPOLFG and TCGA), PIK3CA mutation frequencies in IDH-mutant and IDH-wild-type GBM were similar (IPOLFG: 9% and 3%; TCGA: 8% and 2%). These mutations were not mutually exclusive with PTEN deletion and EGFR amplification. Despite their reduced frequency, we discovered PIK3CA mutations were maintained during glioma recurrence regardless of administered therapies. The immune microenvironment might not contribute to this phenotype as PIK3CA mutations did not influence immune cell infiltration. Conclusions: Despite the absence of a predominant effect in glioma stratification, PIK3CA mutations were maintained during glioma recurrence, possibly contributing to glioma cell survival, representing promising therapeutic targets in recurrent glioma. Nevertheless, understanding the potential synergistic effects between PIK3CA mutations, PTEN deletion, and EGFR amplification is pivotal to targeted therapies’ efficiency.
Collapse
Affiliation(s)
- Cheila Brito
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E., Lisboa, Portugal
| | - Ana Tomás
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E., Lisboa, Portugal
| | - Ana Azevedo
- Serviço de Neurologia, Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E., Lisboa, Portugal
- Faculty of Health Sciences, University of Beira Interior, Covilhã, Portugal
| | - Susana Esteves
- Unidade de Investigação Clínica (UIC), Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E., Lisboa, Portugal
| | - Manuela Mafra
- Serviço de Anatomia Patológica, Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E., Lisboa, Portugal
| | - Lúcia Roque
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E., Lisboa, Portugal
| | - Marta Pojo
- Unidade de Investigação em Patobiologia Molecular (UIPM), Instituto Português de Oncologia de Lisboa Francisco Gentil E.P.E., Lisboa, Portugal
| |
Collapse
|
291
|
Li JF, Tian GL, Pan H, Zhang WT, Li DC, Liu JD, Zhao L, Li HL. An Analysis of the Pathogenic Genes and Mutation Sites of Macrodactyly. Pharmgenomics Pers Med 2022; 15:55-64. [PMID: 35125881 PMCID: PMC8809672 DOI: 10.2147/pgpm.s346373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/20/2022] [Indexed: 11/23/2022] Open
Abstract
Objective This study aimed to explore the pathogenic genes and mutation sites of macrodactyly. Methods Whole-exome sequencing was performed on the pathological tissue and peripheral blood of 12 patients with macrodactyly who were operated in our hospital between June 2018 and May 2020. In order to conduct comprehensive bioinformatics analysis and screen the pathogenic genes of macrodactyly, the patients were divided into four groups: macrodactyly of finger group, macrodactyly of foot group, macrodactyly and syndactyly of finger group, and macrodactyly and syndactyly of foot group. The results of the whole-exome sequencing were verified using Sanger sequencing in order to clarify the pathogenic genes and mutation sites of macrodactyly, and immunohistochemical analysis of the protein signaling pathways encoded by the pathogenic genes was performed to observe the protein expression and further verify the mutant genes. Results In the comprehensive bioinformatics analysis and Sanger verification of the whole-exome sequencing, the PIK3CA gene mutation was screened as the pathogenic gene of macrodactyly. The mutation sites were identified as the p.E542K (c.G1624A) and p.E545K (c.G1633A) sites of exon10 and the p.H1047R (c.A3140G) and p.G1049R (c.G3145C) sites of exon21. Among these, the p.G1049R (c.G3145C) locus was found in macrodactyly for the first time. The mutation of the PIK3CA gene was also found to lead to increased expression of serine-threonine kinase (AKT) in adipocytes in the PI3K-AKT-mTOR signaling pathway. Conclusion Mutation of the PIK3CA gene leads to the enhancement of the PI3K-AKT-mTOR signaling pathway, which is the cause of macrodactyly. There is also some diversity in PIK3CA gene mutation sites.
