1
|
Bellani MA, Shaik A, Majumdar I, Ling C, Seidman MM. Repair of genomic interstrand crosslinks. DNA Repair (Amst) 2024; 141:103739. [PMID: 39106540 PMCID: PMC11423799 DOI: 10.1016/j.dnarep.2024.103739] [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: 03/12/2024] [Revised: 07/11/2024] [Accepted: 07/25/2024] [Indexed: 08/09/2024]
Abstract
Genomic interstrand crosslinks (ICLs) are formed by reactive species generated during normal cellular metabolism, produced by the microbiome, and employed in cancer chemotherapy. While there are multiple options for replication dependent and independent ICL repair, the crucial step for each is unhooking one DNA strand from the other. Much of our insight into mechanisms of unhooking comes from powerful model systems based on plasmids with defined ICLs introduced into cells or cell free extracts. Here we describe the properties of exogenous and endogenous ICL forming compounds and provide an historical perspective on early work on ICL repair. We discuss the modes of unhooking elucidated in the model systems, the concordance or lack thereof in drug resistant tumors, and the evolving view of DNA adducts, including ICLs, formed by metabolic aldehydes.
Collapse
Affiliation(s)
- Marina A Bellani
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Althaf Shaik
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Ishani Majumdar
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Chen Ling
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Michael M Seidman
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
2
|
Newly Synthesized Melphalan Analogs Induce DNA Damage and Mitotic Catastrophe in Hematological Malignant Cancer Cells. Int J Mol Sci 2022; 23:ijms232214258. [PMID: 36430734 PMCID: PMC9693175 DOI: 10.3390/ijms232214258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/12/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
Myeloablative therapy with highdoses of the cytostatic drug melphalan (MEL) in preparation for hematopoietic cell transplantation is the standard of care for multiple myeloma (MM) patients. Melphalan is a bifunctional alkylating agent that covalently binds to nucleophilic sites in the DNA and effective in the treatment, but unfortunately has limited therapeutic benefit. Therefore, new approaches are urgently needed for patients who are resistant to existing standard treatment with MEL. Regulating the pharmacological activity of drug molecules by modifying their structure is one method for improving their effectiveness. The purpose of this work was to analyze the physicochemical and biological properties of newly synthesized melphalan derivatives (EE-MEL, EM-MEL, EM-MOR-MEL, EM-I-MEL, EM-T-MEL) obtained through the esterification of the carboxyl group and the replacement of the the amino group with an amidine group. Compounds were selected based on our previous studies for their improved anticancer properties in comparison with the original drug. For this, we first evaluated the physicochemical properties using the circular dichroism technique, then analyzed the zeta potential and the hydrodynamic diameters of the particles. Then, the in vitro biological properties of the analogs were tested on multiple myeloma (RPMI8226), acute monocytic leukemia (THP1), and promyelocytic leukemia (HL60) cells as model systems for hematological malignant cells. DNA damage was assessed by immunostaining γH2AX, cell cycle distribution changes by propidium iodide (PI) staining, and cell death by the activation of caspase 2. We proved that the newly synthesized derivatives, in particular EM-MOR-MEL and EM-T-MEL, affected the B-DNA conformation, thus increasing the DNA damage. As a result of the DNA changes, the cell cycle was arrested in the S and G2/M phases. The cell death occurred by activating a mitotic catastrophe. Our investigations suggest that the analogs EM-MOR-MEL and EM-T-MEL have better anti-cancer activity in multiple myeloma cells than the currently used melphalan.
Collapse
|
3
|
Aksenova AY, Zhuk AS, Lada AG, Zotova IV, Stepchenkova EI, Kostroma II, Gritsaev SV, Pavlov YI. Genome Instability in Multiple Myeloma: Facts and Factors. Cancers (Basel) 2021; 13:5949. [PMID: 34885058 PMCID: PMC8656811 DOI: 10.3390/cancers13235949] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/20/2021] [Accepted: 11/22/2021] [Indexed: 02/06/2023] Open
Abstract
Multiple myeloma (MM) is a malignant neoplasm of terminally differentiated immunoglobulin-producing B lymphocytes called plasma cells. MM is the second most common hematologic malignancy, and it poses a heavy economic and social burden because it remains incurable and confers a profound disability to patients. Despite current progress in MM treatment, the disease invariably recurs, even after the transplantation of autologous hematopoietic stem cells (ASCT). Biological processes leading to a pathological myeloma clone and the mechanisms of further evolution of the disease are far from complete understanding. Genetically, MM is a complex disease that demonstrates a high level of heterogeneity. Myeloma genomes carry numerous genetic changes, including structural genome variations and chromosomal gains and losses, and these changes occur in combinations with point mutations affecting various cellular pathways, including genome maintenance. MM genome instability in its extreme is manifested in mutation kataegis and complex genomic rearrangements: chromothripsis, templated insertions, and chromoplexy. Chemotherapeutic agents used to treat MM add another level of complexity because many of them exacerbate genome instability. Genome abnormalities are driver events and deciphering their mechanisms will help understand the causes of MM and play a pivotal role in developing new therapies.
Collapse
Affiliation(s)
- Anna Y. Aksenova
- Laboratory of Amyloid Biology, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Anna S. Zhuk
- International Laboratory “Computer Technologies”, ITMO University, 197101 St. Petersburg, Russia;
| | - Artem G. Lada
- Department of Microbiology and Molecular Genetics, University of California, Davis, CA 95616, USA;
| | - Irina V. Zotova
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia; (I.V.Z.); (E.I.S.)
- Vavilov Institute of General Genetics, St. Petersburg Branch, Russian Academy of Sciences, 199034 St. Petersburg, Russia
| | - Elena I. Stepchenkova
- Department of Genetics and Biotechnology, St. Petersburg State University, 199034 St. Petersburg, Russia; (I.V.Z.); (E.I.S.)
