1
|
Wang Z, Li W, Jiang Y, Tran TB, Chung J, Kim M, Scott AJ, Lu J. Camptothesome-based combination nanotherapeutic regimen for improved colorectal cancer immunochemotherapy. Biomaterials 2024; 306:122477. [PMID: 38309054 PMCID: PMC10922823 DOI: 10.1016/j.biomaterials.2024.122477] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 12/21/2023] [Accepted: 01/18/2024] [Indexed: 02/05/2024]
Abstract
Camptothesome is a sphingomyelin-conjugated camptothecin (SM-CSS-CPT) nanovesicle that fortified the therapeutic delivery of CPT in diverse cancer types. To mitigate the Camptothesome-induced IDO1 negative feedback mechanism, we had co-encapsulated, indoximod (IND, IDO1 inhibitor) into Camptothesome using doxorubicin-derived IND (DOX-IND). To maximize the therapeutic potential of DOX-IND/Camptothesome, herein, we first dissected the synergistic drug ratio (DOX-IND/SM-CSS-CPT) via systematical in vitro screening. DOX-IND/Camptothesome with optimal drug ratio synchronized in vivo drug delivery with significantly higher tumor uptake compared to free drugs. This optimum DOX-IND/Camptothesome outperformed the combination of Camptothesome, Doxil and IND or other IDO1 inhibitors (BMS-986205 or epacadostat) in treating mice bearing late-stage MC38 tumors, and combination with immune checkpoint blockade (ICB) enabled it to eradicate 60 % of large tumors. Further, this optimized co-delivery Camptothesome beat Folfox and Folfiri, two first-line combination chemotherapies for colorectal cancer in antitumor efficacy and exhibited no side effects as compared to the severe systemic toxicities associated with Folfox and Folfiri. Finally, we demonstrated that the synergistic DOX-IND/Camptothesome was superior to the combined use of Onivyde + Doxil + IND in curbing the advanced orthotopic CT26-Luc tumors and eliminated 40 % tumors with complete metastasis remission when cooperated with ICB, eliciting stronger anti-CRC immune responses and greater reversal of immunosuppression. These results corroborated that with precise optimal synergistic drug ratio, the therapeutic potential of DOX-IND/Camptothesome can be fully unleased, which warrants further clinical investigation to benefit the cancer patients.
Collapse
Affiliation(s)
- Zhiren Wang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, United States
| | - Wenpan Li
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, United States
| | - Yanhao Jiang
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, United States
| | - Tuyen Ba Tran
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, United States
| | - Jinha Chung
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, United States
| | - Minhyeok Kim
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, United States
| | - Aaron James Scott
- Clinical and Translational Oncology Program, The University of Arizona Cancer Center, Tucson, AZ, 85721, United States; BIO5 Institute, The University of Arizona, Tucson, AZ, 85721, United States
| | - Jianqin Lu
- Skaggs Pharmaceutical Sciences Center, Department of Pharmacology & Toxicology, R. Ken Coit College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, United States; BIO5 Institute, The University of Arizona, Tucson, AZ, 85721, United States; Division of Hematology and Oncology, Department of Medicine, College of Medicine, The University of Arizona, Tucson, AZ 85721, United States; Southwest Environmental Health Sciences Center, The University of Arizona, Tucson, 85721, United States.
| |
Collapse
|
2
|
Liu Z, Georgakopoulos-Soares I, Ahituv N, Wong KC. Risk scoring based on DNA methylation-driven related DEGs for colorectal cancer prognosis with systematic insights. Life Sci 2023; 316:121413. [PMID: 36682524 DOI: 10.1016/j.lfs.2023.121413] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023]
Abstract
Colorectal cancer is a common malignant tumor of the digestive tract. Despite advances in diagnostic techniques and medications. Its prognosis remains challenging. DNA methylation-driven related circulating tumor cells have attracted enormous interest in diagnosing owing to their non-invasive nature and early recognition properties. However, the mechanism through which risk biomarkers act remains elusive. Here, we designed a risk model based on differentially expressed genes, DNA methylation, robust, and survival-related factors in the framework of Cox regression. The model has satisfactory performance and is independently verified by an external and isolated dataset in terms of C-index value, ROC, and tROC. The model was applied to Colorectal cancer patients who were subsequently divided into high- and low-risk groups. Functional annotations, genomic alterations, tumor immune environment, and drug sensitivity were analyzed. We observed that up-regulated genes are associated with epithelial cell differentiation and MAPK signaling pathways. The down-regulated genes are related to IL-7 signaling and apoptosis-induced DNA fragmentation. Interestingly, the immune system was inhibited in high-risk groups. High-frequency mutation genes tend to co-occur. High-risk score patients are related to copy number amplification events. To address the challenges, we suggested eleven and twenty-one drugs that are sensitive to low- and high-risk patients. Finally, an artificial neural network was provided to evaluate the immunotherapeutic efficiency. Taken together, the findings demonstrated that our risk score model is robust and reliable for evaluating the prognosis with novel diagnostic and treatment targets. It also yields benefits for the treatment and provides unique insights into developing therapeutic strategies.
