1
|
Gao W, Wang J, Xu Y, Yu H, Yi S, Bai C, Cong Q, Zhu Y. Research progress in the metabolic reprogramming of hepatocellular carcinoma (Review). Mol Med Rep 2024; 30:131. [PMID: 38818815 PMCID: PMC11148525 DOI: 10.3892/mmr.2024.13255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/03/2024] [Indexed: 06/01/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver malignancy and its morbidity is increasing worldwide due to increasing prevalence. Metabolic reprogramming has been recognized as a hallmark of cancer and serves a role in cancer progression. Glucose, lipids and amino acids are three major components whose altered metabolism can directly affect the energy production of cells, including liver cancer cells. Nutrients and energy are indispensable for the growth and proliferation of cancer cells, thus altering the metabolism of hepatoma cells can inhibit the progression of HCC. The present review summarizes recent studies on tumour regulatory molecules, including numerous noncoding RNAs, oncogenes and tumour suppressors, which regulate the metabolic activities of glucose, lipids and amino acids by targeting key enzymes, signalling pathways or interactions between the two. These regulatory molecules can regulate the rapid proliferation of cancer cells, tumour progression and treatment resistance. It is thought that these tumour regulatory factors may serve as therapeutic targets or valuable biomarkers for HCC, with the potential to mitigate HCC drug resistance. Furthermore, the advantages and disadvantages of metabolic inhibitors as a treatment approach for HCC, as well as possible solutions are discussed, providing insights for developing more effective treatment strategies for HCC.
Collapse
Affiliation(s)
- Wenyue Gao
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R China
| | - Jing Wang
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R China
| | - Yuting Xu
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R China
| | - Hongbo Yu
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R China
| | - Sitong Yi
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R China
| | - Changchuan Bai
- Internal Department of Chinese Medicine, Dalian Hospital of Traditional Chinese Medicine, Dalian, Liaoning 116000, P.R China
| | - Qingwei Cong
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R China
| | - Ying Zhu
- Department of Infectious Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116000, P.R China
| |
Collapse
|
2
|
Wang W, Li Y, Lin K, Wang X, Tu Y, Zhuo Z. Progress in building clinically relevant patient-derived tumor xenograft models for cancer research. Animal Model Exp Med 2023; 6:381-398. [PMID: 37679891 PMCID: PMC10614132 DOI: 10.1002/ame2.12349] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/03/2023] [Indexed: 09/09/2023] Open
Abstract
Patient-derived tumor xenograft (PDX) models, a method involving the surgical extraction of tumor tissues from cancer patients and subsequent transplantation into immunodeficient mice, have emerged as a pivotal approach in translational research, particularly in advancing precision medicine. As the first stage of PDX development, the patient-derived orthotopic xenograft (PDOX) models implant tumor tissue in mice in the corresponding anatomical locations of the patient. The PDOX models have several advantages, including high fidelity to the original tumor, heightened drug sensitivity, and an elevated rate of successful transplantation. However, the PDOX models present significant challenges, requiring advanced surgical techniques and resource-intensive imaging technologies, which limit its application. And then, the humanized mouse models, as well as the zebrafish models, were developed. Humanized mouse models contain a human immune environment resembling the tumor and immune system interplay. The humanized mouse models are a hot topic in PDX model research. Regarding zebrafish patient-derived tumor xenografts (zPDX) and patient-derived organoids (PDO) as promising models for studying cancer and drug discovery, zPDX models are used to transplant tumors into zebrafish as novel personalized medical animal models with the advantage of reducing patient waiting time. PDO models provide a cost-effective approach for drug testing that replicates the in vivo environment and preserves important tumor-related information for patients. The present review highlights the functional characteristics of each new phase of PDX and provides insights into the challenges and prospective developments in this rapidly evolving field.
Collapse
Affiliation(s)
- Weijing Wang
- Department of Clinical MedicineShantou University Medical CollegeShantouChina
| | - Yongshu Li
- College of Life SciencesHubei Normal UniversityHuangshiChina
- Shenzhen Institute for Technology InnovationNational Institute of MetrologyShenzhenChina
| | - Kaida Lin
- Department of Clinical MedicineShantou University Medical CollegeShantouChina
| | - Xiaokang Wang
- Department of PharmacyShenzhen Longhua District Central HospitalShenzhenChina
| | - Yanyang Tu
- Research Center, Huizhou Central People's HospitalGuangdong Medical UniversityHuizhou CityChina
| | - Zhenjian Zhuo
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
- Laboratory Animal Center, School of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate SchoolShenzhenChina
| |
Collapse
|
3
|
Nong S, Han X, Xiang Y, Qian Y, Wei Y, Zhang T, Tian K, Shen K, Yang J, Ma X. Metabolic reprogramming in cancer: Mechanisms and therapeutics. MedComm (Beijing) 2023; 4:e218. [PMID: 36994237 PMCID: PMC10041388 DOI: 10.1002/mco2.218] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 01/22/2023] [Accepted: 01/30/2023] [Indexed: 03/29/2023] Open
Abstract
Cancer cells characterized by uncontrolled growth and proliferation require altered metabolic processes to maintain this characteristic. Metabolic reprogramming is a process mediated by various factors, including oncogenes, tumor suppressor genes, changes in growth factors, and tumor-host cell interactions, which help to meet the needs of cancer cell anabolism and promote tumor development. Metabolic reprogramming in tumor cells is dynamically variable, depending on the tumor type and microenvironment, and reprogramming involves multiple metabolic pathways. These metabolic pathways have complex mechanisms and involve the coordination of various signaling molecules, proteins, and enzymes, which increases the resistance of tumor cells to traditional antitumor therapies. With the development of cancer therapies, metabolic reprogramming has been recognized as a new therapeutic target for metabolic changes in tumor cells. Therefore, understanding how multiple metabolic pathways in cancer cells change can provide a reference for the development of new therapies for tumor treatment. Here, we systemically reviewed the metabolic changes and their alteration factors, together with the current tumor regulation treatments and other possible treatments that are still under investigation. Continuous efforts are needed to further explore the mechanism of cancer metabolism reprogramming and corresponding metabolic treatments.
