1
|
Lin K, Zhou E, Shi T, Zhang S, Zhang J, Zheng Z, Pan Y, Gao W, Yu Y. m6A eraser FTO impairs gemcitabine resistance in pancreatic cancer through influencing NEDD4 mRNA stability by regulating the PTEN/PI3K/AKT pathway. J Exp Clin Cancer Res 2023; 42:217. [PMID: 37605223 PMCID: PMC10464189 DOI: 10.1186/s13046-023-02792-0] [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: 03/22/2023] [Accepted: 08/10/2023] [Indexed: 08/23/2023] Open
Abstract
BACKGROUND Gemcitabine resistance has brought great challenges to the treatment of pancreatic cancer. The N6-methyladenosine (m6A) mutation has been shown to have a significant regulatory role in chemosensitivity; however, it is not apparent whether gemcitabine resistance can be regulated by fat mass and obesity-associated protein (FTO). METHODS Cells with established gemcitabine resistance and tissues from pancreatic cancer patients were used to evaluate FTO expression. The biological mechanisms of the effects of FTO on gemcitabine resistant cells were investigated using CCK-8, colony formation assay, flow cytometry, and inhibitory concentration 50. Immunoprecipitation/mass spectrometry, MeRIP-seq, RNA sequencing and RIP assays, RNA stability, luciferase reporter, and RNA pull down assays were employed to examine the mechanism of FTO affecting gemcitabine resistant pancreatic cancer cells. RESULTS The results revealed that FTO was substantially expressed in cells and tissues that were resistant to gemcitabine. Functionally, the gemcitabine resistance of pancreatic cancer could be enhanced by FTO, while its depletion inhibited the growth of gemcitabine resistant tumor cells in vivo. Immunoprecipitation/mass spectrometry showed that the FTO protein can be bound to USP7 and deubiquitinated by USP7, leading to the upregulation of FTO. At the same time, FTO knockdown significantly decreased the expression level of NEDD4 in an m6A-dependent manner. RNA pull down and RNA immunoprecipitation verified YTHDF2 as the reader of NEDD4, which promoted the chemoresistance of gemcitabine resistant cells. FTO knockdown markedly increased the PTEN expression level in an NEDD4-dependent manner and influenced the chemosensitivity to gemcitabine through the PI3K/AKT pathway in pancreatic cancer cells. CONCLUSION In conclusion, we found that gemcitabine resistance in pancreatic cancer can be influenced by FTO that demethylates NEDD4 RNA in a m6A-dependent manner, which then influences the PTEN expression level and thereby affects the PI3K/AKT pathway. We also identified that the FTO level can be upregulated by USP7.
Collapse
Affiliation(s)
- Kai Lin
- Department of Gastrointestinal Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Endi Zhou
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ting Shi
- Department of Hepatobiliary Surgery, The Afliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Siqing Zhang
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jinfan Zhang
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ziruo Zheng
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuetian Pan
- Medical Faculty of Ludwig Maximilians, University of Munich-Munich, Bayern, Germany
| | - Wentao Gao
- Pancreas Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Yabin Yu
- Department of Hepatobiliary Surgery, The Afliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China.
