1
|
Maione F, Oddo D, Galvagno F, Falcomatà C, Pandini M, Macagno M, Pessei V, Barault L, Gigliotti C, Mira A, Corti G, Lamba S, Riganti C, Castella B, Massaia M, Rad R, Saur D, Bardelli A, Di Nicolantonio F. Preclinical efficacy of carfilzomib in BRAF-mutant colorectal cancer models. Mol Oncol 2024; 18:1552-1570. [PMID: 38348572 PMCID: PMC11161726 DOI: 10.1002/1878-0261.13595] [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: 07/27/2023] [Revised: 11/28/2023] [Accepted: 01/18/2024] [Indexed: 06/09/2024] Open
Abstract
Serine/threonine-protein kinase B-raf (BRAF) mutations are found in 8-15% of colorectal cancer patients and identify a subset of tumors with poor outcome in the metastatic setting. We have previously reported that BRAF-mutant human cells display a high rate of protein production, causing proteotoxic stress, and are selectively sensitive to the proteasome inhibitors bortezomib and carfilzomib. In this work, we tested whether carfilzomib could restrain the growth of BRAF-mutant colorectal tumors not only by targeting cancer cells directly, but also by promoting an immune-mediated antitumor response. In human and mouse colorectal cancer cells, carfilzomib triggered robust endoplasmic reticulum stress and autophagy, followed by the emission of immunogenic-damage-associated molecules. Intravenous administration of carfilzomib delayed the growth of BRAF-mutant murine tumors and mobilized the danger-signal proteins calreticulin and high mobility group box 1 (HMGB1). Analyses of drug-treated samples revealed increased intratumor recruitment of activated cytotoxic T cells and natural killers, concomitant with the downregulation of forkhead box protein P3 (Foxp3)+ T-cell surface glycoprotein CD4 (CD4)+ T cells, indicating that carfilzomib promotes reshaping of the immune microenvironment of BRAF-mutant murine colorectal tumors. These results will inform the design of clinical trials in BRAF-mutant colorectal cancer patients.
Collapse
Affiliation(s)
- Federica Maione
- Department of OncologyUniversity of TorinoTorinoItaly
- Candiolo Cancer InstituteFPO‐IRCCSCandioloItaly
| | - Daniele Oddo
- Department of OncologyUniversity of TorinoTorinoItaly
| | - Federica Galvagno
- Department of OncologyUniversity of TorinoTorinoItaly
- Candiolo Cancer InstituteFPO‐IRCCSCandioloItaly
| | - Chiara Falcomatà
- Institute of Molecular Oncology and Functional GenomicsSchool of Medicine, Technical University of MunichMunichGermany
- Center for Translational Cancer Research (TranslaTUM), School of MedicineTechnical University of MunichMunichGermany
| | - Marta Pandini
- Tumor Microenvironment UnitIstituto di Ricovero e Cura a Carattere Scientifico Humanitas Research HospitalMilanItaly
- Department of Biomedical SciencesHumanitas UniversityMilanItaly
| | | | | | | | | | - Alessia Mira
- Department of OncologyUniversity of TorinoTorinoItaly
| | | | - Simona Lamba
- Department of OncologyUniversity of TorinoTorinoItaly
- Candiolo Cancer InstituteFPO‐IRCCSCandioloItaly
| | | | - Barbara Castella
- Laboratory of Blood Tumor Immunology (LBTI), Molecular Biotechnology Center “Guido Tarone” (MBC)University of TurinTurinItaly
| | - Massimo Massaia
- Laboratory of Blood Tumor Immunology (LBTI), Molecular Biotechnology Center “Guido Tarone” (MBC)University of TurinTurinItaly
- SC EmatologiaAzienda Ospedaliera S. Croce e CarleCuneoItaly
| | - Roland Rad
- Institute of Molecular Oncology and Functional GenomicsSchool of Medicine, Technical University of MunichMunichGermany
- Tumor Microenvironment UnitIstituto di Ricovero e Cura a Carattere Scientifico Humanitas Research HospitalMilanItaly
- German Cancer ConsortiumHeidelbergGermany
| | - Dieter Saur
- Institute of Molecular Oncology and Functional GenomicsSchool of Medicine, Technical University of MunichMunichGermany
- Tumor Microenvironment UnitIstituto di Ricovero e Cura a Carattere Scientifico Humanitas Research HospitalMilanItaly
- German Cancer ConsortiumHeidelbergGermany
- Department of Internal Medicine II, Klinikum rechts der IsarTechnische Universität MünchenMunichGermany
| | - Alberto Bardelli
- Department of OncologyUniversity of TorinoTorinoItaly
- IFOM ETSThe AIRC Institute of Molecular OncologyMilanItaly
| | - Federica Di Nicolantonio