Collapse
Affiliation(s)
- Zhe Liu
- Department of Computer Science, City University of Hong Kong, Hong Kong, China
| | - Ilias Georgakopoulos-Soares
- Institute for Personalized Medicine, Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA; Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
3
|
Lucchetta M, Pellegrini M. Drug repositioning by merging active subnetworks validated in cancer and COVID-19. Sci Rep 2021; 11:19839. [PMID: 34615934 PMCID: PMC8494853 DOI: 10.1038/s41598-021-99399-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 09/23/2021] [Indexed: 02/08/2023] Open
Abstract
Computational drug repositioning aims at ranking and selecting existing drugs for novel diseases or novel use in old diseases. In silico drug screening has the potential for speeding up considerably the shortlisting of promising candidates in response to outbreaks of diseases such as COVID-19 for which no satisfactory cure has yet been found. We describe DrugMerge as a methodology for preclinical computational drug repositioning based on merging multiple drug rankings obtained with an ensemble of disease active subnetworks. DrugMerge uses differential transcriptomic data on drugs and diseases in the context of a large gene co-expression network. Experiments with four benchmark diseases demonstrate that our method detects in first position drugs in clinical use for the specified disease, in all four cases. Application of DrugMerge to COVID-19 found rankings with many drugs currently in clinical trials for COVID-19 in top positions, thus showing that DrugMerge can mimic human expert judgment.
Collapse
Affiliation(s)
- Marta Lucchetta
- Institute of Informatics and Telematics (IIT), CNR, Pisa, 56124, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, 53100, Italy
| | - Marco Pellegrini
- Institute of Informatics and Telematics (IIT), CNR, Pisa, 56124, Italy.
| |
Collapse
|
4
|
Trivedi ND, Armstrong S, Wang H, Hartley M, Deeken J, Ruth He A, Subramaniam D, Melville H, Albanese C, Marshall JL, Hwang J, Pishvaian MJ. A phase I trial of the mTOR inhibitor temsirolimus in combination with capecitabine in patients with advanced malignancies. Cancer Med 2021; 10:1944-1954. [PMID: 33638305 PMCID: PMC7957175 DOI: 10.1002/cam4.3672] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/18/2020] [Accepted: 11/10/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Temsirolimus is an mTOR antagonist with proven anticancer efficacy. Preclinical data suggest greater anticancer effect when mTOR inhibitors are combined with cytotoxic chemotherapy. We performed a Phase I assessment of the combination of temsirolimus and capecitabine in patients with advanced solid tumors. METHODS Patients were enrolled in an alternating dose escalation of temsirolimus (at 15 or 25 mg IV weekly) and capecitabine (at 750, 1000, and 1250 mg/m2 twice daily) in separate Q2-week and Q3-week cohorts. At the recommended Phase II doses (RP2Ds) of temsirolimus and capecitabine (Q2), seven patients were also treated with oxaliplatin (85 mg/m2 , day 1) to determine triplet combination safety and efficacy. RESULTS Forty-five patients were enrolled and 41 were evaluable for dose-limiting toxicities (DLTs). The most common adverse events (AEs) were mucositis, fatigue, and thrombocytopenia. The most common grade 3/4 AEs were hypophosphatemia and anemia. Five patients had DLTs, including hypophosphatemia, mucositis, and thrombocytopenia. The RP2Ds were temsirolimus 25 mg +capecitabine 1000 mg/m2 (Q2); and temsirolimus 25 mg +capecitabine 750 mg/m2 (Q3). Of the 38 patients evaluable for response, one had a partial response (PR) and 19 had stable disease (SD). The overall disease control rate was 52%. Five of the 20 patients with SD/PR maintained disease control for >6 months. CONCLUSIONS The combination of temsirolimus and capecitabine is safe on both a Q2-week and a Q3-week schedule. The combination demonstrated promising evidence of disease control in this highly refractory population and could be considered for testing in disease-specific phase II trials.
