1
|
Linscott MP, Ren JR, Gestl SA, Gunther EJ. Different Oncogenes and Reproductive Histories Shape the Progression of Distinct Premalignant Clones in Multistage Mouse Breast Cancer Models. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1329-1345. [PMID: 38537934 PMCID: PMC11220927 DOI: 10.1016/j.ajpath.2024.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/06/2024] [Accepted: 02/16/2024] [Indexed: 04/10/2024]
Abstract
A remote carcinogen exposure can predispose to breast cancer onset decades later, suggesting that carcinogen-induced mutations generate long-lived premalignant clones. How subsequent events influence the progression of specific premalignant clones remains poorly understood. Herein, multistage mouse models of mammary carcinogenesis were generated by combining chemical carcinogen exposure [using 7,12-dimethylbenzanthracene (DMBA)] with transgenes that enable inducible expression of one of two clinically relevant mammary oncogenes: c-MYC (MYC) or PIK3CAH1047R (PIK). In prior work, DMBA exposure generated mammary clones bearing signature HrasQ61L mutations, which only progressed to mammary cancer after inducible Wnt1 oncogene expression. Here, after an identical DMBA exposure, MYC versus PIK drove cancer progression from mammary clones bearing mutations in distinct Ras family paralogs. For example, MYC drove cancer progression from either Kras- or Nras-mutant clones, whereas PIK transformed Kras-mutant clones only. These Ras mutation patterns were maintained whether oncogenic transgenes were induced within days of DMBA exposure or months later. Completing a full-term pregnancy (parity) failed to protect against either MYC- or PIK-driven tumor progression. Instead, a postpartum increase in mammary tumor predisposition was observed in the context of PIK-driven progression. However, parity decreased the overall prevalence of tumors bearing Krasmut, and the magnitude of this decrease depended on both the number and timing of pregnancies. These multistage models may be useful for elucidating biological features of premalignant mammary neoplasia.
Collapse
Affiliation(s)
- Maryknoll P Linscott
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, Pennsylvania; Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Jerry R Ren
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, Pennsylvania; Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Shelley A Gestl
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, Pennsylvania; Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Edward J Gunther
- The Jake Gittlen Laboratories for Cancer Research, Pennsylvania State University College of Medicine, Hershey, Pennsylvania; Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, Pennsylvania; Department of Medicine, Pennsylvania State University College of Medicine, Hershey, Pennsylvania.
| |
Collapse
|
2
|
Chen J, Zhang X, Tan X, Liu P. Somatic gain-of-function mutations in BUD13 promote oncogenesis by disrupting Fbw7 function. J Exp Med 2023; 220:e20222056. [PMID: 37382881 PMCID: PMC10309187 DOI: 10.1084/jem.20222056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/13/2023] [Accepted: 05/19/2023] [Indexed: 06/30/2023] Open
Abstract
Somatic mutations occurring on key enzymes are extensively studied and targeted therapies are developed with clinical promises. However, context-dependent enzyme function through distinct substrates complicated targeting a given enzyme. Here, we develop an algorithm to elucidate a new class of somatic mutations occurring on enzyme-recognizing motifs that cancer may hijack to facilitate tumorigenesis. We validate BUD13-R156C and -R230Q mutations evading RSK3-mediated phosphorylation with enhanced oncogenicity in promoting colon cancer growth. Further mechanistic studies reveal BUD13 as an endogenous Fbw7 inhibitor that stabilizes Fbw7 oncogenic substrates, while cancerous BUD13-R156C or -R230Q interferes with Fbw7Cul1 complex formation. We also find this BUD13 regulation plays a critical role in responding to mTOR inhibition, which can be used to guide therapy selections. We hope our studies reveal the landscape of enzyme-recognizing motif mutations with a publicly available resource and provide novel insights for somatic mutations cancer hijacks to promote tumorigenesis with the potential for patient stratification and cancer treatment.
Collapse
Affiliation(s)
- Jianfeng Chen
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xinyi Zhang
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Xianming Tan
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Pengda Liu
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
3
|
Cheng X, Sun Y, Highkin M, Vemalapally N, Jin X, Zhou B, Prior JL, Tipton AR, Li S, Iliuk A, Achilefu S, Hagemann IS, Edwards JR, Bose R. Breast Cancer Mutations HER2V777L and PIK3CAH1047R Activate the p21-CDK4/6-Cyclin D1 Axis to Drive Tumorigenesis and Drug Resistance. Cancer Res 2023; 83:2839-2857. [PMID: 37272756 PMCID: PMC10527017 DOI: 10.1158/0008-5472.can-22-3558] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 04/12/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023]
Abstract
In metastatic breast cancer, HER2-activating mutations frequently co-occur with mutations in PIK3CA, TP53, or CDH1. Of these co-occurring mutations, HER2 and PIK3CA are the most commonly comutated gene pair, with approximately 40% of HER2-mutated breast cancers also having activating mutations in PIK3CA. To study the effects of co-occurring HER2 and PIK3CA mutations, we generated genetically engineered mice with the HER2V777L; PIK3CAH1047R transgenes (HP mice) and studied the resulting breast cancers both in vivo as well as ex vivo using cancer organoids. HP breast cancers showed accelerated tumor formation in vivo and increased invasion and migration in in vitro assays. HP breast cancer cells were resistant to the pan-HER tyrosine kinase inhibitor, neratinib, but were effectively treated with neratinib plus the HER2-targeted antibody-drug conjugate trastuzumab deruxtecan. Proteomic and RNA-seq analysis of HP breast cancers identified increased gene expression of cyclin D1 and p21WAF1/Cip1 and changes in cell-cycle markers. Combining neratinib with CDK4/6 inhibitors was another effective strategy for treating HP breast cancers, with neratinib plus palbociclib showing a statistically significant reduction in development of mouse HP tumors as compared to either drug alone. The efficacy of both the neratinib plus trastuzumab deruxtecan and neratinib plus palbociclib combinations was validated using a human breast cancer patient-derived xenograft with very similar HER2 and PIK3CA mutations to the HP mice. Further, these two drug combinations effectively treated spontaneous lung metastasis in syngeneic mice transplanted with HP breast cancer organoids. This study provides valuable preclinical data to support the ongoing phase 1 clinical trials of these drug combinations in breast cancer. SIGNIFICANCE In HER2-mutated breast cancer, PIK3CA mutation activates p21-CDK4/6-cyclin D1 signaling to drive resistance to HER2-targeted therapies, which can be overcome using CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Xiaoqing Cheng
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Yirui Sun
- Center for Pharmacogenomics, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Maureen Highkin
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Nagalaxmi Vemalapally
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Xiaohua Jin
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Brandon Zhou
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Julie L. Prior
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Ashley R. Tipton
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Shunqiang Li
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Anton Iliuk
- Tymora Analytical Operations, 1201 Cumberland Ave. West Lafayette, IN 47906
| | - Samuel Achilefu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Ian S. Hagemann
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110
| | - John R. Edwards
- Center for Pharmacogenomics, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110
| | - Ron Bose
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
4
|
Zhang J, Croft J, Le A. Familial CCM Genes Might Not Be Main Drivers for Pathogenesis of Sporadic CCMs-Genetic Similarity between Cancers and Vascular Malformations. J Pers Med 2023; 13:jpm13040673. [PMID: 37109059 PMCID: PMC10143507 DOI: 10.3390/jpm13040673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/05/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
Cerebral cavernous malformations (CCMs) are abnormally dilated intracranial capillaries that form cerebrovascular lesions with a high risk of hemorrhagic stroke. Recently, several somatic "activating" gain-of-function (GOF) point mutations in PIK3CA (phosphatidylinositol-4, 5-bisphosphate 3-kinase catalytic subunit p110α) were discovered as a dominant mutation in the lesions of sporadic forms of cerebral cavernous malformation (sCCM), raising the possibility that CCMs, like other types of vascular malformations, fall in the PIK3CA-related overgrowth spectrum (PROS). However, this possibility has been challenged with different interpretations. In this review, we will continue our efforts to expound the phenomenon of the coexistence of gain-of-function (GOF) point mutations in the PIK3CA gene and loss-of-function (LOF) mutations in CCM genes in the CCM lesions of sCCM and try to delineate the relationship between mutagenic events with CCM lesions in a temporospatial manner. Since GOF PIK3CA point mutations have been well studied in reproductive cancers, especially breast cancer as a driver oncogene, we will perform a comparative meta-analysis for GOF PIK3CA point mutations in an attempt to demonstrate the genetic similarities shared by both cancers and vascular anomalies.
