201
|
Chandra S, Goswami A, Mandal P. Molecular Heterogeneity of Cervical Cancer Among Different Ethnic/Racial Populations. J Racial Ethn Health Disparities 2022; 9:2441-2450. [PMID: 34741276 DOI: 10.1007/s40615-021-01180-8] [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: 09/15/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 12/29/2022]
Abstract
The study aimed to find differential gene mutation profile and gene expression status among different ethnic/racial human populations relevant for cervical cancer pathogenesis. The study was based on freely available datasets of The Cancer Genome Atlas (TCGA) of cervical cancer samples in Genomic Data Commons (GDC) data portal. We identified that choline metabolism in cancer and Ras signaling pathways were significantly associated with the Hispanic and Latino group of cervical cancer patients. In these pathways, mutations in the PIK3CA gene, especially E545K, were significantly associated with the Hispanic and LATINO group. We found that AFF3 gene mutation was associated with downregulation of its expression only among the White racial category of cervical cancer cases. Additionally, hypomethylation of the CpG position in the S shore region of the PM20D1 gene was associated with overexpression among the Asian category of cervical cancer cases. Heterogeneity of the molecular profile of AFF3 and PM20D1 gene among racial groups reflects the potential of differential targeted therapy of cervical cancer.
Collapse
Affiliation(s)
- Sanchita Chandra
- Biomedical Genetics Laboratory, Department of Zoology, The University of Burdwan, Burdwan, 713104, West Bengal, India
| | - Anindita Goswami
- Biomedical Genetics Laboratory, Department of Zoology, The University of Burdwan, Burdwan, 713104, West Bengal, India
| | - Paramita Mandal
- Biomedical Genetics Laboratory, Department of Zoology, The University of Burdwan, Burdwan, 713104, West Bengal, India.
| |
Collapse
|
202
|
Apurva, Abdul Sattar RS, Ali A, Nimisha, Kumar Sharma A, Kumar A, Santoshi S, Saluja SS. Molecular pathways in periampullary cancer: An overview. Cell Signal 2022; 100:110461. [PMID: 36096460 DOI: 10.1016/j.cellsig.2022.110461] [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: 05/18/2022] [Revised: 08/19/2022] [Accepted: 08/29/2022] [Indexed: 11/22/2022]
Abstract
Molecular alterations in oncogenes and tumor suppressors in various signaling pathways are basis for personalized therapy in cancer. Periampullary carcinoma behaves differently from pancreatic carcinoma both in prognosis and outcome, therefore it needs special attention. Pancreatic cancer have higher incidence of nodal spread and perineural &lymphovascular invasion suggesting it biologically more aggressive tumor compared to periampullary cancer. Since PAC tumors consist of heterogenous tissue of origin, they might contain different mutations in tumor associated genes and other changes in tissue composition among different subgroups clubbed together. Significant progress has been made in understanding the molecular nature of PAC in the previous two decades, and a large number of mutations and other genetic changes have been identified as being responsible for the disease. This review article targets to collate and discuss the molecular evolution of PAC and their implication in its outcome. As per literature, mitogen-activated protein kinase (MAPK), phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K), and Wnt signaling are the most common pathways involved in PAC. Mutations in KRAS, TP53, CTNNB1, SMAD4 and APC genes were the most frequently reported. I-subtype resembles colorectal cancer while the morphology of PB-type shows close resemblance to pancreatic tumors. The frequency of driver gene mutations is higher in I-type compared to PB-type of PAC indicating I-type to be genetically more unstable. The genetic landscape of PAC obtained from WES data highlighted PI3/AKT pathway to be a primary target in I-type and RAS/RAF in PB-type.
Collapse
Affiliation(s)
- Apurva
- Central Molecular Lab, GovindBallabhPant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India; Amity University, Noida, India
| | - Real Sumayya Abdul Sattar
- Central Molecular Lab, GovindBallabhPant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Asgar Ali
- Central Molecular Lab, GovindBallabhPant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Nimisha
- Central Molecular Lab, GovindBallabhPant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Abhay Kumar Sharma
- Central Molecular Lab, GovindBallabhPant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | - Arun Kumar
- Central Molecular Lab, GovindBallabhPant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India
| | | | - Sundeep Singh Saluja
- Central Molecular Lab, GovindBallabhPant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India; Department of GI Surgery, GovindBallabh Pant Institute of Postgraduate Medical Education and Research (GIPMER), New Delhi, India.
| |
Collapse
|
203
|
Mukherjee P, Bagchi A, Banerjee A, Roy H, Bhattacharya A, Biswas A, Chatterji U. PDE4 inhibitor eliminates breast cancer stem cells via noncanonical activation of mTOR. J Cell Biochem 2022; 123:1980-1996. [PMID: 36063486 DOI: 10.1002/jcb.30325] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 05/25/2022] [Accepted: 08/17/2022] [Indexed: 12/24/2022]
Abstract
Ineffective cancer treatment is implicated in metastasis, recurrence, resistance to chemotherapy and radiotherapy, and evasion of immune surveillance. All these failures occur due to the persistence of cancer stem cells (CSCs) even after rigorous therapy, thereby rendering them as essential targets for cancer management. Contrary to the quiescent nature of CSCs, a gene profiler array disclosed that phosphatidylinositol-3-kinase (PI3K), which is known to be crucial for cell proliferation, differentiation, and survival, was significantly upregulated in CSCs. Since PI3K is modulated by cyclic adenosine 3',5' monophosphate (cAMP), analyses of cAMP regulation revealed that breast CSCs expressed increased levels of phosphodiesterase 4 (PDE4) in contrast to normal stem cells. In accordance, the effects of rolipram, a PDE4 inhibitor, were evaluated on PI3K regulators and signaling. The efficacy of rolipram was compared with paclitaxel, an anticancer drug that is ineffective in obliterating breast CSCs. Analyses of downstream signaling components revealed a switch between cell survival and death, in response to rolipram, specifically of the CSCs. Rolipram-mediated downregulation of PDE4A levels in breast CSCs led to an increase in cAMP levels and protein kinase A (PKA) expression. Subsequently, PKA-mediated upregulation of phosphatase and tensin homolog antagonized the PI3K/AKT/mTOR pathway and led to cell cycle arrest. Interestingly, direct yet noncanonical activation of mTOR by PKA, circumventing the influence of PI3K and AKT, temporally shifted the fate of CSCs toward apoptosis. Rolipram in combination with paclitaxel indicated synergistic consequences, which effectively obliterated CSCs within a tumor, thereby suggesting combinatorial therapy as a sustainable and effective strategy to abrogate breast CSCs for better patient prognosis.
Collapse
Affiliation(s)
- Pritha Mukherjee
- Cancer Research Laboratory, Department of Zoology, University of Calcutta, Kolkata, India
| | - Arka Bagchi
- Molecular Cell Biology Laboratory, Department of Zoology, University of Kalyani, Kalyani, India
| | - Ananya Banerjee
- Cancer Research Laboratory, Department of Zoology, University of Calcutta, Kolkata, India
| | - Himansu Roy
- Department of Surgery, Calcutta Medical College, Kolkata, India
| | | | - Arunima Biswas
- Molecular Cell Biology Laboratory, Department of Zoology, University of Kalyani, Kalyani, India
| | - Urmi Chatterji
- Cancer Research Laboratory, Department of Zoology, University of Calcutta, Kolkata, India.,Centre for Research in Nanoscience and Nanotechnology, University of Calcutta, Kolkata, India
| |
Collapse
|
204
|
Colombo M, Passarelli F, Corsetto PA, Rizzo AM, Marabese M, De Simone G, Pastorelli R, Broggini M, Brunelli L, Caiola E. NSCLC Cells Resistance to PI3K/mTOR Inhibitors Is Mediated by Delta-6 Fatty Acid Desaturase (FADS2). Cells 2022; 11:cells11233719. [PMID: 36496978 PMCID: PMC9736998 DOI: 10.3390/cells11233719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/18/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Hyperactivation of the phosphatidylinositol-3-kinase (PI3K) pathway is one of the most common events in human cancers. Several efforts have been made toward the identification of selective PI3K pathway inhibitors. However, the success of these molecules has been partially limited due to unexpected toxicities, the selection of potentially responsive patients, and intrinsic resistance to treatments. Metabolic alterations are intimately linked to drug resistance; altered metabolic pathways can help cancer cells adapt to continuous drug exposure and develop resistant phenotypes. Here we report the metabolic alterations underlying the non-small cell lung cancer (NSCLC) cell lines resistant to the usual PI3K-mTOR inhibitor BEZ235. In this study, we identified that an increased unsaturation degree of lipid species is associated with increased plasma membrane fluidity in cells with the resistant phenotype and that fatty acid desaturase FADS2 mediates the acquisition of chemoresistance. Therefore, new studies focused on reversing drug resistance based on membrane lipid modifications should consider the contribution of desaturase activity.
Collapse
Affiliation(s)
- Marika Colombo
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Federico Passarelli
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Paola A. Corsetto
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Angela M. Rizzo
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Mirko Marabese
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Giulia De Simone
- Protein and Metabolite Biomarkers Unit, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Roberta Pastorelli
- Protein and Metabolite Biomarkers Unit, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| | - Massimo Broggini
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
- Correspondence: (M.B.); (L.B.)
| | - Laura Brunelli
- Protein and Metabolite Biomarkers Unit, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
- Correspondence: (M.B.); (L.B.)
| | - Elisa Caiola
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milan, Italy
| |
Collapse
|
205
|
Giannoni-Luza S, Acosta O, Murillo Carrasco AG, Danos P, Cotrina Concha JM, Miller HG, Pinto JA, Aguilar A, Araujo JM, Fujita R, Buleje J. Chip-based digital Polymerase Chain Reaction as quantitative technique for the detection of PIK3CA mutations in breast cancer patients. Heliyon 2022; 8:e11396. [DOI: 10.1016/j.heliyon.2022.e11396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 09/15/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
|
206
|
Oscorbin IP, Beginyazova OP, Khlistun IV, Shamovskaya DV, Oskina NA, Filipenko ML. Development of a multiplex allele-specific qPCR approach for testing PIK3CA mutations in patients with colorectal cancer. Heliyon 2022; 8:e11804. [DOI: 10.1016/j.heliyon.2022.e11804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 11/08/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022] Open
|
207
|
Becker J, Gross UC, Weber DM, Weibel L, Theiler M, Brandt S, Bode PK. PIK3CA Mutational Analysis in Patients With Macrodactyly. Pediatr Dev Pathol 2022; 25:624-634. [PMID: 36314082 DOI: 10.1177/10935266221080155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Somatic mosaicism for PIK3CA mutations causes various types of growth disorders, which have been summarized under the term PROS (PIK3CA related overgrowth spectrum). Targeted therapy with PI3K inhibitors seems to be a promising alternative for severe PROS cases. Therefore, PIK3CA testing may become more relevant in the future. METHODS We report on 14 PROS patients, who had surgery for macrodactyly in the majority of cases. Clinical data were retrieved from the patient's records. Macroscopic and microscopic findings were retrospectively reviewed. Mutational analysis was performed on formalin-fixed paraffin-embedded (FFPE) material. RESULTS Patient age ranged from 7 months to 35 years. Five patients showed additional anomalies. One patient had CLOVES syndrome. The majority of the specimens were ray resections characterized by hypertrophic fat tissue. Overall, microscopy was subtle. The abnormal adipose tissue showed lobules exhibiting at least focally fibrous septa. In each case, we could detect a PIK3CA mutation. CONCLUSION Histology of affected fat tissue in PROS patients is overall nonspecific. Therefore, mutational analysis represents the key to the diagnosis, especially in unclear clinical cases. We demonstrated that FFPE material is suitable for PIK3CA testing, which can be considered as basis for targeted therapy with PI3K inhibitors.
Collapse
Affiliation(s)
- Jakob Becker
- Department of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland
| | - Ulrike Camenisch Gross
- Department of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland
| | - Daniel M Weber
- Division of Hand Surgery, Department of Pediatric Surgery, 30995University Children's Hospital Zürich, Zürich, Switzerland
| | - Lisa Weibel
- Pediatric Skin Center, Department of Dermatology, University Children's Hospital Zürich, Zürich, Switzerland
| | - Martin Theiler
- Pediatric Skin Center, Department of Dermatology, University Children's Hospital Zürich, Zürich, Switzerland
| | - Simone Brandt
- Department of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland.,Institute of Pathology Medica, Zürich, Switzerland
| | - Peter K Bode
- Department of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
208
|
Chen JW, Murugesan K, Newberg JY, Sokol ES, Savage HM, Stout TJ, Maund SL, Hutchinson KE. Comparison of PIK3CA Mutation Prevalence in Breast Cancer Across Predicted Ancestry Populations. JCO Precis Oncol 2022; 6:e2200341. [PMID: 36446041 PMCID: PMC9812634 DOI: 10.1200/po.22.00341] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/31/2022] [Accepted: 10/11/2022] [Indexed: 11/30/2022] Open
Abstract
PURPOSE Understanding the differences in biomarker prevalence that may exist among diverse populations is invaluable to accurately forecast biomarker-driven clinical trial enrollment metrics and to advance inclusive research and health equity. This study evaluated the frequency and types of PIK3CA mutations (PIK3CAmut) detected in predicted genetic ancestry subgroups across breast cancer (BC) subtypes. METHODS Analyses were conducted using real-world genomic data from adult patients with BC treated in an academic or community setting in the United States and whose tumor tissue was submitted for comprehensive genomic profiling. RESULTS Of 36,151 patients with BC (median age, 58 years; 99% female), the breakdown by predicted genetic ancestry was 75% European, 14% African, 6% Central/South American, 3% East Asian, and 1% South Asian. We demonstrated that patients of African ancestry are less likely to have tumors that harbor PIK3CAmut compared with patients of European ancestry with estrogen receptor-positive/human epidermal growth factor receptor 2-negative (ER+/HER2-) BC (37% [949/2,593] v 44% [7,706/17,637]; q = 4.39E-11) and triple-negative breast cancer (8% [179/2,199] v 14% [991/7,072]; q = 6.07E-13). Moreover, we found that PIK3CAmut were predominantly composed of hotspot mutations, of which mutations at H1047 were the most prevalent across BC subtypes (35%-41% ER+/HER2- BC; 43%-61% HER2+ BC; 40%-59% triple-negative breast cancer). CONCLUSION This analysis established that tumor PIK3CAmut prevalence can differ among predicted genetic ancestries across BC subtypes on the basis of the largest comprehensive genomic profiling data set of patients with cancer treated in the United States. This study highlights the need for equitable representation in research studies, which is imperative to ensuring better health outcomes for all.