Collapse
Affiliation(s)
- Neel D Trivedi
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Samantha Armstrong
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Hongkun Wang
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Marion Hartley
- The Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - John Deeken
- Inova Schar Cancer Institute, Inova Health System, Falls Church, VA, USA
| | - A Ruth He
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Deepa Subramaniam
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Heather Melville
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Chris Albanese
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - John L Marshall
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Jimmy Hwang
- Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC, USA
| | - Michael J Pishvaian
- Department of Oncology, Johns Hopkins University School of Medicine, SKCC, Washington, DC, USA
| |
Collapse
|
5
|
Chang CW, Lee HC, Li LH, Chiang Chiau JS, Wang TE, Chuang WH, Chen MJ, Wang HY, Shih SC, Liu CY, Tsai TH, Chen YJ. Fecal Microbiota Transplantation Prevents Intestinal Injury, Upregulation of Toll-Like Receptors, and 5-Fluorouracil/Oxaliplatin-Induced Toxicity in Colorectal Cancer. Int J Mol Sci 2020; 21:386. [PMID: 31936237 PMCID: PMC7013718 DOI: 10.3390/ijms21020386] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/18/2019] [Accepted: 01/06/2020] [Indexed: 02/07/2023] Open
Abstract
FOLFOX (5-fluorouracil, leucovorin, and oxaliplatin), a 5-fluorouracil (5-FU)-based chemotherapy regimen, is one of most common therapeutic regimens for colorectal cancer. However, intestinal mucositis is a common adverse effect for which no effective preventive strategies exist. Moreover, the efficacy and the safety of fecal microbiota transplants (FMT) in cancer patients treated with anti-neoplastic agents are still scant. We investigated the effect of FMT on FOLFOX-induced mucosal injury. BALB/c mice implanted with syngeneic CT26 colorectal adenocarcinoma cells were orally administered FMT daily during and two days after five-day injection of FOLFOX regimen for seven days. Administration of FOLFOX significantly induced marked levels of diarrhea and intestinal injury. FMT reduced the severity of diarrhea and intestinal mucositis. Additionally, the number of goblet cells and zonula occludens-1 decreased, while apoptotic and NF-κB-positive cells increased following FOLFOX treatment. The expression of toll-like receptors (TLRs), MyD88, and serum IL-6 were upregulated following FOLFOX treatment. These responses were attenuated following FMT. The disrupted fecal gut microbiota composition was also restored by FMT after FOLFOX treatment. Importantly, FMT did not cause bacteremia and safely alleviated FOLFOX-induced intestinal mucositis in colorectal cancer-bearing mice. The putative mechanism may involve the gut microbiota TLR-MyD88-NF-κB signaling pathway in mice with implanted colorectal carcinoma cells.
Collapse
Affiliation(s)
- Ching-Wei Chang
- Institute of Traditional Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
| | - Hung-Chang Lee
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
- MacKay Children’s Hospital, Taipei 10449, Taiwan
| | - Li-Hui Li
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
| | - Jen-Shiu Chiang Chiau
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
| | - Tsang-En Wang
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
| | - Wei-Hung Chuang
- Institute of Biomedical Informatics, Center for Systems and Synthetic Biology, National Yang-Ming University, Taipei 11221, Taiwan;
| | - Ming-Jen Chen
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
| | - Horng-Yuan Wang
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
| | - Shou-Chuan Shih
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
| | - Chia-Yuan Liu
- Institute of Traditional Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei 10449, Taiwan; (T.-E.W.); (M.-J.C.); (H.-Y.W.); (S.-C.S.)
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
| | - Tung-Hu Tsai
- Institute of Traditional Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Department of Chemical Engineering, National United University, Miaoli 36063, Taiwan
| | - Yu-Jen Chen
- Institute of Traditional Medicine, National Yang-Ming University, Taipei 11221, Taiwan;
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City 25173, Taiwan; (H.-C.L.); (L.-H.L.); (J.-S.C.C.)
- Department of Medicine, MacKay Medical College, New Taipei City 25245, Taiwan
- MacKay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan
- Department of Radiation Oncology, MacKay Memorial Hospital, Taipei 10449, Taiwan
| |
Collapse
|
6
|
Zhao P, Ma YG, Zhao Y, Liu D, Dai ZJ, Yan CY, Guan HT. MicroRNA-552 deficiency mediates 5-fluorouracil resistance by targeting SMAD2 signaling in DNA-mismatch-repair-deficient colorectal cancer. Cancer Chemother Pharmacol 2019; 84:427-439. [PMID: 31087138 DOI: 10.1007/s00280-019-03866-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 05/04/2019] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Although DNA-mismatch-repair-deficient (dMMR) status and aberrant expression of miRNAs are both critically implicated in the pathogenesis of resistance to 5-fluorouracil (5-FU) in colorectal cancer (CRC), whether these two factors regulate tumor response to 5-FU in a coordinated manner remains unknown. This study is designed to elucidate whether changes in miR-552 expression levels correlate to 5-FU-based chemoresistance in CRC, and to further identify the putative targets of miR-552 using multiple approaches. METHODS miR-552 expression was assessed in 5-FU-resistant CRC tissues and cells using real-time PCR. Effects of miR-552 dysregulation on 5-FU resistance in CRC cells were determined by measuring cell viability, apoptosis and in vivo oncogenic capacity. Finally, we studied the posttranscriptional regulation of SMAD2 by miR-552 using multiple approaches including luciferase reporter assay, site-directed mutagenesis and transient/stable transfection, at molecular and functional levels. RESULTS Expression of miR-552 was significantly downregulated in 5-FU-resistant CRC tissues and cells, and this downregulation, regulated by dMMR, was associated with poor postchemotherapy prognosis. Functionally, forced expression of miR-552 exhibited a proapoptotic effect and attenuated 5-FU resistance, whereas inhibition of miR-552 expression potentiated 5-FU resistance in CRC cells. Mechanically, miR-552 directly targeted the 3'-UTR of SMAD2, and stable ablation of SMAD2 neutralized the promoting effects of miR-552 deficiency-induced 5-FU resistance. CONCLUSIONS Overall, our findings have revealed a critical role of miR-552/SMAD2 cascade in modulating cellular response to 5-FU chemotherapy. miR-552 may act as an efficient mechanistic link synchronizing dMMR and 5-FU resistance in CRC.
