1
|
Zhang R, Zhang D, Han F, Song X, Zhang Y, Zhang J, Zhu Q, Qin Y. The deubiquitinase USP7 and E3 ligase TRIM21 regulate vasculogenic mimicry and malignant progression of RMS by balancing SNAI2 homeostasis. J Exp Clin Cancer Res 2024; 43:135. [PMID: 38702792 PMCID: PMC11069146 DOI: 10.1186/s13046-024-03056-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Rhabdomyosarcoma (RMS) is a rare malignancy and the most common soft tissue sarcoma in children. Vasculogenic mimicry (VM) is a novel tumor microcirculation model different from traditional tumor angiogenesis, which does not rely on endothelial cells to provide sufficient blood supply for tumor growth. In recent years, VM has been confirmed to be closely associated with tumor progression. However, the ability of RMS to form VM has not yet been reported. METHODS Immunohistochemistry, RT-qPCR and western blot were used to test the expression level of SNAI2 and its clinical significance. The biological function in regulating vasculogenic mimicry and malignant progression of SNAI2 was examined both in vitro and in vivo. Mass spectrometry, co-immunohistochemistry, immunofluorescence staining, and ubiquitin assays were performed to explore the regulatory mechanism of SNAI2. RESULTS Our study indicated that SNAI2 was abnormally expressed in patients with RMS and RMS cell lines and promoted the proliferation and metastasis of RMS. Through cell tubule formation experiments, nude mice Matrigel plug experiments, and immunohistochemistry (IHC), we confirmed that RMS can form VM and that SNAI2 promotes the formation of VM. Due to SNAI2 is a transcription factor that is not easily drugged, we used Co-IP combined with mass spectrometry to screen for the SNAI2-binding protein USP7 and TRIM21. USP7 depletion inhibited RMS VM formation, proliferation and metastasis by promoting SNAI2 degradation. We further demonstrated that TRIM21 is expressed at low levels in human RMS tissues and inhibits VM in RMS cells. TRIM21 promotes SNAI2 protein degradation through ubiquitination in the RMS. The deubiquitinase USP7 and E3 ligase TRIM21 function in an antagonistic rather than competitive mode and play a key role in controlling the stability of SNAI2 to determine the VM formation and progression of RMS. CONCLUSION Our findings reveal a previously unknown mechanism by which USP7 and TRIM21 balance the level of SNAI2 ubiquitination, determining RMS vasculogenic mimicry, proliferation, and migration. This new mechanism may provide new targeted therapies to inhibit the development of RMS by restoring TRIM21 expression or inhibiting USP7 expression in RMS patients with high SNAI2 protein levels.
Collapse
Affiliation(s)
- Ruyue Zhang
- Department of Clinical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Daidi Zhang
- Department of Clinical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Fugen Han
- Department of Otorhinolaryngology Head and Neck surgery, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaorui Song
- Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China
| | - Yaodong Zhang
- Henan Province Engineering Research Center of Diagnosis and Treatment of Pediatric Infection and Critical Care, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China
| | - Jie Zhang
- Department of Otorhinolaryngology Head and Neck surgery, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China.
- Department of Otorhinolaryngology Head and Neck surgery, National Center for Children's Health, Beijing Children's Hospital Capital Medical University, Beijing, 10045, China.
| | - Qingwen Zhu
- Department of Otorhinolaryngology Head and Neck surgery, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450052, China.
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Yanru Qin
- Department of Clinical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
2
|
Thapa R, Afzal O, Afzal M, Gupta G, Bhat AA, Hassan Almalki W, Kazmi I, Alzarea SI, Saleem S, Arora P, Singh SK, Dua K. From LncRNA to metastasis: The MALAT1-EMT axis in cancer progression. Pathol Res Pract 2024; 253:154959. [PMID: 38029713 DOI: 10.1016/j.prp.2023.154959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/11/2023] [Accepted: 11/16/2023] [Indexed: 12/01/2023]
Abstract
Cancer is a complex disease that causes abnormal genetic changes and unchecked cellular growth. It also causes a disruption in the normal regulatory processes that leads to the creation of malignant tissue. The complex interplay of genetic, environmental, and epigenetic variables influences its etiology. Long non-coding RNAs (LncRNAs) have emerged as pivotal contributors within the intricate landscape of cancer biology, orchestrating an array of multifaceted cellular processes that substantiate the processes of carcinogenesis and metastasis. Metastasis is a crucial driver of cancer mortality. Among these, MALAT1 (Metastasis-Associated Lung Adenocarcinoma Transcript 1) has drawn a lot of interest for its function in encouraging metastasis via controlling the Epithelial-Mesenchymal Transition (EMT) procedure. MALAT1 exerts a pivotal influence on the process of EMT, thereby promoting metastasis to distant organs. The mechanistic underpinning of this phenomenon involves the orchestration of an intricate regulatory network encompassing transcription factors, signalling cascades, and genes intricately associated with the EMT process by MALAT1. Its crucial function in transforming tumor cells into an aggressive phenotype is highlighted by its capacity to influence the expression of essential EMT effectors such as N-cadherin, E-cadherin, and Snail. An understanding of the MALAT1-EMT axis provides potential therapeutic approaches for cancer intervention. Targeting MALAT1 or its downstream EMT effectors may reduce the spread of metastatic disease and improve the effectiveness of already available therapies. Understanding the MALAT1-EMT axis holds significant clinical implications. Therefore, directing attention towards MALAT1 or its downstream mediators could present innovative therapeutic strategies for mitigating metastasis and improving patient prognosis. This study highlights the importance of MALAT1 in cancer biology and its potential for cutting back on metastatic disease with novel treatment strategies.