Collapse
Affiliation(s)
- Jessica W. Chen
- Oncology Biomarker Development, Genentech, Inc, South San Francisco, CA
| | | | | | - Ethan S. Sokol
- Cancer Genomics Research, Foundation Medicine, Inc, Cambridge, MA
| | - Heidi M. Savage
- Oncology Biomarker Development, Genentech, Inc, South San Francisco, CA
| | - Thomas J. Stout
- Product Development Oncology, Genentech, Inc, South San Francisco, CA
| | - Sophia L. Maund
- Oncology Biomarker Development, Genentech, Inc, South San Francisco, CA
| | | |
Collapse
|
209
|
Expression of AKT1 Related with Clinicopathological Parameters in Clear Cell Renal Cell Carcinoma. Curr Issues Mol Biol 2022; 44:4921-4929. [PMID: 36286049 PMCID: PMC9600081 DOI: 10.3390/cimb44100334] [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: 09/30/2022] [Revised: 10/14/2022] [Accepted: 10/14/2022] [Indexed: 11/22/2022] Open
Abstract
Pathways such as VEGF, EGF and mTOR are known to be one of the major mechanisms of tumorigenesis including kidney cancer. To identify potential signaling pathway proteins, we performed differential/correlation analyses of mTOR-associated genes from three public datasets. AKT1 protein, one of the PI3K/AKT/mTOR pathways, turned out to be the potential by showing a consistent discrepancy between ccRCC-associated conditions as well as strong correlation with other mTOR-associated genes across the datasets. Then, we analyzed how AKT1 alteration affects clear cell renal cell carcinoma. The pathology of 58 kidney cancer patients was constructed to analyze the relationship between the expression level of AKT1 through immunohistochemical staining and their clinicopathological data. Gender, age and TNM stage did not show significant results. AKT1 is a known oncogene. However, in this study, high expression of AKT1 showed a slight correlation with lower WHO/ISUP grade, longer recurrence-free and progression-free survival rates.
Collapse
|
210
|
Fang H, Li H, Zhang H, Wang S, Xu S, Chang L, Yang Y, Cui R. Short-chain L-3-hydroxyacyl-CoA dehydrogenase: A novel vital oncogene or tumor suppressor gene in cancers. Front Pharmacol 2022; 13:1019312. [PMID: 36313354 PMCID: PMC9614034 DOI: 10.3389/fphar.2022.1019312] [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: 08/15/2022] [Accepted: 09/23/2022] [Indexed: 08/22/2023] Open
Abstract
The reprogramming of cellular metabolism is frequently linked to tumorigenesis. Glucose, fatty acids, and amino acids are the specific substrates involved in how an organism maintains metabolic equilibrium. The HADH gene codes for the short-chain L-3-hydroxyacyl-CoA dehydrogenase (HADH), a crucial enzyme in fatty acid oxidation that catalyzes the third phase of fatty acid oxidation in mitochondria. Increasing data suggest that HADH is differentially expressed in various types of malignancies and is linked to cancer development and progression. The significance of HADH expression in tumors and its potential mechanisms of action in the onset and progression of certain cancers are summarized in this article. The possible roles of HADH as a target and/or biomarker for the detection and treatment of various malignancies is also described here.
Collapse
Affiliation(s)
- He Fang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Hanyang Li
- Department of Thyroid Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Hang Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Shu Wang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Shuang Xu
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, China
| | - Li Chang
- Department of Pathology, The Second Hospital of Jilin University, Changchun, China
| | - Yongsheng Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
211
|
Transcription profiling of feline mammary carcinomas and derived cell lines reveals biomarkers and drug targets associated with metabolic and cell cycle pathways. Sci Rep 2022; 12:17025. [PMID: 36220861 PMCID: PMC9553959 DOI: 10.1038/s41598-022-20874-5] [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: 06/16/2022] [Accepted: 09/20/2022] [Indexed: 12/29/2022] Open
Abstract
The molecular heterogeneity of feline mammary carcinomas (FMCs) represents a prognostic and therapeutic challenge. RNA-Seq-based comparative transcriptomic profiling serves to identify recurrent and exclusive differentially expressed genes (DEGs) across sample types and molecular subtypes. Using mass-parallel RNA-Seq, we identified DEGs and performed comparative function-based analysis across 15 tumours (four basal-like triple-negative [TN], eight normal-like TN, and three luminal B fHER2 negative [LB fHER2-]), two cell lines (CL, TiHo-0906, and TiHo-1403) isolated from the primary tumours (LB fHER2-) of two cats included in this study, and 13 healthy mammary tissue controls. DEGs in tumours were predominantly upregulated; dysregulation of CLs transcriptome was more extensive, including mostly downregulated genes. Cell-cycle and metabolic-related DEGs were upregulated in both tumours and CLs, including therapeutically-targetable cell cycle regulators (e.g. CCNB1, CCNB2, CDK1, CDK4, GTSE1, MCM4, and MCM5), metabolic-related genes (e.g. FADS2 and SLC16A3), heat-shock proteins (e.g. HSPH1, HSP90B1, and HSPA5), genes controlling centrosome disjunction (e.g. RACGAP1 and NEK2), and collagen molecules (e.g. COL2A1). DEGs specifically upregulated in basal-like TN tumours were involved in antigen processing and presentation, in normal-like TN tumours encoded G protein-coupled receptors (GPCRs), and in LB fHER2- tumours were associated with lysosomes, phagosomes, and endosomes formation. Downregulated DEGs in CLs were associated with structural and signalling cell surface components. Hence, our results suggest that upregulation of genes enhancing proliferation and metabolism is a common feature among FMCs and derived CLs. In contrast, the dissimilarities observed in dysregulation of membrane components highlight CLs' disconnection with the tumour microenvironment. Furthermore, recurrent and exclusive DEGs associated with dysregulated pathways might be useful for the development of prognostically and therapeutically-relevant targeted panels.
Collapse
|
212
|
Lei ZN, Teng QX, Tian Q, Chen W, Xie Y, Wu K, Zeng Q, Zeng L, Pan Y, Chen ZS, He Y. Signaling pathways and therapeutic interventions in gastric cancer. Signal Transduct Target Ther 2022; 7:358. [PMID: 36209270 PMCID: PMC9547882 DOI: 10.1038/s41392-022-01190-w] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/14/2022] [Accepted: 09/07/2022] [Indexed: 11/23/2022] Open
Abstract
Gastric cancer (GC) ranks fifth in global cancer diagnosis and fourth in cancer-related death. Despite tremendous progress in diagnosis and therapeutic strategies and significant improvements in patient survival, the low malignancy stage is relatively asymptomatic and many GC cases are diagnosed at advanced stages, which leads to unsatisfactory prognosis and high recurrence rates. With the recent advances in genome analysis, biomarkers have been identified that have clinical importance for GC diagnosis, treatment, and prognosis. Modern molecular classifications have uncovered the vital roles that signaling pathways, including EGFR/HER2, p53, PI3K, immune checkpoint pathways, and cell adhesion signaling molecules, play in GC tumorigenesis, progression, metastasis, and therapeutic responsiveness. These biomarkers and molecular classifications open the way for more precise diagnoses and treatments for GC patients. Nevertheless, the relative significance, temporal activation, interaction with GC risk factors, and crosstalk between these signaling pathways in GC are not well understood. Here, we review the regulatory roles of signaling pathways in GC potential biomarkers, and therapeutic targets with an emphasis on recent discoveries. Current therapies, including signaling-based and immunotherapies exploited in the past decade, and the development of treatment for GC, particularly the challenges in developing precision medications, are discussed. These advances provide a direction for the integration of clinical, molecular, and genomic profiles to improve GC diagnosis and treatments.
Collapse
Affiliation(s)
- Zi-Ning Lei
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Qin Tian
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China
| | - Wei Chen
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China
| | - Yuhao Xie
- Institute for Biotechnology, St. John's University, Queens, NY, 11439, USA
| | - Kaiming Wu
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China
| | - Qianlin Zeng
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China
| | - Leli Zeng
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China.
| | - Yihang Pan
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
- Institute for Biotechnology, St. John's University, Queens, NY, 11439, USA.
| | - Yulong He
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China.
| |
Collapse
|
213
|
Brinzan CS, Aschie M, Cozaru GC, Deacu M, Dumitru E, Burlacu I, Mitroi A. KRAS, NRAS, BRAF, PIK3CA, and AKT1 signatures in colorectal cancer patients in south-eastern Romania. Medicine (Baltimore) 2022; 101:e30979. [PMID: 36221415 PMCID: PMC9542653 DOI: 10.1097/md.0000000000030979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Somatic mutations in the oncogenes of the epidermal growth factor receptor signaling pathway play vital roles in colorectal carcinogenesis and have been closely linked with clinical resistance to monoclonal therapy. In this study, we have analyzed the mutation frequencies of 5 genes and compared the genetic findings with clinicopathological variables in order to determine diagnostically relevant alterations and compare these findings with those of other studies In our Sanger sequencings, KRAS (exons 2, 3, and 4), NRAS (exons 2, 3, and 4), PIK3CA (exons 9 and 20), BRAF (exon 15), AKT1 (exon 2) genes, and microsatellite instability (MSI) status were analyzed using an ABI 3500 analyzer in a cohort of 58 Romanian colorectal cancer (CRC) patients who underwent surgical resection at Emergency County Clinical Hospital in Constanța, Romania. In our series, mutation rates of KRAS, BRAF, PIK3CA, and AKT1 genes were 39.63%, 8.62%, 6.88%, and 3.44%, respectively. By contrast, we did not find any tumor harboring mutation in the NRAS gene. Notably, the KRAS and PIK3CA mutations were not mutually exclusive, 1 patient harbored 2 mutations in exon2, codon 12 (Gly12Val) of KRAS and exon 20, codon 1047 (His1047Arg) of PIK3CA. The finding of our study are generally consistent with data found in the literature. Regarding to clinicopathological variables, mutation of KRAS was associated with distant metastasis at the time of diagnosis, while mutation of BRAF was significantly associated with MSI-H in contrast with MSI-L/MSS tumors. Moreover, PIK3CA mutation tends to be located in the proximal segment of the colon and to be well/moderately differentiated compared to wild-type tumors. In conclusion, the assessment of these mutations suggests that CRC patients from southeast Romania exhibit a mutation profile similar to other populations. These results could contribute to creating a better method of qualifying patients for molecularly targeted therapies and obtaining better screening strategies.
Collapse
Affiliation(s)
- Costel Stelian Brinzan
- Pathology Department, Sf. Apostol Andrei Clinical Emergency County Hospital, Constanta, Romania
- CEDMOG Center, Ovidius University, Constanta, Romania
| | - Mariana Aschie
- Pathology Department, Sf. Apostol Andrei Clinical Emergency County Hospital, Constanta, Romania
- CEDMOG Center, Ovidius University, Constanta, Romania
- Faculty of Medicine, Ovidius University, Constanta, Romania
| | - Georgeta Camelia Cozaru
- Pathology Department, Sf. Apostol Andrei Clinical Emergency County Hospital, Constanta, Romania
- CEDMOG Center, Ovidius University, Constanta, Romania
| | - Mariana Deacu
- Pathology Department, Sf. Apostol Andrei Clinical Emergency County Hospital, Constanta, Romania
- Faculty of Medicine, Ovidius University, Constanta, Romania
| | - Eugen Dumitru
- CEDMOG Center, Ovidius University, Constanta, Romania
- Faculty of Medicine, Ovidius University, Constanta, Romania
| | - Ionut Burlacu
- Pathology Department, Sf. Apostol Andrei Clinical Emergency County Hospital, Constanta, Romania
| | - Anca Mitroi
- Pathology Department, Sf. Apostol Andrei Clinical Emergency County Hospital, Constanta, Romania
- CEDMOG Center, Ovidius University, Constanta, Romania
- *Correspondence: Anca Mitroi, Pathology Department, Sf. Apostol Andrei Clinical Emergency County Hospital, 145 Tomis Blvd, Constanta 900591, Romania (e-mail: )
| |
Collapse
|
214
|
Signaling pathways and targeted therapies in lung squamous cell carcinoma: mechanisms and clinical trials. Signal Transduct Target Ther 2022; 7:353. [PMID: 36198685 PMCID: PMC9535022 DOI: 10.1038/s41392-022-01200-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/03/2022] [Accepted: 09/18/2022] [Indexed: 11/08/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death across the world. Unlike lung adenocarcinoma, patients with lung squamous cell carcinoma (LSCC) have not benefitted from targeted therapies. Although immunotherapy has significantly improved cancer patients' outcomes, the relatively low response rate and severe adverse events hinder the clinical application of this promising treatment in LSCC. Therefore, it is of vital importance to have a better understanding of the mechanisms underlying the pathogenesis of LSCC as well as the inner connection among different signaling pathways, which will surely provide opportunities for more effective therapeutic interventions for LSCC. In this review, new insights were given about classical signaling pathways which have been proved in other cancer types but not in LSCC, including PI3K signaling pathway, VEGF/VEGFR signaling, and CDK4/6 pathway. Other signaling pathways which may have therapeutic potentials in LSCC were also discussed, including the FGFR1 pathway, EGFR pathway, and KEAP1/NRF2 pathway. Next, chromosome 3q, which harbors two key squamous differentiation markers SOX2 and TP63 is discussed as well as its related potential therapeutic targets. We also provided some progress of LSCC in epigenetic therapies and immune checkpoints blockade (ICB) therapies. Subsequently, we outlined some combination strategies of ICB therapies and other targeted therapies. Finally, prospects and challenges were given related to the exploration and application of novel therapeutic strategies for LSCC.
Collapse
|
215
|
Lv W, Du C, Zhang Y, Wu F, Jin Y, Chen X, Liu X, Feng C, Ma X, Zhang S. Clinicopathological characteristics and prognostic analysis of PIK3CA mutation in breast cancer patients in Northwest China. Pathol Res Pract 2022; 238:154063. [PMID: 35994807 DOI: 10.1016/j.prp.2022.154063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 07/31/2022] [Accepted: 08/07/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE Multiple studies on PIK3CA mutations in breast cancer (BC) had been performed, which showed the controversial results among different countries and races even those from the same country. The present study aimed to explore the PIK3CA gene mutation status in BC patients in Northwest China and reveal the relationship between PIK3CA mutations and clinicopathological features along with prognosis. MATERIALS AND METHODS 1002 BC patients from Northwest China were recruited in this study, genomic DNA was extracted from formalin-fixed paraffin-embedded (FFPE) tumor tissues, and hotspot mutations in the exon 9 and 20 of PIK3CA gene were detected by ARMS-PCR. RESULTS PIK3CA mutations were found in 31.2% (313/1002) of BC patients, among them 66.1% were mutations in exon 20% and 32.6% were mutations in exon 9. H1047R was the most common mutation type, accounting for 56.5% of the total mutated samples. Significant correlations were observed between PIK3CA mutation status and age (P = 0.035), histopathologic types (P = 0.004), pathological grade (P = 0.013), ER positivity (P < 0.001), PR positivity (P < 0.001), molecular subtypes (P = 0.004) and family history (P = 0.007). Cox multivariate analysis showed that patients with mutations in exon 9 or 20 had shorter DFS and OS than wild-type patients. Those with exon 9 mutations subgroup had the worst prognosis. Interestingly, patients with H1047L mutation had the best prognosis than others. CONCLUSION PIK3CA mutations could be used as an indicator of clinical outcome or targeted therapy for multiple breast cancer subgroups in Northwest China.