Collapse
Affiliation(s)
- Jian-Feng Li
- Department of Hand Surgery, Beijing Shunyi District Hospital; Shunyi Teaching Hospital of Capital Medical University, Beijing, 101300, People’s Republic of China
- Correspondence: Jian-Feng Li, Department of Hand Surgery, Beijing Shunyi District Hospital; Shunyi Teaching Hospital of Capital Medical University, No. 3 Guangming South Street, Shunyi District, Beijing, 101300, People’s Republic of China, Tel +86 10 69423220, Email
| | - Guang-Lei Tian
- Department of Hand Surgery, Beijing Jishuitan Hospital; Peking University Fourth School of Clinical Medicine, Beijing, 100035, People’s Republic of China
| | - Hui Pan
- Department of Pathology, Beijing Shunyi District Hospital; Shunyi Teaching Hospital of Capital Medical University, Beijing, 101300, People’s Republic of China
| | - Wen-Tong Zhang
- Department of Hand Surgery, Beijing Shunyi District Hospital; Shunyi Teaching Hospital of Capital Medical University, Beijing, 101300, People’s Republic of China
| | - Da-Cun Li
- Department of Hand Surgery, Beijing Shunyi District Hospital; Shunyi Teaching Hospital of Capital Medical University, Beijing, 101300, People’s Republic of China
| | - Jing-Da Liu
- Department of Hand Surgery, Beijing Shunyi District Hospital; Shunyi Teaching Hospital of Capital Medical University, Beijing, 101300, People’s Republic of China
| | - Liang Zhao
- Department of Hand Surgery, Beijing Shunyi District Hospital; Shunyi Teaching Hospital of Capital Medical University, Beijing, 101300, People’s Republic of China
| | - Hai-Lei Li
- Department of Hand Surgery, Beijing Shunyi District Hospital; Shunyi Teaching Hospital of Capital Medical University, Beijing, 101300, People’s Republic of China
| |
Collapse
|
292
|
Zhang X, Qi W, Xu Q, Li X, Zhou L, Ye L. Di(2-ethylhexyl) phthalate (DEHP) and thyroid: biological mechanisms of interference and possible clinical implications. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:1634-1644. [PMID: 34677768 DOI: 10.1007/s11356-021-17027-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 10/10/2021] [Indexed: 05/15/2023]
Abstract
Di(2-ethylhexyl) phthalate (DEHP) is a ubiquitous environmental endocrine disruptor. DEHP can be absorbed into the human body through the air, food, water, and skin. After entering the human body, DEHP is rapidly converted to mono(2-ethylhexyl) phthalate (MEHP) with greater toxicity than DEHP. An increasing number of studies indicates that DEHP or MEHP can damage the thyroid tissue and disrupt the function, but the mechanisms remain unclear. This article reviews the toxicity of DEHP on thyroid structures and functions and summarizes the potential mechanisms to provide evidence for preventing the thyroid-related diseases.
Collapse
Affiliation(s)
- Xueting Zhang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, 130021, China
| | - Wen Qi
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, 130021, China
| | - Qi Xu
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, 130021, China
| | - Xu Li
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, 130021, China
| | - Liting Zhou
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, 130021, China
| | - Lin Ye
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, 130021, China.
| |
Collapse
|
293
|
Chen F, Liu J, Song X, DuCote TJ, Byrd AL, Wang C, Brainson CF. EZH2 inhibition confers PIK3CA-driven lung tumors enhanced sensitivity to PI3K inhibition. Cancer Lett 2022; 524:151-160. [PMID: 34655667 PMCID: PMC8743034 DOI: 10.1016/j.canlet.2021.10.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/16/2021] [Accepted: 10/06/2021] [Indexed: 01/03/2023]
Abstract
Members of the PI3K signaling pathway, especially PIK3CA, the gene encoding the catalytic subunit of the PI3K complex, are highly mutated and amplified in various cancer types, including non-small cell lung cancer. Although PI3K inhibitors have been used in clinics for follicular lymphoma and chronic lymphocytic leukemia, no agents targeting PI3K aberrations in lung cancer have been approved by the FDA so far. In this study, we observed that PIK3CA-E545K, the most common mutation in lung cancer, harbored a modest induction of stem-like properties in lung epithelial cells, and drove development of adenocarcinoma autochthonously when paired with p53 loss in a murine mouse model. We also found that PIK3CA-mutant of amplified lung cancer cells were sensitive to EZH2 inhibition. EZH2 inhibition synergized with PI3K inhibition in human cancer cells in vitro and worked together efficiently in vivo. Mechanistically, EZH2 inhibition cooperated with PI3K inhibition to produce a more potent suppression of phospho-AKT downstream of PI3K. This study suggests a promising combination therapy to combat lung cancers with PIK3CA mutation or amplification. Both copanlisib, the PI3K inhibitor, and tazemetostat, the EZH2 inhibitor, are FDA-approved, which should enhance the clinical translation of this work.
Collapse
Affiliation(s)
- Fan Chen
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - Jinpeng Liu
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA,Department of Internal Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Xiulong Song
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - Tanner J. DuCote
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - Aria L. Byrd
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - Chi Wang
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA,Department of Internal Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Christine F. Brainson
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA,Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA,Corresponding author. Department of Toxicology and Cancer Biology Markey Cancer Center University of Kentucky, 1095 VA Drive, HSRB 456, Lexington, KY, 40536, USA.
