1
|
Nakazawa Y, Miyano M, Tsukamoto S, Kogai H, Yamamoto A, Iso K, Inoue S, Yamane Y, Yabe Y, Umihara H, Taguchi J, Akagi T, Yamaguchi A, Koga M, Toshimitsu K, Hirayama T, Mukai Y, Machinaga A. Delivery of a BET protein degrader via a CEACAM6-targeted antibody-drug conjugate inhibits tumour growth in pancreatic cancer models. Nat Commun 2024; 15:2192. [PMID: 38467634 PMCID: PMC10928091 DOI: 10.1038/s41467-024-46167-1] [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: 03/12/2023] [Accepted: 02/16/2024] [Indexed: 03/13/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has the worst prognosis of all cancers. To improve PDAC therapy, we establish screening systems based on organoid and co-culture technologies and find a payload of antibody-drug conjugate (ADC), a bromodomain and extra-terminal (BET) protein degrader named EBET. We select CEACAM6/CD66c as an ADC target and developed an antibody, #84.7, with minimal reactivity to CEACAM6-expressing normal cells. EBET-conjugated #84.7 (84-EBET) has lethal effects on various PDAC organoids and bystander efficacy on CEACAM6-negative PDAC cells and cancer-associated fibroblasts. In mouse studies, a single injection of 84-EBET induces marked tumor regression in various PDAC-patient-derived xenografts, with a decrease in the inflammatory phenotype of stromal cells and without significant body weight loss. Combination with standard chemotherapy or PD-1 antibody induces more profound and sustained regression without toxicity enhancement. Our preclinical evidence demonstrates potential efficacy by delivering BET protein degrader to PDAC and its microenvironment via CEACAM6-targeted ADC.
Collapse
Affiliation(s)
- Youya Nakazawa
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan.
| | - Masayuki Miyano
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
| | | | - Hiroyuki Kogai
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
| | | | - Kentaro Iso
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
| | - Satoshi Inoue
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
| | | | - Yuki Yabe
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
| | | | - Junichi Taguchi
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
| | - Tsuyoshi Akagi
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
- KAN Research Institute, Inc., Kobe, Japan
| | | | - Minaho Koga
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
| | | | | | | | - Akihito Machinaga
- Tsukuba Research Laboratory, Eisai Co., Ltd., Ibaraki, Japan
- KAN Research Institute, Inc., Kobe, Japan
| |
Collapse
|
2
|
Xu J, Roe J, Lee E, Tonelli C, Ji KY, Younis OW, Somervile TD, Yao M, Milazzo JP, Tiriac H, Kolarzyk AM, Lee E, Grem JL, Lazenby AJ, Grunkemeyer JA, Hollingsworth MA, Grandgenett PM, Borowsky AD, Park Y, Vakoc CR, Tuveson DA, Hwang C. Engrailed-1 Promotes Pancreatic Cancer Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308537. [PMID: 38110836 PMCID: PMC10853725 DOI: 10.1002/advs.202308537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Indexed: 12/20/2023]
Abstract
Engrailed-1 (EN1) is a critical homeodomain transcription factor (TF) required for neuronal survival, and EN1 expression has been shown to promote aggressive forms of triple negative breast cancer. Here, it is reported that EN1 is aberrantly expressed in a subset of pancreatic ductal adenocarcinoma (PDA) patients with poor outcomes. EN1 predominantly repressed its target genes through direct binding to gene enhancers and promoters, implicating roles in the activation of MAPK pathways and the acquisition of mesenchymal cell properties. Gain- and loss-of-function experiments demonstrated that EN1 promoted PDA transformation and metastasis in vitro and in vivo. The findings nominate the targeting of EN1 and downstream pathways in aggressive PDA.
Collapse
Affiliation(s)
- Jihao Xu
- Department of Microbiology and Molecular GeneticsUniversity of California DavisDavisCA95616USA
- Comprehensive Cancer CenterUniversity of California DavisSacramentoCA95817USA
| | - Jae‐Seok Roe
- Department of BiochemistryYonsei UniversitySeoul03722South Korea
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
| | - EunJung Lee
- Department of Microbiology and Molecular GeneticsUniversity of California DavisDavisCA95616USA
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
- Lustgarten Foundation Pancreatic Cancer Research LaboratoryCold Spring HarborNY11724USA
| | - Claudia Tonelli
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
- Lustgarten Foundation Pancreatic Cancer Research LaboratoryCold Spring HarborNY11724USA
| | - Keely Y. Ji
- Department of Microbiology and Molecular GeneticsUniversity of California DavisDavisCA95616USA
| | - Omar W. Younis
- Department of Microbiology and Molecular GeneticsUniversity of California DavisDavisCA95616USA
| | | | - Melissa Yao
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
- Lustgarten Foundation Pancreatic Cancer Research LaboratoryCold Spring HarborNY11724USA
| | | | - Herve Tiriac
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
- Lustgarten Foundation Pancreatic Cancer Research LaboratoryCold Spring HarborNY11724USA
| | - Anna M. Kolarzyk
- Nancy E. and Peter C. Meinig School of Biomedical EngineeringCornell UniversityIthacaNY14853USA
| | - Esak Lee
- Nancy E. and Peter C. Meinig School of Biomedical EngineeringCornell UniversityIthacaNY14853USA
| | - Jean L. Grem
- Department of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Audrey J. Lazenby
- Department of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | | | | | | | - Alexander D. Borowsky
- Department of PathologySchool of MedicineUniversity of California DavisSacramentoCA95817USA
| | - Youngkyu Park
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
- Lustgarten Foundation Pancreatic Cancer Research LaboratoryCold Spring HarborNY11724USA
| | | | - David A. Tuveson
- Cold Spring Harbor LaboratoryCold Spring HarborNY11724USA
- Lustgarten Foundation Pancreatic Cancer Research LaboratoryCold Spring HarborNY11724USA
| | - Chang‐Il Hwang
- Department of Microbiology and Molecular GeneticsUniversity of California DavisDavisCA95616USA
- Comprehensive Cancer CenterUniversity of California DavisSacramentoCA95817USA
| |
Collapse
|
3
|
Song T, Kong B, Liu R, Luo Y, Wang Y, Zhao Y. Bioengineering Approaches for the Pancreatic Tumor Organoids Research and Application. Adv Healthc Mater 2024; 13:e2300984. [PMID: 37694339 DOI: 10.1002/adhm.202300984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 08/29/2023] [Indexed: 09/12/2023]
Abstract
Pancreatic cancer is a highly lethal form of digestive malignancy that poses significant health risks to individuals worldwide. Chemotherapy-based comprehensive treatment is the primary therapeutic approach for midlife and late-life patients. Nevertheless, the heterogeneity of the tumor and individual genetic backgrounds result in substantial variations in drug sensitivity among patients, rendering a single treatment regimen unsuitable for all patients. Conventional pancreatic cancer tumor organoid models are capable of emulating the biological traits of pancreatic cancer and are utilized in drug development and screening. However, these tumor organoids can still not mimic the tumor microenvironment (TME) in vivo, and the poor controllability in the preparation process hinders translation from essential drug screening to clinical pharmacological therapy. In recent years, many engineering methods with remarkable results have been used to develop pancreatic cancer organoid models, including bio-hydrogel, co-culture, microfluidic, and gene editing. Here, this work summarizes and analyzes the recent developments in engineering pancreatic tumor organoid models. In addition, the future direction of improving engineered pancreatic cancer organoids is discussed for their application prospects in clinical treatment.
Collapse
Affiliation(s)
- Taiyu Song
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Bin Kong
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Rui Liu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210002, China
| | - Yuan Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210002, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| |
Collapse
|
4
|
Krauß L, Schneider C, Hessmann E, Saur D, Schneider G. Epigenetic control of pancreatic cancer metastasis. Cancer Metastasis Rev 2023; 42:1113-1131. [PMID: 37659057 PMCID: PMC10713713 DOI: 10.1007/s10555-023-10132-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/10/2023] [Indexed: 09/05/2023]
Abstract
Surgical resection, when combined with chemotherapy, has been shown to significantly improve the survival rate of patients with pancreatic ductal adenocarcinoma (PDAC). However, this treatment option is only feasible for a fraction of patients, as more than 50% of cases are diagnosed with metastasis. The multifaceted process of metastasis is still not fully understood, but recent data suggest that transcriptional and epigenetic plasticity play significant roles. Interfering with epigenetic reprogramming can potentially control the adaptive processes responsible for metastatic progression and therapy resistance, thereby enhancing treatment responses and preventing recurrence. This review will focus on the relevance of histone-modifying enzymes in pancreatic cancer, specifically on their impact on the metastatic cascade. Additionally, it will also provide a brief update on the current clinical developments in epigenetic therapies.
Collapse
Affiliation(s)
- 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
| | - Elisabeth Hessmann
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Göttingen, 37075, Göttingen, Germany
- Clinical Research Unit 5002, KFO5002, University Medical Center Göttingen, 37075, Göttingen, Germany
- CCC-N (Comprehensive Cancer Center Lower Saxony), 37075, Göttingen, Germany
| | - Dieter Saur
- Institute for Translational Cancer Research and Experimental Cancer Therapy, Technical University Munich, 81675, Munich, Germany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
| | - Günter Schneider
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, 37075, Göttingen, Germany.
- CCC-N (Comprehensive Cancer Center Lower Saxony), 37075, Göttingen, Germany.
| |
Collapse
|
5
|
Martinez-Ruiz L, López-Rodríguez A, Florido J, Rodríguez-Santana C, Rodríguez Ferrer JM, Acuña-Castroviejo D, Escames G. Patient-derived tumor models in cancer research: Evaluation of the oncostatic effects of melatonin. Biomed Pharmacother 2023; 167:115581. [PMID: 37748411 DOI: 10.1016/j.biopha.2023.115581] [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: 07/12/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 09/27/2023] Open
Abstract
The development of new anticancer therapies tends to be very slow. Although their impact on potential candidates is confirmed in preclinical studies, ∼95 % of these new therapies are not approved when tested in clinical trials. One of the main reasons for this is the lack of accurate preclinical models. In this context, there are different patient-derived models, which have emerged as a powerful oncological tool: patient-derived xenografts (PDXs), patient-derived organoids (PDOs), and patient-derived cells (PDCs). Although all these models are widely applied, PDXs, which are created by engraftment of patient tumor tissues into mice, is considered more reliable. In fundamental research, the PDX model is used to evaluate drug-sensitive markers and, in clinical practice, to select a personalized therapeutic strategy. Melatonin is of particular importance in the development of innovative cancer treatments due to its oncostatic impact and lack of adverse effects. However, the literature regarding the oncostatic effect of melatonin in patient-derived tumor models is scant. This review aims to describe the important role of patient-derived models in the development of anticancer treatments, focusing, in particular, on PDX models, as well as their use in cancer research. This review also summarizes the existing literature on the anti-tumoral effect of melatonin in patient-derived models in order to propose future anti-neoplastic clinical applications.
Collapse
Affiliation(s)
- Laura Martinez-Ruiz
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - Alba López-Rodríguez
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - Javier Florido
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - Cesar Rodríguez-Santana
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - José M Rodríguez Ferrer
- Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - Darío Acuña-Castroviejo
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain
| | - Germaine Escames
- Institute of Biotechnology, Biomedical Research Center, Health Sciences Technology Park, University of Granada, Granada, Spain; Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain; Department of Biochemistry and Molecular Biology I, Faculty of Science, University of Granada, Granada, Spain.
| |
Collapse
|
6
|
Bararia A, Das A, Mitra S, Banerjee S, Chatterjee A, Sikdar N. Deoxyribonucleic acid methylation driven aberrations in pancreatic cancer-related pathways. World J Gastrointest Oncol 2023; 15:1505-1519. [PMID: 37746645 PMCID: PMC10514732 DOI: 10.4251/wjgo.v15.i9.1505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/29/2023] [Accepted: 08/01/2023] [Indexed: 09/13/2023] Open
Abstract
Pancreatic cancer (PanCa) presents a catastrophic disease with poor overall survival at advanced stages, with immediate requirement of new and effective treatment options. Besides genetic mutations, epigenetic dysregulation of signaling pathway-associated enriched genes are considered as novel therapeutic target. Mechanisms beneath the deoxyribonucleic acid methylation and its utility in developing of epi-drugs in PanCa are under trails. Combinations of epigenetic medicines with conventional cytotoxic treatments or targeted therapy are promising options to improving the dismal response and survival rate of PanCa patients. Recent studies have identified potentially valid pathways that support the prediction that future PanCa clinical trials will include vigorous testing of epigenomic therapies. Epigenetics thus promises to generate a significant amount of new knowledge of biological and medical importance. Our review could identify various components of epigenetic mechanisms known to be involved in the initiation and development of pancreatic ductal adenocarcinoma and related precancerous lesions, and novel pharmacological strategies that target these components could potentially lead to breakthroughs. We aim to highlight the possibilities that exist and the potential therapeutic interventions.