| |
Collapse
|
2
|
Yan C, Niu Y, Li F, Zhao W, Ma L. System analysis based on the pyroptosis-related genes identifies GSDMC as a novel therapy target for pancreatic adenocarcinoma. J Transl Med 2022; 20:455. [PMID: 36199146 PMCID: PMC9533512 DOI: 10.1186/s12967-022-03632-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/07/2022] [Indexed: 11/12/2022] Open
Abstract
Background Pancreatic adenocarcinoma (PAAD) is one of the most common malignant tumors of the digestive tract. Pyroptosis is a newly discovered programmed cell death that highly correlated with the prognosis of tumors. However, the prognostic value of pyroptosis in PAAD remains unclear. Methods A total of 178 pancreatic cancer PAAD samples and 167 normal samples were obtained from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases. The “DESeq2” R package was used to identify differntially expressed pyroptosis-related genes between normal pancreatic samples and PAAD samples. The prognostic model was established in TCGA cohort based on univariate Cox and the least absolute shrinkage and selection operator (LASSO) Cox regression analyses, which was validated in test set from Gene Expression Omnibus (GEO) cohort. Univariate independent prognostic analysis and multivariate independent prognostic analysis were used to determine whether the risk score can be used as an independent prognostic factor to predict the clinicopathological features of PAAD patients. A nomogram was used to predict the survival probability of PAAD patients, which could help in clinical decision-making. The R package "pRRophetic" was applied to calculate the drug sensitivity of each samples from high- and low-risk group. Tumor immune infiltration was investigated using an ESTIMATE algorithm. Finally, the pro‐tumor phenotype of GSDMC was explored in PANC-1 and CFPAC-1 cells. Result On the basis of univariate Cox and LASSO regression analyses, we constructed a risk model with identified five pyroptosis-related genes (IL18, CASP4, NLRP1, GSDMC, and NLRP2), which was validated in the test set. The PAAD samples were divided into high-risk and low-risk groups on the basis of the risk score's median. According to Kaplan Meier curve analysis, samples from high-risk groups had worse outcomes than those from low-risk groups. The time-dependent receiver operating characteristics (ROC) analysis revealed that the risk model could predict the prognosis of PAAD accurately. A nomogram accompanied by calibration curves was presented for predicting 1-, 2-, and 3-year survival in PAAD patients. More importantly, 4 small molecular compounds (A.443654, PD.173074, Epothilone. B, Lapatinib) were identified, which might be potential drugs for the treatment of PAAD patients. Finally, the depletion of GSDMC inhibits the proliferation, invasion, and migration of pancreatic adenocarcinoma cells. Conclusion In this study, we developed a pyroptosis-related prognostic model based on IL18, CASP4, NLRP1, NLRP2, and GSDMC , which may be helpful for clinicians to make clinical decisions for PAAD patients and provide valuable insights for individualized treatment. Our result suggest that GSDMC may promote the proliferation and migration of PAAD cell lines. These findings may provide new insights into the roles of pyroptosis-related genes in PAAD, and offer new therapeutic targets for the treatment of PAAD. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03632-z.
Collapse
Affiliation(s)
- Cheng Yan
- School of Pharmacy, Key Laboratory of Nano-Carbon Modified Film Technology of Henan Province, Diagnostic Laboratory of Animal Diseases, Xinxiang University, Xinxiang, 453000, Henan, China
| | - Yandie Niu
- School of Pharmacy, Key Laboratory of Nano-Carbon Modified Film Technology of Henan Province, Diagnostic Laboratory of Animal Diseases, Xinxiang University, Xinxiang, 453000, Henan, China
| | - Feng Li
- School of Pharmacy, Key Laboratory of Nano-Carbon Modified Film Technology of Henan Province, Diagnostic Laboratory of Animal Diseases, Xinxiang University, Xinxiang, 453000, Henan, China
| | - Wei Zhao
- School of Pharmacy, Key Laboratory of Nano-Carbon Modified Film Technology of Henan Province, Diagnostic Laboratory of Animal Diseases, Xinxiang University, Xinxiang, 453000, Henan, China
| | - Liukai Ma
- School of Pharmacy, Key Laboratory of Nano-Carbon Modified Film Technology of Henan Province, Diagnostic Laboratory of Animal Diseases, Xinxiang University, Xinxiang, 453000, Henan, China.
| |
Collapse
|
3
|
Wu W, Wen K, Zhong Y. Research progress in the establishment of pancreatic cancer models and preclinical applications. CANCER INNOVATION 2022; 1:207-219. [PMID: 38089760 PMCID: PMC10686130 DOI: 10.1002/cai2.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 10/15/2024]
Abstract
Pancreatic cancer (PC) is a highly malignant tumor in the digestive system. The transformation of tissue from normal to pancreatic intraepithelial neoplasm is driven by certain oncogenes, among which the mutation rate of the KRAS gene is as high as 90%. Currently, PC has limited treatment options, low therapeutic effects, and poor prognosis. Thus, more effective methods to combat PC are urgently needed. Some models that can more accurately reflect the biological behaviors and genomic characteristics of PC, such as its morphology, pathology, proliferation, and invasion, are being continuously developed. These include genetic engineering models, orthotopic xenograft models, and heterotopic xenograft models. Using these PC models, scientists have further verified promising drugs and potential therapeutic targets for PC treatment. This is of great significance for limiting the progression of PC with clinical intervention, improving patient outcomes, and improving survival rates.