- Vavilov Institute of General Genetics, St. Petersburg Branch, Russian Academy of Sciences, 199034 St. Petersburg, Russia
| | - Ivan I. Kostroma
- Russian Research Institute of Hematology and Transfusiology, 191024 St. Petersburg, Russia; (I.I.K.); (S.V.G.)
| | - Sergey V. Gritsaev
- Russian Research Institute of Hematology and Transfusiology, 191024 St. Petersburg, Russia; (I.I.K.); (S.V.G.)
| | - Youri I. Pavlov
- Eppley Institute for Research in Cancer, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Departments of Biochemistry and Molecular Biology, Microbiology and Pathology, Genetics Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
4
|
Lai W, Zhu W, Li X, Han Y, Wang Y, Leng Q, Li M, Wen X. GTSE1 promotes prostate cancer cell proliferation via the SP1/FOXM1 signaling pathway. J Transl Med 2021; 101:554-563. [PMID: 33328578 DOI: 10.1038/s41374-020-00510-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 10/23/2020] [Accepted: 10/24/2020] [Indexed: 11/08/2022] Open
Abstract
G2 and S phase-expressed-1 (GTSE1) has been implicated in the pathogenesis of several malignant tumors. However, its specific role in prostate cancer (PCa) remains unclear. In this study, RNA-Seq data from patients with PCa and controls were downloaded from the FIREBROWSE database, and it was found that the GTSE1 mRNA level was significantly upregulated in PCa. Moreover, patients with higher GTSE1 mRNA levels had higher Gleason scores (P < 0.001), a more advanced pT stage (P = 0.011), and a more advanced pN stage (P = 0.006) as well as a shorter time to biochemical recurrence (P = 0.005). In addition, overexpression of GTSE1 could promote proliferation in LNCaP cells, whereas silencing GTSE1 could inhibit the growth of C4-2 cells in vitro and in vivo. Mechanistically, GTSE1 enhanced the expression of FOXM1 by upregulating the SP1 protein level, a transcription factor of FOXM1, which ultimately promoted PCa cell proliferation. In summary, GTSE1 is a new candidate oncogene in the development and progression of PCa, and it can promote PCa cell proliferation via the SP1/FOXM1 signaling pathway.
Collapse
Affiliation(s)
- Wenjie Lai
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weian Zhu
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaojuan Li
- Department of Health Care, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Yuefu Han
- Department of Urology, Yue Bei People's Hospital, Shaoguan, China
| | - Yu Wang
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qu Leng
- Department of Urology, The Affiliated Zhongshan Hospital, Sun Yat-Sen University, Zhongshan, China
| | - Mingzhao Li
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xingqiao Wen
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
5
|
Corbett S, Huang S, Zammarchi F, Howard PW, van Berkel PH, Hartley JA. The Role of Specific ATP-Binding Cassette Transporters in the Acquired Resistance to Pyrrolobenzodiazepine Dimer-Containing Antibody-Drug Conjugates. Mol Cancer Ther 2020; 19:1856-1865. [PMID: 32669316 DOI: 10.1158/1535-7163.mct-20-0222] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/14/2020] [Accepted: 07/02/2020] [Indexed: 01/01/2023]
Abstract
Antibody-drug conjugates (ADC) containing pyrrolobenzodiazepine (PBD) dimers are being evaluated clinically in both hematologic and solid tumors. These include ADCT-301 (camidanlumab tesirine) and ADCT-402 (loncastuximab tesirine) in pivotal phase II trials that contain the payload tesirine, which releases the PBD dimer warhead SG3199. An important consideration in future clinical development is acquired resistance. The aim was to generate and characterize PBD acquired resistant cell lines in both hematologic and solid tumor settings. Human Karpas-299 (ALCL) and NCI-N87 (gastric cancer) cells were incubated with increasing IC50 doses of ADC (targeting CD25 and HER2, respectively) or SG3199 in a pulsed manner until stable acquired resistance was established. The level of resistance achieved was approximately 3,000-fold for ADCT-301 and 3-fold for SG3199 in Karpas-299, and 8-fold for ADCT-502 and 4-fold for SG3199 in NCI-N87. Cross-resistance between ADC and SG3199, and with an alternative PBD-containing ADC or PBD dimer was observed. The acquired resistant lines produced fewer DNA interstrand cross-links, indicating an upstream mechanism of resistance. Loss of antibody binding or internalization was not observed. A human drug transporter PCR Array revealed several genes upregulated in all the resistant cell lines, including ABCG2 and ABCC2, but not ABCB1(MDR1). These findings were confirmed by RT-PCR and Western blot, and inhibitors and siRNA knockdown of ABCG2 and ABCC2 recovered drug sensitivity. These data show that acquired resistance to PBD-ADCs and SG3199 can involve specific ATP-binding cassette drug transporters. This has clinical implications as potential biomarkers of resistance and for the rational design of drug combinations.
Collapse
Affiliation(s)
- Simon Corbett
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, London, United Kingdom
| | - Shiran Huang
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, London, United Kingdom
| | - Francesca Zammarchi
- ADC Therapeutics (UK) Limited, QMB Innovation Centre, London, United Kingdom
| | - Philip W Howard
- AstraZeneca/Spirogen, QMB Innovation Centre, London, United Kingdom
| | | | - John A Hartley
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, London, United Kingdom. .,ADC Therapeutics (UK) Limited, QMB Innovation Centre, London, United Kingdom
| |
Collapse
|
6
|
Patel PR, Senyuk V, Sweiss K, Calip GS, Pan D, Rodriguez N, Oh A, Mahmud N, Rondelli D. PARP Inhibition Synergizes with Melphalan but Does not Reverse Resistance Completely. Biol Blood Marrow Transplant 2020; 26:1273-1279. [PMID: 32194286 DOI: 10.1016/j.bbmt.2020.03.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/21/2020] [Accepted: 03/06/2020] [Indexed: 12/20/2022]
Abstract