Collapse
Affiliation(s)
- Wei Lv
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| | - Chong Du
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| | - Yinbing Zhang
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| | - Fei Wu
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| | - Yaofeng Jin
- Department of Pathology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| | - Xi Chen
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| | - Xuan Liu
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| | - Cong Feng
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China
| | - Xingcong Ma
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China.
| | - Shuqun Zhang
- Department of Oncology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province 710004, China.
| |
Collapse
|
216
|
Minematsu H, Afify SM, Sugihara Y, Hassan G, Zahra MH, Seno A, Adachi M, Seno M. Cancer stem cells induced by chronic stimulation with prostaglandin E2 exhibited constitutively activated PI3K axis. Sci Rep 2022; 12:15628. [PMID: 36115905 PMCID: PMC9482612 DOI: 10.1038/s41598-022-19265-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 08/26/2022] [Indexed: 12/04/2022] Open
Abstract
Previously, our group has demonstrated establishment of Cancer Stem Cell (CSC) models from stem cells in the presence of conditioned medium of cancer cell lines. In this study, we tried to identify the factors responsible for the induction of CSCs. Since we found the lipid composition could be traced to arachidonic acid cascade in the CSC model, we assessed prostaglandin E2 (PGE2) as a candidate for the ability to induce CSCs from induced pluripotent stem cells (iPSCs). Mouse iPSCs acquired the characteristics of CSCs in the presence of 10 ng/mL of PGE2 after 4 weeks. Since constitutive Akt activation and pik3cg overexpression were found in the resultant CSCs, of which growth was found independent of PGE2, chronic stimulation of the receptors EP-2/4 by PGE2 was supposed to induce CSCs from iPSCs through epigenetic effect. The bioinformatics analysis of the next generation sequence data of the obtained CSCs proposed not only receptor tyrosine kinase activation by growth factors but also extracellular matrix and focal adhesion enhanced PI3K pathway. Collectively, chronic stimulation of stem cells with PGE2 was implied responsible for cancer initiation enhancing PI3K/Akt axis.
Collapse
|
217
|
Chaves-Almagro C, Auriau J, Dortignac A, Clerc P, Lulka H, Deleruyelle S, Projetti F, Nakhlé J, Frances A, Berta J, Gigoux V, Fourmy D, Dufresne M, Gomez-Brouchet A, Guillermet-Guibert J, Cordelier P, Knibiehler B, Jockers R, Valet P, Audigier Y, Masri B. Upregulated Apelin Signaling in Pancreatic Cancer Activates Oncogenic Signaling Pathways to Promote Tumor Development. Int J Mol Sci 2022; 23:ijms231810600. [PMID: 36142542 PMCID: PMC9503500 DOI: 10.3390/ijms231810600] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Despite decades of effort in understanding pancreatic ductal adenocarcinoma (PDAC), there is still a lack of innovative targeted therapies for this devastating disease. Herein, we report the expression of apelin and its receptor, APJ, in human pancreatic adenocarcinoma and its protumoral function. Apelin and APJ protein expression in tumor tissues from patients with PDAC and their spatiotemporal pattern of expression in engineered mouse models of PDAC were investigated by immunohistochemistry. Apelin signaling function in tumor cells was characterized in pancreatic tumor cell lines by Western blot as well as proliferation, migration assays and in murine orthotopic xenograft experiments. In premalignant lesions, apelin was expressed in epithelial lesions whereas APJ was found in isolated cells tightly attached to premalignant lesions. However, in the invasive stage, apelin and APJ were co-expressed by tumor cells. In human tumor cells, apelin induced a long-lasting activation of PI3K/Akt, upregulated β-catenin and the oncogenes c-myc and cyclin D1 and promoted proliferation, migration and glucose uptake. Apelin receptor blockades reduced cancer cell proliferation along with a reduction in pancreatic tumor burden. These findings identify the apelin signaling pathway as a new actor for PDAC development and a novel therapeutic target for this incurable disease.
Collapse
Affiliation(s)
- Carline Chaves-Almagro
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048, Université de Toulouse, UPS, Toulouse III, 31432 Toulouse, France
| | - Johanna Auriau
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048, Université de Toulouse, UPS, Toulouse III, 31432 Toulouse, France
| | - Alizée Dortignac
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048, Université de Toulouse, UPS, Toulouse III, 31432 Toulouse, France
| | - Pascal Clerc
- INSERM ERL1226, CNRS UMR 5215, Université de Toulouse, UPS, Toulouse III, 31432 Toulouse, France
| | - Hubert Lulka
- Centre de Recherches en Cancérologie de Toulouse, INSERM, CNRS, Université Paul Sabatier, Université de Toulouse, 31037 Toulouse, France
| | - Simon Deleruyelle
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048, Université de Toulouse, UPS, Toulouse III, 31432 Toulouse, France
| | | | - Jessica Nakhlé
- Centre de Recherches en Cancérologie de Toulouse, INSERM, CNRS, Université Paul Sabatier, Université de Toulouse, 31037 Toulouse, France
| | - Audrey Frances
- Centre de Recherches en Cancérologie de Toulouse, INSERM, CNRS, Université Paul Sabatier, Université de Toulouse, 31037 Toulouse, France
| | - Judit Berta
- Department of Tumor Biology, National Koranyi Institute of Pulmonology, 1121 Budapest, Hungary
| | - Véronique Gigoux
- INSERM ERL1226, CNRS UMR 5215, Université de Toulouse, UPS, Toulouse III, 31432 Toulouse, France
| | - Daniel Fourmy
- INSERM ERL1226, CNRS UMR 5215, Université de Toulouse, UPS, Toulouse III, 31432 Toulouse, France
| | - Marlène Dufresne
- Centre de Recherches en Cancérologie de Toulouse, INSERM, CNRS, Université Paul Sabatier, Université de Toulouse, 31037 Toulouse, France
| | | | - Julie Guillermet-Guibert
- Centre de Recherches en Cancérologie de Toulouse, INSERM, CNRS, Université Paul Sabatier, Université de Toulouse, 31037 Toulouse, France
| | - Pierre Cordelier
- Centre de Recherches en Cancérologie de Toulouse, INSERM, CNRS, Université Paul Sabatier, Université de Toulouse, 31037 Toulouse, France
| | - Bernard Knibiehler
- Centre de Recherches en Cancérologie de Toulouse, INSERM, CNRS, Université Paul Sabatier, Université de Toulouse, 31037 Toulouse, France
| | - Ralf Jockers
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Cité, 75014 Paris, France
| | - Philippe Valet
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048, Université de Toulouse, UPS, Toulouse III, 31432 Toulouse, France
- RESTORE, UMR 1301-Inserm 5070-CNRS EFS, Université de Toulouse, 31100 Toulouse, France
| | - Yves Audigier
- Centre de Recherches en Cancérologie de Toulouse, INSERM, CNRS, Université Paul Sabatier, Université de Toulouse, 31037 Toulouse, France
| | - Bernard Masri
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048, Université de Toulouse, UPS, Toulouse III, 31432 Toulouse, France
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Cité, 75014 Paris, France
- Correspondence: ; Tel.: +33-1-40-51-64-87
| |
Collapse
|
218
|
Detection of breast cancer-related point-mutations using screen-printed and gold-plated electrochemical sensor arrays suitable for point-of-care applications. TALANTA OPEN 2022. [DOI: 10.1016/j.talo.2022.100150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
219
|
Sharma V, Kumar D, Kumar S, Singh H, Sharma N, Gupta S. Impact of tobacco smoking on oral cancer genetics-A next-generation sequencing perspective. IMETA 2022; 1:e44. [PMID: 38868711 PMCID: PMC10989814 DOI: 10.1002/imt2.44] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 06/22/2022] [Accepted: 07/16/2022] [Indexed: 06/14/2024]
Abstract
This study identified a total of 28 genetic loci (comprising 31 genes), which were found to be altered in oral cancer patients having a habit of tobacco smoking. Three loci, that is, 17p13.1 (TP53), 9p21.3 (CDKN2A), and 9q34.3 (NOTCH1) were found to be modified and common in three records whereas one locus, that is, 3q26.32 (PIK3CA) was found to be modified and common in two records. This study suggests that oral cancer patients should be categorized into different subgroups based on (i) genetic signatures, and (ii) smoking status, then the treatment strategies for each group should be precisely defined.
Collapse
Affiliation(s)
- Vishwas Sharma
- Division of CytopathologyICMR—National Institute of Cancer Prevention and ResearchNoidaUttar PradeshIndia
| | - Dinesh Kumar
- WHO FCTC Global Knowledge Hub on Smokeless TobaccoICMR‐ National Institute of Cancer Prevention and ResearchNoidaUttar PradeshIndia
| | - Shravan Kumar
- WHO FCTC Global Knowledge Hub on Smokeless TobaccoICMR‐ National Institute of Cancer Prevention and ResearchNoidaUttar PradeshIndia
| | - Harpreet Singh
- Indian Council of Medical ResearchInformatics, Systems & Research Management (ISRM)New DelhiIndia
| | - Naveen Sharma
- Biomedical Informatics DivisionIndian Council of Medical ResearchNew DelhiIndia
| | - Sanjay Gupta
- Division of CytopathologyICMR—National Institute of Cancer Prevention and ResearchNoidaUttar PradeshIndia
| |
Collapse
|
220
|
Liu X, Deng Y, Huang Y, Ye J, Xie S, He Q, Chen Y, Lin Y, Liang R, Wei J, Li Y, Zhang J. Nasopharyngeal Carcinoma Progression: Accumulating Genomic Instability and Persistent Epstein–Barr Virus Infection. Curr Oncol 2022; 29:6035-6052. [PMID: 36135044 PMCID: PMC9498130 DOI: 10.3390/curroncol29090475] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/09/2022] [Accepted: 08/19/2022] [Indexed: 12/01/2022] Open
Abstract
Genomic instability facilitates the evolution of cells, tissues, organs, and species. The progression of human malignancies can be regarded as the accumulation of genomic instability, which confers a high evolutionary potential for tumor cells to adapt to continuous changes in the tumor microenvironment. Nasopharyngeal carcinoma (NPC) is a head-and-neck squamous-cell carcinoma closely associated with Epstein–Barr virus (EBV) infection. NPC progression is driven by a combination of accumulated genomic instability and persistent EBV infection. Here, we present a review of the key characteristics of genomic instability in NPC and the profound implications of EBV infection. We further discuss the significance of profiling genomic instability for the assessment of disease progression and treatment efficacy, as well as the opportunities and challenges of targeted therapies for NPC based on its unique genomic instability.
Collapse
Affiliation(s)
- Xue Liu
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning 530021, China
| | - Yayan Deng
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning 530021, China
| | - Yujuan Huang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning 530021, China
| | - Jiaxiang Ye
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning 530021, China
| | - Sifang Xie
- Department of Otolaryngology & Head and Neck, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, 6 Taoyuan Road, Nanning 530021, China
| | - Qian He
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Yong Chen
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning 530021, China
| | - Yan Lin
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning 530021, China
| | - Rong Liang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning 530021, China
| | - Jiazhang Wei
- Department of Otolaryngology & Head and Neck, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, 6 Taoyuan Road, Nanning 530021, China
| | - Yongqiang Li
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning 530021, China
- Correspondence: (Y.L.); (J.Z.)
| | - Jinyan Zhang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, 71 Hedi Road, Nanning 530021, China
- Correspondence: (Y.L.); (J.Z.)
| |
Collapse
|
221
|
González-Moles MÁ, Warnakulasuriya S, López-Ansio M, Ramos-García P. Hallmarks of Cancer Applied to Oral and Oropharyngeal Carcinogenesis: A Scoping Review of the Evidence Gaps Found in Published Systematic Reviews. Cancers (Basel) 2022; 14:3834. [PMID: 35954497 PMCID: PMC9367256 DOI: 10.3390/cancers14153834] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/04/2022] [Accepted: 08/04/2022] [Indexed: 02/07/2023] Open
Abstract
In 2000 and 2011, Hanahan and Weinberg published two papers in which they defined the characteristics that cells must fulfil in order to be considered neoplastic cells in all types of tumours that affect humans, which the authors called "hallmarks of cancer". These papers have represented a milestone in our understanding of the biology of many types of cancers and have made it possible to reach high levels of scientific evidence in relation to the prognostic impact that these hallmarks have on different tumour types. However, to date, there is no study that globally analyses evidence-based knowledge on the importance of these hallmarks in oral and oropharyngeal squamous cell carcinomas. For this reason, we set out to conduct this scoping review of systematic reviews with the aim of detecting evidence gaps in relation to the relevance of the cancer hallmarks proposed by Hanahan and Weinberg in oral and oropharyngeal cancer, and oral potentially malignant disorders, and to point out future lines of research in this field.
Collapse
Affiliation(s)
- Miguel Ángel González-Moles
- School of Dentistry, University of Granada, 18011 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Saman Warnakulasuriya
- Faculty of Dentistry, Oral and Craniofacial Sciences, King’s College London, London SE1 9RT, UK
- WHO Collaborating for Oral Cancer, King’s College London, London SE1 9RT, UK
| | - María López-Ansio
- School of Dentistry, University of Granada, 18011 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Pablo Ramos-García
- School of Dentistry, University of Granada, 18011 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| |
Collapse
|
222
|
Caliceti C, Punzo A, Silla A, Simoni P, Roda G, Hrelia S. New Insights into Bile Acids Related Signaling Pathways in the Onset of Colorectal Cancer. Nutrients 2022; 14:nu14142964. [PMID: 35889921 PMCID: PMC9317521 DOI: 10.3390/nu14142964] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/12/2022] [Accepted: 07/19/2022] [Indexed: 02/01/2023] Open
Abstract
Colorectal cancer (CRC) ranks as the second among the causes of tumor death worldwide, with an estimation of 1.9 million new cases in 2020 and more than 900,000 deaths. This rate might increase by 60% over the next 10 years. These data are unacceptable considering that CRC could be successfully treated if diagnosed in the early stages. A high-fat diet promotes the hepatic synthesis of bile acids (BAs) increasing their delivery to the colonic lumen and numerous scientific reports correlate BAs, especially secondary BAs, with CRC incidence. We reviewed the physicochemical and biological characteristics of BAs, focusing on the major pathways involved in CRC risk and progression. We specifically pointed out the role of BAs as signaling molecules and the tangled relationships among their nuclear and membrane receptors with the big bang of molecular and cellular events that trigger CRC occurrence.