Collapse
Affiliation(s)
- Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India.
| | - Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Shakir Saleem
- Department of Public Health. College of Health Sciences, Saudi Electronic University, Riyadh, Saudi Arabia
| | - Poonam Arora
- SGT College of Pharmacy, SGT University, Gurugram, Haryana, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| |
Collapse
|
3
|
Berta D, Gehrke S, Nyíri K, Vértessy BG, Rosta E. Mechanism-Based Redesign of GAP to Activate Oncogenic Ras. J Am Chem Soc 2023; 145:20302-20310. [PMID: 37682266 PMCID: PMC10515638 DOI: 10.1021/jacs.3c04330] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Indexed: 09/09/2023]
Abstract
Ras GTPases play a crucial role in cell signaling pathways. Mutations of the Ras gene occur in about one third of cancerous cell lines and are often associated with detrimental clinical prognosis. Hot spot residues Gly12, Gly13, and Gln61 cover 97% of oncogenic mutations, which impair the enzymatic activity in Ras. Using QM/MM free energy calculations, we present a two-step mechanism for the GTP hydrolysis catalyzed by the wild-type Ras.GAP complex. We found that the deprotonation of the catalytic water takes place via the Gln61 as a transient Brønsted base. We also determined the reaction profiles for key oncogenic Ras mutants G12D and G12C using QM/MM minimizations, matching the experimentally observed loss of catalytic activity, thereby validating our reaction mechanism. Using the optimized reaction paths, we devised a fast and accurate procedure to design GAP mutants that activate G12D Ras. We replaced GAP residues near the active site and determined the activation barrier for 190 single mutants. We furthermore built a machine learning for ultrafast screening, by fast prediction of the barrier heights, tested both on the single and double mutations. This work demonstrates that fast and accurate screening can be accomplished via QM/MM reaction path optimizations to design protein sequences with increased catalytic activity. Several GAP mutations are predicted to re-enable catalysis in oncogenic G12D, offering a promising avenue to overcome aberrant Ras-driven signal transduction by activating enzymatic activity instead of inhibition. The outlined computational screening protocol is readily applicable for designing ligands and cofactors analogously.
Collapse
Affiliation(s)
- Dénes Berta
- Department
of Physics and Astronomy, University College
London, Gower Street, London WC1E
6BT, United Kingdom
| | - Sascha Gehrke
- Department
of Physics and Astronomy, University College
London, Gower Street, London WC1E
6BT, United Kingdom
| | - Kinga Nyíri
- Institute
of Enzymology, Research Centre for Natural Sciences, Magyar tudósok körútja
2, Budapest 1117, Hungary
- Department
of Applied Biotechnology and Food Science, Budapest University of Technology and Economics, Budafoki út 6-8, Budapest 1111, Hungary
| | - Beáta G. Vértessy
- Institute
of Enzymology, Research Centre for Natural Sciences, Magyar tudósok körútja
2, Budapest 1117, Hungary
- Department
of Applied Biotechnology and Food Science, Budapest University of Technology and Economics, Budafoki út 6-8, Budapest 1111, Hungary
| | - Edina Rosta
- Department
of Physics and Astronomy, University College
London, Gower Street, London WC1E
6BT, United Kingdom
| |
Collapse
|
4
|
Entrialgo-Cadierno R, Cueto-Ureña C, Welch C, Feliu I, Macaya I, Vera L, Morales X, Michelina SV, Scaparone P, Lopez I, Darbo E, Erice O, Vallejo A, Moreno H, Goñi-Salaverri A, Lara-Astiaso D, Halberg N, Cortes-Dominguez I, Guruceaga E, Ambrogio C, Lecanda F, Vicent S. The phospholipid transporter PITPNC1 links KRAS to MYC to prevent autophagy in lung and pancreatic cancer. Mol Cancer 2023; 22:86. [PMID: 37210549 DOI: 10.1186/s12943-023-01788-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 05/11/2023] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND The discovery of functionally relevant KRAS effectors in lung and pancreatic ductal adenocarcinoma (LUAD and PDAC) may yield novel molecular targets or mechanisms amenable to inhibition strategies. Phospholipids availability has been appreciated as a mechanism to modulate KRAS oncogenic potential. Thus, phospholipid transporters may play a functional role in KRAS-driven oncogenesis. Here, we identified and systematically studied the phospholipid transporter PITPNC1 and its controlled network in LUAD and PDAC. METHODS Genetic modulation of KRAS expression as well as pharmacological inhibition of canonical effectors was completed. PITPNC1 genetic