Collapse
Affiliation(s)
- Akash Bararia
- Human Genetics Unit, Indian Statistical Institute, Kolkata 700108, India
| | - Amlan Das
- Department of Biochemistry, Royal Global University, Assam 781035, India
| | - Sangeeta Mitra
- Department of Biochemistry and Biophysics, University of Kalyani, West Bengal 741235, India
| | - Sudeep Banerjee
- Department of Gastrointestinal Surgery, Tata Medical Center, Kolkata 700160, India
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9054, New Zealand
- School of Health Sciences and Technology, University of Petroleum and Energy Studies, Dehradun 248007, India
| | - Nilabja Sikdar
- Human Genetics Unit, Indian Statistical Institute, Kolkata 700108, India
| |
Collapse
|
7
|
Färber B, Lapshyna O, Künstner A, Kohl M, Sauer T, Bichmann K, Heckelmann B, Watzelt J, Honselmann K, Bolm L, ten Winkel M, Busch H, Ungefroren H, Keck T, Gemoll T, Wellner UF, Braun R. Molecular profiling and specific targeting of gemcitabine-resistant subclones in heterogeneous pancreatic cancer cell populations. Front Oncol 2023; 13:1230382. [PMID: 37719017 PMCID: PMC10502231 DOI: 10.3389/fonc.2023.1230382] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/08/2023] [Indexed: 09/19/2023] Open
Abstract
Purpose Chemotherapy is pivotal in the multimodal treatment of pancreatic ductal adenocarcinoma (PDAC). Technical advances unveiled a high degree of inter- and intratumoral heterogeneity. We hypothesized that intratumoral heterogeneity (ITH) impacts response to gemcitabine treatment and demands specific targeting of resistant subclones. Methods Using single cell-derived cell lines (SCDCLs) from the classical cell line BxPC3 and the basal-like cell line Panc-1, we addressed the effect of ITH on response to gemcitabine treatment. Results Individual SCDCLs of both parental tumor cell populations showed considerable heterogeneity in response to gemcitabine. Unsupervised PCA including the 1,000 most variably expressed genes showed a clustering of the SCDCLs according to their respective sensitivity to gemcitabine treatment for BxPC3, while this was less clear for Panc-1. In BxPC3 SCDCLs, enriched signaling pathways EMT, TNF signaling via NfKB, and IL2STAT5 signaling correlated with more resistant behavior to gemcitabine. In Panc-1 SCDCLs MYC targets V1 and V2 as well as E2F targets were associated with stronger resistance. We used recursive feature elimination for Feature Selection in order to compute sets of proteins that showed strong association with the response to gemcitabine. The optimal protein set calculated for Panc-1 comprised fewer proteins in comparison to the protein set determined for BxPC3. Based on molecular profiles, we could show that the gemcitabine-resistant SCDCLs of both BxPC3 and Panc-1 are more sensitive to the BET inhibitor JQ1 compared to the respective gemcitabine-sensitive SCDCLs. Conclusion Our model system of SCDCLs identified gemcitabine-resistant subclones and provides evidence for the critical role of ITH for treatment response in PDAC. We exploited molecular differences as the basis for differential response and used these for more targeted therapy of resistant subclones.
Collapse
Affiliation(s)
- Benedikt Färber
- Department of Surgery, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Olga Lapshyna
- Department of Surgery, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Axel Künstner
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Michael Kohl
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
- Section for Translational Surgical Oncology & Biobanking, Department of Surgery, University Hospital Schleswig-Holstein, University of Lübeck, Lübeck, Germany
| | - Thorben Sauer
- Section for Translational Surgical Oncology & Biobanking, Department of Surgery, University Hospital Schleswig-Holstein, University of Lübeck, Lübeck, Germany
| | - Kira Bichmann
- Department of Surgery, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Benjamin Heckelmann
- Department of Surgery, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Jessica Watzelt
- Department of Surgery, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Kim Honselmann
- Department of Surgery, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Louisa Bolm
- Department of Surgery, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Meike ten Winkel
- Department of Surgery, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Hauke Busch
- Medical Systems Biology Group, Lübeck Institute of Experimental Dermatology, University of Lübeck, Lübeck, Germany
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Hendrik Ungefroren
- First Department of Medicine, University Medical Center Schleswig-Holstein, Lübeck, Germany
- Institute of Pathology, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Tobias Keck
- Department of Surgery, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Timo Gemoll
- Section for Translational Surgical Oncology & Biobanking, Department of Surgery, University Hospital Schleswig-Holstein, University of Lübeck, Lübeck, Germany
| | - Ulrich F. Wellner
- Department of Surgery, University Medical Center Schleswig-Holstein, Lübeck, Germany
| | - Rüdiger Braun
- Department of Surgery, University Medical Center Schleswig-Holstein, Lübeck, Germany
| |
Collapse
|
8
|
Reyes-Castellanos G, Abdel Hadi N, Gallardo-Arriaga S, Masoud R, Garcia J, Lac S, El Kaoutari A, Gicquel T, Planque M, Fendt SM, Linares LK, Gayet O, Guillaumond F, Dusetti N, Iovanna J, Carrier A. Combining the antianginal drug perhexiline with chemotherapy induces complete pancreatic cancer regression in vivo. iScience 2023; 26:106899. [PMID: 37305702 PMCID: PMC10250830 DOI: 10.1016/j.isci.2023.106899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 02/06/2023] [Accepted: 05/12/2023] [Indexed: 06/13/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the human cancers with the poorest prognosis. Interestingly, we found that mitochondrial respiration in primary human PDAC cells depends mainly on the fatty acid oxidation (FAO) to meet basic energy requirements. Therefore, we treated PDAC cells with perhexiline, a well-recognized FAO inhibitor used in cardiac diseases. Some PDAC cells respond efficiently to perhexiline, which acts synergistically with chemotherapy (gemcitabine) in vitro and in two xenografts in vivo. Importantly, perhexiline in combination with gemcitabine induces complete tumor regression in one PDAC xenograft. Mechanistically, this co-treatment causes energy and oxidative stress promoting apoptosis but does not exert inhibition of FAO. Yet, our molecular analysis indicates that the carnitine palmitoyltransferase 1C (CPT1C) isoform is a key player in the response to perhexiline and that patients with high CPT1C expression have better prognosis. Our study reveals that repurposing perhexiline in combination with chemotherapy is a promising approach to treat PDAC.
Collapse
Affiliation(s)
| | - Nadine Abdel Hadi
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | | | - Rawand Masoud
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Julie Garcia
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Sophie Lac
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | | | - Tristan Gicquel
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Mélanie Planque
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Laetitia Karine Linares
- INSERM, Université de Montpellier, IRCM, Institut Régional Du Cancer de Montpellier, Montpellier, France
| | - Odile Gayet
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Fabienne Guillaumond
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Nelson Dusetti
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Juan Iovanna
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Alice Carrier
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| |
Collapse
|
9
|
El Kaoutari A, Fraunhoffer NA, Audebert S, Camoin L, Berthois Y, Gayet O, Roques J, Bigonnet M, Bongrain C, Ciccolini J, Iovanna JL, Dusetti NJ, Soubeyran P. Pancreatic ductal adenocarcinoma ubiquitination profiling reveals specific prognostic and theranostic markers. EBioMedicine 2023; 92:104634. [PMID: 37257316 DOI: 10.1016/j.ebiom.2023.104634] [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: 12/06/2022] [Revised: 05/04/2023] [Accepted: 05/14/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has been widely studied at multiomics level. However, little is known about its specific ubiquitination, a major post-translational modification (PTM). As PTMs regulate the final function of any gene, we decided to establish the ubiquitination profiles of 60 PDAC. METHODS We used specific proteomic tools to establish the ubiquitin dependent proteome (ubiquitinome) of frozen PDXs (Patients' derived xenographs). Then, we performed bioinformatics analysis to identify the possible associations of these ubiquitination profiles with tumour phenotype, patient survival and resistance to chemotherapies. Finally, we used proximity ligation assays (PLA) to detect and quantify the ubiquitination level of one identified marker. FINDINGS We identified 38 ubiquitination site profiles correlating with the transcriptomic phenotype of tumours and four had notable prognostic capabilities. Seventeen ubiquitination profiles displayed potential theranostic marker for gemcitabine, seven for 5-FU, six for oxaliplatin and thirteen for irinotecan. Using PLA, we confirmed the use of one ubiquitination profile as a drug-response marker, directly on paraffin embedded tissues, supporting the possible application of these biomarkers in the clinical setting. INTERPRETATION These findings bring new and important insights on the relationship between ubiquitination levels of proteins and different molecular and clinical features of PDAC patients. Markers identified in this study could have a potential application in clinical settings to help to predict response to chemotherapies thereby allowing the personalization of treatments. FUNDING Fondation ARC (PJA 20181208270 and PGA 12021010002840_3562); INCa; Canceropôle PACA; DGOS; Amidex Foundation; Fondation de France; and INSERM.
Collapse
Affiliation(s)
- Abdessamad El Kaoutari
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, 13288, Marseille, France; COMPO Unit, Inria Sophia Antipolis, 13385, Marseille, France
| | - Nicolas A Fraunhoffer
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, 13288, Marseille, France
| | - Stéphane Audebert
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, 13288, Marseille, France
| | - Luc Camoin
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, 13288, Marseille, France
| | - Yolande Berthois
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, 13288, Marseille, France
| | - Odile Gayet
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, 13288, Marseille, France
| | - Julie Roques
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, 13288, Marseille, France
| | - Martin Bigonnet
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, 13288, Marseille, France
| | - Claire Bongrain
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, 13288, Marseille, France
| | - Joseph Ciccolini
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, 13288, Marseille, France; COMPO Unit, Inria Sophia Antipolis, 13385, Marseille, France
| | - Juan L Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, 13288, Marseille, France; Paoli-Calmettes Institut, 13009, Marseille, France
| | - Nelson J Dusetti
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, 13288, Marseille, France.
| | - Philippe Soubeyran
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, 13288, Marseille, France.
| |
Collapse
|
10
|
Agrawal R, Natarajan KN. Oncogenic signaling pathways in pancreatic ductal adenocarcinoma. Adv Cancer Res 2023; 159:251-283. [PMID: 37268398 DOI: 10.1016/bs.acr.2023.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common (∼90% cases) pancreatic neoplasm and one of the most lethal cancer among all malignances. PDAC harbor aberrant oncogenic signaling that may result from the multiple genetic and epigenetic alterations such as the mutation in driver genes (KRAS, CDKN2A, p53), genomic amplification of regulatory genes (MYC, IGF2BP2, ROIK3), deregulation of chromatin-modifying proteins (HDAC, WDR5) among others. A key event is the formation of Pancreatic Intraepithelial Neoplasia (PanIN) that often results from the activating mutation in KRAS. Mutated KRAS can direct a variety of signaling pathways and modulate downstream targets including MYC, which play an important role in cancer progression. In this review, we discuss recent literature shedding light on the origins of PDAC from the perspective of major oncogenic signaling pathways. We highlight how MYC directly and indirectly, with cooperation with KRAS, affect epigenetic reprogramming and metastasis. Additionally, we summarize the recent findings from single cell genomic approaches that highlight heterogeneity in PDAC and tumor microenvironment, and provide molecular avenues for PDAC treatment in the future.
Collapse
Affiliation(s)
- Rahul Agrawal
- DTU Bioengineering, Technical University of Denmark, Kongens Lyngby, Denmark
| | | |
Collapse
|
11
|
Tong T, Zhang C, Li J, Deng M, Wang X. Preclinical models derived from endoscopic ultrasound-guided tissue acquisition for individualized treatment of pancreatic ductal adenocarcinoma. Front Med (Lausanne) 2023; 9:934974. [PMID: 36687406 PMCID: PMC9849774 DOI: 10.3389/fmed.2022.934974] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 12/12/2022] [Indexed: 01/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with poor outcomes. Although the management strategies have evolved in recent years, the PDAC 5-year survival rate remains at only 9%; it may become the second leading cause of cancer death in the USA by 2030. Only 15-20% of PDAC patients are eligible to undergo surgery; diagnostic biopsies and individualized treatment present a more significant challenge for the remaining group. Endoscopic ultrasound-guided tissue acquisition (EUS-TA) has been widely used in the diagnosis of pancreatic masses. With the advancement of this sampling technique, adequate specimens can be obtained from all patients with PDAC in both early and late clinical stages. Recent data suggest that the specimens obtained from EUS-TA might be used to establish viable preclinical models, which conserve the genetic mutation and preserve the heterogeneity of the original tumors. Additionally, any drug sensitivity evident in the EUS-TA-derived preclinical models might predict the clinical response, thus guiding the prospective therapeutic selection. As we move toward the era of precision medicine, this review provides an update on the role of EUS-TA as a method for obtaining genetic material used in preclinical models that can assess and stratify individuals according to their individual cancer biology.
Collapse
Affiliation(s)
- Ting Tong
- Endoscopic Center, The First Affiliated Hospital of Xiamen University, Xiamen, China,Endoscopic Center, Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China,Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Central South University, Changsha, China
| | - Chao Zhang
- Endoscopic Center, Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China,Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Central South University, Changsha, China
| | - Jingbo Li
- Endoscopic Center, Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China,Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Central South University, Changsha, China
| | - Minzi Deng
- Endoscopic Center, Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China,Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Central South University, Changsha, China,*Correspondence: Minzi Deng,
| | - Xiaoyan Wang
- Endoscopic Center, Department of Gastroenterology, The Third Xiangya Hospital, Central South University, Changsha, China,Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Central South University, Changsha, China,Xiaoyan Wang,
| |
Collapse
|
12
|
Chen X, Zhang H, Xiao B. C9orf16 represents the aberrant genetic programs and drives the progression of PDAC. BMC Cancer 2022; 22:1102. [PMID: 36307773 PMCID: PMC9615161 DOI: 10.1186/s12885-022-10202-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 10/19/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Pancreatic ductal adenocarcinoma (PDAC), constituting 90% of pancreatic cancers, is the fourth leading cause of cancer-related deaths in the world. Lack of early detection of PDAC contributes to its poor prognosis as patients are often diagnosed at an advanced stage of disease. This is mostly due to the lack of promising diagnostic and therapeutic targets and corresponding drugs.
Methods and results
Here, by bioinformatic analysis of single cell RNA-sequencing data on normal pancreas tissues, primary and metastatic PDAC tumors, we identified a promising PDAC biomarker, C9orf16. The expression of C9orf16, rarely detectable in normal epithelial cells, was upregulated in primary PDAC cancer cells and was further elevated in metastatic PDAC cancer cells. Gain or loss of function of C9orf16 demonstrated its critical functions in regulating the cell proliferation, invasion and chemotherapy resistance of cancer cells. Pathway analysis and functional studies identified MYC signaling pathways as the most activated pathways in regulating C9orf16 expression and in mediating the development and progression of PDAC.
Conclusions
These data suggested a crucial gene regulation system, MYC-C9orf16, which is actively involved in PDAC development and progression, and targeting this system should be a novel diagnostic and therapeutic target for PDAC.