Collapse
Affiliation(s)
- Cristiana Caliceti
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
- Interdepartmental Centre for Renewable Sources, Environment, Sea and Energy (CIRI FRAME), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
- Biostructures and Biosystems National Institute (INBB), 00136 Rome, Italy;
- Correspondence:
| | - Angela Punzo
- Department of Chemistry “Giacomo Ciamician” Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy;
| | - Alessia Silla
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy; (A.S.); (S.H.)
| | - Patrizia Simoni
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy;
| | - Giulia Roda
- Biostructures and Biosystems National Institute (INBB), 00136 Rome, Italy;
| | - Silvana Hrelia
- Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy; (A.S.); (S.H.)
| |
Collapse
|
223
|
Seol MY, Choi SH, Yoon HI. Combining radiation with PI3K isoform-selective inhibitor administration increases radiosensitivity and suppresses tumor growth in non-small cell lung cancer. JOURNAL OF RADIATION RESEARCH 2022; 63:591-601. [PMID: 35536306 PMCID: PMC9303607 DOI: 10.1093/jrr/rrac018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/14/2022] [Indexed: 06/14/2023]
Abstract
Non-small cell lung cancer (NSCLC) is a malignant lung tumor with a dismal prognosis. The activation of the phosphoinositide 3-kinase (PI3K)/AKT signaling pathway is common in many tumor types including NSCLC, which results in radioresistance and changes in the tumor microenvironment. Although pan-PI3K inhibitors have been tested in clinical trials to overcome radioresistance, concerns regarding their excessive side effects led to the consideration of selective inhibition of PI3K isoforms. In this study, we assessed whether combining radiation with the administration of the PI3K isoform-selective inhibitors reduces radioresistance and tumor growth in NSCLC. Inhibition of the PI3K/AKT pathway enhanced radiosensitivity substantially, and PI3K-α inhibitor showed superior radiosensitizing effect similar to PI3K pan-inhibitor, both in vitro and in vivo. Additionally, a significant increase in DNA double-strand breaks (DSB) and a decrease in migration ability were observed. Our study revealed that combining radiation and the PI3K-α isoform improved radiosensitivity that resulted in a significant delay in tumor growth and improved survival rate.
Collapse
Affiliation(s)
- Mi Youn Seol
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seo Hee Choi
- Department of Radiation Oncology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Gyeonggi-do, 16995, Republic of Korea
| | - Hong In Yoon
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| |
Collapse
|
224
|
Shaheen MF, Tse JY, Sokol ES, Masterson M, Bansal P, Rabinowitz I, Tarleton CA, Dobroff AS, Smith TL, Bocklage TJ, Mannakee BK, Gutenkunst RN, Bischoff J, Ness SA, Riedlinger GM, Groisberg R, Pasqualini R, Ganesan S, Arap W. Genomic landscape of lymphatic malformations: a case series and response to the PI3Kα inhibitor alpelisib in an N-of-1 clinical trial. eLife 2022; 11:e74510. [PMID: 35787784 PMCID: PMC9255965 DOI: 10.7554/elife.74510] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background Lymphatic malformations (LMs) often pose treatment challenges due to a large size or a critical location that could lead to disfigurement, and there are no standardized treatment approaches for either refractory or unresectable cases. Methods We examined the genomic landscape of a patient cohort of LMs (n = 30 cases) that underwent comprehensive genomic profiling using a large-panel next-generation sequencing assay. Immunohistochemical analyses were completed in parallel. Results These LMs had low mutational burden with hotspot PIK3CA mutations (n = 20) and NRAS (n = 5) mutations being most frequent, and mutually exclusive. All LM cases with Kaposi sarcoma-like (kaposiform) histology had NRAS mutations. One index patient presented with subacute abdominal pain and was diagnosed with a large retroperitoneal LM harboring a somatic PIK3CA gain-of-function mutation (H1047R). The patient achieved a rapid and durable radiologic complete response, as defined in RECIST1.1, to the PI3Kα inhibitor alpelisib within the context of a personalized N-of-1 clinical trial (NCT03941782). In translational correlative studies, canonical PI3Kα pathway activation was confirmed by immunohistochemistry and human LM-derived lymphatic endothelial cells carrying an allele with an activating mutation at the same locus were sensitive to alpelisib treatment in vitro, which was demonstrated by a concentration-dependent drop in measurable impedance, an assessment of cell status. Conclusions Our findings establish that LM patients with conventional or kaposiform histology have distinct, yet targetable, driver mutations. Funding R.P. and W.A. are supported by awards from the Levy-Longenbaugh Fund. S.G. is supported by awards from the Hugs for Brady Foundation. This work has been funded in part by the NCI Cancer Center Support Grants (CCSG; P30) to the University of Arizona Cancer Center (CA023074), the University of New Mexico Comprehensive Cancer Center (CA118100), and the Rutgers Cancer Institute of New Jersey (CA072720). B.K.M. was supported by National Science Foundation via Graduate Research Fellowship DGE-1143953. Clinical trial number NCT03941782.
Collapse
Affiliation(s)
- Montaser F Shaheen
- University of Arizona Cancer CenterTucsonUnited States
- Division of Hematology/Oncology, Department of Medicine, University of Arizona College of MedicineTucsonUnited States
| | - Julie Y Tse
- Foundation Medicine, IncCambridgeUnited States
| | | | - Margaret Masterson
- Rutgers Cancer Institute of New JerseyNew BrunswickUnited States
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical SchoolNew BrunswickUnited States
| | - Pranshu Bansal
- University of New Mexico Comprehensive Cancer CenterAlbuquerqueUnited States
- Division of Hematology/Oncology, Department of Internal Medicine, University of New Mexico School of MedicineAlbuquerqueUnited States
| | - Ian Rabinowitz
- University of New Mexico Comprehensive Cancer CenterAlbuquerqueUnited States
- Division of Hematology/Oncology, Department of Internal Medicine, University of New Mexico School of MedicineAlbuquerqueUnited States
| | - Christy A Tarleton
- University of New Mexico Comprehensive Cancer CenterAlbuquerqueUnited States
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico School of MedicineAlbuquerqueUnited States
| | - Andrey S Dobroff
- University of New Mexico Comprehensive Cancer CenterAlbuquerqueUnited States
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico School of MedicineAlbuquerqueUnited States
| | - Tracey L Smith
- Rutgers Cancer Institute of New JerseyNewarkUnited States
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical SchoolNewarkUnited States
| | - Thèrése J Bocklage
- University of New Mexico Comprehensive Cancer CenterAlbuquerqueUnited States
- Department of Pathology, University of Kentucky College of Medicine and Markey Cancer CenterLexingtonUnited States
| | - Brian K Mannakee
- University of Arizona Cancer CenterTucsonUnited States
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of ArizonaTucsonUnited States
| | - Ryan N Gutenkunst
- University of Arizona Cancer CenterTucsonUnited States
- Department of Molecular and Cellular Biology, College of Science, University of ArizonaTucsonUnited States
| | - Joyce Bischoff
- Vascular Biology Program, Boston Children’s HospitalBostonUnited States
- Department of Surgery, Harvard Medical SchoolBostonUnited States
| | - Scott A Ness
- University of New Mexico Comprehensive Cancer CenterAlbuquerqueUnited States
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico School of MedicineAlbuquerqueUnited States
| | - Gregory M Riedlinger
- Rutgers Cancer Institute of New JerseyNew BrunswickUnited States
- Department of Pathology, Rutgers Robert Wood Johnson Medical SchoolNew BrunswickUnited States
| | - Roman Groisberg
- Rutgers Cancer Institute of New JerseyNew BrunswickUnited States
- Division of Medical Oncology, Department of Medicine, Rutgers Robert Wood Johnson Medical SchoolNew BrunswickUnited States
| | - Renata Pasqualini
- Rutgers Cancer Institute of New JerseyNewarkUnited States
- Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical SchoolNewarkUnited States
| | - Shridar Ganesan
- Rutgers Cancer Institute of New JerseyNew BrunswickUnited States
- Division of Medical Oncology, Department of Medicine, Rutgers Robert Wood Johnson Medical SchoolNew BrunswickUnited States
| | - Wadih Arap
- Rutgers Cancer Institute of New JerseyNewarkUnited States
- Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical SchoolNewarkUnited States
| |
Collapse
|
225
|
Hall DCN, Benndorf RA. Aspirin sensitivity of PIK3CA-mutated Colorectal Cancer: potential mechanisms revisited. Cell Mol Life Sci 2022; 79:393. [PMID: 35780223 PMCID: PMC9250486 DOI: 10.1007/s00018-022-04430-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/01/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022]
Abstract
PIK3CA mutations are amongst the most prevalent somatic mutations in cancer and are associated with resistance to first-line treatment along with low survival rates in a variety of malignancies. There is evidence that patients carrying PIK3CA mutations may benefit from treatment with acetylsalicylic acid, commonly known as aspirin, particularly in the setting of colorectal cancer. In this regard, it has been clarified that Class IA Phosphatidylinositol 3-kinases (PI3K), whose catalytic subunit p110α is encoded by the PIK3CA gene, are involved in signal transduction that regulates cell cycle, cell growth, and metabolism and, if disturbed, induces carcinogenic effects. Although PI3K is associated with pro-inflammatory cyclooxygenase-2 (COX-2) expression and signaling, and COX-2 is among the best-studied targets of aspirin, the mechanisms behind this clinically relevant phenomenon are still unclear. Indeed, there is further evidence that the protective, anti-carcinogenic effect of aspirin in this setting may be mediated in a COX-independent manner. However, until now the understanding of aspirin's prostaglandin-independent mode of action is poor. This review will provide an overview of the current literature on this topic and aims to analyze possible mechanisms and targets behind the aspirin sensitivity of PIK3CA-mutated cancers.
Collapse
Affiliation(s)
- Daniella C N Hall
- Department of Clinical Pharmacy and Pharmacotherapy, Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120, Halle (Saale), Germany
| | - Ralf A Benndorf
- Department of Clinical Pharmacy and Pharmacotherapy, Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120, Halle (Saale), Germany.
| |
Collapse
|
226
|
Vasan N, Cantley LC. At a crossroads: how to translate the roles of PI3K in oncogenic and metabolic signalling into improvements in cancer therapy. Nat Rev Clin Oncol 2022; 19:471-485. [PMID: 35484287 PMCID: PMC11215755 DOI: 10.1038/s41571-022-00633-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2022] [Indexed: 12/14/2022]
Abstract
Numerous agents targeting various phosphatidylinositol 3-kinase (PI3K) pathway components, including PI3K, AKT and mTOR, have been tested in oncology clinical trials, resulting in regulatory approvals for the treatment of selected patients with breast cancer, certain other solid tumours or particular haematological malignancies. However, given the prominence of PI3K signalling in cancer and the crucial role of this pathway in linking cancer growth with metabolism, these clinical results could arguably be improved upon. In this Review, we discuss past and present efforts to overcome the somewhat limited clinical efficacy of PI3Kα pathway inhibitors, including optimization of inhibitor specificity, patient selection and biomarkers across cancer types, with a focus on breast cancer, as well as identification and abrogation of signalling-related and metabolic mechanisms of resistance, and interventions to improve management of prohibitive adverse events. We highlight the advantages and limitations of laboratory-based model systems used to study the PI3K pathway, and propose technologies and experimental inquiries to guide the future clinical deployment of PI3K pathway inhibitors in the treatment of cancer.
Collapse
Affiliation(s)
- Neil Vasan
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Lewis C Cantley
- Meyer Cancer Center, Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
227
|
Ciardiello F, Ciardiello D, Martini G, Napolitano S, Tabernero J, Cervantes A. Clinical management of metastatic colorectal cancer in the era of precision medicine. CA Cancer J Clin 2022; 72:372-401. [PMID: 35472088 DOI: 10.3322/caac.21728] [Citation(s) in RCA: 279] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) represents approximately 10% of all cancers and is the second most common cause of cancer deaths. Initial clinical presentation as metastatic CRC (mCRC) occurs in approximately 20% of patients. Moreover, up to 50% of patients with localized disease eventually develop metastases. Appropriate clinical management of these patients is still a challenging medical issue. Major efforts have been made to unveil the molecular landscape of mCRC. This has resulted in the identification of several druggable tumor molecular targets with the aim of developing personalized treatments for each patient. This review summarizes the improvements in the clinical management of patients with mCRC in the emerging era of precision medicine. In fact, molecular stratification, on which the current treatment algorithm for mCRC is based, although it does not completely represent the complexity of this disease, has been the first significant step toward clinically informative genetic profiling for implementing more effective therapeutic approaches. This has resulted in a clinically relevant increase in mCRC disease control and patient survival. The next steps in the clinical management of mCRC will be to integrate the comprehensive knowledge of tumor gene alterations, of tumor and microenvironment gene and protein expression profiling, of host immune competence as well as the application of the resulting dynamic changes to a precision medicine-based continuum of care for each patient. This approach could result in the identification of individual prognostic and predictive parameters, which could help the clinician in choosing the most appropriate therapeutic program(s) throughout the entire disease journey for each patient with mCRC. CA Cancer J Clin. 2022;72:000-000.
Collapse
Affiliation(s)
- Fortunato Ciardiello
- Division of Medical Oncology, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Davide Ciardiello
- Division of Medical Oncology, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
- Division of Medical Oncology, IRCCS Foundation Home for the Relief of Suffering, San Giovanni Rotondo, Italy
| | - Giulia Martini
- Division of Medical Oncology, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Stefania Napolitano
- Division of Medical Oncology, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Josep Tabernero
- Medical Oncology Department, Vall d'Hebron Hospital Campus, Barcelona, Spain
- Institute of Oncology, University of Vic/Central University of Catalonia, Barcelona, Spain
- Oncology Institute of Barcelona-Quironsalud, Biomedical Research Center in Cancer, Barcelona, Spain
| | - Andres Cervantes
- Medical Oncology Department, Instituto de Investigación Sanitaria Valencia Biomedical Research Institute, University of Valencia, Valencia, Spain
- Carlos III Institute of Health, Biomedical Research Center in Cancer, Madrid, Spain
| |
Collapse
|
228
|
Shetty O, Vengurlekar V, Kapoor A, Kamble V, Gurav M, Bhargava P, Srinivas S, Ramaswamy A, Ramadwar M, Saklani AP, Desouza A, Ostwal V. The Prevalence of BRAF, PIK3CA, and RAS Mutations in Indian Patients with Colorectal Cancer. South Asian J Cancer 2022; 11:190-194. [PMID: 36620501 PMCID: PMC9822779 DOI: 10.1055/s-0041-1740244] [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] [Indexed: 01/11/2023] Open
Abstract
Omshree ShettyVikas OstwalIntroduction The present study evaluates the mutation pattern and frequency of BRAF , PIK3CA and RAS in colorectal carcinoma observed in the tertiary cancer center in India. Materials and Methods Consecutive cases of colorectal adenocarcinoma ( n = 330) registered from January 2015 to December 2019 (5-year duration) were selected for the study. Molecular analysis for BRAF . PIK3CA (exon 9 and 20) and RAS ( KRAS & NRAS ) was performed on representative formalin-fixed paraffin-embedded tissues by Sanger sequencing. Results were correlated with clinicopathological features. Patient overall survival (OS) was obtained using Kaplan-Meier method. Results The study cohort was in the age range of 22 to 81 years (median age: 52 years) that included 202 males and 96 females (male: female ratio 2.1:1). BRAF V600E mutation was observed in three cases (1%), while 17 cases (5.7%) had mutations in the PIK3CA gene (exon 9 or exon 20). Mutation analysis for RAS gene ( KRAS & NRAS ) was observed among 42 (15.4%) cases with KRAS mutation and 11 (4%) cases were positive for NRAS mutations. Among RAS, KRAS G12D was the predominant mutation. Median OS with wild-type RAS was 46.6 months (95% confidence interval [CI]: 22.4-70.8), while for RAS mutated patients, it was 25.6 months (95% CI: 16.7-34.5), hazard ratio: 1.7 (95% CI: 1.1-2.7, p = 0.025). Conclusion This study evaluated the prevalence of BRAF, PIK3CA and RAS mutations in the Indian cohort and its impact on clinical behavior. There was lower incidence of BRAF mutations in this cohort and PIK3CA mutation (single) did not impact survival of the patients.