Collapse
Affiliation(s)
- Ping Zhao
- Department of Gastroenterology, The Second Affiliated Hospital of Xian Jiaotong University, Xi'an, 710004, Shaanxi, People's Republic of China
| | - Yu-Guang Ma
- Department of Surgical Oncology, The Second Affiliated Hospital of Xian Jiaotong University, No. 157, West Five Road, Xi'an, 710004, Shaanxi, People's Republic of China
| | - Yang Zhao
- Department of Surgical Oncology, The Second Affiliated Hospital of Xian Jiaotong University, No. 157, West Five Road, Xi'an, 710004, Shaanxi, People's Republic of China
| | - Di Liu
- Department of Surgical Oncology, The Second Affiliated Hospital of Xian Jiaotong University, No. 157, West Five Road, Xi'an, 710004, Shaanxi, People's Republic of China
| | - Zhi-Jun Dai
- Department of Surgical Oncology, The Second Affiliated Hospital of Xian Jiaotong University, No. 157, West Five Road, Xi'an, 710004, Shaanxi, People's Republic of China
| | - Chang-You Yan
- Health and Family Planning Commission of Chengcheng County, Weinan, 714000, Shaanxi, People's Republic of China
| | - Hai-Tao Guan
- Department of Surgical Oncology, The Second Affiliated Hospital of Xian Jiaotong University, No. 157, West Five Road, Xi'an, 710004, Shaanxi, People's Republic of China.
| |
Collapse
|
7
|
Spyridopoulou K, Aindelis G, Lampri E, Giorgalli M, Lamprianidou E, Kotsianidis I, Tsingotjidou A, Pappa A, Kalogirou O, Chlichlia K. Improving the Subcutaneous Mouse Tumor Model by Effective Manipulation of Magnetic Nanoparticles-Treated Implanted Cancer Cells. Ann Biomed Eng 2018; 46:1975-1987. [PMID: 30076502 DOI: 10.1007/s10439-018-2107-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 07/21/2018] [Indexed: 12/12/2022]
Abstract
Murine tumor models have played a fundamental role in the development of novel therapeutic interventions and are currently widely used in translational research. Specifically, strategies that aim at reducing inter-animal variability of tumor size in transplantable mouse tumor models are of particular importance. In our approach, we used magnetic nanoparticles to label and manipulate colon cancer cells for the improvement of the standard syngeneic subcutaneous mouse tumor model. Following subcutaneous injection on the scruff of the neck, magnetically-tagged implanted cancer cells were manipulated by applying an external magnetic field towards localized tumor formation. Our data provide evidence that this approach can facilitate the formation of localized tumors of similar shape, reducing thereby the tumor size's variability. For validating the proof-of-principle, a low-dose of 5-FU was administered in small animal groups as a representative anticancer therapy. Under these experimental conditions, the 5-FU-induced tumor growth inhibition was statistically significant only after the implementation of the proposed method. The presented approach is a promising strategy for studying accurately therapeutic interventions in subcutaneous experimental solid tumor models allowing for the detection of statistically significant differences between smaller experimental groups.
Collapse
Affiliation(s)
- Katerina Spyridopoulou
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus-Dragana, 68100, Alexandroupolis, Greece
| | - Georgios Aindelis
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus-Dragana, 68100, Alexandroupolis, Greece
| | - Evangeli Lampri
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus-Dragana, 68100, Alexandroupolis, Greece
| | - Maria Giorgalli
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus-Dragana, 68100, Alexandroupolis, Greece
| | - Eleftheria Lamprianidou
- Department of Hematology, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Ioannis Kotsianidis
- Department of Hematology, School of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Anastasia Tsingotjidou
- Laboratory of Anatomy, Histology and Embryology, School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus-Dragana, 68100, Alexandroupolis, Greece
| | - Orestis Kalogirou
- Department of Physics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Katerina Chlichlia
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus-Dragana, 68100, Alexandroupolis, Greece.