Collapse
Affiliation(s)
- Jun Zhang
- Departments of Molecular & Translational Medicine (MTM), Texas Tech University Health Science Center El Paso (TTUHSCEP), El Paso, TX 79905, USA
| | - Jacob Croft
- Departments of Molecular & Translational Medicine (MTM), Texas Tech University Health Science Center El Paso (TTUHSCEP), El Paso, TX 79905, USA
| | - Alexander Le
- Departments of Molecular & Translational Medicine (MTM), Texas Tech University Health Science Center El Paso (TTUHSCEP), El Paso, TX 79905, USA
| |
Collapse
|
5
|
Simond AM, Bui T, Zuo D, Sanguin-Gendreau V, Rao T, Phillips WA, Cardiff RD, Muller WJ. Physiological expression of PI3K H1047R mutation reveals its anti-metastatic potential in ErbB2-driven breast cancer. Oncogene 2022; 41:3445-3451. [PMID: 35538223 DOI: 10.1038/s41388-022-02323-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 03/11/2022] [Accepted: 04/12/2022] [Indexed: 12/24/2022]
Abstract
p110α is a catalytic subunit of phosphoinositide 3-kinase (PI3K), a major downstream effector of receptor tyrosine kinase ErbB2, that is amplified and overexpressed in 20-30% of breast cancers, 40% of which have an activating mutation in p110α. Despite the high frequency of PIK3CA gain-of-function mutations, their prognostic value is controversial. Here, we employ a knock-in transgenic strategy to restrict the expression of an activated form of ErbB2 and p110α kinase domain mutation (p110αHR) in the mammary epithelium. Physiological levels of transgene expression under the control of their endogenous promoters did not result in a major synergistic effect. However, tumors arising in ErbB2/p110αHR bi-genic strain metastasized to the lung with significantly reduced capacity compared to tumors expressing ErbB2 alone. The reduced metastasis was further associated with retention of the myoepithelial layer reminiscent of ductal carcinoma in situ (DCIS), a non-invasive stage of human breast cancer. Molecular and biochemical analyses revealed that these poorly metastatic tumors exhibited a significant decrease in phospho-myosin light chain 2 (MLC2) associated with cellular contractility and migration. Examination of human samples for MLC2 activity revealed a progressive increase in cellular contractility between non-invasive DCIS and invasive ductal carcinoma. Collectively, these data argue that p110αHR mutation attenuates metastatic behavior in the context of ErbB2-driven breast cancer.
Collapse
Affiliation(s)
- Alexandra M Simond
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Tung Bui
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada.,Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Dongmei Zuo
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada
| | | | - Trisha Rao
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada
| | - Wayne A Phillips
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.,The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Robert D Cardiff
- Center for Comparative Medicine, University of California, Davis, CA, USA
| | - William J Muller
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montreal, QC, Canada. .,Department of Biochemistry, McGill University, Montreal, QC, Canada. .,Faculty of Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
6
|
Zilli F, Marques Ramos P, Auf der Maur P, Jehanno C, Sethi A, Coissieux M, Eichlisberger T, Sauteur L, Rouchon A, Bonapace L, Pinto Couto J, Rad R, Jensen MR, Banfi A, Stadler MB, Bentires‐Alj M. The NFIB-ERO1A axis promotes breast cancer metastatic colonization of disseminated tumour cells. EMBO Mol Med 2021; 13:e13162. [PMID: 33751828 PMCID: PMC8033524 DOI: 10.15252/emmm.202013162] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
Metastasis is the main cause of deaths related to solid cancers. Active transcriptional programmes are known to regulate the metastatic cascade but the molecular determinants of metastatic colonization remain elusive. Using an inducible piggyBac (PB) transposon mutagenesis screen, we have shown that overexpression of the transcription factor nuclear factor IB (NFIB) alone is sufficient to enhance primary mammary tumour growth and lung metastatic colonization. Mechanistically and functionally, NFIB directly increases expression of the oxidoreductase ERO1A, which enhances HIF1α-VEGFA-mediated angiogenesis and colonization, the last and fatal step of the metastatic cascade. NFIB is thus clinically relevant: it is preferentially expressed in the poor-prognostic group of basal-like breast cancers, and high expression of the NFIB/ERO1A/VEGFA pathway correlates with reduced breast cancer patient survival.
Collapse
Affiliation(s)
- Federica Zilli
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Pedro Marques Ramos
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
- Novartis Institutes for Biomedical ResearchBaselSwitzerland
| | - Priska Auf der Maur
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
| | - Charly Jehanno
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
| | - Atul Sethi
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
- Swiss Institute of BioinformaticsBaselSwitzerland
| | - Marie‐May Coissieux
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | | | - Loïc Sauteur
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
| | - Adelin Rouchon
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
| | - Laura Bonapace
- Novartis Institutes for Biomedical ResearchBaselSwitzerland
| | - Joana Pinto Couto
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
- Novartis Institutes for Biomedical ResearchBaselSwitzerland
| | - Roland Rad
- Department of Medicine IITUM School of MedicineInstitute of Molecular Oncology and Functional GenomicsCenter for Translational Cancer Research (TranslaTUM)Technische Universität MünchenMünchenGermany
- German Cancer Consortium (DKTK)German Cancer Research Center (DKFZ)HeidelbergGermany
| | | | - Andrea Banfi
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
| | - Michael B Stadler
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
- Swiss Institute of BioinformaticsBaselSwitzerland
| | - Mohamed Bentires‐Alj
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| |
Collapse
|
7
|
PIK3CA Mutation Assessment in HR+/HER2− Metastatic Breast Cancer: Overview for Oncology Clinical Practice. JOURNAL OF MOLECULAR PATHOLOGY 2021. [DOI: 10.3390/jmp2010005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Activation of the PI3K–AKT–mTOR pathway occurs in several human cancers, including hormone receptor (HR)-positive breast cancer (BC) where is associated with resistance to endocrine therapy and disease progression. In BC, the most common PI3K–AKT–mTOR pathway alteration is represented by PIK3CA oncogenic mutations. These mutations can occur throughout several domains of the p110α catalytic subunit, but the majority are found in the helical and kinase domains (exon 9 and 20) that represent the “hotspots”. Considering the central role of the PI3K–AKT–mTOR pathway in HR-positive BC, several inhibitors (both pan-PI3K and isoform-specific) have been developed and tested in clinical trials. Recently, the PI3Kα-selective inhibitor alpelisib was the first PI3K inhibitor approved for clinical use in HR-positive metastatic BC based on the results of the phase III SOLAR-1 trial. Several methods to assess PIK3CA mutational status in tumor samples have been developed and validated, including real-time polymerase chain reaction (PCR), digital droplet PCR (ddPCR), BEAMing assays, Sanger sequencing, and next-generation sequencing (NGS) panels. Several new challenges will be expected once alpelisib is widely available in a clinical setting, including the harmonization of testing procedures for the detection of PI3K–AKT–mTOR pathway alterations. Herein, we provide an overview on PI3K–AKT–mTOR pathway alterations in HR-positive BC, discuss their role in determining prognosis and resistance to endocrine therapy and highlight practical considerations about diagnostic methods for the detection of PI3K–AKT–mTOR pathway activation status.