Collapse
Affiliation(s)
- Omshree Shetty
- Molecular Pathology Laboratory, Department of Pathology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Vaibhavi Vengurlekar
- Molecular Pathology Laboratory, Department of Pathology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Akhil Kapoor
- Department of Medical Oncology, Mahamana Pandit Madan Mohan Malviya Cancer Center & Homi Bhabha Cancer Hospital, Tata Memorial Hospital, Varanasi, Uttar Pradesh, India
| | - Vishakha Kamble
- Molecular Pathology Laboratory, Department of Pathology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Mamta Gurav
- Molecular Pathology Laboratory, Department of Pathology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Prabhat Bhargava
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Sujay Srinivas
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Anant Ramaswamy
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Mukta Ramadwar
- Department of Pathology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Avanish P. Saklani
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Ashwin Desouza
- Department of Surgical Oncology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Vikas Ostwal
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Center, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| |
Collapse
|
229
|
ErbB3-Targeting Oncolytic Adenovirus Causes Potent Tumor Suppression by Induction of Apoptosis in Cancer Cells. Int J Mol Sci 2022; 23:ijms23137127. [PMID: 35806132 PMCID: PMC9266575 DOI: 10.3390/ijms23137127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/23/2022] [Accepted: 06/23/2022] [Indexed: 12/10/2022] Open
Abstract
Cancer is a multifactorial and deadly disease. Despite major advancements in cancer therapy in the last two decades, cancer incidence is on the rise and disease prognosis still remains poor. Furthermore, molecular mechanisms of cancer invasiveness, metastasis, and drug resistance remain largely elusive. Targeted cancer therapy involving the silencing of specific cancer-enriched proteins by small interfering RNA (siRNA) offers a powerful tool. However, its application in clinic is limited by the short half-life of siRNA and warrants the development of efficient and stable siRNA delivery systems. Oncolytic adenovirus-mediated therapy offers an attractive alternative to the chemical drugs that often suffer from innate and acquired drug resistance. In continuation to our reports on the development of oncolytic adenovirus-mediated delivery of shRNA, we report here the replication-incompetent (dAd/shErbB3) and replication-competent (oAd/shErbB3) oncolytic adenovirus systems that caused efficient and persistent targeting of ErbB3. We demonstrate that the E1A coded by oAd/shErbB, in contrast to dAd/shErbB, caused downregulation of ErbB2 and ErbB3, yielding stronger downregulation of the ErbB3-oncogenic signaling axis in in vitro models of lung and breast cancer. These results were validated by in vivo antitumor efficacy of dAd/shErbB3 and oAd/shErbB3.
Collapse
|
230
|
Shen WQ, Guo Y, Ru WE, Li C, Zhang GC, Liao N, Du GQ. Using an Improved Residual Network to Identify PIK3CA Mutation Status in Breast Cancer on Ultrasound Image. Front Oncol 2022; 12:850515. [PMID: 35719907 PMCID: PMC9204315 DOI: 10.3389/fonc.2022.850515] [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: 01/07/2022] [Accepted: 04/11/2022] [Indexed: 11/30/2022] Open
Abstract
Background The detection of phosphatidylinositol-3 kinase catalytic alpha (PIK3CA) gene mutations in breast cancer is a key step to design personalizing an optimal treatment strategy. Traditional genetic testing methods are invasive and time-consuming. It is urgent to find a non-invasive method to estimate the PIK3CA mutation status. Ultrasound (US), one of the most common methods for breast cancer screening, has the advantages of being non-invasive, fast imaging, and inexpensive. In this study, we propose to develop a deep convolutional neural network (DCNN) to identify PIK3CA mutations in breast cancer based on US images. Materials and Methods We retrospectively collected 312 patients with pathologically confirmed breast cancer who underwent genetic testing. All US images (n=800) of breast cancer patients were collected and divided into the training set (n=600) and test set (n=200). A DCNN-Improved Residual Network (ImResNet) was designed to identify the PIK3CA mutations. We also compared the ImResNet model with the original ResNet50 model, classical machine learning models, and other deep learning models. Results The proposed ImResNet model has the ability to identify PIK3CA mutations in breast cancer based on US images. Notably, our ImResNet model outperforms the original ResNet50, DenseNet201, Xception, MobileNetv2, and two machine learning models (SVM and KNN), with an average area under the curve (AUC) of 0.775. Moreover, the overall accuracy, average precision, recall rate, and F1-score of the ImResNet model achieved 74.50%, 74.17%, 73.35%, and 73.76%, respectively. All of these measures were significantly higher than other models. Conclusion The ImResNet model gives an encouraging performance in predicting PIK3CA mutations based on breast US images, providing a new method for noninvasive gene prediction. In addition, this model could provide the basis for clinical adjustments and precision treatment.
Collapse
Affiliation(s)
- Wen-Qian Shen
- Department of Ultrasound, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Department of Ultrasound, The Second Affifiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanhui Guo
- Department of Computer Science, University of Illinois Springfield, Springfield, IL, United States
| | - Wan-Er Ru
- Department of Ultrasound, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,College of Medicine, Shantou University, Shantou, China
| | - Cheukfai Li
- Department of Breast Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Guo-Chun Zhang
- Department of Breast Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ning Liao
- Department of Breast Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Guo-Qing Du
- Department of Ultrasound, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
231
|
Nakashima Y, Tsukahara M. Laminin-511 activates the human induced pluripotent stem cell survival via α6β1 integrin-Fyn-RhoA-ROCK signaling. Stem Cells Dev 2022; 31:706-719. [PMID: 35726387 DOI: 10.1089/scd.2022.0010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
In human induced pluripotent stem cells (hiPSCs), laminin-511/α6β1 integrin interacts with E-cadherin, an intercellular adhesion molecule, to induce the activation of the PI3K-dependent signaling pathway. The interaction between laminin-511/α6β1 integrin and E-cadherin, an intercellular adhesion molecule, results in protection against apoptosis via the proto-oncogene tyrosine-protein kinase Fyn(Fyn)-RhoA-ROCK signaling pathway and the Ras homolog gene family member A (RhoA)/Rho kinase (ROCK) signaling pathway (the major pathway for cell death). In this paper, the impact of laminin-511 on hiPSC on α6β1 integrin-Fyn-RhoA-ROCK signaling is discussed and explored along with validation experiments. PIK3CA mRNA (mean [standard deviation {SD}]: iMatrix-511, 1.00 [0. 61]; collagen+MFGE8, 0.023 [0.02]; **P<0.01; n=6) and PIK3R1 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 79]; collagen+MFGE8, 0.040 [0.06]; *P<0.05; n=6) were upregulated by iMatrix-511 resulting from an increased expression of Integrin α6 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 42]; collagen+MFGE8, 0.23 [0.05]; **P<0.01; n=6). iMatrix-511 increased the expression of p120-Catenin mRNA (mean [SD]: iMatrix-511, 1.00 [0. 71]; collagen+MFGE8, 0.025 [0.03]; **P<0.01; n=6) and RAC1 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 28]; collagen+MFGE8, 0.39 [0.15]; **P<0.01; n=6) by increasing the expression of E-cadherin mRNA (mean [SD]: iMatrix-511, 1.00 [0. 38]; collagen+MFGE8, 0.16 [0.11]; **P<0.01; n=6). As a result, iMatrix-511 increased the expression of P190 RhoGAP (GTPase-activating proteins) mRNA, such as ARHGAP1 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 57]; collagen+MFGE8, 0.032 [0.03]; **P<0.01; n=6), ARHGAP4 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 56]; collagen+MFGE8, 0.039 [0.049]; **P<0.01; n=6), and ARHGAP5 mRNA (mean [SD]: iMatrix-511, 1.00 [0. 39]; collagen+MFGE8, 0.063 [0.043]; **P<0.01; n=6). Western blotting showed that phospho-Rac1 remained in the cytoplasm and phospho-Fyn showed nuclear transition in iPSCs cultured on iMatrix-511. Proteome analysis showed that PI3K signaling was enhanced and cytoskeletal actin was activated in iPSCs cultured on iMatrix-511. In conclusion, laminin-511 strongly activated the cell survival by promoting α6β1 integrin-Fyn-RhoA-ROCK signaling in hiPSCs.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), Kyoto Research Park KISTIC Building Room 501, 5th floor, KISTIC building,, Kyoto Research Park KISTIC Building Room 501, 5th floor, KISTIC building, 134 Chudoji Minami-cho,, Shimogyo-ku,, Kyoto, Kyoto, Japan, 600-8813;
| | - Masayoshi Tsukahara
- Kyoto University Center for iPS Cell Research and Application Foundation (CiRA Foundation), Facility for iPS Cell Therapy (FiT), kyoto, Kyoto, Japan;
| |
Collapse
|
232
|
Diociaiuti A, Rotunno R, Pisaneschi E, Cesario C, Carnevale C, Condorelli AG, Rollo M, Di Cecca S, Quintarelli C, Novelli A, Zambruno G, El Hachem M. Clinical and Molecular Spectrum of Sporadic Vascular Malformations: A Single-Center Study. Biomedicines 2022; 10:biomedicines10061460. [PMID: 35740480 PMCID: PMC9220263 DOI: 10.3390/biomedicines10061460] [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: 01/29/2022] [Revised: 05/22/2022] [Accepted: 06/16/2022] [Indexed: 01/04/2023] Open
Abstract
Sporadic vascular malformations (VMs) are a large group of disorders of the blood and lymphatic vessels caused by somatic mutations in several genes—mainly regulating the RAS/MAPK/ERK and PI3K/AKT/mTOR pathways. We performed a cross-sectional study of 43 patients affected with sporadic VMs, who had received molecular diagnosis by high-depth targeted next-generation sequencing in our center. Clinical and imaging features were correlated with the sequence variants identified in lesional tissues. Six of nine patients with capillary malformation and overgrowth (CMO) carried the recurrent GNAQ somatic mutation p.Arg183Gln, while two had PIK3CA mutations. Unexpectedly, 8 of 11 cases of diffuse CM with overgrowth (DCMO) carried known PIK3CA mutations, and the remaining 3 had pathogenic GNA11 variants. Recurrent PIK3CA mutations were identified in the patients with megalencephaly–CM–polymicrogyria (MCAP), CLOVES, and Klippel–Trenaunay syndrome. Interestingly, PIK3CA somatic mutations were associated with hand/foot anomalies not only in MCAP and CLOVES, but also in CMO and DCMO. Two patients with blue rubber bleb nevus syndrome carried double somatic TEK mutations, two of which were previously undescribed. In addition, a novel sporadic case of Parkes Weber syndrome (PWS) due to an RASA1 mosaic pathogenic variant was described. Finally, a girl with a mild PWS and another diagnosed with CMO carried pathogenic KRAS somatic variants, showing the variability of phenotypic features associated with KRAS mutations. Overall, our findings expand the clinical and molecular spectrum of sporadic VMs, and show the relevance of genetic testing for accurate diagnosis and emerging targeted therapies.
Collapse
Affiliation(s)
- Andrea Diociaiuti
- Dermatology Unit and Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (R.R.); (C.C.); (M.E.H.)
- Correspondence: ; Tel.: +39-0668592509
| | - Roberta Rotunno
- Dermatology Unit and Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (R.R.); (C.C.); (M.E.H.)
| | - Elisa Pisaneschi
- Translational Cytogenomics Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (E.P.); (C.C.); (A.N.)
| | - Claudia Cesario
- Translational Cytogenomics Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (E.P.); (C.C.); (A.N.)
| | - Claudia Carnevale
- Dermatology Unit and Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (R.R.); (C.C.); (M.E.H.)
| | - Angelo Giuseppe Condorelli
- Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (A.G.C.); (G.Z.)
| | - Massimo Rollo
- Interventional Radiology Unit, Department of Imaging, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy;
| | - Stefano Di Cecca
- Department Onco-Haematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (S.D.C.); (C.Q.)
| | - Concetta Quintarelli
- Department Onco-Haematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (S.D.C.); (C.Q.)
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Antonio Novelli
- Translational Cytogenomics Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (E.P.); (C.C.); (A.N.)
| | - Giovanna Zambruno
- Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (A.G.C.); (G.Z.)
| | - May El Hachem
- Dermatology Unit and Genodermatosis Unit, Genetics and Rare Diseases Research Division, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio 4, 00165 Rome, Italy; (R.R.); (C.C.); (M.E.H.)
| |
Collapse
|
233
|
Huang YS, Wu CC, Chang CC, Huang SF, Kuo HY, Shih HM. Reciprocal regulation of Daxx and PIK3CA promotes colorectal cancer cell growth. Cell Mol Life Sci 2022; 79:367. [PMID: 35718818 PMCID: PMC11072676 DOI: 10.1007/s00018-022-04399-8] [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] [Received: 02/23/2022] [Revised: 05/16/2022] [Accepted: 05/23/2022] [Indexed: 02/06/2023]
Abstract
Upregulation of death-domain-associated protein (Daxx) is strongly associated with diverse cancer types. Among these, the clinicopathological significance and molecular mechanisms of Daxx overexpression in colorectal cancer (CRC) remain unknown. Here, we showed that Daxx expression was increased in both clinical CRC samples and CRC cell lines. Daxx knockdown significantly reduced proliferation activity in CRC cells and tumor growth in a xenograft model. Further studies revealed that Daxx expression could be attenuated by either treatment with the PIK3CA inhibitor PIK-75 or PIK3CA depletion in CRC cells. Conversely, expression of PIK3CA constitutively active mutants could increase Daxx expression. These data suggest that PIK3CA positively regulates Daxx expression. Consistently, the expression levels of PIK3CA and Daxx were positively correlated in sporadic CRC samples. Interestingly, Daxx knockdown or overexpression yielded decreased or increased levels of PIK3CA, respectively, in CRC cells. We further demonstrated that Daxx activates the promoter activity and expression of PIK3CA. Altogether, our results identify a mechanistic pathway of Daxx overexpression in CRC and suggest a reciprocal regulation between Daxx and PIK3CA for CRC cell growth.
Collapse
Affiliation(s)
- Yen-Sung Huang
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan.
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan.
| | - Chang-Chieh Wu
- Department of Surgery, Tri-Service General Hospital Keelung Branch, National Defense Medical Center, Keelung, 20244, Taiwan
| | - Che-Chang Chang
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan
| | - Shiu-Feng Huang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County, 35053, Taiwan
| | - Hong-Yi Kuo
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County, 35053, Taiwan
| | - Hsiu-Ming Shih
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 11529, Taiwan.