Collapse
|
13
|
Orben F, Lankes K, Schneeweis C, Hassan Z, Jakubowsky H, Krauß L, Boniolo F, Schneider C, Schäfer A, Murr J, Schlag C, Kong B, Öllinger R, Wang C, Beyer G, Mahajan UM, Xue Y, Mayerle J, Schmid RM, Kuster B, Rad R, Braun CJ, Wirth M, Reichert M, Saur D, Schneider G. Epigenetic drug screening defines a PRMT5 inhibitor-sensitive pancreatic cancer subtype. JCI Insight 2022; 7:e151353. [PMID: 35439169 PMCID: PMC9220834 DOI: 10.1172/jci.insight.151353] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 04/12/2022] [Indexed: 11/17/2022] Open
Abstract
Systemic therapies for pancreatic ductal adenocarcinoma (PDAC) remain unsatisfactory. Clinical prognosis is particularly poor for tumor subtypes with activating aberrations in the MYC pathway, creating an urgent need for novel therapeutic targets. To unbiasedly find MYC-associated epigenetic dependencies, we conducted a drug screen in pancreatic cancer cell lines. Here, we found that protein arginine N-methyltransferase 5 (PRMT5) inhibitors triggered an MYC-associated dependency. In human and murine PDACs, a robust connection of MYC and PRMT5 was detected. By the use of gain- and loss-of-function models, we confirmed the increased efficacy of PRMT5 inhibitors in MYC-deregulated PDACs. Although inhibition of PRMT5 was inducing DNA damage and arresting PDAC cells in the G2/M phase of the cell cycle, apoptotic cell death was executed predominantly in cells with high MYC expression. Experiments in primary patient-derived PDAC models demonstrated the existence of a highly PRMT5 inhibitor-sensitive subtype. Our work suggests developing PRMT5 inhibitor-based therapies for PDAC.
Collapse
Affiliation(s)
- Felix Orben
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar and
| | | | - Christian Schneeweis
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar and
- Institute for Translational Cancer Research and Experimental Cancer Therapy, Technical University Munich (TUM), Munich, Germany
| | - Zonera Hassan
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar and
| | - Hannah Jakubowsky
- Institute for Translational Cancer Research and Experimental Cancer Therapy, Technical University Munich (TUM), Munich, Germany
| | - Lukas Krauß
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar and
- University Medical Center Göttingen, Department of General, Visceral and Pediatric Surgery, Göttingen, Germany
| | - Fabio Boniolo
- Institute for Translational Cancer Research and Experimental Cancer Therapy, Technical University Munich (TUM), Munich, Germany
| | - Carolin Schneider
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar and
- University Medical Center Göttingen, Department of General, Visceral and Pediatric Surgery, Göttingen, Germany
| | - Arlett Schäfer
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar and
| | - Janine Murr
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar and
| | | | - Bo Kong
- Department of Surgery, Klinikum rechts der Isar, TUM, Munich, Germany
- Department of General Surgery, University of Ulm, Ulm, Germany
| | - Rupert Öllinger
- Institute of Molecular Oncology and Functional Genomics, TUM School of Medicine and
| | - Chengdong Wang
- Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, TUM, Freising, Germany
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- Department of Surgery, Children’s Hospital of Soochow University, Suzhou, China
| | - Georg Beyer
- Department of Medicine II, LMU University Hospital, Ludwig-Maximilians-Universität München (LMU Munich), Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Ujjwal M. Mahajan
- Department of Medicine II, LMU University Hospital, Ludwig-Maximilians-Universität München (LMU Munich), Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Yonggan Xue
- Department of Medicine II, LMU University Hospital, Ludwig-Maximilians-Universität München (LMU Munich), Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Julia Mayerle
- Department of Medicine II, LMU University Hospital, Ludwig-Maximilians-Universität München (LMU Munich), Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Roland M. Schmid
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar and
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, TUM, Freising, Germany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), TUM, Freising, Germany
| | - Roland Rad
- Institute of Molecular Oncology and Functional Genomics, TUM School of Medicine and
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Christian J. Braun
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Matthias Wirth
- Department of Hematology, Oncology and Tumor Immunology, Campus Benjamin Franklin, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Maximilian Reichert
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar and
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Center for Protein Assemblies (CPA), TUM, Garching, Germany
- Translational Pancreatic Research Cancer Center, Medical Clinic and Polyclinic II, Klinikum rechts der Isar, TUM, Munich, Germany
| | - Dieter Saur
- Institute for Translational Cancer Research and Experimental Cancer Therapy, Technical University Munich (TUM), Munich, Germany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Günter Schneider
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar and
- University Medical Center Göttingen, Department of General, Visceral and Pediatric Surgery, Göttingen, Germany
| |
Collapse
|
14
|
Choi HI, An GY, Yoo E, Baek M, Binas B, Chai JC, Lee YS, Jung KH, Chai YG. The bromodomain inhibitor JQ1 up-regulates the long non-coding RNA MALAT1 in cultured human hepatic carcinoma cells. Sci Rep 2022; 12:7779. [PMID: 35546353 PMCID: PMC9095596 DOI: 10.1038/s41598-022-11868-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 04/29/2022] [Indexed: 11/27/2022] Open
Abstract
The epigenetic reader, bromodomain-containing 4 (BRD4), is overexpressed in hepatocellular carcinoma (HCC), and BRD4 inhibition is considered as a new therapeutic approach. The BRD inhibitor JQ1 is known to inhibit the enrichment of BRD4 at enhancer sites. Gene network analyses have implicated long non-coding RNAs (lncRNAs) in the effects of JQ1, but the precise molecular events remain unexplored. Here, we report that in HepG2 cells, JQ1 significantly reduced various proliferation-related lncRNAs, but up-regulated the known liver tumor marker, MALAT1. Using ChIP-sequencing data, ChIP-qPCR, luciferase reporter assays, and chromatin conformation capture (3C), we characterized the MALAT1 gene locus. We found that JQ1 elicited a rearrangement of its chromatin looping conformation, which involved the putative enhancers E1, E2, E3, the gene body, and the promoter. We further found that the forkhead box protein A2 (FOXA2) binds to E2 and the promoter; suppression of FOXA2 expression resulted in MALAT1 up-regulation and increased cell proliferation. These results suggest that the inhibition of MALAT1 may improve the effect of BET inhibitors as an anti-cancer therapy and that FOXA2 would be a suitable target for that approach.
Collapse
Affiliation(s)
- Hae In Choi
- Department of Bionanotechnology, Hanyang University, Seoul, 04673, Republic of Korea
| | - Ga Yeong An
- Department of Bionanotechnology, Hanyang University, Seoul, 04673, Republic of Korea
| | - Eunyoung Yoo
- Department of Bionanotechnology, Hanyang University, Seoul, 04673, Republic of Korea
| | - Mina Baek
- Department of Molecular and Life Science, Hanyang University, Ansan, Gyeonggi-do, 15588, Republic of Korea
- Institute of Natural Science and Technology, Hanyang University, Ansan, 15588, Republic of Korea
| | - Bert Binas
- Department of Molecular and Life Science, Hanyang University, Ansan, Gyeonggi-do, 15588, Republic of Korea
| | - Jin Choul Chai
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Young Seek Lee
- College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kyoung Hwa Jung
- Convergence Technology Campus of Korea Polytechnic II, Incheon, 21417, Republic of Korea.
| | - Young Gyu Chai
- Department of Bionanotechnology, Hanyang University, Seoul, 04673, Republic of Korea.
- Department of Molecular and Life Science, Hanyang University, Ansan, Gyeonggi-do, 15588, Republic of Korea.
| |
Collapse
|
15
|
Ganji C, Farran B. Current clinical trials for epigenetic targets and therapeutic inhibitors for pancreatic cancer therapy. Drug Discov Today 2022; 27:1404-1410. [PMID: 34952224 DOI: 10.1016/j.drudis.2021.12.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/08/2021] [Accepted: 12/17/2021] [Indexed: 02/06/2023]
Abstract
Pancreatic cancer (PC) is an aggressive disease characterized by high mortality. Diagnosis at advanced stage, resistance, and recurrence are major hurdles for PC therapy and contribute to poor survival rate. Mutations in tumor-promoting kinases and epigenetic dysregulation in tumor suppressor genes are hallmarks of PC and can be used for diagnosis and therapy. In this review, we highlight dysregulated genes associated with epigenetic mechanisms, including DNA methylation and histone acetylation, involved in PC progression and resistance. We also explore epigenetic drugs currently in clinical trials. Combining epigenetic drugs and targeted therapies might represent a promising approach for PC.
Collapse
Affiliation(s)
| | - Batoul Farran
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
16
|
Lee WK, Cheng SY. Targeting transcriptional regulators for treatment of anaplastic thyroid cancer. JOURNAL OF CANCER METASTASIS AND TREATMENT 2021; 7. [PMID: 34761120 PMCID: PMC8577520 DOI: 10.20517/2394-4722.2021.58] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dysregulation of genes perpetuates cancer progression. During carcinogenesis, cancer cells acquire dependency of aberrant transcriptional programs (known as “transcription addiction”) to meet the high demands for uncontrolled proliferation. The needs for particular transcription programs for cancer growth could be cancer-type-selective. The dependencies of certain transcription regulators could be exploited for therapeutic benefits. Anaplastic thyroid cancer (ATC) is an extremely aggressive human cancer for which new treatment modalities are urgently needed. Its resistance to conventional treatments and the lack of therapeutic options for improving survival might have been attributed to extensive genetic heterogeneity due to subsequent evolving genetic alterations and clonal selections during carcinogenesis. Despite this genetic complexity, mounting evidence has revealed a characteristic transcriptional addiction of ATC cells resulting in evolving diverse oncogenic signaling for cancer cell survival. The transcriptional addiction has presented a huge challenge for effective targeting as shown by the failure of previous targeted therapies. However, an emerging notion is that many different oncogenic signaling pathways activated by multiple upstream driver mutations might ultimately converge on the transcriptional responses, which would provide an opportunity to target transcriptional regulators for treatment of ATC. Here, we review the current understanding of how genetic alterations in cancer distorted the transcription program, leading to acquisition of transcriptional addiction. We also highlight recent findings from studies aiming to exploit the opportunity for targeting transcription regulators as potential therapeutics for ATC.
Collapse
Affiliation(s)
- Woo Kyung Lee
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sheue-Yann Cheng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
17
|
Sankarasubramanian S, Pfohl U, Regenbrecht CRA, Reinhard C, Wedeken L. Context Matters-Why We Need to Change From a One Size Fits all Approach to Made-to-Measure Therapies for Individual Patients With Pancreatic Cancer. Front Cell Dev Biol 2021; 9:760705. [PMID: 34805167 PMCID: PMC8599957 DOI: 10.3389/fcell.2021.760705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer is one of the deadliest cancers and remains a major unsolved health problem. While pancreatic ductal adenocarcinoma (PDAC) is associated with driver mutations in only four major genes (KRAS, TP53, SMAD4, and CDKN2A), every tumor differs in its molecular landscape, histology, and prognosis. It is crucial to understand and consider these differences to be able to tailor treatment regimens specific to the vulnerabilities of the individual tumor to enhance patient outcome. This review focuses on the heterogeneity of pancreatic tumor cells and how in addition to genetic alterations, the subsequent dysregulation of multiple signaling cascades at various levels, epigenetic and metabolic factors contribute to the oncogenesis of PDAC and compensate for each other in driving cancer progression if one is tackled by a therapeutic approach. This implicates that besides the need for new combinatorial therapies for PDAC, a personalized approach for treating this highly complex cancer is required. A strategy that combines both a target-based and phenotypic approach to identify an effective treatment, like Reverse Clinical Engineering® using patient-derived organoids, is discussed as a promising way forward in the field of personalized medicine to tackle this deadly disease.
Collapse
Affiliation(s)
| | - Ulrike Pfohl
- CELLphenomics GmbH, Berlin, Germany
- ASC Oncology GmbH, Berlin, Germany
- Institute for Molecular Bio Science, Goethe University Frankfurt Am Main, Frankfurt, Germany
| | - Christian R. A. Regenbrecht
- CELLphenomics GmbH, Berlin, Germany
- ASC Oncology GmbH, Berlin, Germany
- Institute for Pathology, Universitätsklinikum Göttingen, Göttingen, Germany
| | | | - Lena Wedeken
- CELLphenomics GmbH, Berlin, Germany
- ASC Oncology GmbH, Berlin, Germany
| |
Collapse
|
18
|
Schneider G, Wirth M, Keller U, Saur D. Rationale for MYC imaging and targeting in pancreatic cancer. EJNMMI Res 2021; 11:104. [PMID: 34637026 PMCID: PMC8511206 DOI: 10.1186/s13550-021-00843-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/23/2021] [Indexed: 12/11/2022] Open
Abstract
The incidence and lethality of pancreatic ductal adenocarcinoma (PDAC) will continue to increase in the next decade. For most patients, chemotherapeutic combination therapies remain the standard of care. The development and successful implementation of precision oncology in other gastrointestinal tumor entities point to opportunities also for PDAC. Therefore, markers linked to specific therapeutic responses and important subgroups of the disease are needed. The MYC oncogene is a relevant driver in PDAC and is linked to drug resistance and sensitivity. Here, we update recent insights into MYC biology in PDAC, summarize the connections between MYC and drug responses, and point to an opportunity to image MYC non-invasively. In sum, we propose MYC-associated biology as a basis for the development of concepts for precision oncology in PDAC.
Collapse
Affiliation(s)
- Günter Schneider
- Medical Clinic and Policlinic II, Klinikum Rechts Der Isar, TU Munich, 81675, Munich, Germany. .,German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), 69120, Heidelberg, Germany. .,Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, 37075, Göttingen, Germany.
| | - Matthias Wirth
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), 69120, Heidelberg, Germany. .,Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, 12203, Berlin, Germany.
| | - Ulrich Keller
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), 69120, Heidelberg, Germany.,Department of Hematology, Oncology and Cancer Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, 12203, Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine, 13125, Berlin, Germany
| | - Dieter Saur
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), 69120, Heidelberg, Germany.,Insititute for Translational Cancer Research and Experimental Cancer Therapy, MRI, TU Munich, 81675, Munich, Germany
| |
Collapse
|
19
|
Yin Z, Maswikiti EP, Liu Q, Bai Y, Li X, Qi W, Liu L, Ma Y, Chen H. Current research developments of patient-derived tumour xenograft models (Review). Exp Ther Med 2021; 22:1206. [PMID: 34584551 DOI: 10.3892/etm.2021.10640] [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: 06/01/2020] [Accepted: 05/04/2021] [Indexed: 11/06/2022] Open
Abstract
Patient-derived tumor xenograft (PDTX) models are established by transferring patient tumors into immunodeficient mice. In these murine models, the characteristics of the primary tumor are retained, including the microenvironment of tumor cell growth and histopathology. Due to this, it has become the most reliable in vivo human cancer model. However, the success rates differ by type of tumor, site of transplantation and tumor aggressiveness. Subcutaneous transplantation is a standard method for PDTX, and subrenal capsule transplantation improves the engraftment rate. Recently, PDTX models are frequently used in the fields of precision medicine, predictive biomarkers, evaluation of drug efficacy and preclinical research on tumor immunotherapeutic drugs. The aim of the present article was to review the establishment, clinical applications and limitations of the PDTX model in tumor research.