- Department of OncologyUniversity of TorinoTorinoItaly
- Candiolo Cancer InstituteFPO‐IRCCSCandioloItaly
| |
Collapse
|
2
|
Saeed H, Leibowitz BJ, Zhang L, Yu J. Targeting Myc-driven stress addiction in colorectal cancer. Drug Resist Updat 2023; 69:100963. [PMID: 37119690 DOI: 10.1016/j.drup.2023.100963] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/06/2023] [Accepted: 04/17/2023] [Indexed: 05/01/2023]
Abstract
MYC is a proto-oncogene that encodes a powerful regulator of transcription and cellular programs essential for normal development, as well as the growth and survival of various types of cancer cells. MYC rearrangement and amplification is a common cause of hematologic malignancies. In epithelial cancers such as colorectal cancer, genetic alterations in MYC are rare. Activation of Wnt, ERK/MAPK, and PI3K/mTOR pathways dramatically increases Myc levels through enhanced transcription, translation, and protein stability. Elevated Myc promotes stress adaptation, metabolic reprogramming, and immune evasion to drive cancer development and therapeutic resistance through broad changes in transcriptional and translational landscapes. Despite intense interest and effort, Myc remains a difficult drug target. Deregulation of Myc and its targets has profound effects that vary depending on the type of cancer and the context. Here, we summarize recent advances in the mechanistic understanding of Myc-driven oncogenesis centered around mRNA translation and proteostress. Promising strategies and agents under development to target Myc are also discussed with a focus on colorectal cancer.
Collapse
Affiliation(s)
- Haris Saeed
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA 15213, USA; Dept. of Pathology, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA 15213, USA
| | - Brian J Leibowitz
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA 15213, USA; Dept. of Pathology, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA 15213, USA
| | - Lin Zhang
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA 15213, USA; Dept. of Chemical Biology and Pharmacology, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA 15213, USA
| | - Jian Yu
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA 15213, USA; Dept. of Pathology, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA 15213, USA; Dept. of Radiation Oncology, University of Pittsburgh School of Medicine, 5117 Centre Ave., Pittsburgh, PA 15213, USA.
| |
Collapse
|
3
|
Tong J, Tan X, Risnik D, Gao M, Song X, Ermine K, Shen L, Wang S, Yu J, Zhang L. BET protein degradation triggers DR5-mediated immunogenic cell death to suppress colorectal cancer and potentiate immune checkpoint blockade. Oncogene 2021; 40:6566-6578. [PMID: 34615996 PMCID: PMC8642302 DOI: 10.1038/s41388-021-02041-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/16/2021] [Accepted: 09/27/2021] [Indexed: 12/23/2022]
Abstract
Bromodomain and extra-terminal domain (BET) family proteins are epigenetic readers that play a critical role in oncogenesis by controlling the expression of oncogenes such as c-Myc. Targeting BET family proteins has recently emerged as a promising anticancer strategy. However, the molecular mechanisms by which cancer cells respond to BET inhibition are not well understood. In this study, we found that inducing the degradation of BET proteins by the proteolysis targeting chimeras (PROTAC) approach potently suppressed the growth of colorectal cancer (CRC) including patient-derived tumors. Mechanistically, BET degradation transcriptionally activates Death Receptor 5 (DR5) to trigger immunogenic cell death (ICD) in CRC cells. Enhanced DR5 induction further sensitizes CRC cells with a mutation in Speckle-type POZ protein (SPOP). Furthermore, DR5 is indispensable for a striking antitumor effect of combining BET degradation and anti-PD-1 antibody, which was well tolerated in mice and almost eradicated syngeneic tumors. Our results demonstrate that BET degradation triggers DR5-mediated ICD to potently suppress CRC and potentiate immune checkpoint blockade. These results provide a rationale, mechanistic insights, and potential biomarkers for developing a precision CRC therapy by inducing BET protein degradation.