| |
Collapse
|
8
|
Chang CW, Liu CY, Lee HC, Huang YH, Li LH, Chiau JSC, Wang TE, Chu CH, Shih SC, Tsai TH, Chen YJ. Lactobacillus casei Variety rhamnosus Probiotic Preventively Attenuates 5-Fluorouracil/Oxaliplatin-Induced Intestinal Injury in a Syngeneic Colorectal Cancer Model. Front Microbiol 2018; 9:983. [PMID: 29867884 PMCID: PMC5962742 DOI: 10.3389/fmicb.2018.00983] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/26/2018] [Indexed: 12/12/2022] Open
Abstract
Adjuvant 5-fluorouracil (5-FU)-based chemotherapy, including FOLFOX (5-FU, leucovorin, and oxaliplatin), is recommended for colorectal cancer. However, intestinal mucositis remains a common adverse effect for which no effective preventive strategies are available. To develop a convenient and novel way to alleviate mucositis, we investigated the effect of Lactobacillus casei variety rhamnosus (Lcr35) on FOLFOX-induced mucosal injury. BALB/c mice subcutaneously injected with syngeneic CT26 colorectal adenocarcinoma cells were orally administered Lcr35 daily before, during, and after 5-day injection of FOLFOX regimen, for 14 days. The following methods were used: diarrhea score for toxicity, ELISA for cytokine production, histopathology for intestinal injury, immunohistochemistry for apoptosis/proliferation and regulatory proteins, RT-PCR for cytokine mRNA expression, and DNA sequencing for fecal gut microbiota. FOLFOX administration to colorectal cancer-bearing mice significantly inhibited tumor growth and the accompanying marked diarrhea and intestinal injury histologically characterized by the shortening of villi and destruction of intestinal crypts. Preventive administration of Lcr35 dose-dependently reduced the severity of diarrhea and intestinal mucositis without affecting the anti-tumor effect of FOLFOX. The numbers of apoptotic, NF-κB-, and BAX-activated cells increased after FOLFOX, and these responses were mitigated by Lcr35. TNF-α and IL-6 upregulation by FOLFOX treatment was attenuated by Lcr35. The fecal gut microbiota composition of Firmicutes and Bacteroidetes disturbed by FOLFOX was significantly reversed by Lcr35 toward a preferential profile. In conclusion, the oral probiotic Lcr35 prevented FOLFOX-induced intestinal mucositis in colorectal cancer-bearing mice. The putative mechanism might involve modulation of gut microbiota and proinflammatory responses with suppression of intrinsic apoptosis in intestinal injury.
Collapse
Affiliation(s)
- Ching-Wei Chang
- Institute of Traditional Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, New Taipei City, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Chia-Yuan Liu
- Institute of Traditional Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, New Taipei City, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Hung-Chang Lee
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.,Mackay Junior College of Medicine, Nursing, and Management, New Taipei City, Taiwan.,MacKay Children's Hospital, Taipei, Taiwan
| | - Yen-Hua Huang
- Institute of Biomedical Informatics, Center for Systems and Synthetic Biology, National Yang-Ming University, Taipei, Taiwan
| | - Li-Hui Li
- Department of Medical Research, MacKay Memorial Hospital, New Taipei City, Taiwan
| | | | - Tsang-En Wang
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.,Mackay Junior College of Medicine, Nursing, and Management, New Taipei City, Taiwan
| | - Cheng-Hsin Chu
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.,Mackay Junior College of Medicine, Nursing, and Management, New Taipei City, Taiwan
| | - Shou-Chuan Shih
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.,Mackay Junior College of Medicine, Nursing, and Management, New Taipei City, Taiwan
| | - Tung-Hu Tsai
- Institute of Traditional Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Chemical Engineering, National United University, Miaoli, Taiwan
| | - Yu-Jen Chen
- Institute of Traditional Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Research, MacKay Memorial Hospital, New Taipei City, Taiwan.,Department of Medicine, MacKay Medical College, New Taipei City, Taiwan.,Department of Radiation Oncology, MacKay Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
9
|
Di Gialleonardo V, Aldeborgh HN, Miloushev V, Folkers KM, Granlund K, Tap WD, Lewis JS, Weber WA, Keshari KR. Multinuclear NMR and MRI Reveal an Early Metabolic Response to mTOR Inhibition in Sarcoma. Cancer Res 2017; 77:3113-3120. [PMID: 28386017 DOI: 10.1158/0008-5472.can-16-3310] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 11/30/2016] [Accepted: 03/16/2017] [Indexed: 01/07/2023]
Abstract
Biomarkers predicting rapalog responses in sarcomas where PI3K and mTOR are often hyperactivated could improve the suitable recruitment of responsive patients to clinical trials. PI3K/mTOR pathway activation drives energy production by regulating anaerobic glycolysis in cancer cells, suggesting a route toward a monitoring strategy. In this study, we took a multimodality approach to evaluate the phenotypic effects and metabolic changes that occur with inhibition of the PI3K/mTOR pathway. Its central role in regulating glycolysis in human sarcomas was evaluated by short- and long-term rapamycin treatment in sarcoma cell lines. We observed an overall decrease in lactate production in vitro, followed by cell growth inhibition. In vivo, we observed a similar quantitative reduction in lactate production as monitored by hyperpolarized MRI, also followed by tumor size changes. This noninvasive imaging method could distinguish reduced cell proliferation from induction of cell death. Our results illustrate the use of hyperpolarized MRI as a sensitive technique to monitor drug-induced perturbation of the PI3K/mTOR pathway in sarcomas. Cancer Res; 77(11); 3113-20. ©2017 AACR.