depletion was performed in in vitro and in vivo LUAD and PDAC models. PITPNC1-deficient cells were RNA sequenced, and Gene Ontology and enrichment analyses were applied to the output data. Protein-based biochemical and subcellular localization assays were run to investigate PITPNC1-regulated pathways. A drug repurposing approach was used to predict surrogate PITPNC1 inhibitors that were tested in combination with KRASG12C inhibitors in 2D, 3D, and in vivo models. RESULTS PITPNC1 was increased in human LUAD and PDAC, and associated with poor patients' survival. PITPNC1 was regulated by KRAS through MEK1/2 and JNK1/2. Functional experiments showed PITPNC1 requirement for cell proliferation, cell cycle progression and tumour growth. Furthermore, PITPNC1 overexpression enhanced lung colonization and liver metastasis. PITPNC1 regulated a transcriptional signature which highly overlapped with that of KRAS, and controlled mTOR localization via enhanced MYC protein stability to prevent autophagy. JAK2 inhibitors were predicted as putative PITPNC1 inhibitors with antiproliferative effect and their combination with KRASG12C inhibitors elicited a substantial anti-tumour effect in LUAD and PDAC. CONCLUSIONS Our data highlight the functional and clinical relevance of PITPNC1 in LUAD and PDAC. Moreover, PITPNC1 constitutes a new mechanism linking KRAS to MYC, and controls a druggable transcriptional network for combinatorial treatments.
Collapse
Affiliation(s)
- Rodrigo Entrialgo-Cadierno
- Program in Solid Tumours, University of Navarra, Centre of Applied Medical Research (CIMA), 55 Pio XII Avenue, 31008, Pamplona, Spain
| | - Cristina Cueto-Ureña
- Program in Solid Tumours, University of Navarra, Centre of Applied Medical Research (CIMA), 55 Pio XII Avenue, 31008, Pamplona, Spain
| | - Connor Welch
- Program in Solid Tumours, University of Navarra, Centre of Applied Medical Research (CIMA), 55 Pio XII Avenue, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Iker Feliu
- Program in Solid Tumours, University of Navarra, Centre of Applied Medical Research (CIMA), 55 Pio XII Avenue, 31008, Pamplona, Spain
| | - Irati Macaya
- Program in Solid Tumours, University of Navarra, Centre of Applied Medical Research (CIMA), 55 Pio XII Avenue, 31008, Pamplona, Spain
| | - Laura Vera
- Program in Solid Tumours, University of Navarra, Centre of Applied Medical Research (CIMA), 55 Pio XII Avenue, 31008, Pamplona, Spain
| | - Xabier Morales
- Imaging Unit and Cancer Imaging Laboratory, University of Navarra, CIMA, Pamplona, Spain
| | - Sandra Vietti Michelina
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Centre, University of Torino, Turin, Italy
| | - Pietro Scaparone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Centre, University of Torino, Turin, Italy
| | - Ines Lopez
- Program in Solid Tumours, University of Navarra, Centre of Applied Medical Research (CIMA), 55 Pio XII Avenue, 31008, Pamplona, Spain
| | - Elodie Darbo
- University of Bordeaux, INSERM, BRIC, U 1312, F-33000, Bordeaux, France
| | - Oihane Erice
- Program in Solid Tumours, University of Navarra, Centre of Applied Medical Research (CIMA), 55 Pio XII Avenue, 31008, Pamplona, Spain
| | - Adrian Vallejo
- Program in Solid Tumours, University of Navarra, Centre of Applied Medical Research (CIMA), 55 Pio XII Avenue, 31008, Pamplona, Spain
| | - Haritz Moreno
- Program in Solid Tumours, University of Navarra, Centre of Applied Medical Research (CIMA), 55 Pio XII Avenue, 31008, Pamplona, Spain
| | | | - David Lara-Astiaso
- Molecular Therapies Program, University of Navarra, CIMA, Pamplona, Spain
- Wellcome - MRC Cambridge Stem Cell Institute (CSCI), Cambridge, UK
| | - Nils Halberg
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Ivan Cortes-Dominguez
- Imaging Unit and Cancer Imaging Laboratory, University of Navarra, CIMA, Pamplona, Spain
- Bioinformatics Platform, University of Navarra, CIMA, Pamplona, Spain
| | - Elizabeth Guruceaga
- Bioinformatics Platform, University of Navarra, CIMA, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Centre, University of Torino, Turin, Italy
| | - Fernando Lecanda
- Program in Solid Tumours, University of Navarra, Centre of Applied Medical Research (CIMA), 55 Pio XII Avenue, 31008, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
- Department of Pathology, Anatomy and Physiology, University of Navarra, Pamplona, Spain
| | - Silve Vicent
- Program in Solid Tumours, University of Navarra, Centre of Applied Medical Research (CIMA), 55 Pio XII Avenue, 31008, Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.