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, 11031, Taiwan.
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Miaoli County, 35053, Taiwan.
| |
Collapse
|
234
|
Laka K, Mbita Z. P53-Related Anticancer Activities of Drimia calcarata Bulb Extracts Against Lung Cancer. Front Mol Biosci 2022; 9:876213. [PMID: 35769912 PMCID: PMC9235921 DOI: 10.3389/fmolb.2022.876213] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/02/2022] [Indexed: 01/18/2023] Open
Abstract
Current lung cancer treatment strategies are ineffective, and lung cancer cases continue to soar; thus, novel anticancer drugs and targets are needed, and medicinal plants are promising to offer better alternatives. This study was aimed at analysing two p53 splice variants during the potential anticancer activities of Drimia calcarata (Dc) methanol and water extracts against different human lung cancer cell lines of varying p53 mutation status, and these included mutant H1573 and mutant H1437 and p53-wild type (A549) cells. The anticancer activities of the Dc extracts were assessed by establishing the cytotoxic effect and the apoptosis-inducing capacity of these extracts, using the MTT assay and Annexin V analysis, respectively, with the latter confirmed using fluorescence microscopy. The molecular mechanisms induced by these extracts were further evaluated using cell cycle analysis and RT-PCR. Both extracts demonstrated safety against noncancerous lung MRC-5 fibroblasts and exhibited significant anticancer potency (p < 0.001) against the H1437 (IC50 values: 62.50 μg/ml methanol extract and 125 μg/ml WE), H1573 (IC50 value: 125 μg/ml for both extracts) and A549 (IC50 value: 500 μg/ml ME). The water extract had no effect on the viability of A549 cells. Treated H1437 cells underwent p53-dependent apoptosis and S-phase cell cycle arrest while H1573 treated cells underwent p53-independed apoptosis and G0/G1 cell cycle arrest through upregulation of p21 mRNA expression levels. The expression levels of STAT1, STAT3, STAT5A and STAT5B genes increased significantly (p < 0.001) following the treatment of H1573 cells with ME and WE. Treatment of H1437 cells with ME upregulated the STAT1, STAT3, STAT5A and STAT5B mRNAs. Our results indicate that the proliferative inhibitory effect of D. calcarata extracts on A549 and H1573 cells is correlated with the suppression of Bcl-2, STAT3 and STAT5B while that is not the case in H1437 cells. Thus, our results suggest that the dysregulation of anti-apoptotic molecules Bcl-2, STAT3, STAT5A and STAT5B in H1437 may play a role in cancer cell survival, which may consequently contribute to the development of p53-mutated non-small human lung cancer. Our results indicate that D. calcarata is a promising source of anticancer agents for the treatment of p53-mutant human non-small lung cancer cells than the p53-wild type human non-small lung cancer cells.
Collapse
|
235
|
Kobialka P, Sabata H, Vilalta O, Gouveia L, Angulo-Urarte A, Muixí L, Zanoncello J, Muñoz-Aznar O, Olaciregui NG, Fanlo L, Esteve-Codina A, Lavarino C, Javierre BM, Celis V, Rovira C, López-Fernández S, Baselga E, Mora J, Castillo SD, Graupera M. The onset of PI3K-related vascular malformations occurs during angiogenesis and is prevented by the AKT inhibitor miransertib. EMBO Mol Med 2022; 14:e15619. [PMID: 35695059 PMCID: PMC9260211 DOI: 10.15252/emmm.202115619] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 12/15/2022] Open
Abstract
Low‐flow vascular malformations are congenital overgrowths composed of abnormal blood vessels potentially causing pain, bleeding and obstruction of different organs. These diseases are caused by oncogenic mutations in the endothelium, which result in overactivation of the PI3K/AKT pathway. Lack of robust in vivo preclinical data has prevented the development and translation into clinical trials of specific molecular therapies for these diseases. Here, we demonstrate that the Pik3caH1047R activating mutation in endothelial cells triggers a transcriptome rewiring that leads to enhanced cell proliferation. We describe a new reproducible preclinical in vivo model of PI3K‐driven vascular malformations using the postnatal mouse retina. We show that active angiogenesis is required for the pathogenesis of vascular malformations caused by activating Pik3ca mutations. Using this model, we demonstrate that the AKT inhibitor miransertib both prevents and induces the regression of PI3K‐driven vascular malformations. We confirmed the efficacy of miransertib in isolated human endothelial cells with genotypes spanning most of human low‐flow vascular malformations.
Collapse
Affiliation(s)
- Piotr Kobialka
- Endothelial Pathobiology and Microenvironment, Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Helena Sabata
- Endothelial Pathobiology and Microenvironment, Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Odena Vilalta
- Endothelial Pathobiology and Microenvironment, Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Leonor Gouveia
- Endothelial Pathobiology and Microenvironment, Josep Carreras Leukaemia Research Institute, Barcelona, Spain.,Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Ana Angulo-Urarte
- Endothelial Pathobiology and Microenvironment, Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Laia Muixí
- Endothelial Pathobiology and Microenvironment, Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Jasmina Zanoncello
- Endothelial Pathobiology and Microenvironment, Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Oscar Muñoz-Aznar
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Nagore G Olaciregui
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Lucia Fanlo
- 3D Chromatin Organization, Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Cinzia Lavarino
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Biola M Javierre
- 3D Chromatin Organization, Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Veronica Celis
- Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu Barcelona, Barcelona, Spain
| | - Carlota Rovira
- Department of Pathology, Hospital Sant Joan de Déu Barcelona, Barcelona, Spain
| | - Susana López-Fernández
- Department of Plastic Surgery, Hospital de la Santa Creu i de Sant Pau, Barcelona, Spain
| | - Eulàlia Baselga
- Department of Dermatology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Jaume Mora
- Developmental Tumor Biology Laboratory, Institut de Recerca Sant Joan de Déu, Barcelona, Spain.,Pediatric Cancer Center Barcelona, Hospital Sant Joan de Déu Barcelona, Barcelona, Spain
| | - Sandra D Castillo
- Endothelial Pathobiology and Microenvironment, Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Mariona Graupera
- Endothelial Pathobiology and Microenvironment, Josep Carreras Leukaemia Research Institute, Barcelona, Spain.,CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
236
|
Wolter JK, Valencia-Sama I, Osborn AJ, Propst EJ, Irwin MS, Papsin B, Wolter NE. Combination mTOR and SHP2 inhibitor treatment of lymphatic malformation endothelial cells. Microvasc Res 2022; 143:104397. [PMID: 35671835 DOI: 10.1016/j.mvr.2022.104397] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/27/2022] [Accepted: 06/01/2022] [Indexed: 12/13/2022]
Abstract
Mammalian target of rapamycin (mTOR) inhibitors are clinically effective at treating some complex lymphatic malformations (LM). The mTOR inhibitor rapamycin blocks the phosphoinositide 3-kinase (PI3K) pathway, which is commonly mutated in this condition. Although rapamycin is effective at controlling symptoms of LM, treatment courses are long, not all LMs respond to treatment, and many patients relapse after treatment has stopped. Concurrent rat sarcoma virus (RAS) pathway abnormalities have been identified in LM, which may limit the effectiveness of rapamycin. Protein tyrosine phosphatase-2 (SHP2) controls the RAS pathway upstream, and SHP2 inhibitors are being investigated for treatment of various tumors. The objective of this study was to determine the impact of SHP2 inhibition in combination with rapamycin on LM growth in vitro. Using primary patient cells isolated from a surgically resected LM, we found that combination treatment with rapamycin and the SHP2 inhibitor SHP099 caused a synergistic reduction in cell growth, migration and lymphangiogenesis. These results suggest that combination treatment targeting the PI3K and RAS signaling pathways may result in effective treatment of LMs of the head and neck.
Collapse
Affiliation(s)
- Jennifer K Wolter
- Department of Otolaryngology, Head & Neck Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | | | - Alex J Osborn
- Department of Otolaryngology, Head & Neck Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Evan J Propst
- Department of Otolaryngology, Head & Neck Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Meredith S Irwin
- Cell Biology Program, The Hospital for Sick Children, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada; Department of Pediatrics, The Hospital for Sick Children, Toronto, Canada
| | - Blake Papsin
- Department of Otolaryngology, Head & Neck Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Nikolaus E Wolter
- Department of Otolaryngology, Head & Neck Surgery, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
237
|
Wu X, Lin Z, Zhao C, Liu L, Zhang K, Lai J, Meng QF, Yao G, Huang Q, Zhao XZ, Rao L. Neutrophil membrane-coated immunomagnetic nanoparticles for efficient isolation and analysis of circulating tumor cells. Biosens Bioelectron 2022; 213:114425. [PMID: 35688024 DOI: 10.1016/j.bios.2022.114425] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/30/2022] [Accepted: 05/24/2022] [Indexed: 11/02/2022]
Abstract
The isolation and analysis of scarce circulating tumor cells (CTCs) with immunomagnetic nanoparticles (IMNs) have shown promising outcomes in noninvasive cancer diagnosis. However, the IMNs adsorb nonspecific proteins after entering into biofluids and the formed protein coronas cover surface targeting ligands, limiting the detection efficiency of IMNs. In addition, the interaction between surface targeting ligands and white blood cells (WBCs) significantly limits the purity of CTCs isolated by IMNs. Furthermore, the interfacial collision of nanoparticles and cells has negative effects on the viability of isolated CTCs. All of these limitations synthetically restrict the isolation and analysis of rare CTCs for early diagnosis and precision medicine. Here, we proposed that surface functionalization of IMNs with neutrophil membranes can simultaneously reduce nonspecific protein adsorption, enhance the interaction with CTCs, reduce the distraction from WBCs, and improve the viability of isolated CTCs. In spiked blood samples, our neutrophil membrane-coated IMNs (Neu-IMNs) exhibited a superior separation efficiency from 41.36% to 96.82% and an improved purity from 40.25% to 90.68% when compared to bare IMNs. Additionally, we successfully isolated CTCs in 19 out of total 20 blood samples from breast cancer patients using Neu-IMNs and further confirmed the feasibility of the isolated CTCs for downstream cell sequencing. Our work provides a new perspective on engineered IMNs for efficient isolation and analysis of CTCs, paving the way for early noninvasive diagnosis of cancer.
Collapse
Affiliation(s)
- Xianjia Wu
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, China; Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Zhousheng Lin
- Department of Molecular Pathology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, China; Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chenchen Zhao
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, China; Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Lujie Liu
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Kelin Zhang
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, China
| | - Jialin Lai
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Qian-Fang Meng
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Gaungyu Yao
- Breast Center, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qinqin Huang
- Department of Molecular Pathology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450014, China.
| | - Xing-Zhong Zhao
- Key Laboratory of Artificial Micro- and Nano-Structures of Ministry of Education, School of Physics and Technology, Wuhan University, Wuhan, 430072, China.
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| |
Collapse
|
238
|
Aggarwal S, Bhadana K, Singh B, Rawat M, Mohammad T, Al-Keridis LA, Alshammari N, Hassan MI, Das SN. Cinnamomum zeylanicum Extract and its Bioactive Component Cinnamaldehyde Show Anti-Tumor Effects via Inhibition of Multiple Cellular Pathways. Front Pharmacol 2022; 13:918479. [PMID: 35774603 PMCID: PMC9237655 DOI: 10.3389/fphar.2022.918479] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/09/2022] [Indexed: 01/04/2023] Open
Abstract
Cinnamomum zeylanicum is a tropical plant with traditional medicinal significance that possesses antimicrobial, antifungal, anti-parasitic, and anti-tumor properties. Here, we have elucidated the anti-tumor effects of Cinnamomum zeylanicum extract (CZE) and its bioactive compound cinnamaldehyde (CIN) on oral cancer and elucidated underlying molecular mechanisms. Anti-tumor activities of CZE and CIN were demonstrated by various in vitro experiments on oral cancer cells (SCC-4, SCC-9, SCC-25). The cell proliferation, growth, cell cycle arrest, apoptosis, and autophagy were analyzed by MTT, clonogenic assay, propidium iodide, annexin-V-PI, DAPI, and acridine orange staining, respectively. The binding affinity of CIN towards dihydrofolate reductase and p38-MAP kinase alpha was analyzed by molecular docking. Western blot assay was performed to assess the alteration in the expression of various proteins. CZE and CIN treatment significantly inhibited the growth and proliferation of oral cancer cells in a dose-dependent manner. These treatments further induced apoptosis, cell cycle arrest, and autophagy. CZE and CIN inhibited the invasion and cytoplasmic translocation of NF-κB in these cell lines. CIN showed a high affinity to MAP kinase P38 alpha and dihydrofolate reductase with binding affinities of −6.8 and −5.9 kcal/mol, respectively. The cancer cells showed a decreased expression of various PI3k-AKT-mTOR pathways related to VEGF, COX-2, Bcl-2, NF-κB, and proteins post-treatment.
Collapse
Affiliation(s)
- Sadhna Aggarwal
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Kanchan Bhadana
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Baldeep Singh
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Meenakshi Rawat
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Taj Mohammad
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Lamya Ahmed Al-Keridis
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Nawaf Alshammari
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Md. Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
- *Correspondence: Md. Imtaiyaz Hassan, ; Satya N. Das,
| | - Satya N. Das
- Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
- *Correspondence: Md. Imtaiyaz Hassan, ; Satya N. Das,
| |
Collapse
|
239
|
Starita N, Pezzuto F, Sarno S, Losito NS, Perdonà S, Buonaguro L, Buonaguro FM, Tornesello ML. Mutations in the telomerase reverse transcriptase promoter and PIK3CA gene are common events in penile squamous cell carcinoma of Italian and Ugandan patients. Int J Cancer 2022; 150:1879-1888. [PMID: 35253909 PMCID: PMC9310576 DOI: 10.1002/ijc.33990] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/20/2022] [Accepted: 02/09/2022] [Indexed: 11/16/2022]
Abstract
Penile carcinoma develops either through human papillomavirus (HPV) related or unrelated carcinogenic pathways. Genetic alterations and nucleotide changes in coding regions (ie, TP53, CDKN2A, PIK3CA and NOTCH1) are main cancer driver events either in HPV positive or in HPV negative tumours. We investigated the presence of hotspot nucleotide mutations in TERT promoter (TERTp) and PIK3CA exon 9 and their relationship with HPV status in 69 penile cancer cases from Italian and Ugandan patients. Genetic variations and viral sequences have been characterised by end-point polymerase chain reaction (PCR) and Sanger sequencing. The mutant allele frequencies (MAFs) of TERTp -124A/-146A and PIK3CA E545K have been determined by droplet digital PCR (ddPCR) assays. The results showed that TERTp mutations are highly prevalent in penile carcinoma (53.6%) and significantly more frequent in HPV negative (67.6%) than HPV positive (32.4%) cases (P = .0482). PIK3CA mutations were similarly distributed in virus-related and unrelated cases (25.9% and 26.7%, respectively) and coexisted with TERTp changes in 15.8% of penile carcinoma samples. Notably, MAFs of co-occurring mutations were frequently discordant indicating that PIK3CA E545K nucleotide changes are subsequent genetic events occurring in subclones of TERTp mutated cells. The frequencies of TERTp and PIK3CA mutations were higher among Italian compared to Ugandan cases and inversely correlated with the HPV status. In conclusion, TERTp mutations are very common in penile carcinoma and their coexistence with PIK3CA in a substantial number of cases may represent a novel oncogenic synergy relevant for patient stratification and use of therapeutic strategies against new actionable targets.