Collapse
|
8
|
Abstract
One of the hallmarks of hormone receptor (HR)-positive breast cancer is its dependence on the phosphatidylinositol-3-kinase (PI3K) pathway. Here, we review the epidemiologic, functional, and pharmacologic interactions between oncogenic PI3K and the estrogen receptor (ER). We discuss the epidemiology of PI3K pathway alterations, mechanisms of resistance to PI3K inhibitors, and the current mechanistic landscape of crosstalk between PI3K and ER, which provide the rationale for dual ER and PI3K inhibition and is now a standard of care in the treatment of ER+ PIK3CA-mutant metastatic breast cancer. We outline newer studies in this field that delineate the clinically relevant overlaps between PI3K and parallel signaling pathways, insulin signaling, and ER epigenetic modifiers. We also identify several caveats with the current data and propose new strategies to overcome these bottlenecks.
Collapse
Affiliation(s)
- N Vasan
- Human Oncology and Pathogenesis Program, New York, USA
- Departments of Medicine, New York, USA
| | - E Toska
- Human Oncology and Pathogenesis Program, New York, USA
| | - M Scaltriti
- Human Oncology and Pathogenesis Program, New York, USA
- Departments of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| |
Collapse
|
9
|
Shin YJ, Sa JK, Lee Y, Kim D, Chang N, Cho HJ, Son M, Oh MYT, Shin K, Lee JK, Park J, Jo YK, Kim M, Paddison PJ, Tergaonkar V, Lee J, Nam DH. PIP4K2A as a negative regulator of PI3K in PTEN -deficient glioblastoma. J Exp Med 2019; 216:1120-1134. [PMID: 30898893 PMCID: PMC6504209 DOI: 10.1084/jem.20172170] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 08/20/2018] [Accepted: 02/27/2019] [Indexed: 01/01/2023] Open
Abstract
Glioblastoma (GBM) is the most malignant brain tumor with profound genomic alterations. Tumor suppressor genes regulate multiple signaling networks that restrict cellular proliferation and present barriers to malignant transformation. While bona fide tumor suppressors such as PTEN and TP53 often undergo inactivation due to mutations, there are several genes for which genomic deletion is the primary route for tumor progression. To functionally identify putative tumor suppressors in GBM, we employed in vivo RNAi screening using patient-derived xenograft models. Here, we identified PIP4K2A, whose functional role and clinical relevance remain unexplored in GBM. We discovered that PIP4K2A negatively regulates phosphoinositide 3-kinase (PI3K) signaling via p85/p110 component degradation in PTEN-deficient GBMs and specifically targets p85 for proteasome-mediated degradation. Overexpression of PIP4K2A suppressed cellular and clonogenic growth in vitro and impeded tumor growth in vivo. Our results unravel a novel tumor-suppressive role of PIP4K2A for the first time and support the feasibility of combining oncogenomics with in vivo RNAi screen.
Collapse
Affiliation(s)
- Yong Jae Shin
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jason K Sa
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Yeri Lee
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Donggeon Kim
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | | | - Hee Jin Cho
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Miseol Son
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
- Division of Cancer Cell Signaling, Institute of Molecular and Cell Biology, Singapore
| | - Michael Y T Oh
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY
| | - Kayoung Shin
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University, Seoul, Korea
| | - Jin-Ku Lee
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Jiwon Park
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
| | - Yoon Kyung Jo
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Misuk Kim
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
| | - Patrick J Paddison
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Vinay Tergaonkar
- Division of Cancer Cell Signaling, Institute of Molecular and Cell Biology, Singapore
- Department of Pathology, National University of Singapore, Singapore
| | - Jeongwu Lee
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Do-Hyun Nam
- Institute for Refractory Cancer Research, Samsung Medical Center, Seoul, Korea
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, Korea
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
10
|
Papa A, Pandolfi PP. The PTEN⁻PI3K Axis in Cancer. Biomolecules 2019; 9:biom9040153. [PMID: 30999672 PMCID: PMC6523724 DOI: 10.3390/biom9040153] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/25/2022] Open
Abstract
The PI3K-AKT-mTOR signal transduction pathway regulates a variety of biological processes including cell growth, cell cycle progression and proliferation, cellular metabolism, and cytoskeleton reorganization. Fine-tuning of the phosphatidylinositol 3-kinase (PI3K) pathway signaling output is essential for the maintenance of tissue homeostasis and uncontrolled activation of this cascade leads to a number of human pathologies including cancer. Inactivation of the tumor suppressor phosphatase and tensin homologue deleted on Chromosome 10 (PTEN) and/or activating mutations in the proto-typical lipid kinase PI3K have emerged as some of the most frequent events associated with human cancer and as a result the PI3K pathway has become a highly sought-after target for cancer therapies. In this review we summarize the essential role of the PTEN-PI3K axis in controlling cellular behaviors by modulating activation of key proto-oncogenic molecular nodes and functional targets. Further, we highlight important functional redundancies and peculiarities of these two critical enzymes that over the last few decades have become a central part of the cancer research field and have instructed hundreds of pre-clinical and clinical trials to better cancer treatments.
Collapse
Affiliation(s)
- Antonella Papa
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria 3800, Australia.
| | - Pier Paolo Pandolfi
- Cancer Research Institute, Beth Israel Deaconess Cancer Center, Department of Medicine and Pathology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
11
|
Cell Reprogramming in Tumorigenesis and Its Therapeutic Implications for Breast Cancer. Int J Mol Sci 2019; 20:ijms20081827. [PMID: 31013830 PMCID: PMC6515165 DOI: 10.3390/ijms20081827] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/01/2019] [Accepted: 04/10/2019] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most common malignancy in women worldwide and can be categorized into several subtypes according to histopathological parameters or genomic signatures. Such heterogeneity of breast cancer can arise from the reactivation of mammary stem cells in situ during tumorigenesis. Moreover, different breast cancer subtypes exhibit varieties of cancer incidence, therapeutic response, and patient prognosis, suggesting that a specific therapeutic protocol is required for each breast cancer subtype. Recent studies using molecular and cellular assays identified a link between specific genetic/epigenetic alterations and distinct cells of origin of breast cancer subtypes. These alterations include oncogenes, tumor suppressor genes, and cell-lineage determinants, which can induce cell reprogramming (dedifferentiation and transdifferentiation) among two lineage-committed mammary epithelial cells, namely basal and luminal cells. The interconversion of cell states through cell reprogramming into the intermediates of mammary stem cells can give rise to heterogeneous breast cancers that complicate effective therapies of breast cancer. A better understanding of mechanisms underlying cell reprogramming in breast cancer can help in not only elucidating tumorigenesis but also developing therapeutics for breast cancer. This review introduces recent findings on cancer gene-mediated cell reprogramming in breast cancer and discusses the therapeutic potential of targeting cell reprogramming.