Collapse
Affiliation(s)
- Weizheng Wu
- Departments of General SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Kunming Wen
- Departments of General SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Yuxin Zhong
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
4
|
Weegh N, Zentrich E, Zechner D, Struve B, Wassermann L, Talbot SR, Kumstel S, Heider M, Vollmar B, Bleich A, Häger C. Voluntary wheel running behaviour as a tool to assess the severity in a mouse pancreatic cancer model. PLoS One 2021; 16:e0261662. [PMID: 34941923 PMCID: PMC8699632 DOI: 10.1371/journal.pone.0261662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 12/08/2021] [Indexed: 01/07/2023] Open
Abstract
Laboratory animals frequently undergo routine experimental procedures such as handling, restraining and injections. However, as a known source of stress, these procedures potentially impact study outcome and data quality. In the present study, we, therefore, performed an evidence-based severity assessment of experimental procedures used in a pancreatic cancer model including surgical tumour induction and subsequent chemotherapeutic treatment via repeated intraperitoneal injections. Cancer cell injection into the pancreas was performed during a laparotomy under general anaesthesia. After a four-day recovery phase, mice received either drug treatment (galloflavin and metformin) or the respective vehicle substances via daily intraperitoneal injections. In addition to clinical scoring, an automated home-cage monitoring system was used to assess voluntary wheel running (VWR) behaviour as an indicator of impaired well-being. After surgery, slightly elevated clinical scores and minimal body weight reductions, but significantly decreased VWR behaviour were observed. During therapy, body weight declined in response to chemotherapy, but not after vehicle substance injection, while VWR activity was decreased in both cases. VWR behaviour differed between treatment groups and revealed altered nightly activity patterns. In summary, by monitoring VWR a high impact of repeated injections on the well-being of mice was revealed and substance effects on well-being were distinguishable. However, no differences in tumour growth between treatment groups were observed. This might be due to the severity of the procedures uncovered in this study, as exaggerated stress responses are potentially confounding factors in preclinical studies. Finally, VWR was a more sensitive indicator of impairment than clinical scoring in this model.
Collapse
Affiliation(s)
- Nora Weegh
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Eva Zentrich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Dietmar Zechner
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center, Rostock, Germany
| | - Birgitta Struve
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Laura Wassermann
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Steven Roger Talbot
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Simone Kumstel
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center, Rostock, Germany
| | - Miriam Heider
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Brigitte Vollmar
- Rudolf-Zenker-Institute of Experimental Surgery, University Medical Center, Rostock, Germany
| | - André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Christine Häger
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
5
|
Miquel M, Zhang S, Pilarsky C. Pre-clinical Models of Metastasis in Pancreatic Cancer. Front Cell Dev Biol 2021; 9:748631. [PMID: 34778259 PMCID: PMC8578999 DOI: 10.3389/fcell.2021.748631] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a hostile solid malignancy coupled with an extremely high mortality rate. Metastatic disease is already found in most patients at the time of diagnosis, resulting in a 5-year survival rate below 5%. Improved comprehension of the mechanisms leading to metastasis is pivotal for the development of new targeted therapies. A key field to be improved are modeling strategies applied in assessing cancer progression, since traditional platforms fail in recapitulating the complexity of PDAC. Consequently, there is a compelling demand for new preclinical models that mirror tumor progression incorporating the pressure of the immune system, tumor microenvironment, as well as molecular aspects of PDAC. We suggest the incorporation of 3D organoids derived from genetically engineered mouse models or patients as promising new tools capable to transform PDAC pre-clinical modeling and access new frontiers in personalized medicine.