| |
Collapse
|
294
|
OUP accepted manuscript. J Pharm Pharmacol 2022; 74:869-886. [DOI: 10.1093/jpp/rgac002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 02/03/2022] [Indexed: 11/14/2022]
|
295
|
Anticancer effects of veratramine via the phosphatidylinositol-3-kinase/serine-threonine kinase/mechanistic target of rapamycin and its downstream signaling pathways in human glioblastoma cell lines. Life Sci 2022; 288:120170. [PMID: 34826438 DOI: 10.1016/j.lfs.2021.120170] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 11/10/2021] [Accepted: 11/18/2021] [Indexed: 02/06/2023]
Abstract
AIMS Antitumor effects of veratramine in prostate and liver cancers has been investigated, but it is still unclear whether veratramine can be used as an effective therapeutic agent for glioma. The aim of this study was to evaluate the potential pharmacological mechanism of veratramine in glioma. MAIN METHODS Using four types of human glioblastoma cell lines, including A172, HS-683, T98G, and U-373-MG the dose-dependent antitumor effect of veratramine was evaluated. The cytotoxicity and cell proliferation were examined by CCK-8, and cell proliferation was further confirmed by anchorage-independent colony formation assay. The cell cycle distribution and apoptotic rate was assessed by flow cytometry, and apoptosis was further evaluated by apoptosis assay. The migration and invasiveness capacity were analyzed by using transwell. Protein and mRNA levels of related factors were determined by western blotting and RT-qPCR, respectively. KEY FINDINGS Veratramine markedly induced apoptosis, suppressed the cell proliferation via the cell cycle G0/G1 phase arrest, and reduced the capacity for the migration and invasion in human glioblastoma multiforme cell lines. Moreover, veratramine was sufficient to affect the phosphatidylinositol-3-kinase/serine-threonine kinase/mechanistic target of rapamycin signaling pathway and its downstream Mdm2/p53/p21 pathway in human glioblastoma cell lines. SIGNIFICANCE Antitumor effects of veratramine in suppression of glioma progression was mediated by the regulation of PI3K/Akt/mTOR and Mdm2/p53/p21 signaling pathway.
Collapse
|
296
|
Class I PI3K Biology. Curr Top Microbiol Immunol 2022; 436:3-49. [DOI: 10.1007/978-3-031-06566-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
297
|
Genetic Mutations of Pancreatic Cancer and Genetically Engineered Mouse Models. Cancers (Basel) 2021; 14:cancers14010071. [PMID: 35008235 PMCID: PMC8750056 DOI: 10.3390/cancers14010071] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy. Recent multi-gene analysis approaches such as next-generation sequencing have provided useful information on the molecular characterization of pancreatic tumors. Different types of pancreatic cancer and precursor lesions are characterized by specific molecular alterations. Genetically engineered mouse models (GEMMs) of PDAC are useful tools to understand the roles of altered genes. Most GEMMs are driven by oncogenic Kras, and can recapitulate the histological and molecular hallmarks of human PDAC and comparable precursor lesions. In this review, we summarize the main molecular alterations found in pancreatic neoplasms and GEMMs developed based on these alterations. Abstract Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy, and the seventh leading cause of cancer-related deaths worldwide. An improved understanding of tumor biology and novel therapeutic discoveries are needed to improve overall survival. Recent multi-gene analysis approaches such as next-generation sequencing have provided useful information on the molecular characterization of pancreatic tumors. Different types of pancreatic cancer and precursor lesions are characterized by specific molecular alterations. Genetically engineered mouse models (GEMMs) of PDAC are useful to understand the roles of altered genes. Most GEMMs are driven by oncogenic Kras, and can recapitulate the histological and molecular hallmarks of human PDAC and comparable precursor lesions. Advanced GEMMs permit the temporally and spatially controlled manipulation of multiple target genes using a dual-recombinase system or CRISPR/Cas9 gene editing. GEMMs that express fluorescent proteins allow cell lineage tracing to follow tumor growth and metastasis to understand the contribution of different cell types in cancer progression. GEMMs are widely used for therapeutic optimization. In this review, we summarize the main molecular alterations found in pancreatic neoplasms, developed GEMMs, and the contribution of GEMMs to the current understanding of PDAC pathobiology. Furthermore, we attempted to modify the categorization of altered driver genes according to the most updated findings.
Collapse
|
298
|
Borsari C, Wymann MP. Targeting Phosphoinositide 3-Kinase - Five Decades of Chemical Space Exploration. Chimia (Aarau) 2021; 75:1037-1044. [PMID: 34920774 DOI: 10.2533/chimia.2021.1037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Phosphoinositide 3-kinase (PI3K) plays a key role in a plethora of physiologic processes and controls cell growth, metabolism, immunity, cardiovascular and neurological function, and more. The discovery of wort-mannin as the first potent PI3K inhibitor (PI3Ki) in the 1990s provided rapid identification of PI3K-dependent processes, which drove the discovery of the PI3K/protein kinase B (PKB/Akt)/target of rapamycin (mTOR) pathway. Genetic mouse models and first PI3K isoform-specific inhibitors pinpointed putative therapeutic applications. The recognition of PI3K as target for cancer therapy drove subsequently drug development. Here we provide a brief journey through the emerging roles of PI3K to the development of preclinical and clinical PI3Ki candidates.