Collapse
Affiliation(s)
- Shiqi Nong
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Xiaoyue Han
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Yu Xiang
- Department of BiotherapyCancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| | - Yuran Qian
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Yuhao Wei
- Department of Clinical MedicineWest China School of MedicineWest China HospitalSichuan UniversityChengduSichuanChina
| | - Tingyue Zhang
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Keyue Tian
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
| | - Kai Shen
- Department of OncologyFirst Affiliated Hospital of Nanjing Medical UniversityNanjingChina
| | - Jing Yang
- State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Xuelei Ma
- State Key Laboratory of Oral DiseasesWest China Hospital of StomatologyWest China School of StomatologyNational Clinical Research Center for Oral DiseasesSichuan UniversityChengduSichuanChina
- Department of Biotherapy and Cancer CenterState Key Laboratory of BiotherapyCancer CenterWest China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|
4
|
Wang D, Wan X. Progress in research on the role of amino acid metabolic reprogramming in tumour therapy: A review. Biomed Pharmacother 2022; 156:113923. [DOI: 10.1016/j.biopha.2022.113923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/16/2022] [Accepted: 10/24/2022] [Indexed: 11/26/2022] Open
|
5
|
Zhao L, Su H, Liu X, Wang H, Feng Y, Wang Y, Chen H, Dai L, Lai S, Xu S, Li C, Hao J, Tang B. mTORC1-c-Myc pathway rewires methionine metabolism for HCC progression through suppressing SIRT4 mediated ADP ribosylation of MAT2A. Cell Biosci 2022; 12:183. [DOI: 10.1186/s13578-022-00919-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/24/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
Exploiting cancer metabolism during nutrient availability holds immense potential for the clinical and therapeutic benefits of hepatocellular carcinoma (HCC) patients. Dietary methionine is a metabolic dependence of cancer development, but how the signal transduction integrates methionine status to achieve the physiological demand of cancer cells remains unknown.
Methods
Low or high levels of dietary methionine was fed to mouse models with patient-derived xenograft or diethyl-nitrosamine induced liver cancer. RNA sequence and metabolomics were performed to reveal the profound effect of methionine restriction on gene expression and metabolite changes. Immunostaining, sphere formation assays, in vivo tumourigenicity, migration and self-renewal ability were conducted to demonstrate the efficacy of methionine restriction and sorafenib.
Results
We discovered that mTORC1-c-Myc-SIRT4 axis was abnormally regulated in a methionine-dependent manner and affected the HCC progression. c-Myc rewires methionine metabolism through TRIM32 mediated degradation of SIRT4, which regulates MAT2A activity by ADP-ribosylation on amino acid residue glutamic acid 111. MAT2A is a key enzyme to generate S-adenosylmethionine (SAM). Loss of SIRT4 activates MAT2A, thereby increasing SAM level and dynamically regulating gene expression, which triggers the high proliferation rate of tumour cells. SIRT4 exerts its tumour suppressive function with targeted therapy (sorafenib) by affecting methionine, redox and nucleotide metabolism.
Conclusions
These findings establish a novel characterization of the signaling transduction and the metabolic consequences of dietary methionine restriction in malignant liver tissue of mice. mTORC1, c-Myc, SIRT4 and ADP ribosylation site of MAT2A are promising clinical and therapeutic targets for the HCC treatment.
Collapse
|
6
|
Yamamoto J, Inubushi S, Han Q, Tashiro Y, Sugisawa N, Hamada K, Aoki Y, Miyake K, Matsuyama R, Bouvet M, Clarke SG, Endo I, Hoffman RM. Linkage of methionine addiction, histone lysine hypermethylation, and malignancy. iScience 2022; 25:104162. [PMID: 35434545 PMCID: PMC9010622 DOI: 10.1016/j.isci.2022.104162] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 02/19/2022] [Accepted: 03/23/2022] [Indexed: 11/25/2022] Open
Abstract
Methionine addiction, found in all types of cancer investigated, is because of the overuse of methionine by cancer cells for excess transmethylation reactions. In the present study, we compared the histone H3 lysine-methylation status and degree of malignancy between methionine-addicted cancer cells and their isogenic methionine-independent revertants, selected by their growth in low concentration of methionine. The methionine-independent revertans can grow on low levels of methionine or independently of exogenous methionine using methionine precursors, as do normal cells. In the methionine-independent revertants, the excess levels of trimethylated histone H3 lysine marks found in the methionine-addicted parental cancer cells were reduced or lost, and their tumorigenicity and experimental metastatic potential in nude mice were also highly reduced. Methionine addiction of cancer is linked with malignancy and hypermethylation of histone H3 lysines. The results of the present study thus provide a unique framework to further understand a fundamental basis of malignancy. Methionine(MET)-independent revertants were selected from MET-addicted cancer cells MET-independent revertants had greatly reduced malignancy MET-independent revertants have lost or reduced methylation of H3 lysine marks MET addiction, malignancy, and hypermethylated H3 lysine marks are linked
Collapse
Affiliation(s)
- Jun Yamamoto
- AntiCancer Inc, 7917 Ostrow St, San Diego, CA 92111, USA.,Department of Surgery, University of California, San Diego, 9300 Campus Point Drive #7220, La Jolla, CA 92037-7220, USA.,Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Sachiko Inubushi
- AntiCancer Inc, 7917 Ostrow St, San Diego, CA 92111, USA.,Department of Surgery, University of California, San Diego, 9300 Campus Point Drive #7220, La Jolla, CA 92037-7220, USA
| | - Qinghong Han
- AntiCancer Inc, 7917 Ostrow St, San Diego, CA 92111, USA
| | - Yoshihiko Tashiro
- AntiCancer Inc, 7917 Ostrow St, San Diego, CA 92111, USA.,Department of Surgery, University of California, San Diego, 9300 Campus Point Drive #7220, La Jolla, CA 92037-7220, USA
| | - Norihiko Sugisawa
- AntiCancer Inc, 7917 Ostrow St, San Diego, CA 92111, USA.,Department of Surgery, University of California, San Diego, 9300 Campus Point Drive #7220, La Jolla, CA 92037-7220, USA
| | - Kazuyuki Hamada
- AntiCancer Inc, 7917 Ostrow St, San Diego, CA 92111, USA.,Department of Surgery, University of California, San Diego, 9300 Campus Point Drive #7220, La Jolla, CA 92037-7220, USA
| | - Yusuke Aoki
- AntiCancer Inc, 7917 Ostrow St, San Diego, CA 92111, USA.,Department of Surgery, University of California, San Diego, 9300 Campus Point Drive #7220, La Jolla, CA 92037-7220, USA
| | - Kentaro Miyake
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Ryusei Matsuyama
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Michael Bouvet
- Department of Surgery, University of California, San Diego, 9300 Campus Point Drive #7220, La Jolla, CA 92037-7220, USA
| | - Steven G Clarke
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095-1569, USA
| | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | - Robert M Hoffman
- AntiCancer Inc, 7917 Ostrow St, San Diego, CA 92111, USA.,Department of Surgery, University of California, San Diego, 9300 Campus Point Drive #7220, La Jolla, CA 92037-7220, USA
| |
Collapse
|
7
|
PET imaging of pancreatic cancer. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00207-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
8
|
Yao J, Yang M, Atteh L, Liu P, Mao Y, Meng W, Li X. A pancreas tumor derived organoid study: from drug screen to precision medicine. Cancer Cell Int 2021; 21:398. [PMID: 34315500 PMCID: PMC8314636 DOI: 10.1186/s12935-021-02044-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 06/24/2021] [Indexed: 12/17/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) one of the deadliest malignant tumor. Despite considerable progress in pancreatic cancer treatment in the past 10 years, PDAC mortality has shown no appreciable change, and systemic therapies for PDAC generally lack efficacy. Thus, developing biomarkers for treatment guidance is urgently required. This review focuses on pancreatic tumor organoids (PTOs), which can mimic the characteristics of the original tumor in vitro. As a powerful tool with several applications, PTOs represent a new strategy for targeted therapy in pancreatic cancer and contribute to the advancement of the field of personalized medicine.