Collapse
Affiliation(s)
- Jingshan Tong
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Xiao Tan
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, P.R. China
| | - Denise Risnik
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Man Gao
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Xiangping Song
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, P.R. China
| | - Kaylee Ermine
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, P.R. China
| | - Shaomeng Wang
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI, 48109, USA
| | - Jian Yu
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Lin Zhang
- UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
4
|
Sieber-Frank J, Stark HJ, Kalteis S, Prigge ES, Köhler R, Andresen C, Henkel T, Casari G, Schubert T, Fischl W, Li-Weber M, Krammer PH, von Knebel Doeberitz M, Kopitz J, Kloor M, Ahadova A. Treatment resistance analysis reveals GLUT-1-mediated glucose uptake as a major target of synthetic rocaglates in cancer cells. Cancer Med 2021; 10:6807-6822. [PMID: 34546000 PMCID: PMC8495295 DOI: 10.1002/cam4.4212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/15/2021] [Accepted: 07/21/2021] [Indexed: 12/19/2022] Open
Abstract
Rocaglates are natural compounds that have been extensively studied for their ability to inhibit translation initiation. Rocaglates represent promising drug candidates for tumor treatment due to their growth‐inhibitory effects on neoplastic cells. In contrast to natural rocaglates, synthetic analogues of rocaglates have been less comprehensively characterized, but were also shown to have similar effects on the process of protein translation. Here, we demonstrate an enhanced growth‐inhibitory effect of synthetic rocaglates when combined with glucose anti‐metabolite 2‐deoxy‐D‐glucose (2DG) in different cancer cell lines. Moreover, we unravel a new aspect in the mechanism of action of synthetic rocaglates involving reduction of glucose uptake mediated by downregulation or abrogation of glucose transporter GLUT‐1 expression. Importantly, cells with genetically induced resistance to synthetic rocaglates showed substantially less pronounced treatment effect on glucose metabolism and did not demonstrate GLUT‐1 downregulation, pointing at the crucial role of this mechanism for the anti‐tumor activity of the synthetic rocaglates. Transcriptome profiling revealed glycolysis as one of the major pathways differentially regulated in sensitive and resistant cells. Analysis of synthetic rocaglate efficacy in a 3D tissue context with a co‐culture of tumor and normal cells demonstrated a selective effect on tumor cells and substantiated the mechanistic observations obtained in cancer cell lines. Increased glucose uptake and metabolism is a universal feature across different tumor types. Therefore, targeting this feature by synthetic rocaglates could represent a promising direction for exploitation of rocaglates in novel anti‐tumor therapies.