- Department of Pathology, Anatomy and Physiology, University of Navarra, Pamplona, Spain.
| |
Collapse
|
5
|
Wang Y, Qin C, Zhao B, Li Z, Li T, Yang X, Zhao Y, Wang W. EGR1 induces EMT in pancreatic cancer via a P300/SNAI2 pathway. J Transl Med 2023; 21:201. [PMID: 36932397 PMCID: PMC10021983 DOI: 10.1186/s12967-023-04043-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 03/08/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND The prognosis of pancreatic cancer patients remains relatively poor. Although some patients would receive surgical resection, distant metastasis frequently occurs within one year. Epithelial-mesenchymal transition (EMT), as a pathological mechanism in cancer progression, contributed to the local and distant metastasis of pancreatic cancer. METHODS Tissue microarray analysis and immunohistochemistry assays were used to compare the expression of EGR1 in pancreatic cancer and normal pancreatic tissues. Transwell chambers were used to evaluated the migration and invasion ability of cancer cells. Immunofluorescence was utilized to assess the expression of E-cadherin. ChIP-qPCR assay was applied to verify the combination of EGR1 and SNAI2 promoter sequences. Dual-luciferase reporter assay was used to detect the gene promoter activation. Co-IP assay was conducted to verify the interaction of EGR1 and p300/CBP. RESULTS EGR1 was highly expressed in pancreatic cancer rather than normal pancreatic tissues and correlated with poor prognosis and cancer metastasis. EGR1 was proved to enhance the migration and invasion ability of pancreatic cells. Besides, EGR1 was positively correlated with EMT process in pancreatic cancer, via a SNAI2-dependent pathway. P300/CBP was found to play an auxiliary role in the transcriptional activation of the SNAI2 gene by EGR1. Finally, in vivo experiments also proved that EGR1 promoted liver metastasis of pancreatic cancer. CONCLUSION Our findings implied the EMT-promoting effect of EGR1 in pancreatic cancer and revealed the intrinsic mechanism. Blocking the expression of EGR1 may be a new anticancer strategy for pancreatic cancer.
Collapse
Affiliation(s)
- Yuanyang Wang
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Cheng Qin
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Bangbo Zhao
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zeru Li
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Tianyu Li
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiaoying Yang
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yutong Zhao
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Weibin Wang
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China.
| |
Collapse
|
6
|
Mehta S, Buyanbat A, Orkin S, Nabet B. High-efficiency knock-in of degradable tags (dTAG) at endogenous loci in cell lines. Methods Enzymol 2023; 681:1-22. [PMID: 36764753 DOI: 10.1016/bs.mie.2022.08.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
The dTAG system is a versatile strategy for tunable control of protein abundance and facilitates the time-resolved assessment of disease-associated protein function. A "co-opted" fusion-based degron peptide, the "dTAG" facilitates the study of endogenous protein function when knocked-in at the endogenous genetic loci of proteins of interest. We combine CRISPR/Cas9 mediated induction of double-strand breaks (DSB) with the delivery of a single-stranded DNA HDR-donor-template via crude preparations of recombinant adeno-associated virus (rAAV). Our approach to knock-in of large (1-2kb) DNA fragments via crude-rAAV mediated HDR donor delivery is rapid and inexpensive. It facilitates genetic modification of a variety of human as well as mouse cell lines at high efficiency and precision.
Collapse
Affiliation(s)
- Stuti Mehta
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, United States
| | - Altantsetseg Buyanbat
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, United States
| | - Stuart Orkin
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, United States
| | - Behnam Nabet
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, United States.
| |
Collapse
|
7
|
Hensch NR, Bondra K, Wang L, Sreenivas P, Zhao XR, Modi P, Vaseva AV, Houghton PJ, Ignatius MS. Sensitization to Ionizing Radiation by MEK Inhibition Is Dependent on SNAI2 in Fusion-Negative Rhabdomyosarcoma. Mol Cancer Ther 2023; 22:123-134. [PMID: 36162055 PMCID: PMC10046682 DOI: 10.1158/1535-7163.mct-22-0310] [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: 05/08/2022] [Revised: 07/15/2022] [Accepted: 09/21/2022] [Indexed: 02/03/2023]
Abstract
In fusion-negative rhabdomyosarcoma (FN-RMS), a pediatric malignancy with skeletal muscle characteristics, >90% of high-risk patients have mutations that activate the RAS/MEK signaling pathway. We recently discovered that SNAI2, in addition to blocking myogenic differentiation downstream of MEK signaling in FN-RMS, represses proapoptotic BIM expression to protect RMS tumors from ionizing radiation (IR). As clinically relevant concentrations of the MEK inhibitor trametinib elicit poor responses in preclinical xenograft models, we investigated the utility of low-dose trametinib in combination with IR for the treatment of RAS-mutant FN-RMS. We hypothesized that trametinib would sensitize FN-RMS to IR through its downregulation of SNAI2 expression. While we observed little to no difference in myogenic differentiation or cell survival with trametinib treatment alone, robust differentiation and reduced survival were observed after IR. In addition, IR-induced apoptosis was significantly increased in FN-RMS cells treated concurrently with trametinib, as was increased BIM expression. SNAI2's role in these processes was established using overexpression rescue experiments, where overexpression of SNAI2 prevented IR-induced myogenic differentiation and apoptosis. Moreover, combining MEK inhibitor with IR resulted in complete tumor regression and a 2- to 4-week delay in event-free survival (EFS) in preclinical xenograft and patient-derived xenograft models. Our findings demonstrate that the combination of MEK inhibition and IR results in robust differentiation and apoptosis, due to the reduction of SNAI2, which leads to extended EFS in FN-RMS. SNAI2 thus is a potential biomarker of IR insensitivity and target for future therapies to sensitize aggressive sarcomas to IR.