Collapse
Affiliation(s)
- Jia Yao
- Key Laboratory of Biological Therapy and Regenerative Medicine Transformation of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Man Yang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Lawrence Atteh
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Pinyan Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yongcui Mao
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Wenbo Meng
- Department of General Surgery, The First Hospital of Lanzhou University, The First Clinical Medical School of Lanzhou University, Lanzhou, 730000, Gansu, China.
| | - Xun Li
- Department of General Surgery, The First Hospital of Lanzhou University, The First Clinical Medical School of Lanzhou University, Lanzhou, 730000, Gansu, China
| |
Collapse
|
9
|
Endicott M, Jones M, Hull J. Amino acid metabolism as a therapeutic target in cancer: a review. Amino Acids 2021; 53:1169-1179. [PMID: 34292410 PMCID: PMC8325646 DOI: 10.1007/s00726-021-03052-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 07/16/2021] [Indexed: 02/04/2023]
Abstract
Malignant cells often demonstrate a proliferative advantage when compared to non-malignant cells. However, the rapid growth and metabolism required for survival can also highlight vulnerabilities specific to these malignant cells. One such vulnerability exhibited by cancer is an increased demand for amino acids (AAs), which often results in a dependency on exogenous sources of AAs or requires upregulation of de novo synthesis. These metabolic alterations can be exploited by therapy, which aims to improve treatment outcome and decrease relapse and reoccurrence. One clinically utilised strategy targeting AA dependency is the use of asparaginase in the treatment of acute lymphoblastic leukaemia (ALL), which results in a depletion of exogenous asparagine and subsequent cancer cell death. Examples of other successful strategies include the exploitation of arginine deiminase and methioninase, nutrient restriction of methionine and the inhibition of glutaminase. In this review, we summarise these treatment strategies into three promising avenues: AA restriction, enzymatic depletion and inhibition of metabolism. This review provides an insight into the complexity of metabolism in cancer, whilst highlighting these three current research avenues that have support in both preclinical and clinical settings.
Collapse
Affiliation(s)
- Molly Endicott
- Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Michael Jones
- Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK
| | - Jonathon Hull
- Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK.
| |
Collapse
|
10
|
Han Q, Hoffman RM. Chronic Treatment of an Advanced Prostate-cancer Patient With Oral Methioninase Resulted in Long-term Stabilization of Rapidly Rising PSA Levels. In Vivo 2021; 35:2171-2176. [PMID: 34182494 DOI: 10.21873/invivo.12488] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND/AIM Advanced prostate cancer is a recalcitrant disease with very limited treatment options. Our laboratory discovered methionine addiction, presumably a characteristic of all cancer types, including prostate cancer, which can be targeted by methionine restriction (MR), through treatment with oral recombinant methioninase (o-rMETase). PATIENTS AND METHODS o-rMETase was produced by fermentation of recombinant E. coli containing the Pseudomonas putida methioninase gene, and purified by column chromatography. An advanced prostate cancer patient received o-rMETase as a supplement, 500 units per day, divided into two oral doses of 250 units each. RESULTS Before treatment, the patient had a rapid rise in PSA levels, from 39 to 56 ng/ml, within 6 weeks. At the 15th week of o-rMETase administration, the PSA levels stabilized at 62 ng/ml. No overt side effects were observed. CONCLUSION o-rMETase single treatment can be beneficial for advanced prostate cancer patients.
Collapse
|
11
|
Preparation of Internalizing RGD-Modified Recombinant Methioninase Exosome Active Targeting Vector and Antitumor Effect Evaluation. Dig Dis Sci 2021; 66:1045-1053. [PMID: 32323072 DOI: 10.1007/s10620-020-06262-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 04/08/2020] [Indexed: 01/29/2023]
Abstract
BACKGROUND/AIMS Targeted drug delivery vehicles with low immunogenicity and toxicity are needed for cancer therapy. Here, we prepare an active targeting drug carrier of low immunogenicity and toxicity for targeted therapy. METHODS Immature dendritic cells (imDCs) from BALB/c mice were used as donor cells of exosomes (Exos) that were transfected with the plasmids expressing fusion proteins of a tumor-targeting peptide known as internalizing RGD (iRGD) to construct a type of tumor-targeting iRGD-Exos and observe the interaction between these iRGD-Exos. Also, recombinant methioninase (rMETase) was loaded into the iRGD-Exos by electroporation to construct iRGD-Exos-rMETase and to assess the tumor-targeting function of the iRGD-Exos-rMETase. Finally, 30 BALB/c were randomly divided into five groups (n = 6), to observe tumor growth in vivo. RESULTS The iRGD-Exos-rMETase was 99.58 nm in diameter and presented a unique "goblet" structure under transmission electron microscopy (TEM), with the encapsulation efficiency (EE) of 19.05%. iRGD-Exos-rMETase group has the strongest tumor suppressive effect. Compared to the iRGD-Exos-rMETase group, rMETase group and the blank-Exos-rMETase group were less effective, while the PBS group and the iRGD-Exos group showed no inhibitory effect on tumor growth. After treatment, the iRGD-Exos-rMETase group had gastric tumors significantly smaller and lighter than the other groups (P < 0.05). CONCLUSION The iRGD-Exos-rMETase is an effective antitumor therapy that delivers rMETase to tumor tissue using the iRGD-Exos. With its favorable inhibitory effect and tumor-targeting function, the iRGD-Exos-rMETase shows excellent potential value and exciting prospects in clinical applications.