High-dose melphalan (MEL) and autologous stem cell transplantation (ASCT) is the standard of care in the treatment of multiple myeloma (MM). Resistance to MEL has been linked to increased DNA repair. Here we sought to identify whether inhibition of poly(ADP-ribose) polymerase (PARP) synergizes with MEL and can overcome resistance. We tested the synergistic cytotoxicity of 3 inhibitors of PARP (PARPi)-veliparib (VEL), olaparib (OLA), and niraparib (NIRA)-combined with MEL in RPMI8226 and U266 MM cell lines, as well as in their MEL resistance counterparts, RPMI8226-LR5 (LR5) and U266-LR6 (LR6). The addition of VEL, OLA, and NIRA to MEL reduced the half maximal inhibitory concentration (IC50) in RPMI8226 cells from 27.8 µM to 23.1 µM, 22.5 µM, and 18.0 µM, respectively. Similarly, the IC50 of MEL in U266 cells was decreased from 6.2 µM to 3.2 µM, 3.3 µM, and 3.0 µM, respectively. In LR5 and LR6 cells, PARPi did not reverse MEL resistance. We confirmed this in a NOD/SCID/gamma null xenograft mouse model with either MEL-sensitive (RPMI8226) or MEL-resistant (LR5) MM. Treatment with a MEL-VEL combination prolonged survival compared with MEL alone in RPMI8226 mice (107 days versus 67.5 days; P = .0009), but not in LR5 mice (41 versus 39 days; P = .09). We next tested whether 2 double-stranded DNA repair mechanisms, homologous recombination (HR) and nonhomologous end-joining (NHEJ), cause MEL resistance in LR5 and LR6 cells. In an HR assay, LR6 cells had a 4.5-fold greater HR capability than parent U226 cells (P = .05); however, LR5 cells had an equivalent HR ability as parent RPMI8226 cells. We hypothesized that NHEJ may be a mediator of MEL resistance in LR5 cells. Given that DNA-PK is integral to NHEJ and may be a therapeutic target, we treated LR5 cells with the DNA-PK inhibitor NU7026 in combination with MEL. Although NU7026 alone at 2.5 µM had no cytotoxicity, in combination it completely reversed resistance to MEL (MEL IC50, 46.4 µM versus 14.4 µM). We examined the clinical implications of our findings in a dataset of 414 patients treated with tandem ASCT. High PARP1 expressers had lower survival compared with patients with low expression (median 42.7 months versus median not reached; P = .003). We hypothesized that combined expression of the HR gene BRCA1, the NHEJ gene PRKDC (DNA-PK), and PARP1 may predict survival and found that overexpression of 0 (n = 101), 1 or 2 (n = 287), or all 3 (n = 26) genes had a negative impact on median survival (undefined versus 57.8 months versus 14.8 months; P < .0001). Here we demonstrate that PARPi synergized with MEL, but that resistance (which may be due to HR and NHEJ pathways) is not completely reversed by PARPi. In addition, we observed that a 3-gene analysis may be tested to identify patients resistant or sensitive to high-dose MEL.
Collapse
Affiliation(s)
- Pritesh R Patel
- Division of Hematology/ Oncology, University of Illinois at Chicago, Chicago, Illinois.
| | - Vitalyi Senyuk
- Division of Hematology/ Oncology, University of Illinois at Chicago, Chicago, Illinois
| | - Karen Sweiss
- Department of Pharmacy Practice, University of Illinois at Chicago, Chicago, Illinois
| | - Greg S Calip
- Department of Pharmacy Systems, Outcomes and Policy, University of Illinois at Chicago, Chicago, Illinois
| | - Dipanjan Pan
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois; Biomedical Research Center, Carle Foundation Hospital, Urbana, Illinois
| | - Natalie Rodriguez
- University of Illinois at Chicago, College of Medicine, Chicago, Illinois
| | - Annie Oh
- Division of Hematology/ Oncology, University of Illinois at Chicago, Chicago, Illinois
| | - Nadim Mahmud
- Division of Hematology/ Oncology, University of Illinois at Chicago, Chicago, Illinois
| | - Damiano Rondelli
- Division of Hematology/ Oncology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
7
|
Gourzones C, Bellanger C, Lamure S, Gadacha OK, De Paco EG, Vincent L, Cartron G, Klein B, Moreaux J. Antioxidant Defenses Confer Resistance to High Dose Melphalan in Multiple Myeloma Cells. Cancers (Basel) 2019; 11:cancers11040439. [PMID: 30925767 PMCID: PMC6521290 DOI: 10.3390/cancers11040439] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/15/2019] [Accepted: 03/20/2019] [Indexed: 12/25/2022] Open
Abstract
Background: Multiple myeloma (MM) is the second most common hematological cancer after lymphoma. It is characterized by the accumulation of clonal malignant plasma cells within the bone marrow. The development of drug resistance remains a major problem for effective treatment of MM. Understand the mechanisms underlying drug resistance in MM is a focal point to improve MM treatment. Methods: In the current study, we analyzed further the role of redox imbalance induction in melphalan-induced toxicity both in human myeloma cell lines (HMCLs) and primary myeloma cells from patients. Results: We developed an in-vitro model of short-term resistance to high-dose melphalan and identified that pretreatment with physiological concentration of GSH protects HMCLs from melphalan-induced cell cycle arrest and cytotoxicity. We validated these results using primary MM cells from patients co-cultured with their bone marrow microenvironment. GSH did not affect the ability of melphalan to induce DNA damages in MM cells. Interestingly, melphalan induced reactive oxygen species, a significant decrease in GSH concentration, protein and lipd oxydation together with NRF2 (NF-E2-related factor 2) pathway activation. Conclusions: Our data demonstrate that antioxidant defenses confers resistance to high dose melphalan in MM cells, supporting that redox status in MM cells could be determinant for patients’ response to melphalan.
Collapse
Affiliation(s)
- Claire Gourzones
- IGH, CNRS, University of Montpellier, 34000 Montpellier, France.
| | - Céline Bellanger
- IGH, CNRS, University of Montpellier, 34000 Montpellier, France.
| | - Sylvain Lamure
- Department of Clinical Hematology, CHU Montpellier, 34395 Montpellier, France.
| | | | | | - Laure Vincent
- Department of Clinical Hematology, CHU Montpellier, 34395 Montpellier, France.
| | - Guillaume Cartron
- Department of Clinical Hematology, CHU Montpellier, 34395 Montpellier, France.
- Univ Montpellier, UFR de Médecine, 34000 Montpellier, France.
- Univ Montpellier, UMR CNRS 5235, 34000 Montpellier, France.
| | - Bernard Klein
- IGH, CNRS, University of Montpellier, 34000 Montpellier, France.
- Univ Montpellier, UFR de Médecine, 34000 Montpellier, France.
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France.
| | - Jérôme Moreaux
- IGH, CNRS, University of Montpellier, 34000 Montpellier, France.
- Univ Montpellier, UFR de Médecine, 34000 Montpellier, France.