Collapse
|
12
|
Liu XL, Liu JL, Xu YC, Zhang X, Wang YX, Qing LH, Guo W, Ding J, Meng LH. Membrane metallo-endopeptidase mediates cellular senescence induced by oncogenic PIK3CA
H1047R
accompanied with pro-tumorigenic secretome. Int J Cancer 2019; 145:817-829. [DOI: 10.1002/ijc.32153] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 12/05/2018] [Accepted: 01/08/2019] [Indexed: 01/07/2023]
Affiliation(s)
- Xue-Ling Liu
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research; Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai People's Republic of China
- University of Chinese Academy of Sciences; Beijing People's Republic of China
| | - Jia-Li Liu
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research; Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai People's Republic of China
| | - Yi-Chao Xu
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research; Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai People's Republic of China
| | - Xi Zhang
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research; Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai People's Republic of China
| | - Yu-Xiang Wang
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research; Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai People's Republic of China
| | - Li-Hua Qing
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research; Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai People's Republic of China
| | - Wei Guo
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research; Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai People's Republic of China
| | - Jian Ding
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research; Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai People's Republic of China
- University of Chinese Academy of Sciences; Beijing People's Republic of China
| | - Ling-Hua Meng
- Division of Anti-tumor Pharmacology, State Key Laboratory of Drug Research; Shanghai Institute of Materia Medica, Chinese Academy of Sciences; Shanghai People's Republic of China
- University of Chinese Academy of Sciences; Beijing People's Republic of China
| |
Collapse
|
13
|
McNeill RS, Stroobant EE, Smithberger E, Canoutas DA, Butler MK, Shelton AK, Patel SD, Limas JC, Skinner KR, Bash RE, Schmid RS, Miller CR. PIK3CA missense mutations promote glioblastoma pathogenesis, but do not enhance targeted PI3K inhibition. PLoS One 2018; 13:e0200014. [PMID: 29975751 PMCID: PMC6033446 DOI: 10.1371/journal.pone.0200014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 06/18/2018] [Indexed: 12/12/2022] Open
Abstract
Background Glioblastoma (GBM) is the most common adult primary brain tumor. Multimodal treatment is empiric and prognosis remains poor. Recurrent PIK3CA missense mutations (PIK3CAmut) in GBM are restricted to three functional domains: adaptor binding (ABD), helical, and kinase. Defining how these mutations influence gliomagenesis and response to kinase inhibitors may aid in the clinical development of novel targeted therapies in biomarker-stratified patients. Methods We used normal human astrocytes immortalized via expression of hTERT, E6, and E7 (NHA). We selected two PIK3CAmut from each of 3 mutated domains and induced their expression in NHA with (NHARAS) and without mutant RAS using lentiviral vectors. We then examined the role of PIK3CAmut in gliomagenesis in vitro and in mice, as well as response to targeted PI3K (PI3Ki) and MEK (MEKi) inhibitors in vitro. Results PIK3CAmut, particularly helical and kinase domain mutations, potentiated proximal PI3K signaling and migration of NHA and NHARASin vitro. Only kinase domain mutations promoted NHA colony formation, but both helical and kinase domain mutations promoted NHARAS tumorigenesis in vivo. PIK3CAmut status had minimal effects on PI3Ki and MEKi efficacy. However, PI3Ki/MEKi synergism was pronounced in NHA and NHARAS harboring ABD or helical mutations. Conclusion PIK3CAmut promoted differential gliomagenesis based on the mutated domain. While PIK3CAmut did not influence sensitivity to single agent PI3Ki, they did alter PI3Ki/MEKi synergism. Taken together, our results demonstrate that a subset of PIK3CAmut promote tumorigenesis and suggest that patients with helical domain mutations may be most sensitive to dual PI3Ki/MEKi treatment.
Collapse
Affiliation(s)
- Robert S McNeill
- Pathobiology and Translational Science Graduate Program, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Emily E Stroobant
- Department of Chemistry, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Erin Smithberger
- Pathobiology and Translational Science Graduate Program, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Demitra A Canoutas
- Department of Biology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Madison K Butler
- Department of Biology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Abigail K Shelton
- Pathobiology and Translational Science Graduate Program, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Shrey D Patel
- Department of Chemistry, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Juanita C Limas
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Kasey R Skinner
- Neurosciences Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Ryan E Bash
- Departments of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - Ralf S Schmid
- Neurosciences Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America.,Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| | - C Ryan Miller
- Pathobiology and Translational Science Graduate Program, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America.,Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America.,Neurosciences Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America.,Departments of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America.,Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America.,Department of Neurology, University of North Carolina School of Medicine, Chapel Hill, NC, United States of America
| |
Collapse
|
14
|
Zhang Z, Christin JR, Wang C, Ge K, Oktay MH, Guo W. Mammary-Stem-Cell-Based Somatic Mouse Models Reveal Breast Cancer Drivers Causing Cell Fate Dysregulation. Cell Rep 2018; 16:3146-3156. [PMID: 27653681 DOI: 10.1016/j.celrep.2016.08.048] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 07/27/2016] [Accepted: 08/16/2016] [Indexed: 01/23/2023] Open
Abstract
Cancer genomics has provided an unprecedented opportunity for understanding genetic causes of human cancer. However, distinguishing which mutations are functionally relevant to cancer pathogenesis remains a major challenge. We describe here a mammary stem cell (MaSC) organoid-based approach for rapid generation of somatic genetically engineered mouse models (GEMMs). By using RNAi and CRISPR-mediated genome engineering in MaSC-GEMMs, we have discovered that inactivation of Ptpn22 or Mll3, two genes mutated in human breast cancer, greatly accelerated PI3K-driven mammary tumorigenesis. Using these tumor models, we have also identified genetic alterations promoting tumor metastasis and causing resistance to PI3K-targeted therapy. Both Ptpn22 and Mll3 inactivation resulted in disruption of mammary gland differentiation and an increase in stem cell activity. Mechanistically, Mll3 deletion enhanced stem cell activity through activation of the HIF pathway. Thus, our study has established a robust in vivo platform for functional cancer genomics and has discovered functional breast cancer mutations.
Collapse
Affiliation(s)
- Zheng Zhang
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - John R Christin
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Chunhui Wang
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Kai Ge
- Adipocyte Biology and Gene Regulation Section, Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20814, USA
| | - Maja H Oktay
- Department of Pathology, Albert Einstein College of Medicine/Montefiore Medical Center, Bronx, NY 10467, USA; Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Wenjun Guo
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
15
|
Berenjeno IM, Piñeiro R, Castillo SD, Pearce W, McGranahan N, Dewhurst SM, Meniel V, Birkbak NJ, Lau E, Sansregret L, Morelli D, Kanu N, Srinivas S, Graupera M, Parker VER, Montgomery KG, Moniz LS, Scudamore CL, Phillips WA, Semple RK, Clarke A, Swanton C, Vanhaesebroeck B. Oncogenic PIK3CA induces centrosome amplification and tolerance to genome doubling. Nat Commun 2017; 8:1773. [PMID: 29170395 PMCID: PMC5701070 DOI: 10.1038/s41467-017-02002-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 11/01/2017] [Indexed: 01/01/2023] Open
Abstract
Mutations in PIK3CA are very frequent in cancer and lead to sustained PI3K pathway activation. The impact of acute expression of mutant PIK3CA during early stages of malignancy is unknown. Using a mouse model to activate the Pik3ca H1047R hotspot mutation in the heterozygous state from its endogenous locus, we here report that mutant Pik3ca induces centrosome amplification in cultured cells (through a pathway involving AKT, ROCK and CDK2/Cyclin E-nucleophosmin) and in mouse tissues, and increased in vitro cellular tolerance to spontaneous genome doubling. We also present evidence that the majority of PIK3CA H1047R mutations in the TCGA breast cancer cohort precede genome doubling. These previously unappreciated roles of PIK3CA mutation show that PI3K signalling can contribute to the generation of irreversible genomic changes in cancer. While this can limit the impact of PI3K-targeted therapies, these findings also open the opportunity for therapeutic approaches aimed at limiting tumour heterogeneity and evolution.
Collapse
Affiliation(s)
- Inma M Berenjeno
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London, London, WC1E 6DD, UK.
| | - Roberto Piñeiro
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London, London, WC1E 6DD, UK
- Roche-Chus Joint Unit, Complexo Hospitalario Universitario de Santiago de Compostela, Travesía da Choupana S/N, 15706, Santiago de Compostela, Spain
| | - Sandra D Castillo
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London, London, WC1E 6DD, UK
| | - Wayne Pearce
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London, London, WC1E 6DD, UK
| | - Nicholas McGranahan
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UCL Cancer Institute and Hospitals, 72 Huntley Street, London, WC1E 6DD, UK
| | - Sally M Dewhurst
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UCL Cancer Institute and Hospitals, 72 Huntley Street, London, WC1E 6DD, UK
| | - Valerie Meniel
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Nicolai J Birkbak
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UCL Cancer Institute and Hospitals, 72 Huntley Street, London, WC1E 6DD, UK
| | - Evelyn Lau
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London, London, WC1E 6DD, UK
| | - Laurent Sansregret
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London, London, WC1E 6DD, UK
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UCL Cancer Institute and Hospitals, 72 Huntley Street, London, WC1E 6DD, UK
| | - Daniele Morelli
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London, London, WC1E 6DD, UK
| | - Nnennaya Kanu
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UCL Cancer Institute and Hospitals, 72 Huntley Street, London, WC1E 6DD, UK
| | - Shankar Srinivas
- Department of Physiology Anatomy and Genetics, University of Oxford, Oxford, OX1 2JD, UK
| | - Mariona Graupera
- Vascular Signalling Laboratory, Institut d´Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, 08908, Spain
| | - Victoria E R Parker
- Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Karen G Montgomery
- Cancer Biology and Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
| | - Larissa S Moniz
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London, London, WC1E 6DD, UK
| | | | - Wayne A Phillips
- Cancer Biology and Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, 3000, VIC, Australia
| | - Robert K Semple
- Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Alan Clarke
- European Cancer Stem Cell Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Charles Swanton
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London, London, WC1E 6DD, UK.