Collapse
|
12
|
Abid MA, Abid MB. Commentary: Dietary methionine influences therapy in mouse cancer models and alters human metabolism. Front Oncol 2020; 10:1071. [PMID: 32733800 PMCID: PMC7357428 DOI: 10.3389/fonc.2020.01071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/28/2020] [Indexed: 11/13/2022] Open
Affiliation(s)
- Muhammad Abbas Abid
- Department of Otolaryngology - Head & Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Muhammad Bilal Abid
- Divisions of Hematology/Oncology & Infectious Diseases, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
13
|
Li C, Li Z, Zhang T, Wei P, Li N, Zhang W, Ding X, Li J. 1H NMR-Based Metabolomics Reveals the Antitumor Mechanisms of Triptolide in BALB/c Mice Bearing CT26 Tumors. Front Pharmacol 2019; 10:1175. [PMID: 31680959 PMCID: PMC6798008 DOI: 10.3389/fphar.2019.01175] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 09/12/2019] [Indexed: 11/13/2022] Open
Abstract
Triptolide, the main active ingredient in Tripterygium wilfordii Hook. f. (Celastraceae), has shown promising effects against a variety of tumors. However, the molecular pharmacological mechanisms explaining the action of triptolide remain unknown. In this study, the CT26 colon tumor cell line was inoculated subcutaneously into BALB/c mice, and plasma samples were subjected to 1H NMR metabolomics analysis. The metabolic signature identified five metabolites whose levels were lower and 15 whose levels were higher in CT26 tumor-bearing mice than in normal control mice. Triptolide treatment significantly reversed the levels of nine of these metabolites, including isoleucine, glutamine, methionine, proline, 3-hydroxybutyric acid, 2-hydroxyisovalerate, 2-hydroxyisobutyrate, and low-density lipoprotein/very low-density lipoprotein. Based on the identities of these potential biomarkers, we conclude that the antitumor mechanism of triptolide might rely on correcting perturbations in branched-chain amino acid metabolism, serine/glycine/methionine biosynthesis, and ketone bodies metabolism.
Collapse
Affiliation(s)
- Cheng Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhongfeng Li
- Department of Chemistry, Capital Normal University, Beijing, China
| | | | - Peihuang Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Nuo Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xia Ding
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jian Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
14
|
Xin L, Zhou Q, Yuan YW, Zhou LQ, Liu L, Li SH, Liu C. METase/lncRNA HULC/FoxM1 reduced cisplatin resistance in gastric cancer by suppressing autophagy. J Cancer Res Clin Oncol 2019; 145:2507-2517. [PMID: 31485766 DOI: 10.1007/s00432-019-03015-w] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 08/28/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Autophagy plays an important role in regulating cisplatin (CDDP) resistance in gastric cancer cells. However, the underlying mechanism of methioninase (METase) in the regulation of autophagy and CDDP resistance of gastric cancer cells is still not clear. MATERIALS AND METHODS Western blot was used to detect the levels of autophagy-related proteins, multidrug-resistant 1 (MDR-1), and FoxM1 protein. LncRNA HULC was detected by qRT-PCR. Cell viability was detected using CCK-8 assay. The interaction between lncRNA HULC and FoxM1 was confirmed by RNA pull-down and RIP assay. RESULTS Lentiviral vector carrying METase (LV-METase) suppressed autophagy and CDDP resistance of drug-resistant gastric cancer cells. LncRNA HULC was significantly downregulated in drug-resistant gastric cancer cells transfected with LV-METase. Besides, we found that lncRNA HULC interacted with FoxM1. In addition, METase suppressed autophagy to reduce CDDP resistance of drug-resistant gastric cancer cells through regulating HULC/FoxM1, and interfering HULC suppressed autophagy to reduce CDDP resistance of drug-resistant gastric cancer cells through regulating FoxM1. Finally, interfering HULC inhibited tumor growth in vivo. CONCLUSION METase suppressed autophagy to reduce CDDP resistance of drug-resistant gastric cancer cells through regulating HULC/FoxM1 pathway.
Collapse
Affiliation(s)
- Lin Xin
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| | - Qi Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yi-Wu Yuan
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Li-Qiang Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Li Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Shi-Hao Li
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Chuan Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| |
Collapse
|
15
|
Efficacy of Recombinant Methioninase (rMETase) on Recalcitrant Cancer Patient-Derived Orthotopic Xenograft (PDOX) Mouse Models: A Review. Cells 2019; 8:cells8050410. [PMID: 31052611 PMCID: PMC6562625 DOI: 10.3390/cells8050410] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 04/13/2019] [Accepted: 04/17/2019] [Indexed: 01/10/2023] Open
Abstract
An excessive requirement for methionine (MET), termed MET dependence, appears to be a general metabolic defect in cancer and has been shown to be a very effective therapeutic target. MET restriction (MR) has inhibited the growth of all major cancer types by selectively arresting cancer cells in the late-S/G2 phase, when they also become highly sensitive to cytotoxic agents. Recombinant methioninase (rMETase) has been developed to effect MR. The present review describes the efficacy of rMETase on patient-derived orthotopic xenograft (PDOX) models of recalcitrant cancer, including the surprising result that rMETase administrated orally can be highly effective.