Collapse
Affiliation(s)
- Zhenyu Yin
- The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Ewetse Paul Maswikiti
- The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Qian Liu
- The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Yuping Bai
- The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Xiaomei Li
- The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Wenbo Qi
- The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Le Liu
- The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Yanling Ma
- The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Hao Chen
- Department of Oncology, Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| |
Collapse
|
20
|
Pook H, Pauklin S. Mechanisms of Cancer Cell Death: Therapeutic Implications for Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2021; 13:4834. [PMID: 34638318 PMCID: PMC8508208 DOI: 10.3390/cancers13194834] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 12/20/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a type of cancer that is strongly associated with poor prognosis and short median survival times. In stark contrast to the progress seen in other cancer types in recent decades, discoveries of new treatments in PDAC have been few and far between and there has been little improvement in overall survival (OS). The difficulty in treating this disease is multifactorial, contributed to by late presentation, difficult access to primary tumour sites, an 'immunologically cold' phenotype, and a strong tendency of recurrence likely driven by cancer stem cell (CSC) populations. Furthermore, apparently contrasting roles of tumour components (such as fibrotic stroma) and intracellular pathways (such as autophagy and TGFβ) have made it difficult to distinguish beneficial from detrimental drug targets. Despite this, progress has been made in the field, including the determination of mFOLFIRINOX as the standard-of-care adjuvant therapy and the discovery of KRASG12C mutant inhibitors. Moreover, new research, as outlined in this review, has highlighted promising new approaches including the targeting of the tumour microenvironment, enhancement of immunotherapies, epigenetic modulation, and destruction of CSCs.
Collapse
Affiliation(s)
| | - Siim Pauklin
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Old Road, University of Oxford, Oxford OX3 7LD, UK;
| |
Collapse
|
21
|
Mohamed Abd-El-Halim Y, El Kaoutari A, Silvy F, Rubis M, Bigonnet M, Roques J, Cros J, Nicolle R, Iovanna J, Dusetti N, Mas E. A glycosyltransferase gene signature to detect pancreatic ductal adenocarcinoma patients with poor prognosis. EBioMedicine 2021; 71:103541. [PMID: 34425307 PMCID: PMC8379629 DOI: 10.1016/j.ebiom.2021.103541] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 07/26/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is characterized by an important heterogeneity, reflected by different clinical outcomes and chemoresistance. During carcinogenesis, tumor cells display aberrant glycosylated structures, synthetized by deregulated glycosyltransferases, supporting the tumor progression. In this study, we aimed to determine whether PDAC could be stratified through their glycosyltransferase expression profiles better than the current binary classification (basal-like and classical) in order to improve detection of patients with poor prognosis. Methods Bioinformatic analysis of 169 glycosyltransferase RNA sequencing data were performed for 74 patient-derived xenografts (PDX) of resected and unresectable tumors. The Australian cohort of International Cancer Genome Consortium and the microarray dataset from Puleo patient's cohort were used as independent validation datasets. Findings New PDAC stratification based on glycosyltransferase expression profile allowed to distinguish different groups of patients with distinct clinical outcome (p-value = 0.007). A combination of 19 glycosyltransferases differentially expressed in PDX defined a glyco-signature, whose prognostic value was validated on datasets including resected whole tumor tissues. The glyco-signature was able to discriminate three clusters of PDAC patients on the validation cohorts, two clusters displaying a short overall survival compared to one cluster having a better prognosis. Both poor prognostic clusters having different glyco-profiles in Puleo patient's cohort were correlated with stroma activated or desmoplastic subtypes corresponding to distinct microenvironment features (p-value < 0.0001). Besides, differential expression and enrichment analyses revealed deregulated functional pathways specific to different clusters. Interpretation This study identifies a glyco-signature relevant for a prognostic use, potentially applicable to resected and unresectable PDAC. Furthermore, it provides new potential therapeutic targets. Funding This work was supported by INCa (Grants number 2018-078 and 2018-079), Fondation ARC (Grant number ARCPJA32020070002326), Cancéropôle PACA, DGOS (labelization SIRIC, Grant number 6038), Amidex Foundation and Ligue Nationale Contre le Cancer and by institutional fundings from INSERM and the Aix-Marseille Université.
Collapse
Affiliation(s)
- Yousra Mohamed Abd-El-Halim
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Abdessamad El Kaoutari
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Françoise Silvy
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Marion Rubis
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Martin Bigonnet
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Julie Roques
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Jérôme Cros
- Department of Pathology, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, France
| | - Rémy Nicolle
- Programme Cartes d'Identité des Tumeurs (CIT), Ligue Nationale Contre Le Cancer, Paris, France
| | - Juan Iovanna
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Nelson Dusetti
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France.
| | - Eric Mas
- Cancer Research Center of Marseille, Aix Marseille University, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France.
| |
Collapse
|
22
|
Eyres M, Lanfredini S, Xu H, Burns A, Blake A, Willenbrock F, Goldin R, Hughes D, Hughes S, Thapa A, Vavoulis D, Hubert A, D'Costa Z, Sabbagh A, Abraham AG, Blancher C, Jones S, Verrill C, Silva M, Soonawalla Z, Maughan T, Schuh A, Mukherjee S, O'Neill E. TET2 Drives 5hmc Marking of GATA6 and Epigenetically Defines Pancreatic Ductal Adenocarcinoma Transcriptional Subtypes. Gastroenterology 2021; 161:653-668.e16. [PMID: 33915173 DOI: 10.1053/j.gastro.2021.04.044] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 03/12/2021] [Accepted: 04/07/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Pancreatic ductal adenocarcinoma (PDAC) is characterized by advanced disease stage at presentation, aggressive disease biology, and resistance to therapy, resulting in an extremely poor 5-year survival rate of <10%. PDAC is classified into transcriptional subtypes with distinct survival characteristics, although how these arise is not known. Epigenetic deregulation, rather than genetics, has been proposed to underpin progression, but exactly why is unclear and is hindered by the technical limitations of analyzing clinical samples. METHODS We performed genome-wide epigenetic mapping of DNA modifications 5-methylcytosine and 5-hydroxymethylcytosine (5hmc) using oxidative bisulfite sequencing from formalin-embedded sections. We identified overlap with transcriptional signatures in formalin-fixed, paraffin-embedded tissue from resected patients, via bioinformatics using iCluster and mutational profiling and confirmed them in vivo. RESULTS We found that aggressive squamous-like PDAC subtypes result from epigenetic inactivation of loci, including GATA6, which promote differentiated classical pancreatic subtypes. We showed that squamous-like PDAC transcriptional subtypes are associated with greater loss of 5hmc due to reduced expression of the 5-methylcytosine hydroxylase TET2. Furthermore, we found that SMAD4 directly supports TET2 levels in classical pancreatic tumors, and loss of SMAD4 expression was associated with reduced 5hmc, GATA6, and squamous-like tumors. Importantly, enhancing TET2 stability using metformin and vitamin C/ascorbic acid restores 5hmc and GATA6 levels, reverting squamous-like tumor phenotypes and WNT-dependence in vitro and in vivo. CONCLUSIONS We identified epigenetic deregulation of pancreatic differentiation as an underpinning event behind the emergence of transcriptomic subtypes in PDAC. Our data showed that restoring epigenetic control increases biomarkers of classical pancreatic tumors that are associated with improved therapeutic responses and survival.
Collapse
MESH Headings
- 5-Methylcytosine/analogs & derivatives
- 5-Methylcytosine/metabolism
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Ascorbic Acid/pharmacology
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/enzymology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/pathology
- Cell Differentiation
- Cell Line, Tumor
- DNA Methylation/drug effects
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Dioxygenases/genetics
- Dioxygenases/metabolism
- Epigenesis, Genetic/drug effects
- Epigenome
- Epigenomics
- GATA6 Transcription Factor/genetics
- GATA6 Transcription Factor/metabolism
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Metformin/pharmacology
- Mice, Nude
- Mice, Transgenic
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/enzymology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/pathology
- Retrospective Studies
- Smad4 Protein/genetics
- Smad4 Protein/metabolism
- Transcription, Genetic/drug effects
- Transcriptome
- Wnt Signaling Pathway/genetics
- Xenograft Model Antitumor Assays
- Mice
Collapse
Affiliation(s)
- Michael Eyres
- Department of Oncology, University of Oxford, Oxford, UK
| | | | - Haonan Xu
- Department of Oncology, University of Oxford, Oxford, UK
| | - Adam Burns
- Department of Oncology, University of Oxford, Oxford, UK
| | - Andrew Blake
- Department of Oncology, University of Oxford, Oxford, UK
| | | | - Robert Goldin
- Centre for Pathology, Imperial College, London, United Kingdom
| | - Daniel Hughes
- Department of Oncology, University of Oxford, Oxford, UK; Department of Hepatobiliary and Pancreatic Surgery, Oxford University Hospitals National Health Service Foundation Trust, Oxford, United Kingdom
| | - Sophie Hughes
- Department of Oncology, University of Oxford, Oxford, UK
| | - Asmita Thapa
- Department of Oncology, University of Oxford, Oxford, UK
| | | | - Aline Hubert
- Department of Oncology, University of Oxford, Oxford, UK
| | | | - Ahmad Sabbagh
- Department of Oncology, University of Oxford, Oxford, UK
| | | | - Christine Blancher
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Stephanie Jones
- Oxford Radcliffe Biobank, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| | - Clare Verrill
- Nuffield Department of Surgical Sciences and Oxford National Institute for Health Research Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Michael Silva
- Department of Hepatobiliary and Pancreatic Surgery, Oxford University Hospitals National Health Service Foundation Trust, Oxford, United Kingdom
| | - Zahir Soonawalla
- Department of Hepatobiliary and Pancreatic Surgery, Oxford University Hospitals National Health Service Foundation Trust, Oxford, United Kingdom
| | | | - Anna Schuh
- Department of Oncology, University of Oxford, Oxford, UK
| | | | - Eric O'Neill
- Department of Oncology, University of Oxford, Oxford, UK.
| |
Collapse
|
23
|
Shibasaki H, Kinoh H, Cabral H, Quader S, Mochida Y, Liu X, Toh K, Miyano K, Matsumoto Y, Yamasoba T, Kataoka K. Efficacy of pH-Sensitive Nanomedicines in Tumors with Different c-MYC Expression Depends on the Intratumoral Activation Profile. ACS NANO 2021; 15:5545-5559. [PMID: 33625824 DOI: 10.1021/acsnano.1c00364] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Effective inhibition of the protein derived from cellular myelocytomatosis oncogene (c-Myc) is one of the most sought-after goals in cancer therapy. While several c-Myc inhibitors have demonstrated therapeutic potential, inhibiting c-Myc has proven challenging, since c-Myc is essential for normal tissues and tumors may present heterogeneous c-Myc levels demanding contrasting therapeutic strategies. Herein, we developed tumor-targeted nanomedicines capable of treating both tumors with high and low c-Myc levels by adjusting their ability to spatiotemporally control drug action. These nanomedicines loaded homologues of the bromodomain and extraterminal (BET) motif inhibitor JQ1 as epigenetic c-Myc inhibitors through pH-cleavable bonds engineered for fast or slow drug release at intratumoral pH. In tumors with high c-Myc expression, the fast-releasing (FR) nanomedicines suppressed tumor growth more effectively than the slow-releasing (SR) ones, whereas, in the low c-Myc tumors, the efficacy of the nanomedicines was the opposite. By studying the tumor distribution and intratumoral activation of the nanomedicines, we found that, despite SR nanomedicines achieved higher accumulation than the FR counterparts in both c-Myc high and low tumors, the antitumor activity profiles corresponded with the availability of activated drugs inside the tumors. These results indicate the potential of engineered nanomedicines for c-Myc inhibition and spur the idea of precision pH-sensitive nanomedicine based on cancer biomarker levels.
Collapse
Affiliation(s)
- Hitoshi Shibasaki
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Hiroaki Kinoh
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Horacio Cabral
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Sabina Quader
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Yuki Mochida
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Xueying Liu
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Kazuko Toh
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
| | - Kazuki Miyano
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Department of Otorhinolaryngology, Tokyo Yamate Medical Center, 3-22-1, Hyakunin-cho, Shinjuku-ku, Tokyo 169-0073, Japan
| | - Yu Matsumoto
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Tatsuya Yamasoba
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Kazunori Kataoka
- Innovation Center of NanoMedicine, Kawasaki Institute of Industrial Promotion, 3-25-14, Tonomachi, Kawasaki-ku, Kawasaki 210-0821, Japan
- Policy Alternative Research Institute, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
24
|
Schneeweis C, Hassan Z, Schick M, Keller U, Schneider G. The SUMO pathway in pancreatic cancer: insights and inhibition. Br J Cancer 2021; 124:531-538. [PMID: 33071285 PMCID: PMC7851129 DOI: 10.1038/s41416-020-01119-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 08/31/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
An urgent medical need to develop novel treatment strategies for patients with pancreatic ductal adenocarcinoma (PDAC) exists. However, despite various efforts in the histopathological and molecular subtyping of PDAC, novel targeted or specific therapies have not been established. Posttranslational modifications (PTMs) with ubiquitin-like proteins, including small ubiquitin-like modifiers (SUMOs), mediate numerous processes that can contribute to the fitness and survival of cancer cells. The contribution of SUMOylation to transcriptional control, DNA repair pathways, mitotic progression, and oncogenic signalling has been described. Here we review functions of the SUMO pathway in PDAC, with a special focus on its connection to an aggressive subtype of the disease characterised by high MYC activity, and discuss SUMOylation inhibitors under development for precise PDAC therapies.