Collapse
Affiliation(s)
- Julia Sieber-Frank
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hans-Jürgen Stark
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Simon Kalteis
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elena-Sophie Prigge
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Richard Köhler
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carolin Andresen
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | | | | | - Min Li-Weber
- Tumor Immunology Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter H Krammer
- Tumor Immunology Program, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Magnus von Knebel Doeberitz
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen Kopitz
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Kloor
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Aysel Ahadova
- Department of Applied Tumor Biology, Institute of Pathology, University of Heidelberg, Heidelberg, Germany.,Collaboration Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
5
|
Ruan H, Li X, Xu X, Leibowitz BJ, Tong J, Chen L, Ao L, Xing W, Luo J, Yu Y, Schoen RE, Sonenberg N, Lu X, Zhang L, Yu J. eIF4E S209 phosphorylation licenses myc- and stress-driven oncogenesis. eLife 2020; 9:60151. [PMID: 33135632 PMCID: PMC7665890 DOI: 10.7554/elife.60151] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/31/2020] [Indexed: 02/06/2023] Open
Abstract
To better understand a role of eIF4E S209 in oncogenic translation, we generated EIF4ES209A/+ heterozygous knockin (4EKI) HCT 116 human colorectal cancer (CRC) cells. 4EKI had little impact on total eIF4E levels, cap binding or global translation, but markedly reduced HCT 116 cell growth in spheroids and mice, and CRC organoid growth. 4EKI strongly inhibited Myc and ATF4 translation, the integrated stress response (ISR)-dependent glutamine metabolic signature, AKT activation and proliferation in vivo. 4EKI inhibited polyposis in ApcMin/+ mice by suppressing Myc protein and AKT activation. Furthermore, p-eIF4E was highly elevated in CRC precursor lesions in mouse and human. p-eIF4E cooperated with mutant KRAS to promote Myc and ISR-dependent glutamine addiction in various CRC cell lines, characterized by increased cell death, transcriptomic heterogeneity and immune suppression upon deprivation. These findings demonstrate a critical role of eIF4E S209-dependent translation in Myc and stress-driven oncogenesis and as a potential therapeutic vulnerability.
Collapse
Affiliation(s)
- Hang Ruan
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States.,UPMC Hillman Cancer Center, Pittsburgh, United States
| | - Xiangyun Li
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States.,UPMC Hillman Cancer Center, Pittsburgh, United States.,Department of Stem cell and regenerative medicine, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiang Xu
- Department of Stem cell and regenerative medicine, Daping Hospital, Army Medical University, Chongqing, China.,Central laboratory, State key laboratory of trauma, burn and combined Injury, Daping Hospital, Chongqing, China
| | - Brian J Leibowitz
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States.,UPMC Hillman Cancer Center, Pittsburgh, United States
| | - Jingshan Tong
- UPMC Hillman Cancer Center, Pittsburgh, United States.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Lujia Chen
- UPMC Hillman Cancer Center, Pittsburgh, United States.,Department of Biomedical informatics, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Luoquan Ao
- Department of Stem cell and regenerative medicine, Daping Hospital, Army Medical University, Chongqing, China.,Central laboratory, State key laboratory of trauma, burn and combined Injury, Daping Hospital, Chongqing, China
| | - Wei Xing
- Department of Stem cell and regenerative medicine, Daping Hospital, Army Medical University, Chongqing, China.,Central laboratory, State key laboratory of trauma, burn and combined Injury, Daping Hospital, Chongqing, China
| | - Jianhua Luo
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Yanping Yu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Robert E Schoen
- Departments of Medicine and Epidemiology, University of Pittsburgh, Pittsburgh, United States
| | - Nahum Sonenberg
- Department of Biochemistry, Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Xinghua Lu
- UPMC Hillman Cancer Center, Pittsburgh, United States.,Department of Biomedical informatics, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Lin Zhang
- UPMC Hillman Cancer Center, Pittsburgh, United States.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Jian Yu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States.,UPMC Hillman Cancer Center, Pittsburgh, United States
| |
Collapse
|
6
|
Ruan H, Leibowitz BJ, Zhang L, Yu J. Immunogenic cell death in colon cancer prevention and therapy. Mol Carcinog 2020; 59:783-793. [PMID: 32215970 DOI: 10.1002/mc.23183] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 02/23/2020] [Indexed: 12/15/2022]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related death worldwide. The colonic mucosa constitutes a critical barrier and a major site of immune regulation. The immune system plays important roles in cancer development and treatment, and immune activation caused by chronic infection or inflammation is well-known to increase cancer risk. During tumor development, neoplastic cells continuously interact with and shape the tumor microenvironment (TME), which becomes progressively immunosuppressive. The clinical success of immune checkpoint blockade therapies is limited to a small set of CRCs with high tumor mutational load and tumor-infiltrating T cells. Induction of immunogenic cell death (ICD), a type of cell death eliciting an immune response, can therefore help break the immunosuppressive TME, engage the innate components, and prime T cell-mediated adaptive immunity for long-term tumor control. In this review, we discuss the current understanding of ICD induced by antineoplastic agents, the influence of driver mutations, and recent developments to harness ICD in colon cancer. Mechanism-guided combinations of ICD-inducing agents with immunotherapy and actionable biomarkers will likely offer more tailored and durable benefits to patients with colon cancer.