Collapse
|
16
|
Piotrowska A, Wierzbicka J, Rybarczyk A, Tuckey RC, Slominski AT, Żmijewski MA. Vitamin D and its low calcemic analogs modulate the anticancer properties of cisplatin and dacarbazine in the human melanoma A375 cell line. Int J Oncol 2019; 54:1481-1495. [PMID: 30968156 PMCID: PMC6411347 DOI: 10.3892/ijo.2019.4725] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/14/2019] [Indexed: 12/11/2022] Open
Abstract
Melanoma represents a significant challenge in cancer treatment due to the high drug resistance of melanomas and the patient mortality rate. This study presents data indicating that nanomolar concentrations of the hormonally active form of vitamin D, 1α,25-dihydroxyvitamin D3 [1α,25(OH)2D3], its non-calcemic analogues 20S-hydroxyvitamin D3 and 21-hydroxypregnacalciferol, as well as the low-calcemic synthetic analog calcipotriol, modulate the efficacy of the anticancer drugs cisplatin and dacarbazine. It was observed that vitamin D analogs sensitized melanoma A375 cells to hydrogen peroxide used as an inducer of oxidative stress. On the other hand, only 1α,25(OH)2D3 resulted in a minor, but significant effect on the proliferation of melanoma cells treated simultaneously with dacarbazine, but not cisplatin. Notably, cisplatin (300 µM) exhibited a higher overall antiproliferative activity than dacarbazine. Cisplatin treatment of melanoma cells resulted in an induction of apoptosis as demonstrated by flow cytometry (accumulation of cells at the subG1 phase of the cell cycle), whereas dacarbazine caused G1/G0 cell cycle arrest, with the effects being improved by pre-treatment with vitamin D analogs. Treatment with cisplatin resulted in an initial increase in the level of reactive oxygen species (ROS). Dacarbazine caused transient stimulation of ROS levels and the mitochondrial membrane potential (Δψm) (after 1 or 3 h of treatment, respectively), but the effect was not detectable following prolonged (24 h) incubation with the drug. Vitamin D exhibited modulatory effects on the cells treated with dacarbazine, decreasing the half maximal inhibitory concentration (IC50) for the drug, stimulating G1/G0 arrest and causing a marked decrease in Δψm. Finally, cisplatin, dacarbazine and 1α,25(OH)2D3 displayed modulatory effects on the expression of ROS and vitamin D-associated genes in the melanoma A375 cells. In conclusion, nanomolar concentrations of 1,25(OH)2D3 only had minor effects on the proliferation of melanoma cells treated with dacarbazine, decreasing the relative IC50 value. However, co-treatment with vitamin D analogs resulted in the modulation of cell cycle and ROS responses, and affected gene expression, suggesting possible crosstalk between the signaling pathways of vitamin D and the anticancer drugs used in this study.
Collapse
Affiliation(s)
- Anna Piotrowska
- Department of Histology, Faculty of Medicine, Medical University of Gdansk, 80‑211 Gdansk, Poland
| | - Justyna Wierzbicka
- Department of Histology, Faculty of Medicine, Medical University of Gdansk, 80‑211 Gdansk, Poland
| | - Agnieszka Rybarczyk
- Department of Histology, Faculty of Medicine, Medical University of Gdansk, 80‑211 Gdansk, Poland
| | - Robert C Tuckey
- School of Molecular Sciences, Faculty of Science, The University of Western Australia, Perth, Western Australia 6009, Australia
| | - Andrzej T Slominski
- Department of Dermatology, Cancer Chemoprevention Program, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michał A Żmijewski
- Department of Histology, Faculty of Medicine, Medical University of Gdansk, 80‑211 Gdansk, Poland
| |
Collapse
|
17
|
Methioninase Cell-Cycle Trap Cancer Chemotherapy. Methods Mol Biol 2019; 1866:133-148. [PMID: 30725413 DOI: 10.1007/978-1-4939-8796-2_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Cancer cells are methionine (MET) dependent compared to normal cells as they have an elevated requirement for MET in order to proliferate. MET restriction selectively traps cancer cells in the S/G2 phase of the cell cycle. The cell cycle phase can be visualized by color coding with the fluorescence ubiquitination-based cell cycle indicator (FUCCI). Recombinant methioninase (rMETase) is an enzyme that effectively degrades MET. rMETase induces S/G2-phase blockage of cancer cells which is identified by the cancer cells' green fluorescence with FUCCI imaging. Cancer cells in G1/G0 are the majority of the cells in solid tumors and are resistant to the chemotherapy. Treatment of cancer cells with standard chemotherapy drugs only led to the majority of the cancer cell population being arrested in G0/G1 phase, identified by the cancer cells' red fluorescence in the FUCCI system. The G0/G1-phase cancer cells are chemo-resistant. Tumor targeting Salmonella typhimurium A1-R (S. typhimurium A1-R) was used to decoy quiescent G0/G1 stomach cancer cells growing in nude mice to cycle, with subsequent rMETase treatment to selectively trap the decoyed cancer cells in S/G2 phase, which made them highly sensitive to chemotherapy. Subsequent cisplatinum (CDDP) or paclitaxel (PTX) chemotherapy was then administered to kill the decoyed and trapped cancer cells, which completely prevented or regressed tumor growth. In a subsequent experiment, a patient-derived orthotopic xenograft (PDOX) model of recurrent CDDP-resistant metastatic osteosarcoma was eradicated by the combination of Salmonella typhimurium A1-R decoy, rMETase S/G2-phase cell cycle trap, and CDDP cell kill. Salmonella typhimurium A1-R and rMETase pre-treatment thereby overcame CDDP resistance. These results demonstrate the effectiveness of the new chemotherapy paradigm of "decoy, trap, and kill" chemotherapy.
Collapse
|
18
|
Hoffman RM, Han Q, Kawaguchi K, Li S, Tan Y. Afterword: Oral Methioninase-Answer to Cancer and Fountain of Youth? Methods Mol Biol 2019; 1866:311-322. [PMID: 30725426 DOI: 10.1007/978-1-4939-8796-2_24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The elevated methionine (MET) requirement of cancer cells is termed MET dependence and is possibly the only known general metabolic defect in cancer. Targeting MET by recombinant methioninase (rMETase) can arrest the growth of cancer cells in vitro and in vivo due to their elevated requirement for MET. rMETase can also potentiate chemotherapy drugs active in S phase due to the selective arrest of cancer cells in S/G2 phase during MET restriction (MR). We previously reported that rMETase, administrated by intraperitoneal injection (ip-rMETase), could inhibit tumor growth in mouse models of cancer including patient-derived orthotopic xenograft (PDOX) mouse models. We subsequently compared ip-rMETase and oral rMETase (o-rMETase) on a melanoma PDOX mouse model. o-rMETase was significantly more effective than ip-rMETase to inhibit tumor growth without overt toxicity. The combination of o-rMETase+ip-rMETase was significantly more effective than either monotherapy and completely arrested tumor growth. Thus, o-rMETase is effective as an anticancer agent with the potential of clinical development for chronic cancer therapy as well as for cancer prevention. o-rMETase may also have potential as an antiaging agent for healthy people, since MR has been shown to extend the life span of a variety of different organisms.
Collapse
Affiliation(s)
- Robert M Hoffman
- AntiCancer, Inc., San Diego, CA, USA. .,Department of Surgery, University of California, San Diego, CA, USA.
| | | | - Kei Kawaguchi
- AntiCancer, Inc., San Diego, CA, USA.,Department of Surgery, University of California, San Diego, CA, USA
| | | | | |
Collapse
|
19
|
Abstract
Methionine (MET) dependence is a cancer-specific metabolic abnormality that is due to MET overuse for aberrant transmethylation reactions. [11C]-MET is very useful for positron-emission tomography (PET) due to MET overuse in malignant tumors. Many benefits of MET-PET have been demonstrated. MET-PET can differentiate recurrent glioma and necrosis. [11C]-MET-PET can also predict prognosis in gliomas better than [18F]-FDG PET. [11C]-MET-PET is better than MRI for predicting survival in low-grade glioma (LGG). MET-PET has greater specificity for detecting residual tumor after surgery than MRI.