Collapse
Affiliation(s)
- Maria Miquel
- Department of Surgery, University Hospital, Erlangen, Germany
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Shuman Zhang
- Department of Surgery, University Hospital, Erlangen, Germany
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Pilarsky
- Department of Surgery, University Hospital, Erlangen, Germany
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
6
|
Kim CJ, Terado T, Tambe Y, Mukaisho KI, Kageyama S, Kawauchi A, Inoue H. Cryptotanshinone, a novel PDK 4 inhibitor, suppresses bladder cancer cell invasiveness via the mTOR/β‑catenin/N‑cadherin axis. Int J Oncol 2021; 59:40. [PMID: 33982789 PMCID: PMC8131085 DOI: 10.3892/ijo.2021.5220] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/19/2021] [Indexed: 12/13/2022] Open
Abstract
The phosphorylation of pyruvate dehydrogenase (PDH) by pyruvate dehydrogenase kinase (PDK) 4 inhibits its ability to induce a glycolytic shift. PDK4 expression is upregulated in various types of human cancer. Because PDK4 regulation is critical for metabolic changes in cancer cells, it is an attractive target for cancer therapy given its ability to shift glucose metabolism. It was previously shown that a novel PDK4 inhibitor, cryptotanshinone (CPT), suppressed the three‑dimensional (3D)‑spheroid formation of pancreatic and colorectal cancer cells. In the present study, the effects of CPT on the invasiveness of bladder cancer cells were investigated. CPT significantly suppressed the invasiveness and 3D‑spheroid formation of T24 and J82 bladder cancer cells. CPT also suppressed the phosphorylation of PDH and β‑catenin, as well as the expression of N‑cadherin, which are all critical for inducing epithelial‑mesenchymal transition (EMT). The knockdown of β‑catenin or PDK4 using specific small interfering RNAs suppressed N‑cadherin expression and invasiveness in T24 cells. An mTOR inhibitor also suppressed the phosphorylation of β‑catenin and N‑cadherin expression. Furthermore, CPT injection significantly suppressed pancreatic tumor growth and peritoneal dissemination of highly metastatic SUIT‑2 pancreatic cancer cells in a mouse orthotopic pancreatic cancer model, without evident toxicity. Moreover, immunohistochemistry analyses demonstrated decreased β‑catenin expression in CPT‑treated pancreatic tumors compared with control tumors. Taken together, these results indicate that CPT reduced the invasiveness and metastasis of bladder cancer cells by suppressing EMT via the mTOR/β‑catenin/N‑cadherin pathway.
Collapse
Affiliation(s)
- Chul Jang Kim
- Department of Urology, Kohka Public Hospital, Minakuchi-cho, Kohka, Shiga 528-0074, Japan
- Department of Urology, Shiga University of Medical Science, Setatsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Tokio Terado
- Department of Stem Cell Biology and Regenerative Medicine, Shiga University of Medical Science, Setatsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Yukihiro Tambe
- Division of Microbiology and Infectious Diseases, Shiga University of Medical Science, Setatsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Ken-Ichi Mukaisho
- Division of Human Pathology, Shiga University of Medical Science, Setatsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Susumu Kageyama
- Department of Urology, Shiga University of Medical Science, Setatsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Akihiro Kawauchi
- Department of Urology, Shiga University of Medical Science, Setatsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Hirokazu Inoue
- Division of Microbiology and Infectious Diseases, Shiga University of Medical Science, Setatsukinowa-cho, Otsu, Shiga 520-2192, Japan
| |
Collapse
|
7
|
Matsumoto T, Komori T, Yoshino Y, Ioroi T, Kitahashi T, Kitahara H, Ono K, Higuchi T, Sakabe M, Kori H, Kano M, Hori R, Kato Y, Hagiwara S. A Liposomal Gemcitabine, FF-10832, Improves Plasma Stability, Tumor Targeting, and Antitumor Efficacy of Gemcitabine in Pancreatic Cancer Xenograft Models. Pharm Res 2021; 38:1093-1106. [PMID: 33961188 PMCID: PMC8217058 DOI: 10.1007/s11095-021-03045-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 04/15/2021] [Indexed: 01/02/2023]
Abstract
Purpose The clinical application of gemcitabine (GEM) is limited by its pharmacokinetic properties. The aim of this study was to characterize the stability in circulating plasma, tumor targeting, and payload release of liposome-encapsulated GEM, FF-10832. Methods Antitumor activity was assessed in xenograft mouse models of human pancreatic cancer. The pharmacokinetics of GEM and its active metabolite dFdCTP were also evaluated. Results In mice with Capan-1 tumors, the dose-normalized areas under the curve (AUCs) after FF-10832 administration in plasma and tumor were 672 and 1047 times higher, respectively, than after using unencapsulated GEM. The tumor-to-bone marrow AUC ratio of dFdCTP was approximately eight times higher after FF-10832 administration than after GEM administration. These results indicated that liposomal encapsulation produced long-term stability in circulating plasma and tumor-selective targeting of GEM. In mice with Capan-1, SUIT-2, and BxPC-3 tumors, FF-10832 had better antitumor activity and tolerability than GEM. Internalization of FF-10832 in tumor-associated macrophages (TAMs) was revealed by flow cytometry and confocal laser scanning microscopy, and GEM was efficiently released from isolated macrophages of mice treated with FF-10832. These results suggest that TAMs are one of the potential reservoirs of GEM in tumors. Conclusion This study found that FF-10832 had favorable pharmacokinetic properties. The liposomal formulation was more effective and tolerable than unencapsulated GEM in mouse xenograft tumor models. Hence, FF-10832 is a promising candidate for the treatment of pancreatic cancer. Supplementary Information The online version contains supplementary material available at 10.1007/s11095-021-03045-5.