Collapse
Affiliation(s)
- Hang Ruan
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Brian J Leibowitz
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Lin Zhang
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania.,Chemical Biology and Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jian Yu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.,UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| |
Collapse
|
7
|
Figueiredo Neto M, Liu S, Salameh JW, Yokota H, Figueiredo ML. Interleukin-27 Gene Delivery Targeting IL-6Rα-Expressing Cells as a Stress Response Therapy. Int J Mol Sci 2020; 21:E1108. [PMID: 32046108 PMCID: PMC7038084 DOI: 10.3390/ijms21031108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/28/2020] [Accepted: 02/06/2020] [Indexed: 01/11/2023] Open
Abstract
Interleukin-27 (IL-27) has shown promise in halting tumor growth and mediating tumor regression in several models, including prostate cancer. We describe our findings on the effects of IL-27 on the gene expression changes of TC2R prostate adenocarcinoma cells. We utilized RNAseq to assess profile differences between empty vector control, vector delivering IL-27 modified at its C-terminus with a non-specific peptide, and IL-27 modified at the C-terminus with a peptide targeting the IL-6-Rα. The targeted IL-27 had higher bioactivity and activity in vivo in a recent study by our group, but the mechanisms underlying this effect had not been characterized in detail at the gene expression level on tumor cells. In the present work, we sought to examine potential mechanisms for targeted IL-27 enhanced activity directly on tumor cells. The targeted IL-27 appeared to modulate several changes that would be consistent with an anti-tumor effect, including upregulation in the Interferon (IFN) and Interferon regulatory factor (IRF), oxidative phosphorylation, Janus kinase/Signal transducers and activators of transcription (JAK/STAT), and eukaryotic initiation factor 2 (EIF2) signaling. Of these signaling changes predicted by ingenuity pathway analyses (IPA), the novel form also with the highest significance (-log(Benjamini-Hochberg (B-H)) p-value) was the EIF2 signaling upregulation. We validated this predicted change by assaying for eukaryotic initiation factor 2 alpha (eIF2α), or phosphorylated eIF2α (p-eIF2α), and caspase-3 levels. We detected an increase in the phosphorylated form of eIF2α and in the cleaved caspase-3 fraction, indicating that the EIF2 signaling pathway was upregulated in these prostate tumor cells following targeted IL-27 gene delivery. This approach of targeting cytokines to enhance their activity against cancer cells is a novel approach to help augment IL-27's bioactivity and efficacy against prostate tumors and could be extended to other conditions where it could help interfere with the EIF2α pathway and promote caspase-3 activation.
Collapse
Affiliation(s)
- Manoel Figueiredo Neto
- Department of Basic Medical Sciences and Interdisciplinary Biomedical Sciences Program, Purdue University, 625 Harrison Street, LYNN 2177, West Lafayette, IN 47907, USA; (M.F.N.); (J.W.S.)
| | - Shengzhi Liu
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; (S.L.); (H.Y.)
| | - Janelle Wes Salameh
- Department of Basic Medical Sciences and Interdisciplinary Biomedical Sciences Program, Purdue University, 625 Harrison Street, LYNN 2177, West Lafayette, IN 47907, USA; (M.F.N.); (J.W.S.)
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA; (S.L.); (H.Y.)
| | - Marxa Leão Figueiredo
- Department of Basic Medical Sciences and Interdisciplinary Biomedical Sciences Program, Purdue University, 625 Harrison Street, LYNN 2177, West Lafayette, IN 47907, USA; (M.F.N.); (J.W.S.)