Collapse
Affiliation(s)
- Nicole R. Hensch
- Greehey Children's Cancer Research Institute (GCCRI), Department of Molecular Medicine, UT Health Sciences Center, San Antonio, Texas, USA
| | - Kathryn Bondra
- Greehey Children's Cancer Research Institute (GCCRI), Department of Molecular Medicine, UT Health Sciences Center, San Antonio, Texas, USA
| | - Long Wang
- Greehey Children's Cancer Research Institute (GCCRI), Department of Molecular Medicine, UT Health Sciences Center, San Antonio, Texas, USA
| | - Prethish Sreenivas
- Greehey Children's Cancer Research Institute (GCCRI), Department of Molecular Medicine, UT Health Sciences Center, San Antonio, Texas, USA
| | - Xiang R. Zhao
- Greehey Children's Cancer Research Institute (GCCRI), Department of Molecular Medicine, UT Health Sciences Center, San Antonio, Texas, USA
| | - Paulomi Modi
- Greehey Children's Cancer Research Institute (GCCRI), Department of Molecular Medicine, UT Health Sciences Center, San Antonio, Texas, USA
| | - Angelina V. Vaseva
- Greehey Children's Cancer Research Institute (GCCRI), Department of Molecular Medicine, UT Health Sciences Center, San Antonio, Texas, USA
| | - Peter J. Houghton
- Greehey Children's Cancer Research Institute (GCCRI), Department of Molecular Medicine, UT Health Sciences Center, San Antonio, Texas, USA
| | - Myron S. Ignatius
- Greehey Children's Cancer Research Institute (GCCRI), Department of Molecular Medicine, UT Health Sciences Center, San Antonio, Texas, USA
| |
Collapse
|
8
|
Schneeweis C, Diersch S, Hassan Z, Krauß L, Schneider C, Lucarelli D, Falcomatà C, Steiger K, Öllinger R, Krämer OH, Arlt A, Grade M, Schmidt-Supprian M, Hessmann E, Wirth M, Rad R, Reichert M, Saur D, Schneider G. AP1/Fra1 confers resistance to MAPK cascade inhibition in pancreatic cancer. Cell Mol Life Sci 2023; 80:12. [PMID: 36534167 PMCID: PMC9763154 DOI: 10.1007/s00018-022-04638-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 11/01/2022] [Accepted: 11/16/2022] [Indexed: 12/23/2022]
Abstract
Targeting KRAS downstream signaling remains an important therapeutic approach in pancreatic cancer. We used primary pancreatic ductal epithelial cells and mouse models allowing the conditional expression of oncogenic KrasG12D, to investigate KRAS signaling integrators. We observed that the AP1 family member FRA1 is tightly linked to the KRAS signal and expressed in pre-malignant lesions and the basal-like subtype of pancreatic cancer. However, genetic-loss-of-function experiments revealed that FRA1 is dispensable for KrasG12D-induced pancreatic cancer development in mice. Using FRA1 gain- and loss-of-function models in an unbiased drug screen, we observed that FRA1 is a modulator of the responsiveness of pancreatic cancer to inhibitors of the RAF-MEK-ERK cascade. Mechanistically, context-dependent FRA1-associated adaptive rewiring of oncogenic ERK signaling was observed and correlated with sensitivity to inhibitors of canonical KRAS signaling. Furthermore, pharmacological-induced degradation of FRA1 synergizes with MEK inhibitors. Our studies establish FRA1 as a part of the molecular machinery controlling sensitivity to MAPK cascade inhibition allowing the development of mechanism-based therapies.