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UCL Cancer Institute and Hospitals, 72 Huntley Street, London, WC1E 6DD, UK.
| | - Bart Vanhaesebroeck
- UCL Cancer Institute, Paul O'Gorman Building, University College London, 72 Huntley Street London, London, WC1E 6DD, UK.
| |
Collapse
|
16
|
Merino VF, Cho S, Liang X, Park S, Jin K, Chen Q, Pan D, Zahnow CA, Rein AR, Sukumar S. Inhibitors of STAT3, β-catenin, and IGF-1R sensitize mouse PIK3CA-mutant breast cancer to PI3K inhibitors. Mol Oncol 2017; 11:552-566. [PMID: 28296140 PMCID: PMC5527464 DOI: 10.1002/1878-0261.12053] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 02/02/2017] [Accepted: 02/28/2017] [Indexed: 12/15/2022] Open
Abstract
Although mutations in the phosphoinositide 3‐kinase catalytic subunit (PIK3CA) are common in breast cancer, PI3K inhibitors alone have shown modest efficacy. We sought to identify additional pathways altered in PIK3CA‐mutant tumors that might be targeted in combination with PI3K inhibitors. We generated two transgenic mouse models expressing the human PIK3CA‐H1047R‐ and the ‐E545K hotspot‐mutant genes in the mammary gland and evaluated their effects on development and tumor formation. Molecular analysis identified pathways altered in these mutant tumors, which were also targeted in multiple cell lines derived from the PIK3CA tumors. Finally, public databases were analyzed to determine whether novel pathways identified in the mouse tumors were altered in human tumors harboring mutant PIK3CA. Mutant mice showed increased branching and delayed involution of the mammary gland compared to parental FVB/N mice. Mammary tumors arose in 30% of the MMTV‐PIK3CA‐H1047R and in 13% of ‐E545K mice. Compared to MMTV‐Her‐2 transgenic mouse mammary tumors, H1047R tumors showed increased upregulation of Wnt/β‐catenin/Axin2, hepatocyte growth factor (Hgf)/Stat3, insulin‐like growth factor 2 (Igf‐2), and Igf‐1R pathways. Inhibitors of STAT3, β‐catenin, and IGF‐1R sensitized H1047R‐derived mouse tumor cells and PIK3CA‐H1047R overexpressing human HS578T breast cancer cells to the cytotoxic effects of PI3K inhibitors. Analysis of The Cancer Genome Atlas database showed that, unlike primary PIK3CA‐wild‐type and HER‐2+ breast carcinomas, PIK3CA‐mutant tumors display increased expression of AXIN2, HGF, STAT3, IGF‐1, and IGF‐2 mRNA and activation of AKT, IGF1‐MTOR, and WNT canonical signaling pathways. Drugs targeting additional pathways that are altered in PIK3CA‐mutant tumors may improve treatment regimens using PI3K inhibitors alone.
Collapse
Affiliation(s)
- Vanessa F Merino
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Soonweng Cho
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiaohui Liang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sunju Park
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kideok Jin
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Qian Chen
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Duojia Pan
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cynthia A Zahnow
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alan R Rein
- HIV Dynamics and Replication Program, National Cancer Institute, Frederick, MD, USA
| | - Saraswati Sukumar
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Couto JP, Bentires-Alj M. Mouse Models of Breast Cancer: Deceptions that Reveal the Truth. Breast Cancer 2017. [DOI: 10.1007/978-3-319-48848-6_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
18
|
Li H, Gumbiner BM. Deregulation of the Hippo pathway in mouse mammary stem cells promotes mammary tumorigenesis. Mamm Genome 2016; 27:556-564. [PMID: 27601049 DOI: 10.1007/s00335-016-9662-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 08/30/2016] [Indexed: 01/08/2023]
Abstract
The Hippo-YAP pathway mediates organ size control, contact inhibition, and tumorigenesis. It is a kinase cascade that inhibits the nuclear localization and transcriptional activities of YAP and TAZ. E-cadherin, cell junctions, polarity proteins, and the merlin/NF2 tumor suppressor activate the pathway to inhibit YAP/TAZ activity, while growth factor signaling inhibits the pathway to activate YAP/TAZ in the nucleus. We examined its role in the development of mouse mammary glands and tumor formation using gland reconstitution by transplantation of genetically modified mammary stem cells (MaSCs). Knockdown of YAP and TAZ with shRNA in MaSCs did not inhibit gland reconstitution. In contrast, knockdown of β-catenin blocked gland reconstitution, consistent with the known role of Wnt signaling in mammary gland development. However, we find that Hippo signaling is involved in mammary tumor formation. Expression of a constitutively active form of YAP caused rapid formation of large tumors. Moreover, knockdown of YAP/TAZ slowed the development of tumors in polyoma middle T transgenic mice, a well-studied mammary tumor model involving activation of several signaling pathways. YAP accumulated in nuclei of mammary glands in ErbB2/EGFR-transgenic mice, suggesting that EGFR signaling affects YAP in vivo similar to cell culture. ErbB2/EGFR-transgenic mice develop mammary tumors in 7-8 months, but surprisingly, MaSCs from these mice did not form tumors when transplanted into host mice. Nonetheless, expression of dominant-negative Lats, which inhibits Hippo signaling, leads to tumor formation in ErbB2-transgenic mice, suggesting that Hippo signaling is involved in EGFR-induced mammary tumorigenesis.
Collapse
Affiliation(s)
- Hongbin Li
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Barry M Gumbiner
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA. .,Department of Pediatrics, Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, University of Washington School of Medicine, 1900 9th Ave. Mailstop JMB-5, Seattle, WA, 98101, USA.
| |
Collapse
|
19
|
Wang W, Lv J, Wang L, Wang X, Ye L. The impact of heterogeneity in phosphoinositide 3-kinase pathway in human cancer and possible therapeutic treatments. Semin Cell Dev Biol 2016; 64:116-124. [PMID: 27582428 DOI: 10.1016/j.semcdb.2016.08.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 08/24/2016] [Indexed: 02/07/2023]
Abstract
Phosphatidylinositol 3-kinase catalytic subunit alpha (PIK3CA) plays a crucial role in the initiation and progress of cancerous tumors through the overexpression of the PI3K pathway promoting uncontrollable levels of cell proliferation. In addition only Class I PI3K has been discovered to be involved in human cancer due to its unique ability to produce phosphoinositide 3,4,5 trisphosphate (PIP3), which has been discovered to play a crucial role in human oncogenesis. The role of PIK3CA is lucubrated in breast cancer and gastric cancer, but is not well characterized in lung diseases. In this review, we summarized the common biology and mutations in PIK3CA with its related signaling pathways. Furthermore, we elucidated the PIK3CA heterogeneity in different domains, between various cancers and in different lung cancers. We also take a look at current inhibitors such as KP-372-1 (KP-1), KP-372-2 (KP-2), GSK690693, etc. in order to highlight potential treatment of PIK3CA mutations in human cancer and what directions future research should focus on.