Collapse
Affiliation(s)
- Takeshi Matsumoto
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan.
| | - Takashi Komori
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Yuta Yoshino
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Tadaaki Ioroi
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Tsukasa Kitahashi
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Hiromu Kitahara
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Kohei Ono
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Tamami Higuchi
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Masayo Sakabe
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Hiroshi Kori
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Masahiro Kano
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| | - Ritsuko Hori
- Analysis Technology Center, FUJIFILM Corporation, Nakanuma 210, Minamiashigara, Kanagawa, 250-0193, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Shinji Hagiwara
- Bioscience and Engineering laboratories, FUJIFILM Corporation, 577 Ushijima, Kaisei-machi, Ashigarakami-gun, Kanagawa, 258-8577, Japan
| |
Collapse
|
8
|
Fernandes S, Fernandez T, Metze S, Balakrishnan PB, Mai BT, Conteh J, De Mei C, Turdo A, Di Franco S, Stassi G, Todaro M, Pellegrino T. Magnetic Nanoparticle-Based Hyperthermia Mediates Drug Delivery and Impairs the Tumorigenic Capacity of Quiescent Colorectal Cancer Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2021; 13:15959-15972. [PMID: 33797220 PMCID: PMC8045020 DOI: 10.1021/acsami.0c21349] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/15/2021] [Indexed: 05/27/2023]
Abstract
Cancer stem cells (CSCs) are the tumor cell subpopulation responsible for resistance to chemotherapy, tumor recurrence, and metastasis. An efficient therapy must act on low proliferating quiescent-CSCs (q-CSCs). We here investigate the effect of magnetic hyperthermia (MHT) in combination with local chemotherapy as a dual therapy to inhibit patient-derived colorectal qCR-CSCs. We apply iron oxide nanocubes as MHT heat mediators, coated with a thermoresponsive polymer (TR-Cubes) and loaded with DOXO (TR-DOXO) as a chemotherapeutic agent. The thermoresponsive polymer releases DOXO only at a temperature above 44 °C. In colony-forming assays, the cells exposed to TR-Cubes with MHT reveal that qCR-CSCs struggle to survive the heat damage and, with a due delay, restart the division of dormant cells. The eradication of qCR-CSCs with a complete stop of the colony formation was achieved only with TR-DOXO when exposed to MHT. The in vivo tumor formation study confirms the combined effects of MHT with heat-mediated drug release: only the group of animals that received the CR-CSCs pretreated, in vitro, with TR-DOXO and MHT lacked the formation of tumor even after several months. For DOXO-resistant CR-CSCs cells, the same results were shown, in vitro, when choosing the drug oxaliplatin rather than DOXO and applying MHT. These findings emphasize the potential of our nanoplatforms as an effective patient-personalized cancer treatment against qCR-CSCs.
