1
|
Nakamura A, Tanaka Y, Amano T, Takebayashi A, Takahashi A, Hanada T, Yoneoka Y, Tsuji S, Murakami T. Rapamycin inhibits tamoxifen-induced endometrial proliferation in vitro as a pilot approach for endometrial protection in breast cancer. Sci Rep 2025; 15:2112. [PMID: 39819881 PMCID: PMC11739499 DOI: 10.1038/s41598-025-86586-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/13/2025] [Indexed: 01/19/2025] Open
Abstract
Tamoxifen, a common adjuvant therapy for hormone receptor-positive breast cancer, is associated with an increased risk of endometrial pathologies, such as hyperplasia, polyps, and carcinoma. This study investigates rapamycin, an mTOR inhibitor, as a potential novel strategy for preventing tamoxifen-induced endometrial proliferation. This in vitro study utilised endometrial stromal cells isolated from infertile women. The cells were treated with tamoxifen alone or in combination with rapamycin, and proliferation was assessed using the CCK-8 assay. The activation of the mTOR pathway, as well as apoptosis and cell cycle markers, was evaluated by Western blotting to elucidate the molecular mechanisms underlying these effects. The study design emphasised simulating clinically relevant exposure levels. Tamoxifen significantly increased endometrial cell proliferation in a dose-dependent manner. Rapamycin effectively inhibited this proliferation, even at concentrations lower than those typically observed in clinical settings. Quantitative analysis by Western blotting showed activation of the mTOR pathway and cell cycle in the tamoxifen group, and inhibition of these pathways in the tamoxifen plus rapamycin combination group, whereas there was no change in apoptosis. In conclusion, rapamycin shows promise as a prophylactic agent against tamoxifen-induced endometrial pathologies, with potential implications for fertility preservation and endometrial protection in breast cancer patients.
Collapse
Affiliation(s)
- Akiko Nakamura
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan
| | - Yuji Tanaka
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan.
| | - Tsukuru Amano
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan
| | - Akie Takebayashi
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan
| | - Akimasa Takahashi
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan
| | - Tetsuro Hanada
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan
| | - Yutaka Yoneoka
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan
| | - Shunichiro Tsuji
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan
| | - Takashi Murakami
- Department of Obstetrics and Gynecology, Shiga University of Medical Science, 520-2192/Seta Tsukinowa-cho, Otsu, Shiga, Japan
| |
Collapse
|
2
|
Nakamura A, Tanaka Y, Amano T, Takebayashi A, Takahashi A, Hanada T, Tsuji S, Murakami T. mTOR inhibitors as potential therapeutics for endometriosis: a narrative review. Mol Hum Reprod 2024; 30:gaae041. [PMID: 39579091 PMCID: PMC11634386 DOI: 10.1093/molehr/gaae041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/15/2024] [Indexed: 11/25/2024] Open
Abstract
Mammalian target of rapamycin (mTOR) inhibitors have been used clinically as anticancer and immunosuppressive agents for over 20 years, demonstrating their safety after long-term administration. These inhibitors exhibit various effects, including inhibition of cell proliferation, interaction with the oestrogen and progesterone pathways, immunosuppression, regulation of angiogenesis, and control of autophagy. We evaluated the potential of mTOR inhibitors as therapeutic agents for endometriosis, examined the secondary benefits related to reproductive function, and assessed how their side effects can be managed. We conducted a thorough review of publications on the role of the mTOR pathway and the effectiveness of mTOR inhibitors in endometriosis patients. These results indicate that the mTOR pathway is activated in endometriosis. Additionally, mTOR inhibitors have shown efficacy as monotherapies for endometriosis. They may alleviate resistance to hormonal therapy in endometriosis, suggesting a potential synergistic effect when used in combination with hormonal therapy. The potential reproductive benefits of mTOR inhibitors include decreased miscarriage rates, improved implantation, and prevention of age-related follicular loss and ovarian hyperstimulation syndrome. Activation of the mTOR pathway has also been implicated in the malignant transformation of endometriosis. Preclinical studies suggest that the dosage of mTOR inhibitors needed for treating endometriosis may be lower than that required for anticancer or immunosuppressive therapy, potentially reducing dosage-dependent side effects. In conclusion, while mTOR inhibitors, which allow for pregnancy during oral administration, show potential for clinical use in all stages of endometriosis, current evidence is limited to preclinical studies, and further research is needed to confirm clinical effectiveness.
Collapse
Affiliation(s)
- Akiko Nakamura
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Yuji Tanaka
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Tsukuru Amano
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Akie Takebayashi
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Akimasa Takahashi
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Tetsuro Hanada
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Shunichiro Tsuji
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Takashi Murakami
- Department of Obstetrics and Gynaecology, Shiga University of Medical Science, Otsu, Shiga, Japan
| |
Collapse
|
3
|
Kamath HS, Shukla R, Shah U, Patel S, Das S, Chordia A, Satish P, Ghosh D. Role of Gut Microbiota in Predisposition to Colon Cancer: A Narrative Review. Indian J Microbiol 2024; 64:1-13. [PMID: 39282181 PMCID: PMC11399513 DOI: 10.1007/s12088-024-01242-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 02/28/2024] [Indexed: 09/18/2024] Open
Abstract
Globally, colorectal cancer (CRC) is a leading cause of cancer-related mortality. Dietary habits, inflammation, hereditary characteristics, and gut microbiota are some of its causes. The gut microbiota, a diverse population of bacteria living in the digestive system, has an impact on a variety of parameters, including inflammation, DNA damage, and immune response. The gut microbiome has a significant role in colon cancer susceptibility. Many studies have highlighted dysbiosis, an imbalance in the gut microbiota's makeup, as a major factor in colon cancer susceptibility. Dysbiosis has the potential to produce toxic metabolites and pro-inflammatory substances, which can hasten the growth of tumours. The ability of the gut microbiota to affect the host's immune system can also influence whether cancer develops or not. By better comprehending these complex interactions between colon cancer predisposition and gut flora, new preventive and therapeutic techniques might be developed. Targeting the gut microbiome with dietary modifications, probiotics, or faecal microbiota transplantation may offer cutting-edge approaches to reducing the risk of colon cancer and improving patient outcomes. The complex connection between the makeup of the gut microbiota and the emergence of colorectal cancer is explored in this narrative review.