Collapse
Affiliation(s)
- Christian Schneeweis
- Medical Clinic and Polyclinic II, Klinikum Rechts Der Isar, Technical University Munich, 81675 Munich, Germany ,Institute for Translational Cancer Research and Experimental Cancer Therapy, Technical University Munich, 81675 Munich, Germany
| | - Sandra Diersch
- Medical Clinic and Polyclinic II, Klinikum Rechts Der Isar, Technical University Munich, 81675 Munich, Germany
| | - Zonera Hassan
- Medical Clinic and Polyclinic II, Klinikum Rechts Der Isar, Technical University Munich, 81675 Munich, Germany
| | - Lukas Krauß
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Carolin Schneider
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Daniele Lucarelli
- Institute for Translational Cancer Research and Experimental Cancer Therapy, Technical University Munich, 81675 Munich, Germany
| | - Chiara Falcomatà
- Institute for Translational Cancer Research and Experimental Cancer Therapy, Technical University Munich, 81675 Munich, Germany
| | - Katja Steiger
- Comparative Experimental Pathology, Institute of Pathology, School of Medicine, Technical Universität München, 81675 Munich, Germany ,German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Rupert Öllinger
- Institute of Molecular Oncology and Functional Genomics, TUM School of Medicine, TU München, 81675 Munich, Germany
| | - Oliver H. Krämer
- Department of Toxicology, University of Mainz Medical Center, 55131 Mainz, Germany
| | - Alexander Arlt
- Department for Internal Medicine and Gastroenterology, University Hospital, Klinikum Oldenburg AöR, 26133 Oldenburg, Germany
| | - Marian Grade
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany ,CCC-N (Comprehensive Cancer Center Lower Saxony), Göttingen, Germany
| | - Marc Schmidt-Supprian
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany ,Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Elisabeth Hessmann
- CCC-N (Comprehensive Cancer Center Lower Saxony), Göttingen, Germany ,University Medical Center Göttingen Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, 37075 Göttingen, Germany ,Clinical Research Unit 5002, KFO5002, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Matthias Wirth
- Department of Hematology, Oncology and Tumor Immunology, Campus Benjamin Franklin, Charité—Universitätsmedizin Berlin, 12203 Berlin, Germany
| | - Roland Rad
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany ,Institute of Molecular Oncology and Functional Genomics, TUM School of Medicine, TU München, 81675 Munich, Germany
| | - Maximilian Reichert
- Medical Clinic and Polyclinic II, Klinikum Rechts Der Isar, Technical University Munich, 81675 Munich, Germany ,German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany ,Translational Pancreatic Research Cancer Center, Medical Clinic and Polyclinic II, Klinikum Rechts Der Isar, Technical University Munich, 81675 Munich, Germany
| | - Dieter Saur
- Institute for Translational Cancer Research and Experimental Cancer Therapy, Technical University Munich, 81675 Munich, Germany ,German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Günter Schneider
- Medical Clinic and Polyclinic II, Klinikum Rechts Der Isar, Technical University Munich, 81675 Munich, Germany ,Institute for Translational Cancer Research and Experimental Cancer Therapy, Technical University Munich, 81675 Munich, Germany ,Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany ,CCC-N (Comprehensive Cancer Center Lower Saxony), Göttingen, Germany
| |
Collapse
|
9
|
da Silva-Oliveira RJ, Gomes INF, da Silva LS, Lengert AVH, Laus AC, Melendez ME, Munari CC, Cury FDP, Longato GB, Reis RM. Efficacy of Combined Use of Everolimus and Second-Generation Pan-EGRF Inhibitors in KRAS Mutant Non-Small Cell Lung Cancer Cell Lines. Int J Mol Sci 2022; 23:ijms23147774. [PMID: 35887120 PMCID: PMC9317664 DOI: 10.3390/ijms23147774] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/04/2022] [Accepted: 07/11/2022] [Indexed: 01/27/2023] Open
Abstract
Background: EGFR mutations are present in approximately 15−50% of non-small cell lung cancer (NSCLC), which are predictive of anti-EGFR therapies. At variance, NSCLC patients harboring KRAS mutations are resistant to those anti-EGFR approaches. Afatinib and allitinib are second-generation pan-EGFR drugs, yet no predictive biomarkers are known in the NSCLC context. In the present study, we evaluated the efficacy of pan-EGFR inhibitors in a panel of 15 lung cancer cell lines associated with the KRAS mutations phenotype. Methods: KRAS wild-type sensitive NCI-H292 cell line was further transfected with KRAS mutations (p.G12D and p.G12S). The pan-EGFR inhibitors’ activity and biologic effect of KRAS mutations were evaluated by cytotoxicity, MAPK phospho-protein array, colony formation, migration, invasion, and adhesion. In addition, in vivo chicken chorioallantoic membrane assay was performed in KRAS mutant cell lines. The gene expression profile was evaluated by NanoString. Lastly, everolimus and pan-EGFR combinations were performed to determine the combination index. Results: The GI50 score classified two cell lines treated with afatinib and seven treated with allitinib as high-sensitive phenotypes. All KRAS mutant cell lines demonstrated a resistant profile for both therapies (GI50 < 30%). The protein array of KRAS edited cells indicated a significant increase in AKT, CREB, HSP27, JNK, and, importantly, mTOR protein levels compared with KRAS wild-type cells. The colony formation, migration, invasion, adhesion, tumor perimeter, and mesenchymal phenotype were increased in the H292 KRAS mutated cells. Gene expression analysis showed 18 dysregulated genes associated with the focal adhesion-PI3K-Akt-mTOR-signaling correlated in KRAS mutant cell lines. Moreover, mTOR overexpression in KRAS mutant H292 cells was inhibited after everolimus exposure, and sensitivity to afatinib and allitinib was restored. Conclusions: Our results indicate that allitinib was more effective than afatinib in NSCLC cell lines. KRAS mutations increased aggressive behavior through upregulation of the focal adhesion-PI3K-Akt-mTOR-signaling in NSCLC cells. Significantly, everolimus restored sensibility and improved cytotoxicity of EGFR inhibitors in the KRAS mutant NSCLC cell lines.