Collapse
Affiliation(s)
- Christian Schneeweis
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University Munich, 81675, München, Germany
| | - Zonera Hassan
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University Munich, 81675, München, Germany
| | - Markus Schick
- Department of Hematology, Oncology and Tumor Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, 12203, Berlin, Germany
| | - Ulrich Keller
- Department of Hematology, Oncology and Tumor Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Hindenburgdamm 30, 12203, Berlin, Germany.
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), 69120, Heidelberg, Germany.
- Max-Delbrück-Center for Molecular Medicine, 13092, Berlin, Germany.
| | - Günter Schneider
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University Munich, 81675, München, Germany.
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), 69120, Heidelberg, Germany.
| |
Collapse
|
25
|
Alonso-Curbelo D, Ho YJ, Burdziak C, Maag JLV, Morris JP, Chandwani R, Chen HA, Tsanov KM, Barriga FM, Luan W, Tasdemir N, Livshits G, Azizi E, Chun J, Wilkinson JE, Mazutis L, Leach SD, Koche R, Pe'er D, Lowe SW. A gene-environment-induced epigenetic program initiates tumorigenesis. Nature 2021; 590:642-648. [PMID: 33536616 PMCID: PMC8482641 DOI: 10.1038/s41586-020-03147-x] [Citation(s) in RCA: 150] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023]
Abstract
Tissue damage increases the risk of cancer through poorly understood mechanisms1. In mouse models of pancreatic cancer, pancreatitis associated with tissue injury collaborates with activating mutations in the Kras oncogene to markedly accelerate the formation of early neoplastic lesions and, ultimately, adenocarcinoma2,3. Here, by integrating genomics, single-cell chromatin assays and spatiotemporally controlled functional perturbations in autochthonous mouse models, we show that the combination of Kras mutation and tissue damage promotes a unique chromatin state in the pancreatic epithelium that distinguishes neoplastic transformation from normal regeneration and is selected for throughout malignant evolution. This cancer-associated epigenetic state emerges within 48 hours of pancreatic injury, and involves an 'acinar-to-neoplasia' chromatin switch that contributes to the early dysregulation of genes that define human pancreatic cancer. Among the factors that are most rapidly activated after tissue damage in the pre-malignant pancreatic epithelium is the alarmin cytokine interleukin 33, which recapitulates the effects of injury in cooperating with mutant Kras to unleash the epigenetic remodelling program of early neoplasia and neoplastic transformation. Collectively, our study demonstrates how gene-environment interactions can rapidly produce gene-regulatory programs that dictate early neoplastic commitment, and provides a molecular framework for understanding the interplay between genetic and environmental cues in the initiation of cancer.
Collapse
Affiliation(s)
- Direna Alonso-Curbelo
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yu-Jui Ho
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cassandra Burdziak
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Tri-Institutional Training Program in Computational Biology and Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jesper L V Maag
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John P Morris
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rohit Chandwani
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Surgery, Weill Cornell Medical College, New York, NY, USA
- Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY, USA
| | - Hsuan-An Chen
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kaloyan M Tsanov
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Francisco M Barriga
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wei Luan
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nilgun Tasdemir
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Geulah Livshits
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elham Azizi
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jaeyoung Chun
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Alan and Sandra Gerry Metastasis and Tumor Ecosystems Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John E Wilkinson
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Linas Mazutis
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Alan and Sandra Gerry Metastasis and Tumor Ecosystems Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Steven D Leach
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Dartmouth Norris Cotton Cancer Center, Hanover, NH, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard Koche
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dana Pe'er
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Scott W Lowe
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
26
|
Masoud R, Reyes-Castellanos G, Lac S, Garcia J, Dou S, Shintu L, Abdel Hadi N, Gicquel T, El Kaoutari A, Diémé B, Tranchida F, Cormareche L, Borge L, Gayet O, Pasquier E, Dusetti N, Iovanna J, Carrier A. Targeting Mitochondrial Complex I Overcomes Chemoresistance in High OXPHOS Pancreatic Cancer. Cell Rep Med 2020; 1:100143. [PMID: 33294863 PMCID: PMC7691450 DOI: 10.1016/j.xcrm.2020.100143] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 08/28/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023]
Abstract
Mitochondrial respiration (oxidative phosphorylation, OXPHOS) is an emerging target in currently refractory cancers such as pancreatic ductal adenocarcinoma (PDAC). However, the variability of energetic metabolic adaptations between PDAC patients has not been assessed in functional investigations. In this work, we demonstrate that OXPHOS rates are highly heterogeneous between patient tumors, and that high OXPHOS tumors are enriched in mitochondrial respiratory complex I at protein and mRNA levels. Therefore, we treated PDAC cells with phenformin (complex I inhibitor) in combination with standard chemotherapy (gemcitabine), showing that this treatment is synergistic specifically in high OXPHOS cells. Furthermore, phenformin cooperates with gemcitabine in high OXPHOS tumors in two orthotopic mouse models (xenografts and syngeneic allografts). In conclusion, this work proposes a strategy to identify PDAC patients likely to respond to the targeting of mitochondrial energetic metabolism in combination with chemotherapy, and that phenformin should be clinically tested in appropriate PDAC patient subpopulations.
Collapse
Affiliation(s)
- Rawand Masoud
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Gabriela Reyes-Castellanos
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Sophie Lac
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Julie Garcia
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Samir Dou
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Laetitia Shintu
- Aix Marseille Université, CNRS, Centrale Marseille, ISM2, F-13013 Marseille, France
| | - Nadine Abdel Hadi
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Tristan Gicquel
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Abdessamad El Kaoutari
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Binta Diémé
- Aix Marseille Université, CNRS, Centrale Marseille, ISM2, F-13013 Marseille, France
| | - Fabrice Tranchida
- Aix Marseille Université, CNRS, Centrale Marseille, ISM2, F-13013 Marseille, France
| | - Laurie Cormareche
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Laurence Borge
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Odile Gayet
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Eddy Pasquier
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Nelson Dusetti
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Juan Iovanna
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| | - Alice Carrier
- Aix Marseille Université, CNRS, INSERM, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille (CRCM), F-13009 Marseille, France
| |
Collapse
|
27
|
Lankes K, Hassan Z, Doffo MJ, Schneeweis C, Lier S, Öllinger R, Rad R, Krämer OH, Keller U, Saur D, Reichert M, Schneider G, Wirth M. Targeting the ubiquitin-proteasome system in a pancreatic cancer subtype with hyperactive MYC. Mol Oncol 2020; 14:3048-3064. [PMID: 33099868 PMCID: PMC7718946 DOI: 10.1002/1878-0261.12835] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/11/2020] [Accepted: 10/04/2020] [Indexed: 12/12/2022] Open
Abstract
The myelocytomatosis oncogene (MYC) is an important driver in a subtype of pancreatic ductal adenocarcinoma (PDAC). However, MYC remains a challenging therapeutic target; therefore, identifying druggable synthetic lethal interactions in MYC‐active PDAC may lead to novel precise therapies. First, to identify networks with hyperactive MYC, we profiled transcriptomes of established human cell lines, murine primary PDAC cell lines, and accessed publicly available repositories to analyze transcriptomes of primary human PDAC. Networks active in MYC‐hyperactive subtypes were analyzed by gene set enrichment analysis. Next, we performed an unbiased pharmacological screen to define MYC‐associated vulnerabilities. Hits were validated by analysis of drug response repositories and genetic gain‐ and loss‐of‐function experiments. In these experiments, we discovered that the proteasome inhibitor bortezomib triggers a MYC‐associated vulnerability. In addition, by integrating publicly available data, we found the unfolded protein response as a signature connected to MYC. Furthermore, increased sensitivity of MYC‐hyperactive PDACs to bortezomib was validated in genetically modified PDAC cells. In sum, we provide evidence that perturbing the ubiquitin–proteasome system (UPS) might be an option to target MYC‐hyperactive PDAC cells. Our data provide the rationale to further develop precise targeting of the UPS as a subtype‐specific therapeutic approach.
Collapse
Affiliation(s)
- Katharina Lankes
- Klinik und Poliklinik für Innere Medizin II, Technical University of Munich, Munich, Germany
| | - Zonera Hassan
- Klinik und Poliklinik für Innere Medizin II, Technical University of Munich, Munich, Germany
| | - María Josefina Doffo
- Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Campus Benjamin Franklin, Berlin, Germany
| | - Christian Schneeweis
- Klinik und Poliklinik für Innere Medizin II, Technical University of Munich, Munich, Germany
| | - Svenja Lier
- Klinik und Poliklinik für Innere Medizin II, Technical University of Munich, Munich, Germany
| | - Rupert Öllinger
- Institute of Molecular Oncology and Functional Genomics, Technical University Munich, Munich, Germany.,German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Roland Rad
- Institute of Molecular Oncology and Functional Genomics, Technical University Munich, Munich, Germany.,German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Oliver H Krämer
- Department of Toxicology, University Medical Center, Mainz, Germany
| | - Ulrich Keller
- Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Campus Benjamin Franklin, Berlin, Germany.,German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Dieter Saur
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany.,Institute for Translational Cancer Research and Experimental Cancer Therapy, Technical University Munich, Munich, Germany
| | - Maximilian Reichert
- Klinik und Poliklinik für Innere Medizin II, Technical University of Munich, Munich, Germany.,German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Günter Schneider
- Klinik und Poliklinik für Innere Medizin II, Technical University of Munich, Munich, Germany.,German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Matthias Wirth
- Hematology, Oncology and Tumor Immunology, Charité - Universitätsmedizin Campus Benjamin Franklin, Berlin, Germany
| |
Collapse
|
28
|
Biederstädt A, Hassan Z, Schneeweis C, Schick M, Schneider L, Muckenhuber A, Hong Y, Siegers G, Nilsson L, Wirth M, Dantes Z, Steiger K, Schunck K, Langston S, Lenhof HP, Coluccio A, Orben F, Slawska J, Scherger A, Saur D, Müller S, Rad R, Weichert W, Nilsson J, Reichert M, Schneider G, Keller U. SUMO pathway inhibition targets an aggressive pancreatic cancer subtype. Gut 2020; 69:1472-1482. [PMID: 32001555 PMCID: PMC7398468 DOI: 10.1136/gutjnl-2018-317856] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/20/2019] [Accepted: 12/22/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma (PDAC) still carries a dismal prognosis with an overall 5-year survival rate of 9%. Conventional combination chemotherapies are a clear advance in the treatment of PDAC; however, subtypes of the disease exist, which exhibit extensive resistance to such therapies. Genomic MYC amplifications represent a distinct subset of PDAC with an aggressive tumour biology. It is clear that hyperactivation of MYC generates dependencies that can be exploited therapeutically. The aim of the study was to find and to target MYC-associated dependencies. DESIGN We analysed human PDAC gene expression datasets. Results were corroborated by the analysis of the small ubiquitin-like modifier (SUMO) pathway in a large PDAC cohort using immunohistochemistry. A SUMO inhibitor was used and characterised using human and murine two-dimensional, organoid and in vivo models of PDAC. RESULTS We observed that MYC is connected to the SUMOylation machinery in PDAC. Components of the SUMO pathway characterise a PDAC subtype with a dismal prognosis and we provide evidence that hyperactivation of MYC is connected to an increased sensitivity to pharmacological SUMO inhibition. CONCLUSION SUMO inhibitor-based therapies should be further developed for an aggressive PDAC subtype.
Collapse
Affiliation(s)
- Alexander Biederstädt
- Medical Clinic and Policlinic III, Klinikum rechts der Isar, Technical University Munich, München, Germany
| | - Zonera Hassan
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University Munich, München, Germany
| | - Christian Schneeweis
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University Munich, München, Germany
| | - Markus Schick
- Department of Hematology, Oncology and Tumor Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lara Schneider
- Center for Bioinformatics, Saarland Informatics Campus, Saarland University, Saarbrücken, Germany,Saarbrücken Graduate School of Computer Science, Saarland Informatics Campus, Saarland University, Saarbrücken, Germany
| | | | - Yingfen Hong
- Medical Clinic and Policlinic III, Klinikum rechts der Isar, Technical University Munich, München, Germany
| | - Gerrit Siegers
- Medical Clinic and Policlinic III, Klinikum rechts der Isar, Technical University Munich, München, Germany
| | - Lisa Nilsson
- Department of Surgery, Sahlgrenska Cancer Center, Gothenburg University, Gothenburg, Sweden
| | - Matthias Wirth
- Department of Hematology, Oncology and Tumor Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Zahra Dantes
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University Munich, München, Germany
| | - Katja Steiger
- Institute of Pathology, Technical University Munich, München, Germany,German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Kathrin Schunck
- Goethe University, Medical School, Institute of Biochemistry II, Frankfurt, Germany
| | - Steve Langston
- Oncology Drug Discovery Unit, Takeda Pharmaceuticals International Co, Cambridge, Massachusetts, USA
| | - H-P Lenhof
- Center for Bioinformatics, Saarland Informatics Campus, Saarland University, Saarbrücken, Germany
| | - Andrea Coluccio
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University Munich, München, Germany,Institute for Translational Cancer Research and Experimental Cancer Therapy, Technical University Munich, München, Germany
| | - Felix Orben
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University Munich, München, Germany
| | - Jolanta Slawska
- Medical Clinic and Policlinic III, Klinikum rechts der Isar, Technical University Munich, München, Germany
| | - Anna Scherger
- Medical Clinic and Policlinic III, Klinikum rechts der Isar, Technical University Munich, München, Germany
| | - Dieter Saur
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany,Institute for Translational Cancer Research and Experimental Cancer Therapy, Technical University Munich, München, Germany
| | - Stefan Müller
- Goethe University, Medical School, Institute of Biochemistry II, Frankfurt, Germany
| | - Roland Rad
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany,Institute of Molecular Oncology and Functional Genomics, Technical University Munich, München, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University Munich, München, Germany,German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Jonas Nilsson
- Department of Surgery, Sahlgrenska Cancer Center, Gothenburg University, Gothenburg, Sweden
| | - Maximilian Reichert
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University Munich, München, Germany,German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Günter Schneider
- Medical Clinic and Polyclinic II, Klinikum rechts der Isar, Technical University Munich, München, Germany .,German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Ulrich Keller
- Department of Hematology, Oncology and Tumor Immunology, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany .,German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
29
|
Saraswat A, Patki M, Fu Y, Barot S, Dukhande VV, Patel K. Nanoformulation of PROteolysis TArgeting Chimera targeting ‘undruggable’ c-Myc for the treatment of pancreatic cancer. Nanomedicine (Lond) 2020; 15:1761-1777. [DOI: 10.2217/nnm-2020-0156] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aim: To explore the anticancer activity of a novel BRD4 protein degrader ARV-825 (ARV) and its nanoformulation development (ARV-NP) for treatment of pancreatic cancer. Materials & methods: ARV-NP were prepared using nanoprecipitation method and characterized for their physicochemical properties and various anticancer cell culture assays. Results: ARV-NP (89.63 ± 16.39 nm) demonstrated good physical stability, negligible hemolysis and improved half-life of ARV. ARV-NP showed significant cytotoxicity, apoptosis and anticlonogenic effect in pancreatic cancer cells. Significant downregulation of target proteins BRD4, c-Myc, Bcl-2 and upregulation of apoptotic marker cleaved caspase-3 was observed. Most importantly, ARV-NP treatment significantly inhibited the cell viability of 3D tumor spheroids of pancreatic cancer. Conclusion: ARV-NP represents a novel therapeutic strategy for pancreatic cancer.