- Department of Biological Hematology, CHU Montpellier, 34295 Montpellier, France.
| |
Collapse
|
8
|
Wu C, Nilsson K, Zheng Y, Ekenstierna C, Sugiyama N, Forslund O, Kajitani N, Yu H, Wennerberg J, Ekblad L, Schwartz S. Short half-life of HPV16 E6 and E7 mRNAs sensitizes HPV16-positive tonsillar cancer cell line HN26 to DNA-damaging drugs. Int J Cancer 2019; 144:297-310. [PMID: 30303514 PMCID: PMC6587446 DOI: 10.1002/ijc.31918] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/10/2018] [Accepted: 09/17/2018] [Indexed: 12/12/2022]
Abstract
Here we show that treatment of the HPV16-positive tonsillar cancer cell line HN26 with DNA alkylating cancer drug melphalan-induced p53 and activated apoptosis. Melphalan reduced the levels of RNA polymerase II and cellular transcription factor Sp1 that were associated with HPV16 DNA. The resulting inhibition of transcription caused a rapid loss of the HPV16 early mRNAs encoding E6 and E7 as a result of their inherent instability. As a consequence of HPV16 E6 and E7 down-regulation, the DNA damage inflicted on the cells by melphalan caused induction of p53 and activation of apoptosis in the HN26 cells. The BARD1-negative phenotype of the HN26 cells may have contributed to the failure to repair DNA damage caused by melphalan, as well as to the efficient apoptosis induction. Finally, nude mice carrying the HPV16 positive tonsillar cancer cells responded better to melphalan than to cisplatin, the chemotherapeutic drug of choice for tonsillar cancer. We concluded that the short half-life of the HPV16 E6 and E7 mRNAs renders HPV16-driven tonsillar cancer cells particularly sensitive to DNA damaging agents such as melphalan since melphalan both inhibits transcription and causes DNA damage.
Collapse
Affiliation(s)
- Chengjun Wu
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Kersti Nilsson
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Yunji Zheng
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Camilla Ekenstierna
- Department of Clinical Sciences Lund, Oncology and Pathology, Lund University, Skane University Hospital, Lund, Sweden
| | - Natsuki Sugiyama
- Department of Clinical Sciences Lund, Oncology and Pathology, Lund University, Skane University Hospital, Lund, Sweden
| | - Ola Forslund
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Naoko Kajitani
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Haoran Yu
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Johan Wennerberg
- Department of Clinical Sciences Lund, Oto-rhino-laryngology, Head and Neck Surgery, Lund University, Skane University Hospital, Lund, Sweden
| | - Lars Ekblad
- Department of Clinical Sciences Lund, Oncology and Pathology, Lund University, Skane University Hospital, Lund, Sweden
| | - Stefan Schwartz
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
9
|
Koomen DC, Guingab-Cagmat JD, Oliveira PS, Fang B, Liu M, Welsh EA, Meads MB, Nguyen T, Meke L, Eschrich SA, Shain KH, Garrett TJ, Koomen JM. Proteometabolomics of Melphalan Resistance in Multiple Myeloma. Methods Mol Biol 2019; 1996:273-296. [PMID: 31127562 DOI: 10.1007/978-1-4939-9488-5_21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Drug resistance remains a critical problem for the treatment of multiple myeloma (MM), which can serve as a specific example for a highly prevalent unmet medical need across almost all cancer types. In MM, the therapeutic arsenal has expanded and diversified, yet we still lack in-depth molecular understanding of drug mechanisms of action and cellular pathways to therapeutic escape. For those reasons, preclinical models of drug resistance are developed and characterized using different approaches to gain insights into tumor biology and elucidate mechanisms of drug resistance. For MM, numerous drugs are used for treatment, including conventional chemotherapies (e.g., melphalan or L-phenylalanine nitrogen mustard), proteasome inhibitors (e.g., Bortezomib), and immunomodulators (e.g., Lenalidomide). These agents have diverse effects on the myeloma cells, and several mechanisms of drug resistance have been previously described. The disparity of these mechanisms and the complexity of these biological processes lead to the formation of complicated hypotheses that require omics approaches for efficient and effective analysis of model systems that can then be interpreted for patient benefit. Here, we describe the combination of metabolomics and proteomics to assess melphalan resistance in MM by examining three specific areas: drug metabolism, modulation of endogenous metabolites to assist in therapeutic escape, and changes in protein activity gauged by ATP probe uptake.
Collapse
Affiliation(s)
| | | | | | - Bin Fang
- Moffitt Cancer Center, Tampa, FL, USA
| | - Min Liu
- Moffitt Cancer Center, Tampa, FL, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Liu A, Zeng S, Lu X, Xiong Q, Xue Y, Tong L, Xu W, Sun Y, Zhang Z, Xu C. Overexpression of G2 and S phase-expressed-1 contributes to cell proliferation, migration, and invasion via regulating p53/FoxM1/CCNB1 pathway and predicts poor prognosis in bladder cancer. Int J Biol Macromol 2018; 123:322-334. [PMID: 30414902 DOI: 10.1016/j.ijbiomac.2018.11.032] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/07/2018] [Accepted: 11/07/2018] [Indexed: 12/12/2022]
Abstract
Bladder cancer is one of the most common urogenital tumors worldwide. The specific function and molecular mechanism of GTSE1 in bladder cancer remain unknown. In the present study, real-time quantitative polymerase chain reaction and Western blotting were used to identify GTSE1 expression in bladder cancer tissues and cells, and immunohistochemical assays were conducted to investigate GTSE1 expression in tissue microarray. Regression analyses explored the relationship between GTSE1 expression and pathological characteristics. A series of functional tests were performed to observe the effects of GTSE1 knockdown or overexpression, and the related mechanism was also performed. GTSE1 expression was significantly higher in bladder cancer tissues; overexpression of GTSE1 was positively associated with disease recurrence history, lymph node invasion, and progression. Patients with higher GTSE1 expression were more likely to experience shorter survival time, and GTSE1 expression served as a prognostic factor for the disease progression. Knockdown of GTSE1 obviously suppressed the proliferation, migration, and invasion capacity whereas increasing GTSE1 led to the opposite trend, which suggested that GTSE1 could serve as a potential therapeutic target for bladder cancer. GTSE1 overexpression in bladder cancer might participate in the regulation of FoxM1/CCNB1 expression via the induction of the transfer of p53 to cytoplasm.