Collapse
Affiliation(s)
- Renato José da Silva-Oliveira
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
- Correspondence: (R.J.d.S.-O.); (R.M.R.)
| | - Izabela Natalia Faria Gomes
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
| | - Luciane Sussuchi da Silva
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
| | - André van Helvoort Lengert
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
| | - Ana Carolina Laus
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
| | - Matias Eliseo Melendez
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
| | - Carla Carolina Munari
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
| | - Fernanda de Paula Cury
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
| | - Giovanna Barbarini Longato
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
| | - Rui Manuel Reis
- Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, Brazil; (I.N.F.G.); (L.S.d.S.); (A.v.H.L.); (A.C.L.); (M.E.M.); (C.C.M.); (F.d.P.C.); (G.B.L.)
- Life and Health Sciences Research Institute (ICVS) Medical School, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4710-057 Braga, Portugal
- Correspondence: (R.J.d.S.-O.); (R.M.R.)
| |
Collapse
|
10
|
Xu ZH, Wang WQ, Liu L, Lou WH. A special subtype: Revealing the potential intervention and great value of KRAS wildtype pancreatic cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188751. [PMID: 35732240 DOI: 10.1016/j.bbcan.2022.188751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 11/22/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the predominant form of pancreatic cancer and has devastating consequences on affected families and society. Its dismal prognosis is attributed to poor specificity of symptoms during early stages. It is widely believed that PDAC patients with the wildtype (WT) KRAS gene benefit more from currently available treatments than those with KRAS mutations. The oncogenic genetic changes alternations generally found in KRAS wildtype PDAC are related to either the KRAS pathway or microsatellite instability/mismatch repair deficiency (MSI/dMMR), which enable the application of tailored treatments based on each patient's genetic characteristics. This review focuses on targeted therapies against alternative tumour mechanisms in KRAS WT PDAC.
Collapse
Affiliation(s)
- Zhi-Hang Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Quan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Wen-Hui Lou
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
11
|
Li Z, Wu X, Li J, Yu S, Ke X, Yan T, Zhu Y, Cheng J, Yang J. HMGA2-Snai2 axis regulates tumorigenicity and stemness of head and neck squamous cell carcinoma. Exp Cell Res 2022; 418:113271. [PMID: 35764101 DOI: 10.1016/j.yexcr.2022.113271] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 06/23/2022] [Accepted: 06/23/2022] [Indexed: 11/04/2022]
Abstract
Cancer stem cells (CSCs) are a tumorigenic cell subpopulation, which contributes to treatment resistance, tumor recurrence, and metastasis. This study aimed to investigate the role and underlying molecular targets of high mobility group AT-hook 2 (HMGA2) in the progression and CSCs regulation of head and neck squamous cell carcinoma (HNSCC). HMGA2 mRNA and protein expression levels were examined in HNSCC specimens and cells by qRT-PCR, Western blot, and immunohistochemistry. The roles of HMGA2 were validated via loss-of-function and exogenous overexpression experiments in vitro and in vivo, and CSCs properties were assessed by tumorsphere formation assay. Chromatin immunoprecipitation (ChIP) and luciferase reporter assays provided further insight into the molecular mechanisms by which HMGA2 regulates stemness. HMGA2 was abnormally overexpressed in HNSCC, and it promoted the expression of the CSCs markers including SOX2, CD133, CD44, ALDH1A1, and Bmi1. HMGA2 was correlated with stemness, malignant progression, and reduced survival in HNSCC. Luciferase reporter assay indicated that Snai2 was a direct downstream target gene of HMGA2. Mechanistically, ChIP-qPCR assay showed that HMGA2 was recruited to three binding sites on the Snai2 promoter, directly facilitating the transcription of Snai2 in HNSCC. Snai2 overexpression reversed the inhibitory effect of HMGA2 interference on the proliferation, invasion, and metastasis of HNSCC and CSC marker expression in vitro and in vivo. HMGA2 promoted the malignant progression of HNSCC and acquired CSCs properties through direct regulation of Snai2, thereby suggesting that targeting the HMGA2-Snai2 axis might be a promising therapeutic strategy for HNSCC.