Collapse
Affiliation(s)
- Chiara Borsari
- Department of Biomedicine, University of Basel, Mattenstrasse 28, CH-4058 Basel, Switzerland
| | - Matthias P Wymann
- Department of Biomedicine, University of Basel, Mattenstrasse 28, CH-4058 Basel, Switzerland;,
| |
Collapse
|
299
|
Differential Regulation of the EGFR/PI3K/AKT/PTEN Pathway between Low- and High-Grade Gliomas. Brain Sci 2021; 11:brainsci11121655. [PMID: 34942957 PMCID: PMC8699139 DOI: 10.3390/brainsci11121655] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/09/2021] [Accepted: 12/14/2021] [Indexed: 01/07/2023] Open
Abstract
Gliomas represent 70% of all central system nervous tumors and are classified according to the degree of malignancy as low- or high-grade. The permanent activation of the EGFR/PI3K/AKT pathway by various genetic or post-translational alterations of EGFR, PI3KCA, and PTEN has been associated with increased proliferation and resistance to apoptosis. The present study aimed to analyze the molecular/genetic changes in the EGFR/PI3K/AKT/PTEN pathway between low-grade and high-grade gliomas in a sample of Colombian patients. A total of 30 samples were tested for PI3K and PTEN mutations, EGFR, PI3K, and AKT gene amplification, AKT, PI3K, BAX, Bcl2 expression levels, and phosphorylation of AKT and PTEN, EGFR and/or PI3K gene amplification was found in 50% of low-grade and 45% of high-grade ones. AKT amplification was found in 25% of the low-grade and 13.6% of the high-grade. The expression of PI3K, AKT, Bcl2, and BAX was increased particularly to a high degree. AKT phosphorylation was found in 66% of low-grade and 31.8% of high-grade. Increased phosphorylation of PTEN was found in 77% low-grade and 66% high-grade. Our results indicate that alterations in the EGFR/PI3K/AKT/PTEN pathway could be important in the initiation and malignant progression of this type of tumor.
Collapse
|
300
|
Morin G, Canaud G. Treatment strategies for mosaic overgrowth syndromes of the PI3K-AKT-mTOR pathway. Br Med Bull 2021; 140:36-49. [PMID: 34530449 DOI: 10.1093/bmb/ldab023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/05/2021] [Accepted: 08/27/2021] [Indexed: 11/14/2022]
Abstract
INTRODUCTION OR BACKGROUND Mosaic overgrowth syndromes (OS) are a proteiform ensemble of rare diseases displaying asymmetric overgrowth involving any tissue type, with degrees of severity ranging from isolated malformation to life-threatening conditions such as pulmonary embolism. Despite discordant clinical presentations, all those syndromes share common genetic anomalies: somatic mutations of genes involved in cell growth and proliferation. The PI3K-AKT-mTOR signaling pathway is one of the most prominent regulators of cell homeostasis, and somatic oncogenic mutations affecting this pathway are responsible for mosaic OS. This review aims to describe the clinical and molecular characteristics of the main OS involving the PI3K-AKT-mTOR pathway, along with the treatments available or under development. SOURCES OF DATA This review summarizes available data regarding OS in scientific articles published in peer-reviewed journals. AREAS OF AGREEMENT OS care requires a multidisciplinary approach relying on clinical and radiological follow-up along with symptomatic treatment. However, no specific treatment has yet shown efficacy in randomized control trials. AREAS OF CONTROVERSY Clinical classifications of OS led to frequent misdiagnosis. Moreover, targeted therapies directed at causal mutated proteins are developing in OSs through cancer drugs repositioning, but the evidence of efficacy and tolerance is still lacking for most of them. GROWING POINTS The genetic landscape of OS is constantly widening and molecular classifications tend to increase the accuracy of diagnosis, opening opportunities for targeted therapies. AREAS TIMELY FOR DEVELOPING RESEARCH OS are a dynamic, expanding field of research. Studies focusing on the identification of genetic anomalies and their pharmacological inhibition are needed.
Collapse
Affiliation(s)
- Gabriel Morin
- Université de Paris, Paris, France.,INSERM U1151, Institut Necker-Enfants Malades, Paris, France.,Unité d'hypercroissance dysharmonieuse et centre d'anomalies vasculaires, hôpital Necker Enfants Malades, AP-HP, France
| | - Guillaume Canaud
- Université de Paris, Paris, France.,INSERM U1151, Institut Necker-Enfants Malades, Paris, France.,Unité d'hypercroissance dysharmonieuse et centre d'anomalies vasculaires, hôpital Necker Enfants Malades, AP-HP, France
| |
Collapse
|