Collapse
Affiliation(s)
- William Wang
- Zhongshan Hospital Institute of Clinical Science, Fudan University, Shanghai Institute of Clinical Bioinformatics, Biomedical Research Center, Shanghai, China.
| | - Jiapei Lv
- Zhongshan Hospital Institute of Clinical Science, Fudan University, Shanghai Institute of Clinical Bioinformatics, Biomedical Research Center, Shanghai, China
| | - Lingyan Wang
- Zhongshan Hospital Institute of Clinical Science, Fudan University, Shanghai Institute of Clinical Bioinformatics, Biomedical Research Center, Shanghai, China
| | - Xiangdong Wang
- Zhongshan Hospital Institute of Clinical Science, Fudan University, Shanghai Institute of Clinical Bioinformatics, Biomedical Research Center, Shanghai, China.
| | - Ling Ye
- Zhongshan Hospital Institute of Clinical Science, Fudan University, Shanghai Institute of Clinical Bioinformatics, Biomedical Research Center, Shanghai, China
| |
Collapse
|
20
|
Leroy C, Ramos P, Cornille K, Bonenfant D, Fritsch C, Voshol H, Bentires-Alj M. Activation of IGF1R/p110β/AKT/mTOR confers resistance to α-specific PI3K inhibition. Breast Cancer Res 2016; 18:41. [PMID: 27048245 PMCID: PMC4820873 DOI: 10.1186/s13058-016-0697-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 03/16/2016] [Indexed: 01/26/2023] Open
Abstract
Background The PI3K pathway is hyperactivated in many cancers, including 70 % of breast cancers. Pan- and isoform-specific inhibitors of the PI3K pathway are currently being evaluated in clinical trials. However, the clinical responses to PI3K inhibitors when used as single agents are not as efficient as expected. Methods In order to anticipate potential molecular mechanisms of resistance to the p110α isoform-selective inhibitor BYL719, we developed resistant breast cancer cell lines, assessed the concomitant changes in cellular signaling pathways using unbiased phosphotyrosine proteomics and characterized the mechanism of resistance using pharmacological inhibitors. Results We found an increase in IGF1R, IRS1/IRS2 and p85 phosphorylation in the resistant lines. Co-immunoprecipitation experiments identified an IGF1R/IRS/p85/p110β complex that causes the activation of AKT/mTOR/S6K and stifles the effects of BYL719. Pharmacological inhibition of members of this complex reduced mTOR/S6K activation and restored sensitivity to BYL719. Conclusion Our study demonstrates that the IGF1R/p110β/AKT/mTOR axis confers resistance to BYL719 in PIK3CA mutant breast cancers. This provides a rationale for the combined targeting of p110α with IGF1R or p110β in patients with breast tumors harboring PIK3CA mutations. Electronic supplementary material The online version of this article (doi:10.1186/s13058-016-0697-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cedric Leroy
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstraße 66, 4058, Basel, Switzerland.,Novartis Institutes for Biomedical Research, Postfach, CH-4002, Basel, Switzerland
| | - Pedro Ramos
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstraße 66, 4058, Basel, Switzerland.,Novartis Institutes for Biomedical Research, Postfach, CH-4002, Basel, Switzerland
| | - Karen Cornille
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstraße 66, 4058, Basel, Switzerland
| | - Debora Bonenfant
- Novartis Institutes for Biomedical Research, Postfach, CH-4002, Basel, Switzerland
| | - Christine Fritsch
- Novartis Institutes for Biomedical Research, Postfach, CH-4002, Basel, Switzerland
| | - Hans Voshol
- Novartis Institutes for Biomedical Research, Postfach, CH-4002, Basel, Switzerland
| | - Mohamed Bentires-Alj
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstraße 66, 4058, Basel, Switzerland.
| |
Collapse
|
21
|
Abstract
Tumor heterogeneity impinges on prognosis, response to therapy, and metastasis. As such, heterogeneity is one of the most important and clinically relevant areas of cancer research. Breast cancer displays frequent intra- and inter-tumor heterogeneity as the result of genetic and non-genetic alterations that often enhance the vigor of cancer cells. In-depth characterization and understanding of the origin of this phenotypic and molecular diversity is paramount to improving diagnosis, the definition of prognostic and predictive biomarkers, and the design of therapeutic strategies. Here, we summarize current knowledge about sources of breast cancer heterogeneity, its consequences, and possible counter-measures. We discuss especially the impact on tumor heterogeneity of the differentiation state of the cell-of-origin, cancer cell plasticity, the microenvironment, and genetic evolution. Factors that enhance cancer cell vigor are clearly detrimental for patients.
Collapse
Affiliation(s)
- Shany Koren
- Friedrich Miescher Institute for Biomedical Research (FMI), Basel, Switzerland
| | | |
Collapse
|
22
|
Perryman AL, Stratton TP, Ekins S, Freundlich JS. Predicting Mouse Liver Microsomal Stability with "Pruned" Machine Learning Models and Public Data. Pharm Res 2016; 33:433-49. [PMID: 26415647 PMCID: PMC4712113 DOI: 10.1007/s11095-015-1800-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/22/2015] [Indexed: 02/07/2023]
Abstract
PURPOSE Mouse efficacy studies are a critical hurdle to advance translational research of potential therapeutic compounds for many diseases. Although mouse liver microsomal (MLM) stability studies are not a perfect surrogate for in vivo studies of metabolic clearance, they are the initial model system used to assess metabolic stability. Consequently, we explored the development of machine learning models that can enhance the probability of identifying compounds possessing MLM stability. METHODS Published assays on MLM half-life values were identified in PubChem, reformatted, and curated to create a training set with 894 unique small molecules. These data were used to construct machine learning models assessed with internal cross-validation, external tests with a published set of antitubercular compounds, and independent validation with an additional diverse set of 571 compounds (PubChem data on percent metabolism). RESULTS "Pruning" out the moderately unstable / moderately stable compounds from the training set produced models with superior predictive power. Bayesian models displayed the best predictive power for identifying compounds with a half-life ≥1 h. CONCLUSIONS Our results suggest the pruning strategy may be of general benefit to improve test set enrichment and provide machine learning models with enhanced predictive value for the MLM stability of small organic molecules. This study represents the most exhaustive study to date of using machine learning approaches with MLM data from public sources.
Collapse
Affiliation(s)
- Alexander L Perryman
- Division of Infectious Disease, Department of Medicine, and the Ruy V. Lourenço Center for the Study of Emerging and Re-emerging Pathogens, Rutgers University-New Jersey Medical School, Newark, New Jersey, 07103, USA
| | - Thomas P Stratton
- Department of Pharmacology & Physiology, Rutgers University-New Jersey Medical School, Medical Sciences Building, I-503, 185 South Orange Ave., Newark, New Jersey, 07103, USA
| | - Sean Ekins
- Collaborations in Chemistry, 5616 Hilltop Needmore Road, Fuquay-Varina, NC, 27526, USA
- Collaborative Drug Discovery, 1633 Bayshore Highway, Suite 342, Burlingame, CA, 94010, USA
| | - Joel S Freundlich
- Division of Infectious Disease, Department of Medicine, and the Ruy V. Lourenço Center for the Study of Emerging and Re-emerging Pathogens, Rutgers University-New Jersey Medical School, Newark, New Jersey, 07103, USA.