Collapse
Affiliation(s)
- Zhongwu Li
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China
| | - Xiang Wu
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China
| | - Jin Li
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China
| | - Shijin Yu
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China
| | - Xueping Ke
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China
| | - Tingyuan Yan
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China
| | - Yumin Zhu
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Cheng
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China.
| | - Jianrong Yang
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing, China; Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China; Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
12
|
Peschke K, Jakubowsky H, Schäfer A, Maurer C, Lange S, Orben F, Bernad R, Harder FN, Eiber M, Öllinger R, Steiger K, Schlitter M, Weichert W, Mayr U, Phillip V, Schlag C, Schmid RM, Braren RF, Kong B, Demir IE, Friess H, Rad R, Saur D, Schneider G, Reichert M. Identification of treatment-induced vulnerabilities in pancreatic cancer patients using functional model systems. EMBO Mol Med 2022; 14:e14876. [PMID: 35119792 PMCID: PMC8988213 DOI: 10.15252/emmm.202114876] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 01/12/2022] [Accepted: 01/13/2022] [Indexed: 02/06/2023] Open
Abstract
Despite the advance and success of precision oncology in gastrointestinal cancers, the frequency of molecular-informed therapy decisions in pancreatic ductal adenocarcinoma (PDAC) is currently neglectable. We present a longitudinal precision oncology platform based on functional model systems, including patient-derived organoids, to identify chemotherapy-induced vulnerabilities. We demonstrate that treatment-induced tumor cell plasticity in vivo distinctly changes responsiveness to targeted therapies, without the presence of a selectable genetic marker, indicating that tumor cell plasticity can be functionalized. By adding a mechanistic layer to precision oncology, adaptive processes of tumors under therapy can be exploited, particularly in highly plastic tumors, such as pancreatic cancer.
Collapse
Affiliation(s)
- Katja Peschke
- Medical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of MunichMünchenGermany
| | - Hannah Jakubowsky
- Institute for Translational Cancer Research and Experimental Cancer TherapyTechnical University of MunichMunichGermany
| | - Arlett Schäfer
- Medical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of MunichMünchenGermany
| | - Carlo Maurer
- Medical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of MunichMünchenGermany
| | - Sebastian Lange
- Medical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of MunichMünchenGermany
- Institute of Molecular Oncology and Functional GenomicsTUM School of MedicineTechnical University of MunichMunichGermany
| | - Felix Orben
- Medical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of MunichMünchenGermany
| | - Raquel Bernad
- Medical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of MunichMünchenGermany
- Institute for Translational Cancer Research and Experimental Cancer TherapyTechnical University of MunichMunichGermany
| | - Felix N Harder
- Institute of Diagnostic and Interventional RadiologyTechnical University of MunichMunichGermany
| | - Matthias Eiber
- Department of Nuclear MedicineKlinikum Rechts der IsarTechnical University of MunichMunichGermany
| | - Rupert Öllinger
- Institute of Molecular Oncology and Functional GenomicsTUM School of MedicineTechnical University of MunichMunichGermany
| | - Katja Steiger
- Institute of PathologyTechnical University of MunichMünchenGermany
| | | | - Wilko Weichert
- Institute of PathologyTechnical University of MunichMünchenGermany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK)HeidelbergGermany
| | - Ulrich Mayr
- Medical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of MunichMünchenGermany
| | - Veit Phillip
- Medical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of MunichMünchenGermany
| | - Christoph Schlag
- Medical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of MunichMünchenGermany
| | - Roland M Schmid
- Medical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of MunichMünchenGermany
| | - Rickmer F Braren
- Institute of Diagnostic and Interventional RadiologyTechnical University of MunichMunichGermany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK)HeidelbergGermany
| | - Bo Kong
- Department of SurgeryKlinikum rechts der IsarTechnical University of MunichMunichGermany
- Department of General SurgeryUniversity of UlmUlmGermany
| | - Ihsan Ekin Demir
- Department of SurgeryKlinikum rechts der IsarTechnical University of MunichMunichGermany
| | - Helmut Friess
- Department of SurgeryKlinikum rechts der IsarTechnical University of MunichMunichGermany
| | - Roland Rad
- Institute of Molecular Oncology and Functional GenomicsTUM School of MedicineTechnical University of MunichMunichGermany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK)HeidelbergGermany
| | - Dieter Saur
- Institute for Translational Cancer Research and Experimental Cancer TherapyTechnical University of MunichMunichGermany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK)HeidelbergGermany
| | - Günter Schneider
- Medical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of MunichMünchenGermany
- Department of General, Visceral and Pediatric SurgeryUniversity Medical Center GöttingenGöttingenGermany
| | - Maximilian Reichert
- Medical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of MunichMünchenGermany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK)HeidelbergGermany
- Center for Protein Assemblies (CPA)Technical University of MunichGarchingGermany
- Translational Pancreatic Cancer Research CenterMedical Clinic and Polyclinic IIKlinikum rechts der IsarTechnical University of MunichMünchenGermany
| |
Collapse
|