Collapse
Affiliation(s)
- Valentina Di Gialleonardo
- Radiology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hannah N Aldeborgh
- Radiology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Vesselin Miloushev
- Radiology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kelly M Folkers
- Radiology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kristin Granlund
- Radiology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - William D Tap
- Medicine Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Jason S Lewis
- Radiology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Medicine Memorial Sloan Kettering Cancer Center, New York, New York
| | - Wolfgang A Weber
- Radiology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York.,Weill Cornell Medical College, New York, New York
| | - Kayvan R Keshari
- Radiology and Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York. .,Weill Cornell Medical College, New York, New York
| |
Collapse
|
10
|
Gremonprez F, Willaert W, Ceelen W. Animal models of colorectal peritoneal metastasis. Pleura Peritoneum 2016; 1:23-43. [PMID: 30911606 DOI: 10.1515/pp-2016-0006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 03/04/2016] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer remains an important cause of mortality worldwide. The presence of peritoneal carcinomatosis (PC) causes significant symptoms and is notoriously difficult to treat. Therefore, informative preclinical research into the mechanisms and possible novel treatment options of colorectal PC is essential in order to improve the prognostic outlook in these patients. Several syngeneic and xenograft animal models of colorectal PC were established, studying a wide range of experimental procedures and substances. Regrettably, more sophisticated models such as those giving rise to spontaneous PC or involving genetically engineered mice are lacking. Here, we provide an overview of all reported colorectal PC animal models and briefly discuss their use, strengths, and limitations.
Collapse
Affiliation(s)
- Félix Gremonprez
- Department of Gastrointestinal Surgery, Ghent University Hospital, Ghent, Belgium
| | - Wouter Willaert
- Department of Gastrointestinal Surgery, Ghent University Hospital, Ghent, Belgium
| | - Wim Ceelen
- Department of Gastrointestinal Surgery, Ghent University Hospital, 2K12 IC UZ Gent De Pintelaan 185, 9000 Ghent, Belgium
| |
Collapse
|
11
|
ZHANG BINHAO, LENG CHAO, WU CHAO, ZHANG ZHANGUO, DOU LEI, LUO XIN, ZHANG BIXIANG, CHEN XIAOPING. Smad4 sensitizes colorectal cancer to 5-fluorouracil through cell cycle arrest by inhibiting the PI3K/Akt/CDC2/survivin cascade. Oncol Rep 2015; 35:1807-15. [DOI: 10.3892/or.2015.4479] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/17/2015] [Indexed: 11/06/2022] Open
|
12
|
Chen HJ, Sun J, Huang Z, Hou H, Arcilla M, Rakhilin N, Joe DJ, Choi J, Gadamsetty P, Milsom J, Nandakumar G, Longman R, Zhou XK, Edwards R, Chen J, Chen KY, Bu P, Wang L, Xu Y, Munroe R, Abratte C, Miller AD, Gümüş ZH, Shuler M, Nishimura N, Edelmann W, Shen X, Lipkin SM. Comprehensive models of human primary and metastatic colorectal tumors in immunodeficient and immunocompetent mice by chemokine targeting. Nat Biotechnol 2015; 33:656-60. [PMID: 26006007 PMCID: PMC4532544 DOI: 10.1038/nbt.3239] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 04/15/2015] [Indexed: 11/09/2022]
Abstract
Current orthotopic xenograft models of human colorectal cancer (CRC) require surgery and do not robustly form metastases in the liver, the most common site clinically. CCR9 traffics lymphocytes to intestine and colorectum. We engineered use of the chemokine receptor CCR9 in CRC cell lines and patient-derived cells to create primary gastrointestinal (GI) tumors in immunodeficient mice by tail-vein injection rather than surgery. The tumors metastasize inducibly and robustly to the liver. Metastases have higher DKK4 and NOTCH signaling levels and are more chemoresistant than paired subcutaneous xenografts. Using this approach, we generated 17 chemokine-targeted mouse models (CTMMs) that recapitulate the majority of common human somatic CRC mutations. We also show that primary tumors can be modeled in immunocompetent mice by microinjecting CCR9-expressing cancer cell lines into early-stage mouse blastocysts, which induces central immune tolerance. We expect that CTMMs will facilitate investigation of the biology of CRC metastasis and drug screening.