- Department of Pharmacology & Physiology, Rutgers University-New Jersey Medical School, Medical Sciences Building, I-503, 185 South Orange Ave., Newark, New Jersey, 07103, USA.
| |
Collapse
|
23
|
PIK3CA(H1047R) induces multipotency and multi-lineage mammary tumours. Nature 2015; 525:114-8. [PMID: 26266975 DOI: 10.1038/nature14669] [Citation(s) in RCA: 229] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 06/16/2015] [Indexed: 12/14/2022]
Abstract
The adult mouse mammary epithelium contains self-sustained cell lineages that form the inner luminal and outer basal cell layers, with stem and progenitor cells contributing to its proliferative and regenerative potential. A key issue in breast cancer biology is the effect of genomic lesions in specific mammary cell lineages on tumour heterogeneity and progression. The impact of transforming events on fate conversion in cancer cells of origin and thus their contribution to tumour heterogeneity remains largely elusive. Using in situ genetic lineage tracing and limiting dilution transplantation, we have unravelled the potential of PIK3CA(H1047R), one of the most frequent mutations occurring in human breast cancer, to induce multipotency during tumorigenesis in the mammary gland. Here we show that expression of PIK3CA(H1047R) in lineage-committed basal Lgr5-positive and luminal keratin-8-positive cells of the adult mouse mammary gland evokes cell dedifferentiation into a multipotent stem-like state, suggesting this to be a mechanism involved in the formation of heterogeneous, multi-lineage mammary tumours. Moreover, we show that the tumour cell of origin influences the frequency of malignant mammary tumours. Our results define a key effect of PIK3CA(H1047R) on mammary cell fate in the pre-neoplastic mammary gland and show that the cell of origin of PIK3CA(H1047R) tumours dictates their malignancy, thus revealing a mechanism underlying tumour heterogeneity and aggressiveness.
Collapse
|
24
|
Anticipating mechanisms of resistance to PI3K inhibition in breast cancer: a challenge in the era of precision medicine. Biochem Soc Trans 2015; 42:733-41. [PMID: 25109950 DOI: 10.1042/bst20140034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Frequent subversion of the PI3K (phosphoinositide 3-kinase) pathway during neoplastic transformation contributes to several hallmarks of cancer that result in a competitive advantage for cancer cells. Deregulation of this pathway can be the result of genomic alterations such as PIK3CA mutation, PTEN (phosphatase and tensin homologue deleted on chromosome 10) loss or the activation of upstream protein tyrosine kinases. Not surprisingly, the PI3K signalling pathway has become an attractive therapeutic target, and numerous inhibitors are in clinical trials. Unfortunately, current therapies for advanced cancers that target PI3K often lead to the development of resistance and relapse of the disease. It is therefore important to establish the molecular mechanisms of resistance to PI3K-targeted therapy. With the focus on breast cancer, in the present article, we summarize the different ways of targeting PI3K, review potential mechanisms of resistance to PI3K inhibition and discuss the rationale of combination treatments to reach a balance between efficacy and toxicity.
Collapse
|
25
|
Burke JE, Williams RL. Synergy in activating class I PI3Ks. Trends Biochem Sci 2015; 40:88-100. [PMID: 25573003 DOI: 10.1016/j.tibs.2014.12.003] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 12/03/2014] [Accepted: 12/05/2014] [Indexed: 12/20/2022]
Abstract
The class I phosphoinositide 3-kinases (PI3Ks) are lipid kinases that transduce a host of cellular signals and regulate a broad range of essential functions including growth, proliferation, and migration. As such, PI3Ks have pivotal roles in diseases such as cancer, diabetes, primary immune disorders, and inflammation. These enzymes are activated downstream of numerous activating stimuli including receptor tyrosine kinases, G protein-coupled receptors (GPCRs), and the Ras superfamily of small G proteins. A major challenge is to decipher how each PI3K isoform is able to successfully synergize these inputs into their intended signaling function. This article highlights recent progress in characterizing the molecular mechanisms of PI3K isoform-specific activation pathways, as well as novel roles for PI3Ks in human diseases, specifically cancer and immune diseases.
Collapse
Affiliation(s)
- John E Burke
- Department of Biochemistry and Microbiology, University of Victoria, 3800 Finnerty Drive, Victoria BC, V8P 5C2, Canada.
| | - Roger L Williams
- Medical Research Council (MRC) Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge, CB2 0QH, UK
| |
Collapse
|
26
|
Verbeke S, Richard E, Monceau E, Schmidt X, Rousseau B, Velasco V, Bernard D, Bonnefoi H, MacGrogan G, Iggo RD. Humanization of the mouse mammary gland by replacement of the luminal layer with genetically engineered preneoplastic human cells. Breast Cancer Res 2014; 16:504. [PMID: 25527189 PMCID: PMC4407301 DOI: 10.1186/s13058-014-0504-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 12/11/2014] [Indexed: 01/09/2023] Open
Abstract
Introduction The cell of origin for estrogen receptor α (ERα) positive breast cancer is
probably a luminal stem cell in the terminal duct lobular units. To model these
cells we have used the murine myoepithelial layer in the mouse mammary ducts as a
scaffold upon which to build a human luminal layer. To prevent squamous
metaplasia, a common artifact in genetically engineered breast cancer models, we
sought to limit activation of the epidermal growth factor receptor (EGFR) during
in vitro cell culture before grafting the
cells. Methods Human reduction mammoplasty cells were grown in
vitro in WIT medium. Epidermal growth factor (EGF) in the medium was
replaced with amphiregulin and neuregulin to decrease activation of EGFR and
increase activation of EGFR homologs 3 and 4 (ERBB3 and ERBB4). Lentiviral vectors
were used to express oncogenic transgenes and fluorescent proteins. Human mammary
epithelial cells were mixed with irradiated mouse fibroblasts and matrigel, then
injected through the nipple into the mammary ducts of immunodeficient mice.
Engrafted cells were visualized by stereomicroscopy for fluorescent proteins and
characterized by histology and immunohistochemistry. Results Growth of normal mammary epithelial cells in conditions favoring ERBB3/4
signaling prevented squamous metaplasia in
vitro. Normal human cells were quickly lost after intraductal
injection but cells infected with lentiviruses expressing CCND1, MYC, TERT, BMI1 and a
short hairpin RNA targeting TP53 were able to
engraft and progressively replace the luminal layer in the mouse mammary ducts,
resulting in the formation of an extensive network of humanized ducts. Despite
expressing multiple oncogenes, the human cells formed a morphologically normal
luminal layer. Expression of a single additional oncogene, PIK3CA-H1047R, converted the
cells into invasive cancer cells. The resulting tumors were ERα+, Ki67+ luminal B
adenocarcinomas that were resistant to treatment with fulvestrant. Conclusions Injection of preneoplastic human mammary epithelial cells into the mammary
ducts of immunodeficient mice leads to replacement of the murine luminal layer
with morphologically normal human cells. Genetic manipulation of the injected
cells makes it possible to study defined steps in the transformation of human
mammary epithelial cells in a more physiological environment than has hitherto
been possible. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0504-9) contains supplementary material, which is available to authorized
users.
Collapse
Affiliation(s)
- Stephanie Verbeke
- INSERM U916, Bergonié Cancer Institute, University of Bordeaux, 229 cours de l'Argonne, Bordeaux, 33076, France.
| | - Elodie Richard
- INSERM U916, Bergonié Cancer Institute, University of Bordeaux, 229 cours de l'Argonne, Bordeaux, 33076, France.
| | - Elodie Monceau
- INSERM U916, Bergonié Cancer Institute, University of Bordeaux, 229 cours de l'Argonne, Bordeaux, 33076, France.
| | - Xenia Schmidt
- INSERM U916, Bergonié Cancer Institute, University of Bordeaux, 229 cours de l'Argonne, Bordeaux, 33076, France. .,School of Medicine, University of St Andrews, Medical and Biological Sciences Building, North Haugh, St Andrews, KY16 9TF, UK.
| | - Benoit Rousseau
- Animalerie A2, University of Bordeaux, 146 rue Léo Saignat, 33076, Bordeaux, France.
| | - Valerie Velasco
- INSERM U916, Bergonié Cancer Institute, University of Bordeaux, 229 cours de l'Argonne, Bordeaux, 33076, France. .,Pathology Department, Bergonié Cancer Institute, 229 cours de l'Argonne, 33076, Bordeaux, France.
| | - David Bernard
- INSERM U1052, Centre Leon Berard, University of Lyon, 28 rue Laennec, 69008, Lyon, France.
| | - Herve Bonnefoi
- INSERM U916, Bergonié Cancer Institute, University of Bordeaux, 229 cours de l'Argonne, Bordeaux, 33076, France.