Collapse
Affiliation(s)
- Robert M Hoffman
- AntiCancer, Inc., San Diego, CA, USA. .,Department of Surgery, University of California, San Diego, CA, USA.
| |
Collapse
|
20
|
Abstract
Methionine (MET) restriction (MR) has been shown to arrest cancer growth and sensitizes tumors to chemotherapy. MR total parenteral nutrition (MR TPN) with a chemotherapy-containing amino acid solution ("AO-90") (lacking both MET and L-cysteine[CYS]) showed synergistic effects with 5-fluorouracil (5-FU) in tumor-bearing rats and in a Phase I clinical trial with gastrointestinal tract cancers compared to 5-FU in a MET-containing TPN. All gastric cancer patients underwent gastrectomy. Resected tumors in the AO-90 group showed significant reduction of cancer histologically, while almost no effect was seen in the control group. A Phase II clinical trial of dietary MR combined with cystemustine treatment for melanoma or glioma was carried out. Twenty-two patients (20 with metastatic melanoma and 2 with recurrent gloma) received a median of four cycles of the combination of a 1-day MR diet with cystemustine (60 mg/m2) every 2 weeks. This combination was well tolerated (toxicity and nutritional status). The median disease-free survival was 1.8 months and the median survival was 4.6 months, with two long-duration stabilizations. MET depletion in plasma was 40%. In another study, eight patients with a variety of metastatic solid tumors were enrolled in a Phase I clinical trial of a commercially available MR medical food. Participants remained on the experimental diet for an average of 17.3 weeks. Plasma methionine levels fell from 21.6 to 9 μm within 2 weeks, a 58% decline. The only side effect was weight loss of approximately 0.5 kg per week. A feasibility study combining dietary MR with a FOLFOX regimen in patients with metastatic colorectal cancer was carried out. The plasma MET concentration was reduced by dietary MR by 58% on the first day of the MR diet. Among the four patients evaluable for response, three experienced a partial response and one patient had disease stabilization. The results of the above-described clinical trials indicate the clinical potential of MR.
Collapse
Affiliation(s)
- Robert M Hoffman
- AntiCancer, Inc., San Diego, CA, USA.
- Department of Surgery, University of California, San Diego, CA, USA.
| |
Collapse
|
21
|
Kiyuna T, Tome Y, Murakami T, Miyake K, Igarashi K, Kawaguchi K, Oshiro H, Higuchi T, Miyake M, Sugisawa N, Zhang Z, Razmjooei S, Wangsiricharoen S, Chmielowski B, Nelson SD, Russell TA, Dry SM, Li Y, Eckardt MA, Singh AS, Chawla S, Kanaya F, Eilber FC, Singh SR, Zhao M, Hoffman RM. A combination of irinotecan/cisplatinum and irinotecan/temozolomide or tumor-targeting Salmonella typhimurium A1-R arrest doxorubicin- and temozolomide-resistant myxofibrosarcoma in a PDOX mouse model. Biochem Biophys Res Commun 2018; 505:733-739. [PMID: 30292411 DOI: 10.1016/j.bbrc.2018.09.106] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 09/16/2018] [Indexed: 12/22/2022]
Abstract
Myxofibrosarcoma (MFS) is the most common sarcomas in elderly patients and is either chemo-resistant or recurs with metastasis after chemotherapy. This recalcitrant cancer in need of improved treatment. We have established a patient-derived orthotopic xenograft (PDOX) of MFS. The MFS PDOX model was established in the biceps femoris of nude mice and randomized into 7 groups of 7 mice each: control; doxorubicin (DOX); pazopanib (PAZ); temozolomide (TEM); Irinotecan (IRN); IRN combined with TEM; IRN combined with cisplatinum (CDDP) and Salmonella typhimurium A1-R (S. typhimurium A1-R). Treatment was evaluated by relative tumor volume and relative body weight. The MFS PDOX models were DOX, PAZ, and TEM resistant. IRN combined with TEM and IRN combined with CDDP were most effective on the MFS PDOX. S. typhimurium A1-R arrested the MFS PDOX tumor. There was no significant body weight loss in any group. The present study suggests that the combination of IRN with either TEM or CDDP, and S. typhimurium have clinical potential for MFS.
Collapse
Affiliation(s)
- Tasuku Kiyuna
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA; Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Yasunori Tome
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan.
| | - Takashi Murakami
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Kentaro Miyake
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Kentaro Igarashi
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Kei Kawaguchi
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Hiromichi Oshiro
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Takashi Higuchi
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Masuyo Miyake
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Norihiko Sugisawa
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | - Zhiying Zhang
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA
| | | | | | | | - Scott D Nelson
- Department of Pathology, University of California, Los Angeles, CA, USA
| | - Tara A Russell
- Division of Surgical Oncology, University of California, Los Angeles, CA, USA
| | - Sarah M Dry
- Department of Pathology, University of California, Los Angeles, CA, USA
| | - Yunfeng Li
- Department of Pathology, University of California, Los Angeles, CA, USA
| | - Mark A Eckardt
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Arun S Singh
- Div. of Hematology-Oncology, University of California, Los Angeles, CA, USA
| | - Sant Chawla
- Sarcoma Oncology Center, Santa Monica, CA, USA.
| | - Fuminori Kanaya
- Department of Orthopedic Surgery, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Fritz C Eilber
- Division of Surgical Oncology, University of California, Los Angeles, CA, USA.
| | - Shree Ram Singh
- Basic Research Laboratory, National Cancer Institute, Frederick, MD, USA.
| | | | - Robert M Hoffman
- AntiCancer Inc, San Diego, CA, USA; Department of Surgery, University of California, San Diego, CA, USA.