Collapse
Affiliation(s)
- Noemy Starita
- Molecular Biology and Viral Oncology UnitIstituto Nazionale Tumori IRCCS Fondazione G. PascaleNaplesItaly
| | - Francesca Pezzuto
- Molecular Biology and Viral Oncology UnitIstituto Nazionale Tumori IRCCS Fondazione G. PascaleNaplesItaly
| | - Sabrina Sarno
- Department of PathologyIstituto Nazionale Tumori IRCCS Fondazione G. PascaleNaplesItaly
| | - Nunzia Simona Losito
- Department of PathologyIstituto Nazionale Tumori IRCCS Fondazione G. PascaleNaplesItaly
| | - Sisto Perdonà
- Urology UnitIstituto Nazionale Tumori IRCCS Fondazione G. PascaleNaplesItaly
| | - Luigi Buonaguro
- Innovative Immunological ModelsIstituto Nazionale Tumori IRCCS Fondazione G. PascaleNaplesItaly
| | - Franco M. Buonaguro
- Molecular Biology and Viral Oncology UnitIstituto Nazionale Tumori IRCCS Fondazione G. PascaleNaplesItaly
| | - Maria Lina Tornesello
- Molecular Biology and Viral Oncology UnitIstituto Nazionale Tumori IRCCS Fondazione G. PascaleNaplesItaly
| |
Collapse
|
240
|
Hong N, Sun G, Zuo X, Chen M, Liu L, Wang J, Feng X, Shi W, Gong M, Ma P. Application of informatics in cancer research and clinical practice: Opportunities and challenges. CANCER INNOVATION 2022; 1:80-91. [PMID: 38089452 PMCID: PMC10686161 DOI: 10.1002/cai2.9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 04/24/2022] [Indexed: 10/15/2024]
Abstract
Cancer informatics has significantly progressed in the big data era. We summarize the application of informatics approaches to the cancer domain from both the informatics perspective (e.g., data management and data science) and the clinical perspective (e.g., cancer screening, risk assessment, diagnosis, treatment, and prognosis). We discuss various informatics methods and tools that are widely applied in cancer research and practices, such as cancer databases, data standards, terminologies, high-throughput omics data mining, machine-learning algorithms, artificial intelligence imaging, and intelligent radiation. We also address the informatics challenges within the cancer field that pursue better treatment decisions and patient outcomes, and focus on how informatics can provide opportunities for cancer research and practices. Finally, we conclude that the interdisciplinary nature of cancer informatics and collaborations are major drivers for future research and applications in clinical practices. It is hoped that this review is instrumental for cancer researchers and clinicians with its informatics-specific insights.
Collapse
Affiliation(s)
- Na Hong
- Department of Medical SciencesDigital Health China Technologies Co., Ltd.BeijingChina
| | - Gang Sun
- Xinjiang Cancer Center, Key Laboratory of Oncology of Xinjiang Uyghur Autonomous RegionThe Affiliated Cancer Hospital of Xinjiang Medical UniversityÜrümqiChina
| | - Xiuran Zuo
- Department of Information, Central Hospital of WuhanTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Meng Chen
- National Cancer Center, National Clinical Research Center for Cancer, Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Li Liu
- Big Data Center, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Jiani Wang
- National Cancer Center, National Clinical Research Center for Cancer, Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiaobin Feng
- Hepato‐Pancreato‐Biliary Center, Beijing Tsinghua Changgung HospitalSchool of Clinical Medicine, Tsinghua UniversityBeijingChina
| | - Wenzhao Shi
- Department of Medical SciencesDigital Health China Technologies Co., Ltd.BeijingChina
| | - Mengchun Gong
- Department of Medical SciencesDigital Health China Technologies Co., Ltd.BeijingChina
- Institute of Health ManagementSouthern Medical UniversityGuangzhouChina
| | - Pengcheng Ma
- Big Data Center, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
241
|
Expert consensus on the clinical application of PI3K/AKT/mTOR inhibitors in the treatment of advanced breast cancer. CANCER INNOVATION 2022; 1:25-54. [PMID: 38089455 PMCID: PMC10686175 DOI: 10.1002/cai2.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/05/2022] [Indexed: 04/07/2024]
Abstract
Phosphoinositide 3-kinase (PI3K)/protein kinase B (PKB or AKT)/mammalian target of rapamycin (mTOR) signaling pathway (PAM pathway) plays an important role in the development of breast cancer and are closely associated with the resistance to endocrine therapy in advanced breast cancer. Therefore, anticancer treatment targeting key molecules in this signaling pathway has become a research hotspot in recent years. Randomized clinical trials have demonstrated that PI3K/AKT/mTOR inhibitors bring significant clinical benefit to patients with advanced breast cancer, especially to those with hormone receptor (HR)-positive, human epidermal growth factor receptor (HER) 2-negative advanced breast cancer. Alpelisib, a PI3K inhibitor, and everolimus, an mTOR inhibitor, have been approved by FDA. Based on their high efficacy and relatively good safety profile, an expanded indication of everolimus in breast cancer has been approved by National Medical Products Administration (NMPA). Alpelisib is expected to be approved in China in the near future. The members of the consensus expert panel reached this consensus to comprehensively define the role of PI3K/AKT/mTOR signaling pathway in breast cancer, efficacy and clinical applications of PI3K/AKT/mTOR inhibitors, management of adverse reactions, and PIK3CA mutation detection, to promote the understanding of PI3K/AKT/mTOR inhibitors for Chinese oncologists, improve clinical decision-making, and prolong the survival of target patient population.
Collapse
|
242
|
Hernandez-Prera JC, Saeed-Vafa D, Heidarian A, Gewandter K, Otto K, Wenig BM. Sclerosing Polycystic Adenoma: Conclusive Clinical and Molecular Evidence of Its Neoplastic Nature. Head Neck Pathol 2022; 16:416-426. [PMID: 34410594 PMCID: PMC9187789 DOI: 10.1007/s12105-021-01374-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/14/2021] [Indexed: 02/06/2023]
Abstract
Sclerosing polycystic adenosis, initially considered a non-neoplastic salivary gland lesion and classified as such in the 2017 WHO Classification of Head and Neck Tumors, has been the subject of controversy regarding its possible neoplastic nature. The reporting of recurrent PI3K pathway alteration represents evidence to support these lesions as being neoplastic and more appropriately referred to as sclerosing polycystic adenoma (SPA). Herein, we provide additional evidence that supports the classification of SPA as a true neoplasm. Eight cases of SPA were identified in our database and consultation files. All cases were subjected to PTEN immunohistochemistry (IHC) and next-generation sequencing (NGS). In addition, one patient underwent genetic counseling and germline testing. The cases included 5 men and 3 women with a mean age of 41 years (range 11-78) and all tumors arose in the parotid gland. One patient had multiple recurrences over a period of 2 years. Morphologically the tumors were circumscribed and characterized by an admixture of acini, ducts and cysts embedded in a fibrotic/sclerotic stroma. The cells lining the ducts and cysts showed variable granular, vacuolated, foamy and apocrine cytoplasmic features, as well as acinar cells contained intracytoplasmic brightly eosinophilic granules. The apocrine intraductal proliferations showed mild to moderate atypia in 6 cases. One case showed overt malignant morphology that ranged from intraductal carcinoma to invasive salivary duct carcinoma. Seven cases tested for PTEN IHC showed loss of nuclear expression in the acinar and ductal cells with retained PTEN expression in the myoepithelial cell and stroma. NGS detected PIK3CA or PIK3R1 genetic alterations in 7 cases, including a novel TFG-PIK3CA fusion. Coexisting PTEN mutations were seen in 4 cases, including in a patient with clinical stigmata of Cowden syndrome and confirmed by germline genetic testing. Our findings herein documented including recurrence of tumor, malignant transformation, high prevalence of PI3K pathway oncogenic alterations and the possible heretofore undescribed association with Cowden syndrome add support to classifying SPA as true neoplasms justifying their designation as adenoma, rather than adenosis.
Collapse
Affiliation(s)
- Juan C Hernandez-Prera
- Department of Pathology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
| | - Daryoush Saeed-Vafa
- Department of Pathology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Amin Heidarian
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Kristen Otto
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Bruce M Wenig
- Department of Pathology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| |
Collapse
|
243
|
Gan M, Zhang C, Qiu L, Wang Y, Bao H, Yu R, Liu R, Wu X, Shao Y, Hou P, Fei Z. Molecular landscape and therapeutic alterations in Asian soft-tissue sarcoma patients. Cancer Med 2022; 11:4070-4078. [PMID: 35586877 PMCID: PMC9636498 DOI: 10.1002/cam4.4725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/27/2022] [Accepted: 03/08/2022] [Indexed: 11/18/2022] Open
Abstract
Background Soft‐tissue sarcoma (STS) is a rare solid malignant tumor with numerous histologic subtypes. Current studies on targeted therapy for STS are in preclinical and early‐phase trials. Genomic differences largely influence the prognosis of patients even with the same subtype. To investigate the genomic alterations (GAs) and the potential of targeted therapy in STS, we analyzed the genomic landscape, the therapeutic GAs, and biomarkers of immunotherapy in Chinese STS patients. Methods Targeted sequencing covering 425 genes was performed, from which we obtained the results of tissue samples from 351 Chinese STS patients of all ages covering different histologic subtypes. Bioinformatics analysis of altered genes with nonsynonymous mutations, copy‐number variations, and gene fusions were performed. OncoKB therapeutic GAs and relevant biomarkers including TMB, MSI, and HRD were further examined for potential targeted therapy. Results In total, 2743 GAs were identified in 330 genes with a median of 6 (1–38) per case. The top 11 frequently altered genes were: TP53, MCL1, MDM2, CDK4, MYC, CDKN2A, GNAS, RB1, ATRX, CDKN2B, and FGFR1. OncoKB defined therapeutic GAs were found in 23 genes in 43% of the patients. In general, 9.4% of the patients had high‐TMB, 2.8% had MSI, and 13.7% had HRD. A significant difference in the percentage of patients with OncoKB therapeutic GAs were observed between the most frequent two subtypes, leiomyosarcoma and liposarcoma. Altogether, 54% of the patients had the potential to respond to a targeted therapy. Conclusion This study indicated the potential efficacy of targeted therapy on many STS patients, and also provided insight for novel precision therapy. The clinical efficacy of combining targeted therapy and immunotherapy can be further investigated.
Collapse
Affiliation(s)
- Meifu Gan
- Department of PathologyTaizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical UniversityLinhaiChina
| | - Chen Zhang
- Radiotherapy & Chemotherapy Department 2, HwaMei HospitalUniversity of Chinese Academy of ScienceNingboChina
| | - Liqing Qiu
- Hangzhou Cancer Institution, Affiliated Hangzhou Cancer HospitalZhejiang University School of MedicineHangzhouChina
| | - Yue Wang
- Geneseeq Research InstituteNanjing Geneseeq Technology IncNanjingChina
| | - Hua Bao
- Geneseeq Research InstituteNanjing Geneseeq Technology IncNanjingChina
| | - Ruoying Yu
- Geneseeq Research InstituteNanjing Geneseeq Technology IncNanjingChina
| | - Rui Liu
- Geneseeq Research InstituteNanjing Geneseeq Technology IncNanjingChina
| | - Xue Wu
- Geneseeq Research InstituteNanjing Geneseeq Technology IncNanjingChina
| | - Yang Shao
- Geneseeq Research InstituteNanjing Geneseeq Technology IncNanjingChina
- School of Public HealthNanjing Medical UniversityNanjingChina
| | - Peifeng Hou
- Department of Medical OncologyFujian Medical University Union HospitalFuzhouChina
| | - Zhenglei Fei
- Anorectal SurgeryNingbo Medical Center Lihuili HospitalNingBoChina
| |
Collapse
|
244
|
Sabbah DA, Samarat HH, Al‐Shalabi E, Bardaweel SK, Hajjo R, Sweidan K, Khalaf RA, Al‐Zuheiri AM, Abushaikha G. Design, Synthesis, and Biological Examination of
N‐
Phenyl‐6‐fluoro‐4‐hydroxy‐2‐quinolone‐3‐carboxamides as Anticancer Agents. ChemistrySelect 2022. [DOI: 10.1002/slct.202200662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Dima A. Sabbah
- Department of Pharmacy Faculty of Pharmacy Institution Al-Zaytoonah University of Jordan Address P.O. Box 130 Amman 11733 Jordan
| | - Hla H. Samarat
- Department of Pharmacy Faculty of Pharmacy Institution Al-Zaytoonah University of Jordan Address P.O. Box 130 Amman 11733 Jordan
| | - Eveen Al‐Shalabi
- Department of Pharmacy Faculty of Pharmacy Institution Al-Zaytoonah University of Jordan Address P.O. Box 130 Amman 11733 Jordan
| | - Sanaa K. Bardaweel
- Department of Pharmaceutical Sciences School of Pharmacy Institution The University of Jordan Address Amman 11942 Jordan
| | - Rima Hajjo
- Department of Pharmacy Faculty of Pharmacy Institution Al-Zaytoonah University of Jordan Address P.O. Box 130 Amman 11733 Jordan
- Laboratory for Molecular Modeling Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy The University of North Carlina at Chapel Hill Chapel Hill NC 27515 USA
| | - Kamal Sweidan
- Department of Chemistry Institution The University of Jordan Address Amman 11942 Jordan
| | - Reema Abu Khalaf
- Department of Pharmacy Faculty of Pharmacy Institution Al-Zaytoonah University of Jordan Address P.O. Box 130 Amman 11733 Jordan
| | - Aya M. Al‐Zuheiri
- Department of Pharmacy Faculty of Pharmacy Institution Al-Zaytoonah University of Jordan Address P.O. Box 130 Amman 11733 Jordan
| | - Ghassan Abushaikha
- Department of Medicinal and Biological Chemistry College of Pharmacy and Pharmaceutical Sciences Institution The University of Toledo Address Toledo OH 43606-3390 USA
| |
Collapse
|
245
|
Mohan P, Pasion J, Ciriello G, Lailler N, de Stanchina E, Viale A, van den Berg A, Diepstra A, Wendel HG, Sanghvi VR, Singh K. Frequent 4EBP1 Amplification Induces Synthetic Dependence on FGFR Signaling in Cancer. Cancers (Basel) 2022; 14:2397. [PMID: 35626002 PMCID: PMC9139685 DOI: 10.3390/cancers14102397] [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: 04/19/2022] [Revised: 05/06/2022] [Accepted: 05/11/2022] [Indexed: 02/04/2023] Open
Abstract
The eIF4E translation initiation factor has oncogenic properties and concordantly, the inhibitory eIF4E-binding protein (4EBP1) is considered a tumor suppressor. The exact molecular effects of 4EBP1 activation in cancer are still unknown. Surprisingly, 4EBP1 is a target of genomic copy number gains (Chr. 8p11) in breast and lung cancer. We noticed that 4EBP1 gains are genetically linked to gains in neighboring genes, including WHSC1L1 and FGFR1. Our results show that FGFR1 gains act to attenuate the function of 4EBP1 via PI3K-mediated phosphorylation at Thr37/46, Ser65, and Thr70 sites. This implies that not 4EBP1 but instead FGFR1 is the genetic target of Chr. 8p11 gains in breast and lung cancer. Accordingly, these tumors show increased sensitivity to FGFR1 and PI3K inhibition, and this is a therapeutic vulnerability through restoring the tumor-suppressive function of 4EBP1. Ribosome profiling reveals genes involved in insulin signaling, glucose metabolism, and the inositol pathway to be the relevant translational targets of 4EBP1. These mRNAs are among the top 200 translation targets and are highly enriched for structure and sequence motifs in their 5'UTR, which depends on the 4EBP1-EIF4E activity. In summary, we identified the translational targets of 4EBP1-EIF4E that facilitate the tumor suppressor function of 4EBP1 in cancer.