Collapse
Affiliation(s)
- Aishwarya Saraswat
- College of Pharmacy & Health Sciences, St. John’s University, Queens, NY 11439, USA
| | - Manali Patki
- College of Pharmacy & Health Sciences, St. John’s University, Queens, NY 11439, USA
| | - Yige Fu
- College of Pharmacy & Health Sciences, St. John’s University, Queens, NY 11439, USA
| | - Shrikant Barot
- College of Pharmacy & Health Sciences, St. John’s University, Queens, NY 11439, USA
| | - Vikas V Dukhande
- College of Pharmacy & Health Sciences, St. John’s University, Queens, NY 11439, USA
| | - Ketan Patel
- College of Pharmacy & Health Sciences, St. John’s University, Queens, NY 11439, USA
| |
Collapse
|
30
|
Evidencing a Pancreatic Ductal Adenocarcinoma Subpopulation Sensitive to the Proteasome Inhibitor Carfilzomib. Clin Cancer Res 2020; 26:5506-5519. [DOI: 10.1158/1078-0432.ccr-20-1232] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/27/2020] [Accepted: 07/13/2020] [Indexed: 01/04/2023]
|
31
|
Kandimalla R, Tomihara H, Banwait JK, Yamamura K, Singh G, Baba H, Goel A. A 15-Gene Immune, Stromal, and Proliferation Gene Signature that Significantly Associates with Poor Survival in Patients with Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 2020; 26:3641-3648. [PMID: 32234757 PMCID: PMC7367725 DOI: 10.1158/1078-0432.ccr-19-4044] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 03/03/2020] [Accepted: 03/26/2020] [Indexed: 01/05/2023]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease with dismal survival rates. Tumor microenvironment (TME), comprising of immune cells and cancer-associated fibroblasts, plays a key role in driving poor prognosis and resistance to chemotherapy. Herein, we aimed to identify a TME-associated, risk-stratification gene biomarker signature in PDAC. EXPERIMENTAL DESIGN The initial biomarker discovery was performed in The Cancer Genome Atlas (TCGA, n = 163) transcriptomic data. This was followed by independent validation of the gene signature in the International Cancer Genome Consortium (ICGC, n = 95), E-MTAB-6134 (n = 288), and GSE71729 (n = 123) datasets for predicting overall survival (OS), and for its ability to detect poor molecular subtypes. Clinical validation and nomogram establishment was undertaken by performing multivariate Cox regression analysis. RESULTS Our biomarker discovery effort identified a 15-gene immune, stromal, and proliferation (ISP) gene signature that significantly associated with poor OS [HR, 3.90; 95% confidence interval (CI), 2.36-6.41; P < 0.0001]. This signature also robustly predicted survival in three independent validation cohorts ICGC [HR, 2.63 (1.56-4.41); P < 0.0001], E-MTAB-6134 [HR, 1.53 (1.14-2.04); P = 0.004], and GSE71729 [HR, 2.33 (1.49-3.63); P < 0.0001]. Interestingly, the ISP signature also permitted identification of poor molecular PDAC subtypes with excellent accuracy in all four cohorts; TCGA (AUC = 0.94), ICGC (AUC = 0.91), E-MTAB-6134 (AUC = 0.80), and GSE71729 (AUC = 0.83). The ISP-derived high-risk patients exhibited significantly poor OS in a clinical validation cohort [n = 119; HR, 2.62 (1.50-4.56); P = 0.0004]. A nomogram was established which included the ISP, CA19-9, and T- and N-stage for eventual clinical translation. CONCLUSIONS We report a novel gene signature for risk-stratification and robust identification of patients with PDAC with poor molecular subtypes.
Collapse
Affiliation(s)
- Raju Kandimalla
- Center for Gastrointestinal Research; Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute, Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas
| | - Hideo Tomihara
- Center for Gastrointestinal Research; Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute, Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jasjit K Banwait
- Center for Gastrointestinal Research; Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute, Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas
| | - Kensuke Yamamura
- Department of Gastroenterological Surgery, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Gagandeep Singh
- Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, California
| | - Hideo Baba
- Department of Gastroenterological Surgery, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Ajay Goel
- Center for Gastrointestinal Research; Center for Translational Genomics and Oncology, Baylor Scott & White Research Institute, Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas.
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, California
| |
Collapse
|
32
|
Deng Y, Zhou T, Wu JL, Chen Y, Shen CY, Zeng M, Chen T, Zhang XM. The impact of molecular classification based on the transcriptome of pancreatic cancer: from bench to bedside. CHINESE JOURNAL OF ACADEMIC RADIOLOGY 2020; 3:67-75. [DOI: 10.1007/s42058-020-00037-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 05/16/2020] [Accepted: 05/18/2020] [Indexed: 07/25/2024]
|
33
|
Sodir NM, Kortlever RM, Barthet VJA, Campos T, Pellegrinet L, Kupczak S, Anastasiou P, Swigart LB, Soucek L, Arends MJ, Littlewood TD, Evan GI. MYC Instructs and Maintains Pancreatic Adenocarcinoma Phenotype. Cancer Discov 2020; 10:588-607. [PMID: 31941709 DOI: 10.1158/2159-8290.cd-19-0435] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 11/30/2019] [Accepted: 01/10/2020] [Indexed: 11/16/2022]
Abstract
The signature features of pancreatic ductal adenocarcinoma (PDAC) are its fibroinflammatory stroma, poor immune activity, and dismal prognosis. We show that acute activation of Myc in indolent pancreatic intraepithelial neoplasm (PanIN) epithelial cells in vivo is, alone, sufficient to trigger immediate release of instructive signals that together coordinate changes in multiple stromal and immune-cell types and drive transition to pancreatic adenocarcinomas that share all the characteristic stromal features of their spontaneous human counterpart. We also demonstrate that this Myc-driven PDAC switch is completely and immediately reversible: Myc deactivation/inhibition triggers meticulous disassembly of advanced PDAC tumor and stroma and concomitant death of tumor cells. Hence, both the formation and deconstruction of the complex PDAC phenotype are continuously dependent on a single, reversible Myc switch. SIGNIFICANCE: We show that Myc activation in indolent Kras G12D-induced PanIN epithelium acts as an immediate pleiotropic switch, triggering tissue-specific signals that instruct all the diverse signature stromal features of spontaneous human PDAC. Subsequent Myc deactivation or inhibition immediately triggers a program that coordinately disassembles PDAC back to PanIN.See related commentary by English and Sears, p. 495.
Collapse
Affiliation(s)
- Nicole M Sodir
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Roderik M Kortlever
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | | | - Tania Campos
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Luca Pellegrinet
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Steven Kupczak
- Cambridge Research Institute, Li Ka Shing Centre, Cambridge, United Kingdom
| | | | - Lamorna Brown Swigart
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, California
| | - Laura Soucek
- Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Mark J Arends
- Division of Pathology, Cancer Research UK Edinburgh Centre, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Trevor D Littlewood
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Gerard I Evan
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
34
|
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a rapidly evolving and most frequently fatal disease. Despite the enormous progress in understanding the mechanisms related to PDAC pathogenesis, the impact on patient management has not yet been possible. Pancreatic organoids can be generated from small amounts of tissue. One of the most promising applications of organoids is that they can serve as a platform for selecting the right drugs for each patient. This approach has the potential to identify individual therapeutic vulnerabilities by allowing the personalization of treatments. However, these analyzes require several weeks before obtaining enough organoids from the same individual, to carry out the tests with several drugs, and to analyze the results, which limits its use in current clinical practice for the patients with a PDAC, whose it must be remembered that half die within 6 months of diagnosis. To overcome this obstacle, we assessed the ability of transcriptomic molecular signatures to identify patients with a particular sensitivity profile to a given treatment. The approaches based on transcriptomic profiling have the enormous advantage of using very little biological material and thus significantly reducing the time to arrive at the selection of more effective drugs to each patient.
Collapse
Affiliation(s)
- Nelson Dusetti
- Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm U1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix Marseille Université, Marseille, France
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm U1068, CNRS UMR 7258, Institut Paoli-Calmettes, Aix Marseille Université, Marseille, France
| |
Collapse
|
35
|
Swayden M, Soubeyran P, Iovanna J. Upcoming Revolutionary Paths in Preclinical Modeling of Pancreatic Adenocarcinoma. Front Oncol 2020; 9:1443. [PMID: 32038993 PMCID: PMC6987422 DOI: 10.3389/fonc.2019.01443] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022] Open
Abstract
To date, PDAC remains the cancer having the worst prognosis with mortality rates constantly on the rise. Efficient cures are still absent, despite all attempts to understand the aggressive physiopathology underlying this disease. A major stumbling block is the outdated preclinical modeling strategies applied in assessing effectiveness of novel anticancer therapeutics. Current in vitro preclinical models have a low fidelity to mimic the exact architectural and functional complexity of PDAC tumor found in human set, due to the lack of major components such as immune system and tumor microenvironment with its associated chemical and mechanical signals. The existing PDAC preclinical platforms are still far from being reliable and trustworthy to guarantee the success of a drug in clinical trials. Therefore, there is an urgent demand to innovate novel in vitro preclinical models that mirrors with precision tumor-microenvironment interface, pressure of immune system, and molecular and morphological aspects of the PDAC normally experienced within the living organ. This review outlines the traditional preclinical models of PDAC namely 2D cell lines, genetically engineered mice, and xenografts, and describing the present famous approach of 3D organoids. We offer a detailed narration of the pros and cons of each model system. Finally, we suggest the incorporation of two off-center newly born techniques named 3D bio-printing and organs-on-chip and discuss the potentials of swine models and in silico tools, as powerful new tools able to transform PDAC preclinical modeling to a whole new level and open new gates in personalized medicine.
Collapse
Affiliation(s)
- Mirna Swayden
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Philippe Soubeyran
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| |
Collapse
|
36
|
Urrutia G, Salmonson A, Toro-Zapata J, de Assuncao TM, Mathison A, Dusetti N, Iovanna J, Urrutia R, Lomberk G. Combined Targeting of G9a and Checkpoint Kinase 1 Synergistically Inhibits Pancreatic Cancer Cell Growth by Replication Fork Collapse. Mol Cancer Res 2019; 18:448-462. [PMID: 31822519 DOI: 10.1158/1541-7786.mcr-19-0490] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 10/31/2019] [Accepted: 12/06/2019] [Indexed: 12/11/2022]
Abstract
Because of its dismal outcome, pancreatic ductal adenocarcinoma (PDAC) remains a therapeutic challenge making the testing of new pharmacologic tools a goal of paramount importance. Here, we developed a rational approach for inhibiting PDAC growth based on leveraging cell-cycle arrest of malignant cells at a phase that shows increased sensitivity to distinct epigenomic inhibitors. Specifically, we simultaneously inhibited checkpoint kinase 1 (Chk1) by prexasertib and the G9a histone methyltransferase with BRD4770, thereby targeting two key pathways for replication fork stability. Methodologically, the antitumor effects and molecular mechanisms of the combination were assessed by an extensive battery of assays, utilizing cell lines and patient-derived cells as well as 3D spheroids and xenografts. We find that the prexasertib-BRD4770 combination displays a synergistic effect on replication-associated phenomena, including cell growth, DNA synthesis, cell-cycle progression at S phase, and DNA damage signaling, ultimately leading to a highly efficient induction of cell death. Moreover, cellular and molecular data reveal that the synergistic effect of these pathways can be explained, at least in large part, by the convergence of both Chk1 and G9a functions at the level of the ATR-RPA-checkpoint pathway, which is operational during replication stress. Thus, targeting the epigenetic regulator G9a, which is necessary for replication fork stability, combined with inhibition of the DNA damage checkpoint, offers a novel approach for controlling PDAC growth through replication catastrophe. IMPLICATIONS: This study offers an improved, context-dependent, paradigm for the use of epigenomic inhibitors and provides mechanistic insight into their potential therapeutic use against PDAC.