Collapse
Affiliation(s)
- Soraia Fernandes
- Istituto
Italiano di Tecnologia (IIT), via Morego 30, 16163 Genova, Italy
| | - Tamara Fernandez
- Istituto
Italiano di Tecnologia (IIT), via Morego 30, 16163 Genova, Italy
| | - Sabrina Metze
- Istituto
Italiano di Tecnologia (IIT), via Morego 30, 16163 Genova, Italy
| | | | - Binh T. Mai
- Istituto
Italiano di Tecnologia (IIT), via Morego 30, 16163 Genova, Italy
| | - John Conteh
- Istituto
Italiano di Tecnologia (IIT), via Morego 30, 16163 Genova, Italy
| | - Claudia De Mei
- Istituto
Italiano di Tecnologia (IIT), via Morego 30, 16163 Genova, Italy
| | - Alice Turdo
- PROMISE
Department,Piazza delle Cliniche 2, University
of Palermo, 90133 Palermo, Italy
| | - Simone Di Franco
- DICHIRONS
Department, University of Palermo, Via del Vespro 129, 90133 Palermo, Italy
| | - Giorgio Stassi
- DICHIRONS
Department, University of Palermo, Via del Vespro 129, 90133 Palermo, Italy
| | - Matilde Todaro
- PROMISE
Department,Piazza delle Cliniche 2, University
of Palermo, 90133 Palermo, Italy
| | - Teresa Pellegrino
- Istituto
Italiano di Tecnologia (IIT), via Morego 30, 16163 Genova, Italy
| |
Collapse
|
9
|
Onaciu A, Munteanu R, Munteanu VC, Gulei D, Raduly L, Feder RI, Pirlog R, Atanasov AG, Korban SS, Irimie A, Berindan-Neagoe I. Spontaneous and Induced Animal Models for Cancer Research. Diagnostics (Basel) 2020; 10:E660. [PMID: 32878340 PMCID: PMC7555044 DOI: 10.3390/diagnostics10090660] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 08/24/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022] Open
Abstract
Considering the complexity of the current framework in oncology, the relevance of animal models in biomedical research is critical in light of the capacity to produce valuable data with clinical translation. The laboratory mouse is the most common animal model used in cancer research due to its high adaptation to different environments, genetic variability, and physiological similarities with humans. Beginning with spontaneous mutations arising in mice colonies that allow for pursuing studies of specific pathological conditions, this area of in vivo research has significantly evolved, now capable of generating humanized mice models encompassing the human immune system in biological correlation with human tumor xenografts. Moreover, the era of genetic engineering, especially of the hijacking CRISPR/Cas9 technique, offers powerful tools in designing and developing various mouse strains. Within this article, we will cover the principal mouse models used in oncology research, beginning with behavioral science of animals vs. humans, and continuing on with genetically engineered mice, microsurgical-induced cancer models, and avatar mouse models for personalized cancer therapy. Moreover, the area of spontaneous large animal models for cancer research will be briefly presented.
Collapse
Affiliation(s)
- Anca Onaciu
- Research Center for Advanced Medicine - Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.O.); (R.M.); (R.-I.F.)
| | - Raluca Munteanu
- Research Center for Advanced Medicine - Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.O.); (R.M.); (R.-I.F.)
| | - Vlad Cristian Munteanu
- Department of Urology, The Oncology Institute “Prof Dr. Ion Chiricuta”, 400015 Cluj-Napoca, Romania;
- Department of Anatomy and Embryology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Diana Gulei
- Research Center for Advanced Medicine - Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.O.); (R.M.); (R.-I.F.)
| | - Lajos Raduly
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (L.R.); (R.P.)
| | - Richard-Ionut Feder
- Research Center for Advanced Medicine - Medfuture, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (A.O.); (R.M.); (R.-I.F.)
| | - Radu Pirlog
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (L.R.); (R.P.)
- Department of Morphological Sciences, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Atanas G. Atanasov
- Ludwig Boltzmann Institute for Digital Health and Patient Safety, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria;
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, 05-552 Magdalenka, Poland
- Institute of Neurobiology, Bulgarian Academy of Sciences, 23 Acad. G. Bonchev str., 1113 Sofia, Bulgaria
- Department of Pharmacognosy, University of Vienna, 1090 Vienna, Austria
| | - Schuyler S. Korban
- Department of Natural Resources and Environmental Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA;
| | - Alexandru Irimie
- 11th Department of Surgical Oncology and Gynaecological Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania;
- Department of Surgery, The Oncology Institute Prof. Dr. Ion Chiricuta, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 23 Marinescu Street, 400337 Cluj-Napoca, Romania; (L.R.); (R.P.)