- Purdue Center for Cancer Research and Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
8
|
Tan YJ, Lee YT, Petersen SH, Kaur G, Kono K, Tan SC, Majid AMSA, Oon CE. BZD9L1 sirtuin inhibitor as a potential adjuvant for sensitization of colorectal cancer cells to 5-fluorouracil. Ther Adv Med Oncol 2019; 11:1758835919878977. [PMID: 31632470 PMCID: PMC6767736 DOI: 10.1177/1758835919878977] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 09/06/2019] [Indexed: 02/06/2023] Open
Abstract
Background: This study aims to investigate the combination effect of a novel sirtuin
inhibitor (BZD9L1) with 5-fluorouracil (5-FU) and to determine its molecular
mechanism of action in colorectal cancer (CRC). Methods: BZD9L1 and 5-FU either as single treatment or in combination were tested
against CRC cells to evaluate synergism in cytotoxicity, senescence and
formation of micronucleus, cell cycle and apoptosis, as well as the
regulation of related molecular players. The effects of combined treatments
at different doses on stress and apoptosis, migration, invasion and cell
death mechanism were evaluated through two-dimensional and three-dimensional
cultures. In vivo studies include investigation on the
combination effects of BZD9L1 and 5-FU on colorectal tumour xenograft growth
and an evaluation of tumour proliferation and apoptosis using
immunohistochemistry. Results: Combination treatments exerted synergistic reduction on cell viability on HCT
116 cells but not on HT-29 cells. Combined treatments reduced survival,
induced cell cycle arrest, apoptosis, senescence and micronucleation in HCT
116 cells through modulation of multiple responsible molecular players and
apoptosis pathways, with no effect in epithelial mesenchymal transition
(EMT). Combination treatments regulated SIRT1 and SIRT2 protein expression
levels differently and changed SIRT2 protein localization. Combined
treatment reduced growth, migration, invasion and viability of HCT 116
spheroids through apoptosis, when compared with the single treatment. In
addition, combined treatment was found to reduce tumour growth in
vivo through reduction of tumour proliferation and necrosis
compared with the vehicle control group. This highlights the potential
therapeutic effects of BZD9L1 and 5-FU towards CRC. Conclusion: This study may pave the way for use of BZD9L1 as an adjuvant to 5-FU in
improving the therapeutic efficacy for the treatment of colorectal
cancer.
Collapse
Affiliation(s)
- Yi Jer Tan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, Malaysia
| | - Yeuan Ting Lee
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, Malaysia
| | - Sven H Petersen
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Gurjeet Kaur
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, Malaysia
| | - Koji Kono
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Soo Choon Tan
- USains Biomics Laboratory Testing Services Sdn. Bhd., Universiti Sains Malaysia, Penang, Malaysia
| | - Amin M S Abdul Majid
- EMAN Testing and Research Laboratories, Department of Pharmacology, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | - Chern Ein Oon
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang, 11800, Malaysia
| |
Collapse
|
9
|
Tan X, Tong J, Wang YJ, Fletcher R, Schoen RE, Yu J, Shen L, Zhang L. BET Inhibitors Potentiate Chemotherapy and Killing of SPOP-Mutant Colon Cancer Cells via Induction of DR5. Cancer Res 2019; 79:1191-1203. [PMID: 30674532 DOI: 10.1158/0008-5472.can-18-3223] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 12/22/2018] [Accepted: 01/16/2019] [Indexed: 12/27/2022]
Abstract
Bromodomain and extraterminal domain (BET) family proteins such as BRD4 are epigenetic readers that control expression of a number of oncogenic proteins. Targeting this family of proteins has recently emerged as a promising anticancer approach. BET inhibitors (BETi), either alone or in combination with other anticancer agents, have exhibited efficacy in a variety of tumors. However, the molecular mechanisms underlying differential response to BETi are not well understood. In this study, we report that death receptor 5 (DR5), a key component of the extrinsic apoptotic pathway, is markedly induced in response to BRD4 depletion and BETi treatment in colorectal cancer cells. Induction of DR5, following BET inhibition, was mediated by endoplasmic reticulum stress and CHOP-dependent transcriptional activation. Enhanced DR5 induction was necessary for the chemosensitization and apoptotic effects of BETi and was responsible for increased BETi sensitivity in colorectal cancer cells containing a mutation in speckle-type POZ protein (SPOP), a subunit of BRD4 E3 ubiquitin ligase. In a colorectal cancer xenograft model, BETi combined with chemotherapy suppressed the tumor growth in a DR5-dependent manner and potently inhibited patient-derived xenograft tumor growth with enhanced DR5 induction and apoptosis. These findings suggest that BETi alone or in combination with chemotherapy is effective against colorectal cancer due to enhanced DR5 induction and apoptosis. DR5 induction may also serve as a useful marker for designing personalized treatment and improved colorectal cancer combination therapies.Significance: These findings reveal how BET inhibition sensitizes chemotherapy and kills a subset of colon cancer cells with specific genetic alterations and may provide a new molecular marker for improving colon cancer therapies.