Collapse
Affiliation(s)
- Guillermo Urrutia
- Division of Research, Department of Surgery; Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ann Salmonson
- Division of Research, Department of Surgery; Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jorge Toro-Zapata
- Division of Research, Department of Surgery; Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Thiago M de Assuncao
- Division of Research, Department of Surgery; Medical College of Wisconsin, Milwaukee, Wisconsin.,Genomic Sciences and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Angela Mathison
- Division of Research, Department of Surgery; Medical College of Wisconsin, Milwaukee, Wisconsin.,Genomic Sciences and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Nelson Dusetti
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Raul Urrutia
- Division of Research, Department of Surgery; Medical College of Wisconsin, Milwaukee, Wisconsin.,Genomic Sciences and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Gwen Lomberk
- Division of Research, Department of Surgery; Medical College of Wisconsin, Milwaukee, Wisconsin. .,Genomic Sciences and Precision Medicine Center (GSPMC), Medical College of Wisconsin, Milwaukee, Wisconsin.,Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
37
|
Wang CF, Shi XJ. Generation and application of patient-derived xenograft models in pancreatic cancer research. Chin Med J (Engl) 2019; 132:2729-2736. [PMID: 31725451 PMCID: PMC6940092 DOI: 10.1097/cm9.0000000000000524] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma cancer (PDAC) is one of the leading causes of cancer-related death worldwide. Hence, the development of effective anti-PDAC therapies is urgently required. Patient-derived xenograft (PDX) models are useful models for developing anti-cancer therapies and screening drugs for precision medicine. This review aimed to provide an updated summary of using PDX models in PDAC. DATA SOURCES The author retrieved information from the PubMed database up to June 2019 using various combinations of search terms, including PDAC, pancreatic carcinoma, pancreatic cancer, patient-derived xenografts or PDX, and patient-derived tumor xenografts or PDTX. STUDY SELECTION Original articles and review articles relevant to the review's theme were selected. RESULTS PDX models are better than cell line-derived xenograft and other models. PDX models consistently demonstrate retained tumor morphology and genetic stability, are beneficial in cancer research, could enhance drug discovery and oncologic mechanism development of PDAC, allow an improved understanding of human cancer cell biology, and help guide personalized treatment. CONCLUSIONS In this review, we outline the status and application of PDX models in both basic and pre-clinical pancreatic cancer researches. PDX model is one of the most appropriate pre-clinical tools that can improve the prognosis of patients with pancreatic cancer in the future.
Collapse
Affiliation(s)
- Cheng-Fang Wang
- Department of Hepato-Biliary Surgery, The General Hospital of People's Liberation Army (301 hospital), Beijing 100853, China
| | | |
Collapse
|
38
|
Swayden M, Alzeeb G, Masoud R, Berthois Y, Audebert S, Camoin L, Hannouche L, Vachon H, Gayet O, Bigonnet M, Roques J, Silvy F, Carrier A, Dusetti N, Iovanna JL, Soubeyran P. PML hyposumoylation is responsible for the resistance of pancreatic cancer. FASEB J 2019; 33:12447-12463. [PMID: 31557059 DOI: 10.1096/fj.201901091r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The dismal prognosis of pancreatic ductal adenocarcinoma (PDAC) is mainly due to its rapidly acquired resistance to all conventional treatments. Despite drug-specific mechanisms of resistance, none explains how these cells resist the stress induced by any kind of anticancer treatment. Activation of stress-response pathways relies on the post-translational modifications (PTMs) of involved proteins. Among all PTMs, those mediated by the ubiquitin family of proteins play a central role. Our aim was to identify alterations of ubiquitination, neddylation, and sumoylation associated with the multiresistant phenotype and demonstrate their implications in the survival of PDAC cells undergoing treatment. This approach pointed at an alteration of promyelocytic leukemia (PML) protein sumoylation associated with both gemcitabine and oxaliplatin resistance. We could show that this alteration of PML sumoylation is part of a general mechanism of drug resistance, which in addition involves the abnormal activation of NF-κB and cAMP response element binding pathways. Importantly, using patient-derived tumors and cell lines, we identified a correlation between the levels of PML expression and sumoylation and the sensitivity of tumors to anticancer treatments.-Swayden, M., Alzeeb, G., Masoud, R., Berthois, Y., Audebert, S., Camoin, L., Hannouche, L., Vachon, H., Gayet, O., Bigonnet, M., Roques, J., Silvy, F., Carrier, A., Dusetti, N., Iovanna, J. L., Soubeyran, P. PML hyposumoylation is responsible for the resistance of pancreatic cancer.
Collapse
Affiliation(s)
- Mirna Swayden
- Centre de Recherche en Cancérologie de Marseille (CRCM), Centre de Recherche en Cancérologie de Marseille (CRCM), Centre National de la Recherche Scientifique (CNRS), Institut Paoli-Calmettes, INSERM, Aix-Marseille University, Marseille, France
| | - George Alzeeb
- Centre de Recherche en Cancérologie de Marseille (CRCM), Centre de Recherche en Cancérologie de Marseille (CRCM), Centre National de la Recherche Scientifique (CNRS), Institut Paoli-Calmettes, INSERM, Aix-Marseille University, Marseille, France
| | - Rawand Masoud
- Centre de Recherche en Cancérologie de Marseille (CRCM), Centre de Recherche en Cancérologie de Marseille (CRCM), Centre National de la Recherche Scientifique (CNRS), Institut Paoli-Calmettes, INSERM, Aix-Marseille University, Marseille, France
| | - Yolande Berthois
- Centre de Recherche en Cancérologie de Marseille (CRCM), Centre de Recherche en Cancérologie de Marseille (CRCM), Centre National de la Recherche Scientifique (CNRS), Institut Paoli-Calmettes, INSERM, Aix-Marseille University, Marseille, France
| | - Stéphane Audebert
- Centre de Recherche en Cancérologie de Marseille (CRCM), Centre de Recherche en Cancérologie de Marseille (CRCM), Centre National de la Recherche Scientifique (CNRS), Institut Paoli-Calmettes, INSERM, Aix-Marseille University, Marseille, France
| | - Luc Camoin
- Centre de Recherche en Cancérologie de Marseille (CRCM), Centre de Recherche en Cancérologie de Marseille (CRCM), Centre National de la Recherche Scientifique (CNRS), Institut Paoli-Calmettes, INSERM, Aix-Marseille University, Marseille, France
| | - Laurent Hannouche
- Transcriptomique and Génomique Marseille Luminy (TGML), Théories et Approches de la Complexité Génomique (TAGC), INSERM, Aix-Marseille University, Marseille, France
| | - Hortense Vachon
- Transcriptomique and Génomique Marseille Luminy (TGML), Théories et Approches de la Complexité Génomique (TAGC), INSERM, Aix-Marseille University, Marseille, France
| | - Odile Gayet
- Centre de Recherche en Cancérologie de Marseille (CRCM), Centre de Recherche en Cancérologie de Marseille (CRCM), Centre National de la Recherche Scientifique (CNRS), Institut Paoli-Calmettes, INSERM, Aix-Marseille University, Marseille, France
| | - Martin Bigonnet
- Centre de Recherche en Cancérologie de Marseille (CRCM), Centre de Recherche en Cancérologie de Marseille (CRCM), Centre National de la Recherche Scientifique (CNRS), Institut Paoli-Calmettes, INSERM, Aix-Marseille University, Marseille, France
| | - Julie Roques
- Centre de Recherche en Cancérologie de Marseille (CRCM), Centre de Recherche en Cancérologie de Marseille (CRCM), Centre National de la Recherche Scientifique (CNRS), Institut Paoli-Calmettes, INSERM, Aix-Marseille University, Marseille, France
| | - Françoise Silvy
- Centre de Recherche en Cancérologie de Marseille (CRCM), Centre de Recherche en Cancérologie de Marseille (CRCM), Centre National de la Recherche Scientifique (CNRS), Institut Paoli-Calmettes, INSERM, Aix-Marseille University, Marseille, France
| | - Alice Carrier
- Centre de Recherche en Cancérologie de Marseille (CRCM), Centre de Recherche en Cancérologie de Marseille (CRCM), Centre National de la Recherche Scientifique (CNRS), Institut Paoli-Calmettes, INSERM, Aix-Marseille University, Marseille, France
| | - Nelson Dusetti
- Centre de Recherche en Cancérologie de Marseille (CRCM), Centre de Recherche en Cancérologie de Marseille (CRCM), Centre National de la Recherche Scientifique (CNRS), Institut Paoli-Calmettes, INSERM, Aix-Marseille University, Marseille, France
| | - Juan L Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), Centre de Recherche en Cancérologie de Marseille (CRCM), Centre National de la Recherche Scientifique (CNRS), Institut Paoli-Calmettes, INSERM, Aix-Marseille University, Marseille, France
| | - Philippe Soubeyran
- Centre de Recherche en Cancérologie de Marseille (CRCM), Centre de Recherche en Cancérologie de Marseille (CRCM), Centre National de la Recherche Scientifique (CNRS), Institut Paoli-Calmettes, INSERM, Aix-Marseille University, Marseille, France
| |
Collapse
|
39
|
Emerging epigenomic landscapes of pancreatic cancer in the era of precision medicine. Nat Commun 2019; 10:3875. [PMID: 31462645 PMCID: PMC6713756 DOI: 10.1038/s41467-019-11812-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 08/06/2019] [Indexed: 12/11/2022] Open
Abstract
Genetic studies have advanced our understanding of pancreatic cancer at a mechanistic and translational level. Genetic concepts and tools are increasingly starting to be applied to clinical practice, in particular for precision medicine efforts. However, epigenomics is rapidly emerging as a promising conceptual and methodological paradigm for advancing the knowledge of this disease. More importantly, recent studies have uncovered potentially actionable pathways, which support the prediction that future trials for pancreatic cancer will involve the vigorous testing of epigenomic therapeutics. Thus, epigenomics promises to generate a significant amount of new knowledge of both biological and medical importance. In pancreatic cancer, the epigenomic landscape can strongly impact the disease phenotype. Here, the authors discuss recent advances in our understanding of pancreatic cancer epigenomics, and how this knowledge can integrate with precision medicine approaches in this lethal disease.
Collapse
|
40
|
Bian B, Juiz NA, Gayet O, Bigonnet M, Brandone N, Roques J, Cros J, Wang N, Dusetti N, Iovanna J. Pancreatic Cancer Organoids for Determining Sensitivity to Bromodomain and Extra-Terminal Inhibitors (BETi). Front Oncol 2019; 9:475. [PMID: 31231611 PMCID: PMC6560163 DOI: 10.3389/fonc.2019.00475] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/17/2019] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a heterogeneous disease, therefore stratification of patients is essential to predict their responses to therapies and to choose the best treatment. PDAC-derived organoids were produced from PDTX and Endoscopic Ultrasound-Guided Fine-Needle Aspiration (EUS-FNA) biopsies. A signature based on 16 genes targets of the c-MYC oncogene was applied to classify samples into two sub-groups with distinctive phenotypes named MYC-high and MYC-low. The analysis of 9 PDTXs and the corresponding derived organoids revealed that this signature which was previously designed from PDTX is transferable to the organoid model. Primary organoids from 24 PDAC patients were treated with NHWD-870 or JQ1, two inhibitors of c-MYC transcription. Notably, the comparison of their effect between the two sub-groups showed that both compounds are more efficient in MYC-high than in MYC-low samples, being NHWD-870 the more potent treatment. In conclusion, this study shows that the molecular signatures could be applied to organoids obtained directly from PDAC patients to predict the treatment response and could help to take the more appropriate therapeutic decision for each patient in a clinical timeframe.
Collapse
Affiliation(s)
- Benjamin Bian
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Natalia Anahi Juiz
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Odile Gayet
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Martin Bigonnet
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Nicolas Brandone
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Julie Roques
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Jérôme Cros
- Pathology Department, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, UMR 1149, Inflammation Research Center, INSERM - Paris Diderot University, Paris, France
| | - Nenghui Wang
- Ningbo Wenda Pharma Technology Ltd., Zhejiang, China
| | - Nelson Dusetti
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW Pharmacological inhibition of Bromodomain and Extra-Terminal (BET) domain proteins is a very exciting epigenetic therapeutic modality. Due to the central role of BET proteins in transcription regulation, their inhibition heavily affects lymphoma cells and BET inhibitors show a clear preclinical antitumor activity as single agents and in combination, paired with early reports of clinical activity. RECENT FINDINGS Relevant data have been recently presented on the mechanism of action of the BET inhibitors, on modalities to improve their activity in lymphomas, and their clinical evaluation. SUMMARY There are now plenty of preclinical data sustaining BET proteins as therapeutic targets in lymphomas. Newer compounds and combinations with other agents may be pursued in the future aiming also to identify those patients that they most likely benefit from BET inhibition.
Collapse
|
42
|
Juiz NA, Iovanna J, Dusetti N. Pancreatic Cancer Heterogeneity Can Be Explained Beyond the Genome. Front Oncol 2019; 9:246. [PMID: 31024848 PMCID: PMC6460948 DOI: 10.3389/fonc.2019.00246] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 03/18/2019] [Indexed: 12/23/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a major health problem because it induces almost systematic mortality. Carcinogenesis begins with genetic aberrations which trigger epigenetic modifications. While genetic mutations initiate tumorigenesis, they are unable to explain the vast heterogeneity observed among PDAC patients. Instead, epigenetic changes drive transcriptomic alterations that can regulate the malignant phenotype. The contribution of factors from the environment and tumor microenvironment defines different epigenetic landscapes that outline two clinical subtypes: basal, with the worst prognosis, and classical. The epigenetic nature of PDAC, as a reversible phenomenon, encouraged several studies to test epidrugs. However, these drugs lack specificity and although there are epigenetic patterns shared by all PDAC tumors, there are others that are specific to each subtype. Molecular characterization of the epigenetic mechanisms underlying PDAC heterogeneity could be an invaluable tool to predict personalized therapies, stratify patients and search for novel therapies with more specific phenotype-based targets. Novel therapeutic strategies using current anticancer compounds or existing drugs used in other pathologies, alone or in combination, could be used to kill tumor cells or convert aggressive tumors into a more benign phenotype.