| | - Gaetan MacGrogan
- INSERM U916, Bergonié Cancer Institute, University of Bordeaux, 229 cours de l'Argonne, Bordeaux, 33076, France. .,Pathology Department, Bergonié Cancer Institute, 229 cours de l'Argonne, 33076, Bordeaux, France.
| | - Richard D Iggo
- INSERM U916, Bergonié Cancer Institute, University of Bordeaux, 229 cours de l'Argonne, Bordeaux, 33076, France. .,School of Medicine, University of St Andrews, Medical and Biological Sciences Building, North Haugh, St Andrews, KY16 9TF, UK.
| |
Collapse
|
27
|
Gross C, Bassell GJ. Neuron-specific regulation of class I PI3K catalytic subunits and their dysfunction in brain disorders. Front Mol Neurosci 2014; 7:12. [PMID: 24592210 PMCID: PMC3923137 DOI: 10.3389/fnmol.2014.00012] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 01/28/2014] [Indexed: 11/13/2022] Open
Abstract
The phosphoinositide 3-kinase (PI3K) complex plays important roles in virtually all cells of the body. The enzymatic activity of PI3K to phosphorylate phosphoinositides in the membrane is mediated by a group of catalytic and regulatory subunits. Among those, the class I catalytic subunits, p110α, p110β, p110γ, and p110δ, have recently drawn attention in the neuroscience field due to their specific dysregulation in diverse brain disorders. While in non-neuronal cells these catalytic subunits may have partially redundant functions, there is increasing evidence that in neurons their roles are more specialized, and confined to distinct receptor-dependent pathways. This review will summarize the emerging role of class I PI3K catalytic subunits in neurotransmitter-regulated neuronal signaling, and their dysfunction in a variety of neurological diseases, including fragile X syndrome, schizophrenia, and epilepsy. We will discuss recent literature describing the use of PI3K subunit-selective inhibitors to rescue brain disease-associated phenotypes in in vitro and animal models. These studies give rise to the exciting prospect that these drugs, originally designed for cancer treatment, may be repurposed as therapeutic drugs for brain disorders in the future.
Collapse
Affiliation(s)
- Christina Gross
- Department of Cell Biology, Emory University School of Medicine Atlanta, GA, USA ; Center for Translational Social Neuroscience, Emory University School of Medicine Atlanta, GA, USA
| | - Gary J Bassell
- Department of Cell Biology, Emory University School of Medicine Atlanta, GA, USA ; Center for Translational Social Neuroscience, Emory University School of Medicine Atlanta, GA, USA ; Department of Neurology, Emory University School of Medicine Atlanta, GA, USA
| |
Collapse
|
28
|
Zardavas D, Phillips WA, Loi S. PIK3CA mutations in breast cancer: reconciling findings from preclinical and clinical data. Breast Cancer Res 2014; 16:201. [PMID: 25192370 PMCID: PMC4054885 DOI: 10.1186/bcr3605] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 01/23/2014] [Indexed: 02/08/2023] Open
Abstract
PIK3CA mutations represent one of the most common genetic aberrations in breast cancer. They have been reported to be present in over one-third of cases, with enrichment in the luminal and in human epidermal growth factor receptor 2-positive subtypes. Substantial preclinical data on the oncogenic properties of these mutations have been reported. However, whilst the preclinical data have clearly shown an association with robust activation of the pathway and resistance to common therapies used in breast cancer, the clinical data reported up to now do not support that the PIK3CA mutated genotype is associated with high levels of pathway activation or with a poor prognosis. We speculate that this may be due to the minimal use of transgenic mice models thus far. In this review, we discuss both the preclinical and clinical data associated with PIK3CA mutations and their potential implications. Prospective clinical trials stratifying by PIK3CA genotype will be necessary to determine if the mutation also predicts for increased sensitivity to agents targeting the phosphoinositide 3-kinase pathway.
Collapse
Affiliation(s)
| | - Wayne A Phillips
- Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre, Melbourne,
Victoria 3002, Australia
- Division of Cancer Medicine and Research, Peter MacCallum Cancer Centre, St
Andrews Place, East Melbourne, Victoria 3002, Australia
| | - Sherene Loi
- Division of Cancer Medicine and Research, Peter MacCallum Cancer Centre, St
Andrews Place, East Melbourne, Victoria 3002, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville,
Victoria 3002, Australia
| |
Collapse
|
29
|
Expression of PIK3CA mutant E545K in the mammary gland induces heterogeneous tumors but is less potent than mutant H1047R. Oncogenesis 2013; 2:e74. [PMID: 24080956 PMCID: PMC3816227 DOI: 10.1038/oncsis.2013.38] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2013] [Revised: 08/13/2013] [Accepted: 08/20/2013] [Indexed: 02/07/2023] Open
Abstract
The phosphoinositide 3-kinase (PI3K) signaling cascade is a key mediator of cellular growth, survival and metabolism and is frequently subverted in human cancer. The gene encoding for the alpha catalytic subunit of PI3K (PIK3CA) is mutated and/or amplified in ∼30% of breast cancers. Mutations in either the kinase domain (H1047R) or the helical domain (E545K) are most common and result in a constitutively active enzyme with oncogenic capacity. PIK3CAH1047R was previously demonstrated to induce tumors in transgenic mouse models; however, it was not known whether overexpression of PIK3CAE545K is sufficient to induce mammary tumors and whether tumor initiation by these two types of mutants differs. Here, we demonstrate that expression of PIK3CAE545K in the mouse mammary gland induces heterogenous mammary carcinomas but with a longer latency than PIK3CAH1047R-expressing mice. Our results suggest that the helical domain mutant PIK3CAE545K is a less potent inducer of mammary tumors due to less efficient activation of downstream Akt signaling.
Collapse
|
30
|
Mutant PIK3CA accelerates HER2-driven transgenic mammary tumors and induces resistance to combinations of anti-HER2 therapies. Proc Natl Acad Sci U S A 2013; 110:14372-7. [PMID: 23940356 DOI: 10.1073/pnas.1303204110] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Human epidermal growth factor receptor 2 (HER2; ERBB2) amplification and phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit alpha (PIK3CA) mutations often co-occur in breast cancer. Aberrant activation of the phosphatidylinositol 3-kinase (PI3K) pathway has been shown to correlate with a diminished response to HER2-directed therapies. We generated a mouse model of HER2-overexpressing (HER2(+)), PIK3CA(H1047R)-mutant breast cancer. Mice expressing both human HER2 and mutant PIK3CA in the mammary epithelium developed tumors with shorter latencies compared with mice expressing either oncogene alone. HER2 and mutant PIK3CA also cooperated to promote lung metastases. By microarray analysis, HER2-driven tumors clustered with luminal breast cancers, whereas mutant PIK3CA tumors were associated with claudin-low breast cancers. PIK3CA and HER2(+)/PIK3CA tumors expressed elevated transcripts encoding markers of epithelial-to-mesenchymal transition and stem cells. Cells from HER2(+)/PIK3CA tumors more efficiently formed mammospheres and lung metastases. Finally, HER2(+)/PIK3CA tumors were resistant to trastuzumab alone and in combination with lapatinib or pertuzumab. Both drug resistance and enhanced mammosphere formation were reversed by treatment with a PI3K inhibitor. In sum, PIK3CA(H1047R) accelerates HER2-mediated breast epithelial transformation and metastatic progression, alters the intrinsic phenotype of HER2-overexpressing cancers, and generates resistance to approved combinations of anti-HER2 therapies.
Collapse
|
31
|
Britschgi A, Radimerski T, Bentires-Alj M. Targeting PI3K, HER2 and the IL-8/JAK2 axis in metastatic breast cancer: Which combination makes the whole greater than the sum of its parts? Drug Resist Updat 2013; 16:68-72. [DOI: 10.1016/j.drup.2013.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 09/21/2013] [Indexed: 11/28/2022]
|