- Department of Functional Genomics and Experimental Pathology, The Oncology Institute “Prof. Dr. Ion Chiricuta”, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
| |
Collapse
|
10
|
Naito T, Higuchi T, Shimada Y, Kakinuma C. An improved mouse orthotopic bladder cancer model exhibiting progression and treatment response characteristics of human recurrent bladder cancer. Oncol Lett 2020; 19:833-839. [PMID: 31885717 PMCID: PMC6924206 DOI: 10.3892/ol.2019.11172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 09/10/2019] [Indexed: 01/03/2023] Open
Abstract
Nonmuscle-invasive (superficial) bladder cancer is generally treated via surgical removal, followed by adjuvant therapy (bacillus Calmette-Guerin). However, bladder cancer can often recur, and in a substantial number of recurrent cases, the cancer progresses and metastasizes. Furthermore, residual microtumors following excision may lead to an increased risk of recurrence. An in vivo model mimicking the pattern of urinary bladder microtumor regrowth may provide an effective experimental system for improving postsurgical treatment outcomes. A mouse bladder cancer model established using orthotopic transplant of UM-UC-3 human urinary bladder carcinoma cells has been established, however, to the best of our knowledge, no report has investigated sequential histological changes, including early-phase changes and treatment responses in bladder cancer. In the present study, the efficiency of the model was optimized and the sequential changes were examined using histopathology and in situ imaging. The therapeutic effects of cisplatin (CDDP) and gemcitabine (GEM) were also examined, which are drugs that are often used for follow-up chemotherapy. Tumor-seeding efficiency reached 90-100%, with muscle layer and bladder lumen invasion occurring in ~21 days, using the following modifications: i) Shallow catheter insertion to mitigate bladder wall damage; ii) bladder pretreatment using prewarmed trypsin, followed by light urethral clamping and body temperature maintenance for more efficient removal of transitional epithelium; and iii) seeding with UM-UC-3 cells (rather than HT1376, 5637 or T24 tumor cells) in a medium supplemented with Matrigel. Transplant with UM-UC-3 cells resulted in isolated microlesions that progressed into tumors, invading the bladder lumen and muscle layer to the serosal surface. Tumor growth was markedly reduced by weekly intravenous injections of CDDP and partially suppressed by GEM. Therefore, this model is reliable, and pathological progression and treatment responses recapitulate the features of recurrent human bladder cancer.
Collapse
Affiliation(s)
- Tomoharu Naito
- Pharmaceutical and Healthcare Research Laboratories, FUJIFILM Corporation, Ashigarakami-gun, Kanagawa 258-8577, Japan
- Department of Human Pathology, School of Medicine, Juntendo University, Tokyo 113-0033, Japan
| | - Tamami Higuchi
- Pharmaceutical and Healthcare Research Laboratories, FUJIFILM Corporation, Ashigarakami-gun, Kanagawa 258-8577, Japan
- Department of Oncology Clinical Development, Gunma University, Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Yasuhiro Shimada
- Pharmaceutical and Healthcare Research Laboratories, FUJIFILM Corporation, Ashigarakami-gun, Kanagawa 258-8577, Japan
- Department of Human Pathology, School of Medicine, Juntendo University, Tokyo 113-0033, Japan
| | - Chihaya Kakinuma
- Pharmaceutical and Healthcare Research Laboratories, FUJIFILM Corporation, Ashigarakami-gun, Kanagawa 258-8577, Japan
- Department of Human Pathology, School of Medicine, Juntendo University, Tokyo 113-0033, Japan
| |
Collapse
|
11
|
Rezaee M, Wang J, Razavi M, Ren G, Zheng F, Hussein A, Ullah M, Thakor AS. A Study Comparing the Effects of Targeted Intra-Arterial and Systemic Chemotherapy in an Orthotopic Mouse Model of Pancreatic Cancer. Sci Rep 2019; 9:15929. [PMID: 31685925 PMCID: PMC6828954 DOI: 10.1038/s41598-019-52490-1] [Citation(s) in RCA: 6] [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: 04/16/2019] [Accepted: 10/18/2019] [Indexed: 02/06/2023] Open
Abstract
Systemic chemotherapy is the first line treatment for patients with unresectable pancreatic cancer, however, insufficient drug delivery to the pancreas is a major problem resulting in poor outcomes. We evaluated the therapeutic effects of targeted intra-arterial (IA) delivery of gemcitabine directly into the pancreas in an orthotopic mouse model of pancreatic cancer. Nude mice with orthotopic pancreatic tumors were randomly assigned into 3 groups receiving gemcitabine: systemic intravenous (IV) injection (low: 0.3 mg/kg and high: 100 mg/kg) and direct IA injection (0.3 mg/kg). Treatments were administered weekly for 2 weeks. IA treatment resulted in a significantly greater reduction in tumor growth compared to low IV treatment. To achieve a comparable reduction in tumor growth as seen with IA treatment, gemcitabine had to be given IV at over 300x the dose (high IV treatment) which was associated with some toxicity. After 2 weeks, tumor samples from animals treated with IA gemcitabine had significantly lower residual cancer cells, higher cellular necrosis and evidence of increased apoptosis when compared to animals treated with low IV gemcitabine. Our study shows targeted IA injection of gemcitabine directly into the pancreas, via its arterial blood supply, has a superior therapeutic effect in reducing tumor growth compared to the same concentration administered by conventional systemic injection.