Collapse
Affiliation(s)
- Natalia Anahi Juiz
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Aix-Marseille Université, Marseille, France
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Aix-Marseille Université, Marseille, France
| | - Nelson Dusetti
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Aix-Marseille Université, Marseille, France
| |
Collapse
|
43
|
Bulle A, Dekervel J, Libbrecht L, Nittner D, Deschuttere L, Lambrecht D, Van Cutsem E, Verslype C, van Pelt J. Gemcitabine induces Epithelial-to-Mesenchymal Transition in patient-derived pancreatic ductal adenocarcinoma xenografts. Am J Transl Res 2019; 11:765-779. [PMID: 30899378 PMCID: PMC6413274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 12/23/2018] [Indexed: 06/09/2023]
Abstract
There is a lack of well-characterized models for pancreatic ductal adenocarcinoma (PDAC). PDAC itself is unique because of its pronounced tumor microenvironment that influences tumor progression, behavior and therapeutic resistance. Here we investigated, in patient-derived tumor xenograft (PDTX) models developed from fine needle biopsies, the cancer cells behavior, Epithelial-to-Mesenchymal Transition (EMT) and drug response. For this, we studied two behaviorally distinct PDTX models. Tumor volume measurement, histology, immuno-histochemical staining, RT-qPCR, RNA sequencing and Western blotting were used to further characterize these models and investigate the effect of two classes of drugs (gemcitabine and acriflavine (HIF-inhibitor)). The models recapitulated the corresponding primary tumors. The growth-rate of the poorly differentiated tumor (PAC010) was faster than that of the moderately differentiated tumor (PAC006) (P<0.05). The PAC010 model showed increased cell proliferation (Ki-67 staining) and markers indicating survival (increased p-AKT, p-ERK and p-NF-kB65 and suppression of cleaved PARP). Gene and protein analysis showed higher expression of mesenchymal markers in PAC010 model (e.g. VIM, SNAI2). Pathway analysis demonstrated activation of processes related to EMT, tumor progression and aggressiveness in PAC010. Gemcitabine treatment resulted in shrinking of the tumor volume and reduced proliferation in both models. Importantly, gemcitabine treatment significantly enhanced the expression of mesenchymal marker supportive of metastatic behavior and of survival pathways, particularly in the non-aggressive PAC006 model. Acriflavine had little effect on tumor growth in both models. In conclusion, we observed in this unique model of PDAC, a clear link between EMT and poor tumor differentiation and found that gemcitabine can increase EMT.
Collapse
Affiliation(s)
- Ashenafi Bulle
- Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven & University Hospitals Leuven and Leuven Cancer Institute (LKI)Leuven, Belgium
| | - Jeroen Dekervel
- Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven & University Hospitals Leuven and Leuven Cancer Institute (LKI)Leuven, Belgium
| | - Louis Libbrecht
- Department of Pathology, University Hospital Saint-LucBrussels, Belgium
| | - David Nittner
- Laboratory of Translational Genetics, Department of Oncology, KU Leuven and Vesalius Research Center for Cancer Biology, VIBLeuven, Belgium
| | - Lise Deschuttere
- Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven & University Hospitals Leuven and Leuven Cancer Institute (LKI)Leuven, Belgium
| | - Diether Lambrecht
- Laboratory of Translational Genetics, Department of Oncology, KU Leuven and Vesalius Research Center for Cancer Biology, VIBLeuven, Belgium
| | - Eric Van Cutsem
- Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven & University Hospitals Leuven and Leuven Cancer Institute (LKI)Leuven, Belgium
| | - Chris Verslype
- Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven & University Hospitals Leuven and Leuven Cancer Institute (LKI)Leuven, Belgium
| | - Jos van Pelt
- Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven & University Hospitals Leuven and Leuven Cancer Institute (LKI)Leuven, Belgium
| |
Collapse
|
44
|
Tseng CH, Huang WT, Chew CH, Lai JK, Tu SH, Wei PL, Lee KY, Lai GM, Chen CC. Electrospun Polylactic Acid (PLLA) Microtube Array Membrane (MTAM)-An Advanced Substrate for Anticancer Drug Screening. MATERIALS 2019; 12:ma12040569. [PMID: 30769818 PMCID: PMC6416630 DOI: 10.3390/ma12040569] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 12/14/2022]
Abstract
The advent of personalized cancer treatment resulted in the shift from the administration of cytotoxic drugs with broad activity spectrum to a targeted tumor-specific therapy. Aligned to this development, the focus of this study revolved around the application of our novel and patented microtube array membrane (MTAM) in the US National Cancer Institute (NCI) developed an HFA (hollow fiber assay) assay; hereinafter known as MTAM/HFA. Electrospun poly-L-lactic acid (PLLA) MTAM was sterilized and loaded with cell lines/patient derived tumor cells (PDTC) and subcutaneously implanted into the backs of BALB/C mice. Anticancer drugs were administered at the respective time points and the respective MTAMs were retrieved and the viability tumor cells within were quantified with the MTT assay. Results revealed that the MTAMs were excellent culture substrate for various cancer cell lines and PDTCs (patient derived tumor cells). Compared to traditional HFA systems that utilize traditional hollow fibers, MTAM/HFA revealed superior drug sensitivity for a wide range of anticancer drug classes. Additionally, the duration for each test was <14 days; all this while capable of producing similar trend outcome to the current gold-standard xenograft models. These benefits were observed in both the in vitro and in vivo stages, making it a highly practical phenotypic-based solution that could potentially be applied in personalized medicine.
Collapse
Affiliation(s)
- Chia-Hsuan Tseng
- Graduate Institute of Biomedical Materials & Tissue Engineering, Taipei Medical University, Xinyi District, Taipei 11031, Taiwan.
| | - Wan-Ting Huang
- Graduate Institute of Biomedical Materials & Tissue Engineering, Taipei Medical University, Xinyi District, Taipei 11031, Taiwan.
- MTAMTech corporation, 17th floor, 3rd Yuanqu Street, Nangang District, Taipei 11503, Taiwan.
| | - Chee Ho Chew
- Graduate Institute of Biomedical Materials & Tissue Engineering, Taipei Medical University, Xinyi District, Taipei 11031, Taiwan.
| | - Jun-Kai Lai
- MTAMTech corporation, 17th floor, 3rd Yuanqu Street, Nangang District, Taipei 11503, Taiwan.
| | - Shih-Hsin Tu
- Department of Surgery, Taipei Medical University Hospital, Xinyi District, Taipei 11031, Taiwan.
| | - Po-Li Wei
- Department of Surgery, Taipei Medical University Hospital, Xinyi District, Taipei 11031, Taiwan.
| | - Kang-Yun Lee
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei 235, Taiwan.
- Division of Thoracic Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 250, Taiwan.
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 250, Taiwan.
| | - Gi-Ming Lai
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 250, Taiwan.
| | - Chien-Chung Chen
- Graduate Institute of Biomedical Materials & Tissue Engineering, Taipei Medical University, Xinyi District, Taipei 11031, Taiwan.
- MTAMTech corporation, 17th floor, 3rd Yuanqu Street, Nangang District, Taipei 11503, Taiwan.
- Ph.D Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei 250, Taiwan.
| |
Collapse
|
45
|
Swayden M, Iovanna J, Soubeyran P. Pancreatic cancer chemo-resistance is driven by tumor phenotype rather than tumor genotype. Heliyon 2018; 4:e01055. [PMID: 30582059 PMCID: PMC6299038 DOI: 10.1016/j.heliyon.2018.e01055] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 09/28/2018] [Accepted: 12/12/2018] [Indexed: 12/12/2022] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is one of the deadliest forms of cancer. A major reason for this situation is the fact that these tumors are already resistant or become rapidly resistant to all conventional therapies. Like any transformation process, initiation and development of PDCA are driven by a well known panel of genetic alterations, few of them are shared with most cancers, but many mutations are specific to PDAC and are partially responsible for the great inter-tumor heterogeneity. Importantly, this knowledge has been inefficient in predicting response to anticancer therapy, or in establishing diagnosis and prognosis. Hence, the pre-existing or rapidly acquired resistance of pancreatic cancer cells to therapeutic drugs rely on other parameters and features developed by the cells and/or the micro-environment, that are independent of their genetic profiles. This review sheds light on all major phenotypic, non genetic, alterations known to play important roles in PDAC cells resistance to treatments and therapeutic escape.
Collapse
Affiliation(s)
| | | | - Philippe Soubeyran
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| |
Collapse
|
46
|
Wang B, Fan P, Zhao J, Wu H, Jin X, Wu H. FBP1 loss contributes to BET inhibitors resistance by undermining c-Myc expression in pancreatic ductal adenocarcinoma. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:224. [PMID: 30201002 PMCID: PMC6131902 DOI: 10.1186/s13046-018-0888-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 08/21/2018] [Indexed: 01/05/2023]
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal tumor types worldwide. BET inhibitors display anti-tumor activity in pancreatic cancer, however the cells often develop resistance after a long-term treatment and the underlying molecular basis is not fully understood. Methods Drug screening assay in Fructose-1, 6-biphosphatase (FBP1) knockdown or overexpressing pancreatic cancer cells was performed. Tumor cell motility, FBP1 protein and mRNA changes were investigated after BET inhibitors treatment. The interaction between TRIM28 and FBP1 after BET inhibitors treatment was examined by Co-immunoprecipitation (IP) and GST pull-down. The relationship between FBP1 and c-Myc was examined by western blot, RT-qPCR and immunohistochemistry (IHC). Results The expression of FBP1 protein increased the sensitivity of pancreatic cancer cells to JQ1. Furthermore, we showed that JQ1 stabilized FBP1 protein level by disrupting the interaction between FBP1 and TRIM28 in pancreatic cancer cells. Moreover, we demonstrated that FBP1 promoted c-Myc degradation through disrupting the ERK-c-Myc axis. Conclusions FBP1 modulates the sensitivity of pancreatic cancer cells to BET inhibitors by decreasing the expression of c-Myc. These findings highlight FBP1 could be used as a therapeutic niche for patient-tailored therapies. Electronic supplementary material The online version of this article (10.1186/s13046-018-0888-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bo Wang
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ping Fan
- Department of Digestive Surgical Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jingyuan Zhao
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Heyu Wu
- Operating Room, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Xin Jin
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China. .,Department of Digestive Surgical Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Heshui Wu
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
47
|
Recent Advances in Chromatin Mechanisms Controlling Pancreatic Carcinogenesis. EPIGENOMES 2018. [DOI: 10.3390/epigenomes2020011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
48
|
|
49
|
Lan W, Bian B, Xia Y, Dou S, Gayet O, Bigonnet M, Santofimia-Castaño P, Cong M, Peng L, Dusetti N, Iovanna J. E2F signature is predictive for the pancreatic adenocarcinoma clinical outcome and sensitivity to E2F inhibitors, but not for the response to cytotoxic-based treatments. Sci Rep 2018; 8:8330. [PMID: 29844366 PMCID: PMC5974374 DOI: 10.1038/s41598-018-26613-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 05/14/2018] [Indexed: 12/26/2022] Open
Abstract
The main goal of this study was to find out strategies of clinical relevance to classify patients with a pancreatic ductal adenocarcinoma (PDAC) for individualized treatments. In the present study a set of 55 patient-derived xenografts (PDX) were obtained and their transcriptome were analyzed by using an Affymetrix approach. A supervised bioinformatics-based analysis let us to classify these PDX in two main groups named E2F-highly dependent and E2F-lowly dependent. Afterwards their characterization by using a Kaplan-Meier analysis demonstrated that E2F high patients survived significantly less than E2F low patients (9.5 months vs. 16.8 months; p = 0.0066). Then we tried to establish if E2F transcriptional target levels were associated to the response to cytotoxic treatments by comparing the IC50 values of E2F high and E2F low cells after gemcitabine, 5-fluorouracil, oxaliplatin, docetaxel or irinotecan treatment, and no association was found. Then we identified an E2F inhibitor compound, named ly101-4B, and we observed that E2F-higly dependent cells were more sensitive to its treatment (IC50 of 19.4 ± 1.8 µM vs. 44.1 ± 4.4 µM; p = 0.0061). In conclusion, in this work we describe an E2F target expression-based classification that could be predictive for patient outcome, but more important, for the sensitivity of tumors to the E2F inhibitors as a treatment. Finally, we can assume that phenotypic characterization, essentially by an RNA expression analysis of the PDAC, can help to predict their clinical outcome and their response to some treatments when are rationally selected.
Collapse
Affiliation(s)
- Wenjun Lan
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
- Aix-Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, «Equipe Labellisée Ligue Contre le Cancer», Marseille, France
| | - Benjamin Bian
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Yi Xia
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Samir Dou
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Odile Gayet
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Martin Bigonnet
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Patricia Santofimia-Castaño
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France
| | - Mei Cong
- Aix-Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, «Equipe Labellisée Ligue Contre le Cancer», Marseille, France
| | - Ling Peng
- Aix-Marseille Université, CNRS, Centre Interdisciplinaire de Nanoscience de Marseille, UMR 7325, «Equipe Labellisée Ligue Contre le Cancer», Marseille, France
| | - Nelson Dusetti
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France.
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM U1068, CNRS UMR 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, France.
| |
Collapse
|
50
|
Lai X, Stiff A, Duggan M, Wesolowski R, Carson WE, Friedman A. Modeling combination therapy for breast cancer with BET and immune checkpoint inhibitors. Proc Natl Acad Sci U S A 2018; 115:5534-5539. [PMID: 29735668 PMCID: PMC6003484 DOI: 10.1073/pnas.1721559115] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
CTLA-4 is an immune checkpoint expressed on active anticancer T cells. When it combines with its ligand B7 on dendritic cells, it inhibits the activity of the T cells. The Bromo- and Extra-Terminal (BET) protein family includes proteins that regulate the expression of key oncogenes and antiapoptotic proteins. BET inhibitor (BETi) has been shown to reduce the expression of MYC by suppressing its transcription factors and to down-regulate the hypoxic transcriptome response to VEGF-A. This paper develops a mathematical model of the treatment of cancer by combination therapy of BETi and CTLA-4 inhibitor. The model shows that the two drugs are positively correlated in the sense that the tumor volume decreases as the dose of each of the drugs is increased. The model also considers the effect of the combined therapy on levels of myeloid-derived suppressor cells (MDSCs) and the overexpression of TNF-α, which may predict gastrointestinal side effects of the combination.
Collapse
Affiliation(s)
- Xiulan Lai
- Institute for Mathematical Sciences, Renmin University of China, 100872 Beijing, P. R. China
| | - Andrew Stiff
- Medical Scientist Training Program, The Ohio State University, Columbus, OH 43210
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH 43210
| | - Megan Duggan
- Department of Surgery, The Ohio State University, Columbus, OH 43210
| | - Robert Wesolowski
- Stefanie Spielman Comprehensive Breast Center, Wexner Medical Center, The Ohio State University, Columbus, OH 43212
| | - William E Carson
- Division of Surgical Oncology, Department of Surgery, The Ohio State University, Columbus, OH 43210
- Division of Surgical Oncology, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210
| | - Avner Friedman
- Mathematical Bioscience Institute, The Ohio State University, Columbus, OH 43210;
- Department of Mathematics, The Ohio State University, Columbus, OH 43210
| |
Collapse
|