| |
Collapse
|
22
|
Kawaguchi K, Miyake K, Zhao M, Kiyuna T, Igarashi K, Miyake M, Higuchi T, Oshiro H, Bouvet M, Unno M, Hoffman RM. Tumor targeting Salmonella typhimurium A1-R in combination with gemcitabine (GEM) regresses partially GEM-resistant pancreatic cancer patient-derived orthotopic xenograft (PDOX) nude mouse models. Cell Cycle 2018; 17:2019-2026. [PMID: 29963961 DOI: 10.1080/15384101.2018.1480223] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Gemcitabine (GEM) is first-line therapy for pancreatic cancer but has limited efficacy in most cases. Nanoparticle-albumin bound (nab)-paclitaxel is becoming first-line therapy for pancreatic cancer, but also has limited efficacy for pancreatic cancer. Our goal was to improve the treatment outcome in patient-like models of pancreatic cancer. We previously established patient-derived orthotopic xenografts (PDOX) pancreatic cancers from two patients. The pancreatic tumor was implanted orthotopically in the pancreatic tail of nude mice to establish the PDOX models. Five weeks after implantation, 50 PDOX mouse models were randomized into five groups of 10 mice for each pancreatic cancer PDOX: untreated control; GEM (100 mg/kg, i.p., once a week for 2 weeks); GEM + nab-PTX (GEM: 100 mg/kg, i.p., once a week for 2 weeks, nab-PTX: 10 mg/kg, i.v., twice a week for 2 weeks); S. typhimurium A1-R (5 × 107 CFU/100 μl, i.v., once a week for 2 weeks); GEM + S. typhimurium A1-R (GEM: 100 mg/kg, i.p., once a week for 2 weeks, S. typhimurium A1-R; 5 × 107 CFU/100 μl, i.v., once a week for 2 weeks). GEM + nab-PTX was significantly more effective than GEM alone in one PDOX model (p = 0.0004), but there was no significant difference in the other PDOX model. The combination of GEM + S. typhimurium A1-R regressed both PDOX models. These results show S. typhimurium A1-R can overcome the ineffectiveness or partial effectiveness of GEM in patient-like models of pancreatic cancer and demonstrate clinical potential for this combination.
Collapse
Affiliation(s)
- Kei Kawaguchi
- a AntiCancer, Inc ., San Diego , CA , USA.,b Department of Surgery , University of California , San Diego , CA , USA.,c Department of Surgery, Graduate School of Medicine , Tohoku University , Sendai , Japan
| | - Kentaro Miyake
- a AntiCancer, Inc ., San Diego , CA , USA.,b Department of Surgery , University of California , San Diego , CA , USA
| | - Ming Zhao
- a AntiCancer, Inc ., San Diego , CA , USA
| | - Tasuku Kiyuna
- a AntiCancer, Inc ., San Diego , CA , USA.,b Department of Surgery , University of California , San Diego , CA , USA
| | - Kentaro Igarashi
- a AntiCancer, Inc ., San Diego , CA , USA.,b Department of Surgery , University of California , San Diego , CA , USA
| | - Masuyo Miyake
- a AntiCancer, Inc ., San Diego , CA , USA.,b Department of Surgery , University of California , San Diego , CA , USA
| | - Takashi Higuchi
- a AntiCancer, Inc ., San Diego , CA , USA.,b Department of Surgery , University of California , San Diego , CA , USA
| | - Hiromichi Oshiro
- a AntiCancer, Inc ., San Diego , CA , USA.,b Department of Surgery , University of California , San Diego , CA , USA
| | - Michael Bouvet
- b Department of Surgery , University of California , San Diego , CA , USA
| | - Michiaki Unno
- c Department of Surgery, Graduate School of Medicine , Tohoku University , Sendai , Japan
| | - Robert M Hoffman
- a AntiCancer, Inc ., San Diego , CA , USA.,b Department of Surgery , University of California , San Diego , CA , USA
| |
Collapse
|
23
|
Kawaguchi K, Higuchi T, Li S, Han Q, Tan Y, Igarashi K, Zhao M, Miyake K, Kiyuna T, Miyake M, Ohshiro H, Sugisawa N, Zhang Z, Razmjooei S, Wangsiricharoen S, Chmielowski B, Nelson SD, Russell TA, Dry SM, Li Y, Eckardt MA, Singh AS, Singh SR, Eilber FC, Unno M, Hoffman RM. Combination therapy of tumor-targeting Salmonella typhimurium A1-R and oral recombinant methioninase regresses a BRAF-V600E-negative melanoma. Biochem Biophys Res Commun 2018; 503:3086-3092. [DOI: 10.1016/j.bbrc.2018.08.097] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 08/13/2018] [Indexed: 01/10/2023]
|
24
|
Kawaguchi K, Miyake K, Han Q, Li S, Tan Y, Igarashi K, Kiyuna T, Miyake M, Higuchi T, Oshiro H, Zhang Z, Razmjooei S, Wangsiricharoen S, Bouvet M, Singh SR, Unno M, Hoffman RM. Oral recombinant methioninase (o-rMETase) is superior to injectable rMETase and overcomes acquired gemcitabine resistance in pancreatic cancer. Cancer Lett 2018; 432:251-259. [DOI: 10.1016/j.canlet.2018.06.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/05/2018] [Accepted: 06/12/2018] [Indexed: 01/06/2023]
|
25
|
Kawaguchi K, Miyake K, Han Q, Li S, Tan Y, Igarashi K, Lwin TM, Higuchi T, Kiyuna T, Miyake M, Oshiro H, Bouvet M, Unno M, Hoffman RM. Targeting altered cancer methionine metabolism with recombinant methioninase (rMETase) overcomes partial gemcitabine-resistance and regresses a patient-derived orthotopic xenograft (PDOX) nude mouse model of pancreatic cancer. Cell Cycle 2018; 17:868-873. [PMID: 29623758 PMCID: PMC6056209 DOI: 10.1080/15384101.2018.1445907] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 02/20/2018] [Indexed: 10/17/2022] Open
Abstract
Pancreatic cancer is a recalcitrant disease. Gemcitabine (GEM) is the most widely-used first-line therapy for pancreatic cancer, but most patients eventually fail. Transformative therapy is necessary to significantly improve the outcome of pancreatic cancer patients. Tumors have an elevated requirement for methionine and are susceptible to methionine restriction. The present study used a patient-derived orthotopic xenograft (PDOX) nude mouse model of pancreatic cancer to determine the efficacy of recombinant methioninase (rMETase) to effect methionine restriction and thereby overcome GEM-resistance. A pancreatic cancer obtained from a patient was grown orthotopically in the pancreatic tail of nude mice to establish the PDOX model. Five weeks after implantation, 40 pancreatic cancer PDOX mouse models were randomized into four groups of 10 mice each: untreated control (n = 10); GEM (100 mg/kg, i.p., once a week for 5 weeks, n = 10); rMETase (100 units, i.p., 14 consecutive days, n = 10); GEM+rMETase (GEM: 100 mg/kg, i.p., once a week for 5 weeks, rMETase: 100 units, i.p., 14 consecutive days, n = 10). Although GEM partially inhibited PDOX tumor growth, combination therapy (GEM+rMETase) was significantly more effective than mono therapy (GEM: p = 0.0025, rMETase: p = 0.0010). The present study is the first demonstrating the efficacy of rMETase combination therapy in a pancreatic cancer PDOX model to overcome first-line therapy resistance in this recalcitrant disease.