Collapse
Affiliation(s)
- Anwei Liu
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China
| | - Shuxiong Zeng
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China
| | - Xin Lu
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China
| | - Qiao Xiong
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China
| | - Yongping Xue
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China
| | - Liping Tong
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China
| | - Weidong Xu
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China
| | - Yinghao Sun
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China
| | - Zhensheng Zhang
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China.
| | - Chuanliang Xu
- Department of Urology, Changhai Hospital Affiliated by the Second Military Medical University, Shanghai City, China.
| |
Collapse
|
11
|
Hartley JA, Flynn MJ, Bingham JP, Corbett S, Reinert H, Tiberghien A, Masterson LA, Antonow D, Adams L, Chowdhury S, Williams DG, Mao S, Harper J, Havenith CEG, Zammarchi F, Chivers S, van Berkel PH, Howard PW. Pre-clinical pharmacology and mechanism of action of SG3199, the pyrrolobenzodiazepine (PBD) dimer warhead component of antibody-drug conjugate (ADC) payload tesirine. Sci Rep 2018; 8:10479. [PMID: 29992976 PMCID: PMC6041317 DOI: 10.1038/s41598-018-28533-4] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 06/22/2018] [Indexed: 01/12/2023] Open
Abstract
Synthetic pyrrolobenzodiazepine (PBD) dimers, where two PBD monomers are linked through their aromatic A-ring phenolic C8-positions via a flexible propyldioxy tether, are highly efficient DNA minor groove cross-linking agents with potent cytotoxicity. PBD dimer SG3199 is the released warhead component of the antibody-drug conjugate (ADC) payload tesirine (SG3249), currently being evaluated in several ADC clinical trials. SG3199 was potently cytotoxic against a panel of human solid tumour and haematological cancer cell lines with a mean GI50 of 151.5 pM. Cells defective in DNA repair protein ERCC1 or homologous recombination repair showed increased sensitivity to SG3199 and the drug was only moderately susceptible to multidrug resistance mechanisms. SG3199 was highly efficient at producing DNA interstrand cross-links in naked linear plasmid DNA and dose-dependent cross-linking was observed in cells. Cross-links formed rapidly in cells and persisted over 36 hours. Following intravenous (iv) administration to rats SG3199 showed a very rapid clearance with a half life as short as 8 minutes. These combined properties of cytotoxic potency, rapid formation and persistence of DNA interstrand cross-links and very short half-life contribute to the emerging success of SG3199 as a warhead in clinical stage ADCs.
Collapse
Affiliation(s)
- John A Hartley
- Cancer Research UK Drug DNA Interactions Research Group, UCL Cancer Institute, 72 Huntley Street, London, WC1E 6BT, UK. .,Spirogen Ltd, QMB Innovation Centre, 42 New Road, London, E1 2AX, UK.
| | - Michael J Flynn
- Cancer Research UK Drug DNA Interactions Research Group, UCL Cancer Institute, 72 Huntley Street, London, WC1E 6BT, UK
| | - John P Bingham
- Cancer Research UK Drug DNA Interactions Research Group, UCL Cancer Institute, 72 Huntley Street, London, WC1E 6BT, UK
| | - Simon Corbett
- Cancer Research UK Drug DNA Interactions Research Group, UCL Cancer Institute, 72 Huntley Street, London, WC1E 6BT, UK.,Spirogen Ltd, QMB Innovation Centre, 42 New Road, London, E1 2AX, UK
| | - Halla Reinert
- Cancer Research UK Drug DNA Interactions Research Group, UCL Cancer Institute, 72 Huntley Street, London, WC1E 6BT, UK
| | - Arnaud Tiberghien
- Spirogen Ltd, QMB Innovation Centre, 42 New Road, London, E1 2AX, UK
| | - Luke A Masterson
- Spirogen Ltd, QMB Innovation Centre, 42 New Road, London, E1 2AX, UK
| | - Dyeison Antonow
- Spirogen Ltd, QMB Innovation Centre, 42 New Road, London, E1 2AX, UK
| | - Lauren Adams
- Spirogen Ltd, QMB Innovation Centre, 42 New Road, London, E1 2AX, UK
| | - Sajidah Chowdhury
- Spirogen Ltd, QMB Innovation Centre, 42 New Road, London, E1 2AX, UK
| | - David G Williams
- Spirogen Ltd, QMB Innovation Centre, 42 New Road, London, E1 2AX, UK
| | - Shenlan Mao
- MedImmune, One MedImmune Way, Gaithersburg, MD, 20878, USA
| | - Jay Harper
- MedImmune, One MedImmune Way, Gaithersburg, MD, 20878, USA
| | - Carin E G Havenith
- ADC Therapeutics (UK) Limited, QMB Innovation Centre, 42 New Road, London, E1 2AX, UK
| | - Francesca Zammarchi
- ADC Therapeutics (UK) Limited, QMB Innovation Centre, 42 New Road, London, E1 2AX, UK
| | - Simon Chivers
- ADC Therapeutics (UK) Limited, QMB Innovation Centre, 42 New Road, London, E1 2AX, UK
| | - Patrick H van Berkel
- ADC Therapeutics (UK) Limited, QMB Innovation Centre, 42 New Road, London, E1 2AX, UK
| | - Philip W Howard
- Spirogen Ltd, QMB Innovation Centre, 42 New Road, London, E1 2AX, UK
| |
Collapse
|
12
|
Nilsson K, Wu C, Kajitani N, Yu H, Tsimtsirakis E, Gong L, Winquist EB, Glahder J, Ekblad L, Wennerberg J, Schwartz S. The DNA damage response activates HPV16 late gene expression at the level of RNA processing. Nucleic Acids Res 2018; 46:5029-5049. [PMID: 29596642 PMCID: PMC6007495 DOI: 10.1093/nar/gky227] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 03/15/2018] [Accepted: 03/19/2018] [Indexed: 12/13/2022] Open
Abstract
We show that the alkylating cancer drug melphalan activated the DNA damage response and induced human papillomavirus type 16 (HPV16) late gene expression in an ATM- and Chk1/2-dependent manner. Activation of HPV16 late gene expression included inhibition of the HPV16 early polyadenylation signal that resulted in read-through into the late region of HPV16. This was followed by activation of the exclusively late, HPV16 splice sites SD3632 and SA5639 and production of spliced late L1 mRNAs. Altered HPV16 mRNA processing was paralleled by increased association of phosphorylated BRCA1, BARD1, BCLAF1 and TRAP150 with HPV16 DNA, and increased association of RNA processing factors U2AF65 and hnRNP C with HPV16 mRNAs. These RNA processing factors inhibited HPV16 early polyadenylation and enhanced HPV16 late mRNA splicing, thereby activating HPV16 late gene expression.