Collapse
Affiliation(s)
- Prathibha Mohan
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (P.M.); (J.P.); (H.-G.W.)
| | - Joyce Pasion
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (P.M.); (J.P.); (H.-G.W.)
| | - Giovanni Ciriello
- Department of Computational Biology, University of Lausanne, CH-1005 Lausanne, Switzerland;
| | - Nathalie Lailler
- Integrated Genomics Operation, Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (N.L.); (A.V.)
| | - Elisa de Stanchina
- Molecular Pharmacology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA;
| | - Agnes Viale
- Integrated Genomics Operation, Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (N.L.); (A.V.)
| | - Anke van den Berg
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (A.v.d.B.); (A.D.)
| | - Arjan Diepstra
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands; (A.v.d.B.); (A.D.)
| | - Hans-Guido Wendel
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (P.M.); (J.P.); (H.-G.W.)
| | - Viraj R. Sanghvi
- Department of Molecular and Cellular Pharmacology, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Kamini Singh
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Albert Einstein Cancer Center, Bronx, NY 10461, USA
| |
Collapse
|
246
|
Sibilano M, Tullio V, Adorno G, Savini I, Gasperi V, Catani MV. Platelet-Derived miR-126-3p Directly Targets AKT2 and Exerts Anti-Tumor Effects in Breast Cancer Cells: Further Insights in Platelet-Cancer Interplay. Int J Mol Sci 2022; 23:ijms23105484. [PMID: 35628294 PMCID: PMC9141257 DOI: 10.3390/ijms23105484] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/09/2022] [Accepted: 05/12/2022] [Indexed: 02/07/2023] Open
Abstract
Among the surrounding cells influencing tumor biology, platelets are recognized as novel players as they release microvesicles (MVs) that, once delivered to cancer cells, modulate signaling pathways related to cell growth and dissemination. We have previously shown that physiological delivery of platelet MVs enriched in miR-126 exerted anti-tumor effects in different breast cancer (BC) cell lines. Here, we seek further insight by identifying AKT2 kinase as a novel miR-126-3p direct target, as assessed by bioinformatic analysis and validated by luciferase assay. Both ectopic expression and platelet MV-mediated delivery of miR-126-3p downregulated AKT2 expression, thus suppressing proliferating and invading properties, in either triple negative (BT549 cells) or less aggressive Luminal A (MCF-7 cells) BC subtypes. Accordingly, as shown by bioinformatic analysis, both high miR-126 and low AKT2 levels were associated with favorable long-term prognosis in BC patients. Our results, together with the literature data, indicate that miR-126-3p exerts suppressor activity by specifically targeting components of the PIK3/AKT signaling cascade. Therefore, management of platelet-derived MV production and selective delivery of miR-126-3p to tumor cells may represent a useful tool in multimodal therapeutic approaches in BC patients.
Collapse
Affiliation(s)
- Matteo Sibilano
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.S.); (V.T.); (I.S.)
| | - Valentina Tullio
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.S.); (V.T.); (I.S.)
| | - Gaspare Adorno
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy;
| | - Isabella Savini
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.S.); (V.T.); (I.S.)
| | - Valeria Gasperi
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.S.); (V.T.); (I.S.)
- Correspondence: (V.G.); (M.V.C.); Tel.: +39-06-7259-6465 (V.G.); +39-06-7259-6465 (M.V.C.)
| | - Maria Valeria Catani
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy; (M.S.); (V.T.); (I.S.)
- Correspondence: (V.G.); (M.V.C.); Tel.: +39-06-7259-6465 (V.G.); +39-06-7259-6465 (M.V.C.)
| |
Collapse
|
247
|
Rangwala AM, Mingione VR, Georghiou G, Seeliger MA. Kinases on Double Duty: A Review of UniProtKB Annotated Bifunctionality within the Kinome. Biomolecules 2022; 12:biom12050685. [PMID: 35625613 PMCID: PMC9138534 DOI: 10.3390/biom12050685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/03/2022] [Accepted: 05/09/2022] [Indexed: 01/27/2023] Open
Abstract
Phosphorylation facilitates the regulation of all fundamental biological processes, which has triggered extensive research of protein kinases and their roles in human health and disease. In addition to their phosphotransferase activity, certain kinases have evolved to adopt additional catalytic functions, while others have completely lost all catalytic activity. We searched the Universal Protein Resource Knowledgebase (UniProtKB) database for bifunctional protein kinases and focused on kinases that are critical for bacterial and human cellular homeostasis. These kinases engage in diverse functional roles, ranging from environmental sensing and metabolic regulation to immune-host defense and cell cycle control. Herein, we describe their dual catalytic activities and how they contribute to disease pathogenesis.
Collapse
|
248
|
Damodaran S, Zhao F, Deming DA, Mitchell EP, Wright JJ, Gray RJ, Wang V, McShane LM, Rubinstein LV, Patton DR, Williams PM, Hamilton SR, Suga JM, Conley BA, Arteaga CL, Harris LN, O'Dwyer PJ, Chen AP, Flaherty KT. Phase II Study of Copanlisib in Patients With Tumors With PIK3CA Mutations: Results From the NCI-MATCH ECOG-ACRIN Trial (EAY131) Subprotocol Z1F. J Clin Oncol 2022; 40:1552-1561. [PMID: 35133871 PMCID: PMC9084438 DOI: 10.1200/jco.21.01648] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/15/2021] [Accepted: 01/06/2022] [Indexed: 01/14/2023] Open
Abstract
PURPOSE Activating mutations in PIK3CA are observed across multiple tumor types. The NCI-MATCH (EAY131) is a tumor-agnostic platform trial that enrolls patients to targeted therapies on the basis of matching genomic alterations. Arm Z1F evaluated copanlisib, an α and δ isoform-specific phosphoinositide 3-kinase (PI3K) inhibitor, in patients with PIK3CA mutations (with or without PTEN loss). PATIENTS AND METHODS Patients received copanlisib (60 mg intravenous) once weekly on days 1, 8, and 15 in 28-day cycles until progression or toxicity. Patients with KRAS mutations, human epidermal growth factor receptor 2-positive breast cancers, and lymphomas were excluded. The primary end point was centrally assessed objective response rate (ORR); secondary end points included progression-free survival, 6-month progression-free survival, and overall survival. RESULTS Thirty-five patients were enrolled, and 25 patients were included in the primary efficacy analysis as prespecified in the Protocol. Multiple histologies were enrolled, with gynecologic (n = 6) and gastrointestinal (n = 6) being the most common. Sixty-eight percent of patients had ≥ 3 lines of prior therapy. The ORR was 16% (4 of 25, 90% CI, 6 to 33) with P = .0341 against a null rate of 5%. The most common reason for protocol discontinuation was disease progression (n = 17, 68%). Grade 3/4 toxicities observed were consistent with reported toxicities for PI3K pathway inhibition. Sixteen patients (53%) had grade 3 toxicities, and one patient (3%) had grade 4 toxicity (CTCAE v5.0). Most common toxicities include hyperglycemia (n = 19), fatigue (n = 12), diarrhea (n = 11), hypertension (n = 10), and nausea (n = 10). CONCLUSION The study met its primary end point with an ORR of 16% (P = .0341) with copanlisib showing clinical activity in select tumors with PIK3CA mutation in the refractory setting.
Collapse
Affiliation(s)
| | - Fengmin Zhao
- Dana-Farber Cancer Institute-ECOG-ACRIN Biostatistics Center, Boston, MA
| | | | - Edith P. Mitchell
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - John J. Wright
- Investigational Drug Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Robert J. Gray
- Dana-Farber Cancer Institute-ECOG-ACRIN Biostatistics Center, Boston, MA
| | - Victoria Wang
- Dana-Farber Cancer Institute-ECOG-ACRIN Biostatistics Center, Boston, MA
| | - Lisa M. McShane
- Biometric Research Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Larry V. Rubinstein
- Biometric Research Branch, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - David R. Patton
- National Cancer Institute/Center for Biomedical Informatics & Information Technology, Rockville, MD
| | - P. Mickey Williams
- Molecular Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD
| | | | | | - Barbara A. Conley
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | | | - Lyndsay N. Harris
- Cancer Diagnosis Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Peter J. O'Dwyer
- University of Pennsylvania Abramson Cancer Center, Division of Medical Oncology, Philadelphia, PA
| | - Alice P. Chen
- Developmental Therapeutics Clinic/Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, MD
| | - Keith T. Flaherty
- Dana-Farber Cancer Institute/Harvard Medical School/Massachusetts General Hospital, Boston, MA
| |
Collapse
|
249
|
Gao X, Wang Y, Ribeiro CF, Manokaran C, Chang H, Von T, Rodrigues S, Cizmecioglu O, Jia S, Korpal M, Korn JM, Wang Z, Schmit F, Jiang L, Pagliarini R, Yang Y, Sethi I, Signoretti S, Yuan GC, Loda M, Zhao JJ, Roberts TM. Blocking PI3K p110β Attenuates Development of PTEN-Deficient Castration-Resistant Prostate Cancer. Mol Cancer Res 2022; 20:673-685. [PMID: 35105671 PMCID: PMC9081176 DOI: 10.1158/1541-7786.mcr-21-0322] [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] [Received: 05/05/2021] [Revised: 12/20/2021] [Accepted: 01/26/2022] [Indexed: 11/16/2022]
Abstract
A common outcome of androgen deprivation in prostate cancer therapy is disease relapse and progression to castration-resistant prostate cancer (CRPC) via multiple mechanisms. To gain insight into the recent clinical findings that highlighted genomic alterations leading to hyperactivation of PI3K, we examined the roles of the commonly expressed p110 catalytic isoforms of PI3K in a murine model of Pten-null invasive CRPC. While blocking p110α had negligible effects in the development of Pten-null invasive CRPC, either genetic or pharmacologic perturbation of p110β dramatically slowed CRPC initiation and progression. Once fully established, CRPC tumors became partially resistant to p110β inhibition, indicating the acquisition of new dependencies. Driven by our genomic analyses highlighting potential roles for the p110β/RAC/PAK1 and β-catenin pathways in CRPC, we found that combining p110β with RAC/PAK1 or tankyrase inhibitors significantly reduced the growth of murine and human CRPC organoids in vitro and in vivo. Because p110β activity is dispensable for most physiologic processes, our studies support novel therapeutic strategies both for preventing disease progression into CRPC and for treating CRPC. IMPLICATIONS This work establishes p110β as a promising target for preventing the progression of primary PTEN-deficient prostate tumors to CRPC, and for treating established CRPC in combination with RAC/PAK1 or tankyrase inhibitors.
Collapse
Affiliation(s)
- Xueliang Gao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts USA
- Department of Cell and Molecular Pharmacology & Experimental Therapeutics, Medical University of South Carolina, SC, USA
| | - Yubao Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts USA
| | - Caroline F. Ribeiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
- Department of Pathology, Harvard Medical School, Boston, Massachusetts USA
| | - Cherubin Manokaran
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts USA
| | - Hyeyoun Chang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts USA
| | - Thanh Von
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts USA
| | - Silvia Rodrigues
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
- Department of Pathology, Harvard Medical School, Boston, Massachusetts USA
| | - Onur Cizmecioglu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts USA
| | - Shidong Jia
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
- Department of Pathology, Harvard Medical School, Boston, Massachusetts USA
| | - Manav Korpal
- Oncology Disease Area, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - Joshua M. Korn
- Oncology Disease Area, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - Zhigang Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
| | - Fabienne Schmit
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts USA
| | - Lan Jiang
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
| | - Raymond Pagliarini
- Oncology Disease Area, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - Yi Yang
- Oncology Disease Area, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - Isha Sethi
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
| | - Sabina Signoretti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
- Department of Pathology, Harvard Medical School, Boston, Massachusetts USA
| | - Guo-Cheng Yuan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
| | - Massimo Loda
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
- Department of Pathology, Harvard Medical School, Boston, Massachusetts USA
| | - Jean J. Zhao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts USA
| | - Thomas M. Roberts
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts USA
| |
Collapse
|
250
|
Zhang Q, Shen J, Wu Y, Ruan W, Zhu F, Duan S. LINC00520: A Potential Diagnostic and Prognostic Biomarker in Cancer. Front Immunol 2022; 13:845418. [PMID: 35309319 PMCID: PMC8924041 DOI: 10.3389/fimmu.2022.845418] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/15/2022] [Indexed: 11/13/2022] Open
Abstract
Long non-coding RNA (lncRNA) is important in the study of cancer mechanisms. LINC00520 is located on human chromosome 14q22.3 and is a highly conserved long non-coding RNA. LINC00520 is widely expressed in various tissues. The expression of LINC00520 is regulated by transcription factors such as Sp1, TFAP4, and STAT3. The high expression of LINC00520 is significantly related to the risk of 11 cancers. LINC00520 can competitively bind 10 miRNAs to promote tumor cell proliferation, invasion, and migration. In addition, LINC00520 is involved in the regulation of P13K/AKT and JAK/STAT signaling pathways. The expression of LINC00520 is significantly related to the clinicopathological characteristics and prognosis of tumor patients and is also related to the sensitivity of HNSCC to radiotherapy. Here, this article summarizes the abnormal expression pattern of LINC00520 in cancer and its potential molecular regulation mechanism and points out that LINC00520 can be used as a potential biomarker for cancer diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Qiudan Zhang
- School of Medicine, Zhejiang University City College, Hangzhou, China.,Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, China
| | - Jinze Shen
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Yuchen Wu
- Department of Clinical Medicine, The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Wenjing Ruan
- Department of Respiratory Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Feng Zhu
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Shiwei Duan
- School of Medicine, Zhejiang University City College, Hangzhou, China.,Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, China
| |
Collapse
|