Collapse
Affiliation(s)
- Huanhuan Joyce Chen
- 1] Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA. [2] Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Jian Sun
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Zhiliang Huang
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Harry Hou
- Department of Cell Biology, Albert Einstein College of Medicine, New York, New York, USA
| | - Myra Arcilla
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Nikolai Rakhilin
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Daniel J Joe
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Jiahn Choi
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Poornima Gadamsetty
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Jeff Milsom
- Department of Surgery, Weill Cornell Medical College, New York, New York, USA
| | - Govind Nandakumar
- Department of Surgery, Weill Cornell Medical College, New York, New York, USA
| | - Randy Longman
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Xi Kathy Zhou
- Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, New York, USA
| | - Robert Edwards
- Department of Pathology, University of California, Irvine, Irvine, California, USA
| | - Jonlin Chen
- School of Electrical and Computer Engineering, Cornell University, Ithaca, New York, USA
| | - Kai Yuan Chen
- School of Electrical and Computer Engineering, Cornell University, Ithaca, New York, USA
| | - Pengcheng Bu
- 1] Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA. [2] School of Electrical and Computer Engineering, Cornell University, Ithaca, New York, USA
| | - Lihua Wang
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Yitian Xu
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Robert Munroe
- College of Veterinary Medicine and Biological Sciences, Cornell University, Ithaca, New York, USA
| | - Christian Abratte
- College of Veterinary Medicine and Biological Sciences, Cornell University, Ithaca, New York, USA
| | - Andrew D Miller
- College of Veterinary Medicine and Biological Sciences, Cornell University, Ithaca, New York, USA
| | - Zeynep H Gümüş
- 1] Department of Medicine, Weill Cornell Medical College, New York, New York, USA. [2] Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Michael Shuler
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Nozomi Nishimura
- Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA
| | - Winfried Edelmann
- Department of Cell Biology, Albert Einstein College of Medicine, New York, New York, USA
| | - Xiling Shen
- 1] Department of Biomedical Engineering, Cornell University, Ithaca, New York, USA. [2] School of Electrical and Computer Engineering, Cornell University, Ithaca, New York, USA
| | - Steven M Lipkin
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
13
|
Loss of Smad4 in colorectal cancer induces resistance to 5-fluorouracil through activating Akt pathway. Br J Cancer 2014; 110:946-57. [PMID: 24384683 PMCID: PMC3929873 DOI: 10.1038/bjc.2013.789] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 11/12/2013] [Accepted: 11/15/2013] [Indexed: 02/06/2023] Open
Abstract
Background: Higher frequency of Smad4 inactivation or loss of expression is observed in metastasis of colorectal cancer (CRC) leading to unfavourable survival and contributes to chemoresistance. However, the molecular mechanism of how Smad4 regulates chemosensitivity of CRC is unknown. Methods: We evaluated how the loss of Smad4 in CRC enhanced chemoresistance to 5-fluorouracil (5-FU) using two CRC cell lines in vitro and in vivo. Immunoblotting with cell and tumour lysates and immunohistochemical analyses with tissue microarray were performed. Results: Knockdown or loss of Smad4 induced tumorigenicity, migration, invasion, angiogenesis, metastasis, and 5-FU resistance. Smad4 expression in mouse tumours regulated cell-cycle regulatory proteins leading to Rb phosphorylation. Loss of Smad4 activated Akt pathway that resulted in upregulation of anti-apoptotic proteins, Bcl-2 and Bcl-w, and Survivin. Suppression of phosphatidylinositol-3-kinase (PI3K)/Akt pathway by LY294002 restored chemosensitivity of Smad4-deficient cells to 5-FU. Vascular endothelial growth factor-induced angiogenesis in Smad4-deficient cells might also lead to chemoresistance. Low levels of Smad4 expression in CRC tissues correlated with higher levels of Bcl-2 and Bcl-w and with poor overall survival as observed in immunohistochemical staining of tissue microarrays. Conclusion: Loss of Smad4 in CRC patients induces resistance to 5-FU-based therapy through activation of Akt pathway and inhibitors of this pathway may sensitise these patients to 5-FU.
Collapse
|
14
|
Li SH, Huang EY, Lu HI, Huang WT, Yen CC, Huang WC, Chen CH. Phosphorylated mammalian target of rapamycin expression is associated with the response to chemoradiotherapy in patients with esophageal squamous cell carcinoma. J Thorac Cardiovasc Surg 2012; 144:1352-1359.e1. [PMID: 22841170 DOI: 10.1016/j.jtcvs.2012.06.049] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2012] [Revised: 05/18/2012] [Accepted: 06/18/2012] [Indexed: 02/03/2023]
Abstract
OBJECTIVE The mammalian target of rapamycin signaling pathway has been implicated in therapeutic resistance in several types of cancer. However, the significance of mammalian target of rapamycin activation in chemoradiotherapy sensitivity and its effect on the prognosis of esophageal squamous cell carcinoma treated with chemoradiotherapy remain unknown. However, this pathway is of particular interest because an effective inhibitor is available. METHODS By using immunohistochemistry, phosphorylated mammalian target of rapamycin expression was examined in 77 patients with esophageal squamous cell carcinoma treated with preoperative chemoradiotherapy followed by surgery between 1999 and 2009, and correlated with treatment outcome. With the use of CE81T/VGH and TE2 cell lines, cells were treated with chemotherapy, temsirolimus (mammalian target of rapamycin inhibitor), or a combination of chemotherapy and temsirolimus, and investigated by 3-(4.5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. RESULTS Pathologic complete response rates were 42% and 16% in patients with negative and positive phosphorylated mammalian target of rapamycin expression, respectively (P = .01). The 3-year overall survivals were 57% and 30% in patients with negative and positive phosphorylated mammalian target of rapamycin expression, respectively (P = .005). Positive phosphorylated mammalian target of rapamycin expression was independently associated with inferior overall and disease-free survival. In patients who did not achieve pathologic complete response, postchemoradiotherapy esophagectomy specimens showed significantly higher phosphorylated mammalian target of rapamycin expression than pretreatment biopsy specimens. In cell lines, concomitant administration of temsirolimus enhanced the effect of chemotherapy. CONCLUSIONS Phosphorylated mammalian target of rapamycin expression is independently associated with the response to chemoradiotherapy and prognosis of patients with esophageal squamous cell carcinoma treated with preoperative chemoradiotherapy. Mammalian target of rapamycin inhibition can sensitize esophageal cancer cells to chemotherapy. Our results suggest the potential for mammalian target of rapamycin as a therapeutic target for patients with esophageal squamous cell carcinoma who receive multimodality treatment.