Collapse
Affiliation(s)
- Kersti Nilsson
- Department of Laboratory Medicine, Lund University, BMC-B13, 221 84 Lund, Sweden
| | - Chengjun Wu
- Department of Laboratory Medicine, Lund University, BMC-B13, 221 84 Lund, Sweden
| | - Naoko Kajitani
- Department of Laboratory Medicine, Lund University, BMC-B13, 221 84 Lund, Sweden
| | - Haoran Yu
- Department of Laboratory Medicine, Lund University, BMC-B13, 221 84 Lund, Sweden
| | | | - Lijing Gong
- Department of Laboratory Medicine, Lund University, BMC-B13, 221 84 Lund, Sweden
- China Academy of Sport and Health Sciences, Beijing Sport University, Xinxi Road 48, Haidian District, 100084 Beijing, China
| | - Ellenor B Winquist
- Department of Laboratory Medicine, Lund University, BMC-B13, 221 84 Lund, Sweden
| | - Jacob Glahder
- Department of Laboratory Medicine, Lund University, BMC-B13, 221 84 Lund, Sweden
| | - Lars Ekblad
- Department of Clinical Sciences Lund, Oncology and Pathology, Lund University, Skane University Hospital, 221 85 Lund, Sweden
| | - Johan Wennerberg
- Department of Clinical Sciences Lund, Oto-rhino-laryngology, Head and Neck Surgery, Lund University, Skane University Hospital, 221 85 Lund, Sweden
| | - Stefan Schwartz
- Department of Laboratory Medicine, Lund University, BMC-B13, 221 84 Lund, Sweden
| |
Collapse
|
13
|
Nucleotide excision repair is a potential therapeutic target in multiple myeloma. Leukemia 2017; 32:111-119. [PMID: 28588253 PMCID: PMC5720937 DOI: 10.1038/leu.2017.182] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 04/05/2017] [Accepted: 05/24/2017] [Indexed: 12/20/2022]
Abstract
Despite the development of novel drugs, alkylating agents remain an important component of therapy in multiple myeloma (MM). DNA repair processes contribute towards sensitivity to alkylating agents and therefore we here evaluate the role of nucleotide excision repair (NER), which is involved in the removal of bulky adducts and DNA crosslinks in MM. We first evaluated NER activity using a novel functional assay and observed a heterogeneous NER efficiency in MM cell lines and patient samples. Using next-generation sequencing data, we identified that expression of the canonical NER gene, excision repair cross-complementation group 3 (ERCC3), significantly impacted the outcome in newly diagnosed MM patients treated with alkylating agents. Next, using small RNA interference, stable knockdown and overexpression, and small-molecule inhibitors targeting xeroderma pigmentosum complementation group B (XPB), the DNA helicase encoded by ERCC3, we demonstrate that NER inhibition significantly increases sensitivity and overcomes resistance to alkylating agents in MM. Moreover, inhibiting XPB leads to the dual inhibition of NER and transcription and is particularly efficient in myeloma cells. Altogether, we show that NER impacts alkylating agents sensitivity in myeloma cells and identify ERCC3 as a potential therapeutic target in MM.
Collapse
|
14
|
Valdez BC, Li Y, Murray D, Liu Y, Nieto Y, Champlin RE, Andersson BS. The PARP inhibitor olaparib enhances the cytotoxicity of combined gemcitabine, busulfan and melphalan in lymphoma cells. Leuk Lymphoma 2017; 58:2705-2716. [PMID: 28394191 DOI: 10.1080/10428194.2017.1306647] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The combination of gemcitabine (Gem), busulfan (Bu), and melphalan (Mel) is a promising regimen for autologous stem-cell transplantation (SCT) for lymphomas. To further improve the efficacy of [Gem + Bu + Mel], we added poly(ADP-ribose) polymerase (PARP) inhibitor olaparib (Ola). We hypothesized that Ola would inhibit the repair of damaged DNA caused by [Gem + Bu + Mel]. Exposure of J45.01 and Toledo cell lines to IC10-20 of individual drug inhibited proliferation by 6-16%; [Gem + Bu + Mel] by 20-27%; and [Gem + Bu + Mel + Ola] by 61-67%. The synergistic cytotoxicity of the four-drug combination may be attributed to activation of the DNA-damage response, inhibition of PARP activity and DNA repair, decreased mitochondrial membrane potential, increased production of reactive oxygen species, and activation of the SAPK/JNK stress signaling pathway, all of which may enhance apoptosis. Similar observations were obtained using mononuclear cells isolated from patients with T-cell lymphocytic leukemia. Our results provide a rationale for undertaking clinical trials of this drug combination for lymphoma patients undergoing SCT.
Collapse
Affiliation(s)
- Benigno C Valdez
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Yang Li
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - David Murray
- b Department of Experimental Oncology , Cross Cancer Institute , Edmonton , Canada
| | - Yan Liu
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Yago Nieto
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Richard E Champlin
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Borje S Andersson
- a Department of Stem Cell Transplantation and Cellular Therapy , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
15
|
Guo L, Zhang S, Zhang B, Chen W, Li X, Zhang W, Zhou C, Zhang J, Ren N, Ye Q. Silencing GTSE-1 expression inhibits proliferation and invasion of hepatocellular carcinoma cells. Cell Biol Toxicol 2016; 32:263-74. [PMID: 27240802 PMCID: PMC4945688 DOI: 10.1007/s10565-016-9327-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 04/05/2016] [Indexed: 02/07/2023]
Abstract
G2 and S phase-expressed-1 (GTSE1) was recently reported to upregulate in several types of human cancer, based on negatively regulate p53 expression. However, its expression and functional roles in hepatocellular carcinoma (HCC) remain unknown. In this study, GTSE1 was observed to be highly expressed in HCC specimens and cell lines both at messenger RNA (mRNA) and protein levels. Furthermore, high GTSE1 expression was positively associated with tumor size, venous invasion, advanced tumor stage, and short overall survival. Moreover, we generated stable GTSE1 knockdown HCC cell lines to explore the effects of GTSE1 silencing on the growth and invasion of HCC in vitro. In determining the pathway through which GTSE1 regulated cell proliferation and invasion, GTSE1 silencing was found to inhibit AKT phosphorylation and downregulated cell cycle-related protein. In addition, GTSE1 downregulation decreased the growth of xenografts. In conclusion, these results indicated for the first time that overexpression of GTSE1 was involved in the progress of HCC, enhancing proliferation and promoting cell invasion in HCC cells.