Collapse
MESH Headings
- Administration, Intravenous
- Animals
- Antimetabolites, Antineoplastic/adverse effects
- Antimetabolites, Antineoplastic/therapeutic use
- Cell Line, Tumor
- Deoxycytidine/adverse effects
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/therapeutic use
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Drug Administration Schedule
- Female
- Humans
- Infusions, Intra-Arterial
- Male
- Mice
- Mice, Nude
- Neoplasm, Residual
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Transplantation, Heterologous
- Gemcitabine
Collapse
Affiliation(s)
- Melika Rezaee
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
- Chicago Medical School, Rosalind Franklin University, North Chicago, Illinois, 60064, USA
| | - Jing Wang
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Mehdi Razavi
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Gang Ren
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Fengyan Zheng
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Ahmed Hussein
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Mujib Ullah
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA.
| |
Collapse
|
12
|
Cao X, Hu Y, Luo S, Wang Y, Gong T, Sun X, Fu Y, Zhang Z. Neutrophil-mimicking therapeutic nanoparticles for targeted chemotherapy of pancreatic carcinoma. Acta Pharm Sin B 2019; 9:575-589. [PMID: 31193785 PMCID: PMC6543032 DOI: 10.1016/j.apsb.2018.12.009] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 10/26/2018] [Accepted: 12/20/2018] [Indexed: 12/20/2022] Open
Abstract
Due to the critical correlation between inflammation and carcinogenesis, a therapeutic candidate with anti-inflammatory activity may find application in cancer therapy. Here, we report the therapeutic efficacy of celastrol as a promising candidate compound for treatment of pancreatic carcinoma via naïve neutrophil membrane-coated poly(ethylene glycol) methyl ether-block-poly(lactic-co-glycolic acid) (PEG-PLGA) nanoparticles. Neutrophil membrane-coated nanoparticles (NNPs) are well demonstrated to overcome the blood pancreas barrier to achieve pancreas-specific drug delivery in vivo. Using tumor-bearing mice xenograft model, NNPs showed selective accumulations at the tumor site following systemic administration as compared to nanoparticles without neutrophil membrane coating. In both orthotopic and ectopic tumor models, celastrol-loaded NNPs demonstrated greatly enhanced tumor inhibition which significantly prolonged the survival of tumor bearing mice and minimizing liver metastases. Overall, these results suggest that celastrol-loaded NNPs represent a viable and effective treatment option for pancreatic carcinoma.
Collapse
Key Words
- 5-FU, fluorouracil
- CLT, celastrol
- Celastrol
- DAPI, 4′,6-diamidino-2-phenylindole
- DiD, 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindodicarbocyanine perchlorate
- IKKα, IκB kinase α
- IKKβ, IκB kinase β
- IL-1β, interleukin 1 beta
- IL-6, interleukin 6
- Inflammation
- NF-κB, nuclear factor kappa B
- NIK, NF kappa B inducing kinase
- NNPs, neutrophil membrane-coated nanoparticles
- NPs, nanoparticles without neutrophil membrane coating
- Naïve neutrophils membrane
- PEG-PLGA nanoparticle
- PEG-PLGA, poly(ethylene glycol) methyl ether-block-poly(lactic-co-glycolic acid)
- PI, propidium iodide
- Pancreatic carcinoma
- TAK1, TGF-β-activated kinase 1
- TEM, transmission electronic microscopy
- TNF-α, tumor necrosis factor alpha
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yao Fu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|