Collapse
Affiliation(s)
- Shau-Hsuan Li
- Department of Hematology-Oncology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan, ROC
| | | | | | | | | | | | | |
Collapse
|
15
|
Insulin and glucagon regulate pancreatic α-cell proliferation. PLoS One 2011; 6:e16096. [PMID: 21283589 PMCID: PMC3026810 DOI: 10.1371/journal.pone.0016096] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Accepted: 12/08/2010] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) results from insulin resistance and β-cell dysfunction, in the setting of hyperglucagonemia. Glucagon is a 29 amino acid peptide hormone, which is secreted from pancreatic α cells: excessively high circulating levels of glucagon lead to excessive hepatic glucose output. We investigated if α-cell numbers increase in T2DM and what factor (s) regulate α-cell turnover. Leprdb/Leprdb (db/db) mice were used as a T2DM model and αTC1 cells were used to study potential α-cell trophic factors. Here, we demonstrate that in db/db mice α-cell number and plasma glucagon levels increased as diabetes progressed. Insulin treatment (EC50 = 2 nM) of α cells significantly increased α-cell proliferation in a concentration-dependent manner compared to non-insulin-treated α cells. Insulin up-regulated α-cell proliferation through the IR/IRS2/AKT/mTOR signaling pathway, and increased insulin-mediated proliferation was prevented by pretreatment with rapamycin, a specific mTOR inhibitor. GcgR antagonism resulted in reduced rates of cell proliferation in αTC1 cells. In addition, blockade of GcgRs in db/db mice improved glucose homeostasis, lessened α-cell proliferation, and increased intra-islet insulin content in β cells in db/db mice. These studies illustrate that pancreatic α-cell proliferation increases as diabetes develops, resulting in elevated plasma glucagon levels, and both insulin and glucagon are trophic factors to α-cells. Our current findings suggest that new therapeutic strategies for the treatment of T2DM may include targeting α cells and glucagon.
Collapse
|
16
|
Spier BJ, Walker AJ, Cornett DD, Pfau PR, Halberg RB, Said A. Screening colonoscopy and detection of neoplasia in asymptomatic, average-risk, solid organ transplant recipients: case-control study. Transpl Int 2010; 23:1233-8. [DOI: 10.1111/j.1432-2277.2010.01137.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
17
|
Carozzi VA, Canta A, Oggioni N, Sala B, Chiorazzi A, Meregalli C, Bossi M, Marmiroli P, Cavaletti G. Neurophysiological and neuropathological characterization of new murine models of chemotherapy-induced chronic peripheral neuropathies. Exp Neurol 2010; 226:301-9. [PMID: 20832406 DOI: 10.1016/j.expneurol.2010.09.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 08/31/2010] [Accepted: 09/01/2010] [Indexed: 12/30/2022]
Abstract
Cisplatin, paclitaxel and bortezomib belong to some of the most effective families of chemotherapy drugs for solid and haematological cancers. Epothilones represent a new family of very promising antitubulin agents. The clinical use of all these drugs is limited by their severe peripheral neurotoxicity. Several in vivo rat models have reproduced the characteristics of the peripheral neurotoxicity of these drugs. However, since only a very limited number of cancer types can be studied in immunocompetent rats, these animal models do not represent an effective way to evaluate, at the same time, the antineoplastic activity and the neurotoxic effects of the anticancer compounds. In this study, we characterized the neurophysiological impairment induced by chronic chemotherapy treatment in BALB/c mice, a strain suitable for assessing the activity of anticancer treatments. At the end of a 4-week period of treatment with cisplatin, paclitaxel, epothilone-B or bortezomib, sensory and sensory/motor nerve conduction velocities (NCV) were determined in the caudal and digital nerves and dorsal root ganglia (DRG) and sciatic nerves were collected for histopathological analysis. The electrophysiological studies revealed that all the compounds caused a statistically significant reduction in the caudal NCV, while impairment of the digital NCV was less severe. This functional damage was confirmed by the histopathological observations evidencing axonal degeneration in the sciatic nerve induced by all the drugs associated with pathological changes in DRG induced only by cisplatin and bortezomib. These results confirm the possibility to use our models to combine the study of the antineoplastic activity of anticancer drugs and of their toxic effects on the peripheral nervous system in the BALB/c mouse strain.
Collapse
Affiliation(s)
- V A Carozzi
- Department of Neuroscience and Biomedical Technologies, University of Milano-Bicocca, Monza (MI), Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|