Collapse
Affiliation(s)
- Lei Guo
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fundan University, Ministry of Education, Shanghai, 200032, China
| | - Shumin Zhang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Bo Zhang
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fundan University, Ministry of Education, Shanghai, 200032, China
| | - Wanyong Chen
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fundan University, Ministry of Education, Shanghai, 200032, China
| | - Xiaoqiang Li
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fundan University, Ministry of Education, Shanghai, 200032, China
| | - Wentao Zhang
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fundan University, Ministry of Education, Shanghai, 200032, China
| | - Chenhao Zhou
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fundan University, Ministry of Education, Shanghai, 200032, China
| | - Jubo Zhang
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Key Laboratory of Carcinogenesis and Cancer Invasion, Fundan University, Ministry of Education, Shanghai, 200032, China
| | - Ning Ren
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Key Laboratory of Carcinogenesis and Cancer Invasion, Fundan University, Ministry of Education, Shanghai, 200032, China. .,Department of Liver Surgery, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China.
| | - Qinghai Ye
- Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Key Laboratory of Carcinogenesis and Cancer Invasion, Fundan University, Ministry of Education, Shanghai, 200032, China. .,Department of Liver Surgery, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
16
|
Colmegna B, Uboldi S, Frapolli R, Licandro SA, Panini N, Galmarini CM, Badri N, Spanswick VJ, Bingham JP, Kiakos K, Erba E, Hartley JA, D'Incalci M. Increased sensitivity to platinum drugs of cancer cells with acquired resistance to trabectedin. Br J Cancer 2015; 113:1687-93. [PMID: 26633559 PMCID: PMC4701998 DOI: 10.1038/bjc.2015.407] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 10/29/2015] [Accepted: 11/06/2015] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND In order to investigate the mechanisms of acquired resistance to trabectedin, trabectedin-resistant human myxoid liposarcoma (402-91/T) and ovarian carcinoma (A2780/T) cell lines were derived and characterised in vitro and in vivo. METHODS Resistant cell lines were obtained by repeated exposures to trabectedin. Characterisation was performed by evaluating drug sensitivity, cell cycle perturbations, DNA damage and DNA repair protein expression. In vivo experiments were performed on A2780 and A2780/T xenografts. RESULTS 402-91/T and A2780/T cells were six-fold resistant to trabectedin compared with parental cells. Resistant cells were found to be hypersensitive to UV light and did not express specific proteins involved in the nucleotide excision repair (NER) pathway: XPF and ERCC1 in 402-91/T and XPG in A2780/T. NER deficiency in trabectedin-resistant cells was associated with the absence of a G2/M arrest induced by trabectedin and with enhanced sensitivity (two-fold) to platinum drugs. In A2780/T, this collateral sensitivity, confirmed in vivo, was associated with an increased formation of DNA interstrand crosslinks. CONCLUSIONS Our finding that resistance to trabectedin is associated with the loss of NER function, with a consequent increased sensitivity to platinum drugs, provides the rational for sequential use of these drugs in patients who have acquired resistance to trabectedin.
Collapse
Affiliation(s)
- B Colmegna
- Department of Oncology, IRCCS – Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, Milan 20156, Italy
| | - S Uboldi
- Department of Oncology, IRCCS – Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, Milan 20156, Italy
| | - R Frapolli
- Department of Oncology, IRCCS – Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, Milan 20156, Italy
| | - S A Licandro
- Department of Oncology, IRCCS – Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, Milan 20156, Italy
| | - N Panini
- Department of Oncology, IRCCS – Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, Milan 20156, Italy
| | - C M Galmarini
- Department of Research and Development (R&D), PharmaMar S.A., Colmenar Viejo, Madrid 28770, Spain
| | - Nadia Badri
- Department of Research and Development (R&D), PharmaMar S.A., Colmenar Viejo, Madrid 28770, Spain
| | - V J Spanswick
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | - J P Bingham
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | - Konstantinos Kiakos
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | - E Erba
- Department of Oncology, IRCCS – Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, Milan 20156, Italy
| | - J A Hartley
- Cancer Research UK Drug-DNA Interactions Research Group, UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK
| | - M D'Incalci
- Department of Oncology, IRCCS – Istituto di Ricerche Farmacologiche Mario Negri, via La Masa 19, Milan 20156, Italy
| |
Collapse
|
17
|
Subhash VV, Tan SH, Tan WL, Yeo MS, Xie C, Wong FY, Kiat ZY, Lim R, Yong WP. GTSE1 expression represses apoptotic signaling and confers cisplatin resistance in gastric cancer cells. BMC Cancer 2015. [PMID: 26209226 PMCID: PMC4514980 DOI: 10.1186/s12885-015-1550-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Background Platinum based therapy is commonly used in the treatment of advanced gastric cancer. However, resistance to chemotherapy is a major challenge that causes marked variation in individual response rate and survival rate. In this study, we aimed to identify the expression of GTSE1 and its correlation with cisplatin resistance in gastric cancer cells. Methods Methylation profiling was carried out in tissue samples from gastric cancer patients before undergoing neoadjuvent therapy using docetaxel, cisplatin and 5FU (DCX) and in gastric cancer cell lines. The correlation between GTSE1 expression and methylation in gastric cancer cells was determined by RT-PCR and MSP respectively. GTSE1 expression was knocked-down using shRNA’s and its effects on cisplatin cytotoxicity and cell survival were detected by MTS, proliferation and clonogenic survival assays. Additionally, the effect of GTSE1 knock down in drug induced apoptosis was determined by western blotting and apoptosis assays. Results GTSE1 exhibited a differential methylation index in gastric cancer patients and in cell lines that correlated with DCX treatment response and cisplatin sensitivity, respectively. In-vitro, GTSE1 expression showed a direct correlation with hypomethylation. Interestingly, Cisplatin treatment induced a dose dependent up regulation as well as nuclear translocation of GTSE1 expression in gastric cancer cells. Knock down of GTSE1 enhanced cisplatin cytotoxity and led to a significant reduction in cell proliferation and clonogenic survival. Also, loss of GTSE1 expression caused a significant increase in P53 mediated apoptosis in cisplatin treated cells. Conclusion Our study identifies GTSE1 as a biomarker for cisplatin resistance in gastric cancer cells. This study also suggests the repressive role of GTSE1 in cisplatin induced apoptosis and signifies its potential utility as a therapeutic target for better clinical management of gastric cancer patients. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1550-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vinod Vijay Subhash
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.