Collapse
Affiliation(s)
- Xiao Tan
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jingshan Tong
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yi-Jun Wang
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Rochelle Fletcher
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Robert E Schoen
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jian Yu
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China.
| | - Lin Zhang
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania.
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
10
|
Budhraja A, Turnis ME, Churchman ML, Kothari A, Yang X, Xu H, Kaminska E, Panetta JC, Finkelstein D, Mullighan CG, Opferman JT. Modulation of Navitoclax Sensitivity by Dihydroartemisinin-Mediated MCL-1 Repression in BCR-ABL + B-Lineage Acute Lymphoblastic Leukemia. Clin Cancer Res 2017; 23:7558-7568. [PMID: 28974549 DOI: 10.1158/1078-0432.ccr-17-1231] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 08/31/2017] [Accepted: 09/28/2017] [Indexed: 01/06/2023]
Abstract
Purpose: BCR-ABL+ B-ALL leukemic cells are highly dependent on the expression of endogenous antiapoptotic MCL-1 to promote viability and are resistant to BH3-mimetic agents such as navitoclax (ABT-263) that target BCL-2, BCL-XL, and BCL-W. However, the survival of most normal blood cells and other cell types is also dependent on Mcl-1 Despite the requirement for MCL-1 in these cell types, initial reports of MCL-1-specific BH3-mimetics have not described any overt toxicities associated with single-agent use, but these agents are still early in clinical development. Therefore, we sought to identify approved drugs that could sensitize leukemic cells to ABT-263.Experimental Design: A screen identified dihydroartemisinin (DHA), a water-soluble metabolite of the antimalarial artemisinin. Using mouse and human leukemic cell lines, and primary patient-derived xenografts, the effect of DHA on survival was tested, and mechanistic studies were carried out to discover how DHA functions. We further tested in vitro and in vivo whether combining DHA with ABT-263 could enhance the response of leukemic cells to combination therapy.Results: DHA causes the downmodulation of MCL-1 expression by triggering a cellular stress response that represses translation. The repression of MCL-1 renders leukemic cells highly sensitive to synergistic cell death induced by ABT-263 in a mouse model of BCR-ABL+ B-ALL both in vitro and in vivo Furthermore, DHA synergizes with ABT-263 in human Ph+ ALL cell lines, and primary patient-derived xenografts of Ph+ ALL in culture.Conclusions: Our findings suggest that combining DHA with ABT-263 can improve therapeutic response in BCR-ABL+ B-ALL. Clin Cancer Res; 23(24); 7558-68. ©2017 AACR.
Collapse
Affiliation(s)
- Amit Budhraja
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Meghan E Turnis
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Michelle L Churchman
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Anisha Kothari
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Xue Yang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Haiyan Xu
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Ewa Kaminska
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - John C Panetta
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Joseph T Opferman
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, Tennessee.
| |
Collapse
|
11
|
Koriem KMM. Protective effect of natural products and hormones in colon cancer using metabolome: A physiological overview. Asian Pac J Trop Biomed 2017. [DOI: 10.1016/j.apjtb.2017.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|