Collapse
Affiliation(s)
- Kei Kawaguchi
- AntiCancer, Inc., San Diego, CA
- Department of Surgery, University of California, San Diego, CA
- Department of Surgery, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Kentaro Miyake
- AntiCancer, Inc., San Diego, CA
- Department of Surgery, University of California, San Diego, CA
| | | | | | | | - Kentaro Igarashi
- AntiCancer, Inc., San Diego, CA
- Department of Surgery, University of California, San Diego, CA
| | | | - Takashi Higuchi
- AntiCancer, Inc., San Diego, CA
- Department of Surgery, University of California, San Diego, CA
| | - Tasuku Kiyuna
- AntiCancer, Inc., San Diego, CA
- Department of Surgery, University of California, San Diego, CA
| | - Masuyo Miyake
- AntiCancer, Inc., San Diego, CA
- Department of Surgery, University of California, San Diego, CA
| | - Hiromichi Oshiro
- AntiCancer, Inc., San Diego, CA
- Department of Surgery, University of California, San Diego, CA
| | - Michael Bouvet
- Department of Surgery, University of California, San Diego, CA
| | - Michiaki Unno
- Department of Surgery, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Robert M. Hoffman
- AntiCancer, Inc., San Diego, CA
- Department of Surgery, University of California, San Diego, CA
| |
Collapse
|
26
|
Igarashi K, Kawaguchi K, Li S, Han Q, Tan Y, Gainor E, Kiyuna T, Miyake K, Miyake M, Higuchi T, Oshiro H, Singh AS, Eckardt MA, Nelson SD, Russell TA, Dry SM, Li Y, Yamamoto N, Hayashi K, Kimura H, Miwa S, Tsuchiya H, Eilber FC, Hoffman RM. Recombinant methioninase combined with doxorubicin (DOX) regresses a DOX-resistant synovial sarcoma in a patient-derived orthotopic xenograft (PDOX) mouse model. Oncotarget 2018; 9:19263-19272. [PMID: 29721200 PMCID: PMC5922394 DOI: 10.18632/oncotarget.24996] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 03/15/2018] [Indexed: 01/09/2023] Open
Abstract
Synovial sarcoma (SS) is a recalcitrant subgroup of soft tissue sarcoma (STS). A tumor from a patient with high grade SS from a lower extremity was grown orthotopically in the right biceps femoris muscle of nude mice to establish a patient-derived orthotopic xenograft (PDOX) mouse model. The PDOX mice were randomized into the following groups when tumor volume reached approximately 100 mm3: G1, control without treatment; G2, doxorubicin (DOX) (3 mg/kg, intraperitoneal [i.p.] injection, weekly, for 2 weeks; G3, rMETase (100 unit/mouse, i.p., daily, for 2 weeks); G4 DOX (3mg/kg), i.p. weekly, for 2 weeks) combined with rMETase (100 unit/mouse, i.p., daily, for 2 weeks). On day 14 after treatment initiation, all therapies significantly inhibited tumor growth compared to untreated control, except DOX: (DOX: p = 0.48; rMETase: p < 0.005; DOX combined with rMETase < 0.0001). DOX combined with rMETase was significantly more effective than both DOX alone (p < 0.001) and rMETase alone (p < 0.05). The relative body weight on day 14 compared with day 0 did not significantly differ between any treatment group or untreated control. The results indicate that r-METase can overcome DOX-resistance in this recalcitrant disease.
Collapse
Affiliation(s)
- Kentaro Igarashi
- AntiCancer, Inc., San Diego, California, USA.,Department of Surgery, University of California, San Diego, California, USA.,Department of Orthopaedic Surgery, Kanazawa University, Kanazawa, Japan
| | - Kei Kawaguchi
- AntiCancer, Inc., San Diego, California, USA.,Department of Surgery, University of California, San Diego, California, USA
| | - Shukuan Li
- AntiCancer, Inc., San Diego, California, USA
| | | | - Yuying Tan
- AntiCancer, Inc., San Diego, California, USA
| | | | - Tasuku Kiyuna
- AntiCancer, Inc., San Diego, California, USA.,Department of Surgery, University of California, San Diego, California, USA
| | - Kentaro Miyake
- AntiCancer, Inc., San Diego, California, USA.,Department of Surgery, University of California, San Diego, California, USA
| | - Masuyo Miyake
- AntiCancer, Inc., San Diego, California, USA.,Department of Surgery, University of California, San Diego, California, USA
| | - Takashi Higuchi
- AntiCancer, Inc., San Diego, California, USA.,Department of Surgery, University of California, San Diego, California, USA
| | - Hiromichi Oshiro
- AntiCancer, Inc., San Diego, California, USA.,Department of Surgery, University of California, San Diego, California, USA
| | - Arun S Singh
- Division of Hematology-Oncology, University of California, Los Angeles, CA, USA
| | - Mark A Eckardt
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Scott D Nelson
- Department of Pathology, University of California, Los Angeles, CA, USA
| | - Tara A Russell
- Division of Surgical Oncology, University of California, Los Angeles, CA, USA
| | - Sarah M Dry
- Department of Pathology, University of California, Los Angeles, CA, USA
| | - Yunfeng Li
- Department of Pathology, University of California, Los Angeles, CA, USA
| | - Norio Yamamoto
- Department of Orthopaedic Surgery, Kanazawa University, Kanazawa, Japan
| | - Katsuhiro Hayashi
- Department of Orthopaedic Surgery, Kanazawa University, Kanazawa, Japan
| | - Hiroaki Kimura
- Department of Orthopaedic Surgery, Kanazawa University, Kanazawa, Japan
| | - Shinji Miwa
- Department of Orthopaedic Surgery, Kanazawa University, Kanazawa, Japan
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic Surgery, Kanazawa University, Kanazawa, Japan
| | - Fritz C Eilber
- Division of Surgical Oncology, University of California, Los Angeles, CA, USA
| | - Robert M Hoffman
- AntiCancer, Inc., San Diego, California, USA.,Department of Surgery, University of California, San Diego, California, USA
| |
Collapse
|