| | - Shi Hui Tan
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore, Singapore.
| | - Woei Loon Tan
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore, Singapore.
| | - Mei Shi Yeo
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore, Singapore.
| | - Chen Xie
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore, Singapore.
| | - Foong Ying Wong
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore, Singapore.
| | - Zee Ying Kiat
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore, Singapore.
| | - Robert Lim
- Department of Haematology-Oncology, National University Hospital of Singapore, Singapore, Singapore.
| | - Wei Peng Yong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore. .,Department of Haematology-Oncology, National University Hospital of Singapore, Singapore, Singapore.
| |
Collapse
|
18
|
Xiong T, Wei H, Chen X, Xiao H. PJ34, a poly(ADP-ribose) polymerase (PARP) inhibitor, reverses melphalan-resistance and inhibits repair of DNA double-strand breaks by targeting the FA/BRCA pathway in multidrug resistant multiple myeloma cell line RPMI8226/R. Int J Oncol 2014; 46:223-32. [PMID: 25351371 DOI: 10.3892/ijo.2014.2726] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 10/03/2014] [Indexed: 11/06/2022] Open
Abstract
There is still no ideal treatment for multidrug resistant multiple myeloma, looking for drugs which can reverse chemotherapy resistance and enhance curative effects of chemotherapy drugs becomes a problem that needs to be solved urgently. Poly(ADP-ribose) polymerase inhibitors appear to be an important tool for medical therapy of several malignancies. In the present study, we investigated the potential of the PARP-1 inhibitor PJ34, in vitro, to further enhance the efficacy of the traditional chemotherapy drug melphalan in the multidrug-resistant multiple myeloma cell line RPMI8226/R. The effects of different concentrations of PJ34 and melphalan on cell proliferation were determined by the CCK-8 assay. The expressions of FA/BRCA pathway-related factors were detected by western blotting and RT-PCR. The percentage of cell apoptosis was measured with flow cytometry. DNA double-strand break (DSB) repair was quantified by γH2AX immunofluorescence. In addition, DNA damage repair at the level of the individual cell was determined by comet assay. Co-administration of PJ34 and melphalan had synergistic inhibitory effects on the proliferation of RPMI8226/R cells, suggesting more powerful antitumor activities. The apoptosis percentage also was increased more obviously by the treatment of melphalan plus PJ34. The activation of FA/BRCA pathway was inhibited by downregulation of related factors including FANCD2, BRCA2 and Rad51. PJ34 significantly increased the ratio of γH2AX-positive cells and the number of foci/cells. The comet tail rate of cells, tail length, tail moment and Olive tail moment all increased after PJ34 treatment in RPMI8226/R cells. These results indicate that PJ34 combined treatment with melphalan produces synergistic effects and reverses multidrug resistance of RPMI8226/R cells effectively. PJ34 cannot induce DNA damage directly, but it may increase the DNA damage induced by melphalan through inhibiting DNA damage repair. The suppression of FA/BRCA pathway may be the mechanism. Therefore, we suggest that PARP inhibitors may deserve future investigations as tools for medical treatment of multidrug resistant multiple myeloma.
Collapse
Affiliation(s)
- Ting Xiong
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Heng Wei
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Xiaoqiong Chen
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Hui Xiao
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
19
|
Liu L, Zuo LF, Guo JW. ABCG2 gene amplification and expression in esophageal cancer cells with acquired adriamycin resistance. Mol Med Rep 2014; 9:1299-304. [PMID: 24535197 DOI: 10.3892/mmr.2014.1949] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 01/29/2014] [Indexed: 11/06/2022] Open
Abstract
Resistance to chemotherapeutic agents is the main reason for treatment failure in patients with cancer. The primary mechanism of multidrug resistance (MDR) is the overexpression of drug efflux transporters, including ATP‑binding cassette transporter G2 (ABCG2). To the best of our knowledge, the MDR mechanisms of esophageal cancer have not been described. An adriamycin (ADM)-resistant subline, Eca109/ADM, was generated from the Eca109 esophageal cancer cell line by a stepwise selection in ADM from 0.002 to 0.02 ng/µl. The resulting subline, designated Eca109/ADM, revealed a 3.29-fold resistance against ADM compared with the Eca109 cell line. The ABCG2 gene expression in the Eca109/ADM cells was increased compared with that of the Eca109 cells. The cellular properties of the Eca109/ADM cells were detected by reverse transcription polymerase chain reaction (RT-PCR), flow cytometry and western blotting. The ABCG2 expression levels were detected by RT-PCR and flow cytometry, and the drug efflux effect was detected by flow cytometry. The present study detected the correlation between ABCG2 and the multidrug resistance of esophageal cancer. ABCG2 gene expression and the drug efflux effect of the Eca109/ADM cells were increased compared with those of the Eca109 cells. Collectively, the results of this study indicated that the overexpression of ABCG2 in the Eca109/ADM cells resulted in drug efflux, which may be responsible for the development of esophageal cancer MDR.
Collapse
Affiliation(s)
- Liang Liu
- Department of Flow Cytometry Analysis, Tumor Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Lian Fu Zuo
- Department of Flow Cytometry Analysis, Tumor Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Jian Wen Guo
- Department of Flow Cytometry Analysis, Tumor Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
20
|
KE SHIZHONG, NI XIAOYAN, ZHANG YUEHUA, WANG YINAN, WU BIN, GAO FENGGUANG. Camptothecin and cisplatin upregulate ABCG2 and MRP2 expression by activating the ATM/NF-κB pathway in lung cancer cells. Int J Oncol 2013; 42:1289-96. [DOI: 10.3892/ijo.2013.1805] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 01/04/2013] [Indexed: 11/06/2022] Open
|