Collapse
Affiliation(s)
- Hattiangadi Shruthi Kamath
- Kasturba Medical College, Mangalore, a constituent institution of the Manipal Academy of Higher Education (MAHE), Mangalore, Karnataka India
| | - Rushikesh Shukla
- Kasturba Medical College, Mangalore, a constituent institution of the Manipal Academy of Higher Education (MAHE), Mangalore, Karnataka India
| | - Urmil Shah
- Kasturba Medical College, Mangalore, a constituent institution of the Manipal Academy of Higher Education (MAHE), Mangalore, Karnataka India
| | - Siddhi Patel
- Kasturba Medical College, Mangalore, a constituent institution of the Manipal Academy of Higher Education (MAHE), Mangalore, Karnataka India
| | - Soumyajit Das
- Kasturba Medical College, Mangalore, a constituent institution of the Manipal Academy of Higher Education (MAHE), Mangalore, Karnataka India
| | - Ayush Chordia
- Kasturba Medical College, Mangalore, a constituent institution of the Manipal Academy of Higher Education (MAHE), Mangalore, Karnataka India
| | - Poorvikha Satish
- Kasturba Medical College, Mangalore, a constituent institution of the Manipal Academy of Higher Education (MAHE), Mangalore, Karnataka India
| | - Dibyankita Ghosh
- Kasturba Medical College, Mangalore, a constituent institution of the Manipal Academy of Higher Education (MAHE), Mangalore, Karnataka India
| |
Collapse
|
4
|
Takeda T, Tsubaki M, Genno S, Tokunaga K, Tanaka R, Nishida S. HER3/Akt/mTOR pathway is a key therapeutic target for the reduction of triple‑negative breast cancer metastasis via the inhibition of CXCR4 expression. Int J Mol Med 2023; 52:80. [PMID: 37477145 PMCID: PMC10555474 DOI: 10.3892/ijmm.2023.5283] [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: 03/13/2023] [Accepted: 06/20/2023] [Indexed: 07/22/2023] Open
Abstract
Triple‑negative breast cancer (TNBC), a highly metastatic subtype of breast cancer, and it has the worst prognosis among all subtypes of breast cancer. However, no effective systematic therapy is currently available for TNBC metastasis. Therefore, novel therapies targeting the key molecular mechanisms involved in TNBC metastasis are required. The present study examined whether the expression levels of human epidermal growth factor receptor 3 (HER3) were associated with the metastatic phenotype of TNBC, and evaluated the potential of HER3 as a therapeutic target in vitro and in vivo. A new highly metastatic 4T1 TNBC cell line, termed 4T1‑L8, was established. The protein expression levels in 4T1‑L8 cells were measured using luminex magnetic bead assays and western blot analysis. The HER3 expression levels and distant metastasis‑free survival (DMFS) in TNBC were analyzed using Kaplan‑Meier Plotter. Transwell migration and invasion assays were performed to detect migration and invasion. The anti‑metastatic effects were determined in an experimental mouse model of metastasis. The results revealed that the increased expression of the HER3/Akt/mTOR pathway was associated with a greater level of cell migration, invasion and metastasis of TNBC cells. In addition, it was found that high expression levels of HER3 were associated with a poor DMFS. The inhibition of the HER3/Akt/mammalian target of rapamycin (mTOR) pathway decreased the migration, invasion and metastasis of TNBC cells by decreasing the expression of C‑X‑C chemokine receptor type 4 (CXCR4). Furthermore, treatment of metastatic TNBC cells with everolimus inhibited their migration, invasion and metastasis by decreasing CXCR4 expression. Thus, targeting the HER3/Akt/mTOR pathway opens up a new avenue for the development of therapeutics against TNBC metastasis; in addition, everolimus may prove to be an effective therapeutic agent for the suppression of TNBC metastasis.
Collapse
Affiliation(s)
- Tomoya Takeda
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Shuji Genno
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Kenta Tokunaga
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Remi Tanaka
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Higashi-Osaka, Osaka 577-8502, Japan
| |
Collapse
|
5
|
Davis SL, Hartman SJ, Bagby SM, Schlaepfer M, Yacob BW, Tse T, Simmons DM, Diamond JR, Lieu CH, Leal AD, Cadogan EB, Hughes GD, Durant ST, Messersmith WA, Pitts TM. ATM kinase inhibitor AZD0156 in combination with irinotecan and 5-fluorouracil in preclinical models of colorectal cancer. BMC Cancer 2022; 22:1107. [PMID: 36309653 PMCID: PMC9617348 DOI: 10.1186/s12885-022-10084-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/11/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
AZD0156 is an oral inhibitor of ATM, a serine threonine kinase that plays a key role in DNA damage response (DDR) associated with double-strand breaks. Topoisomerase-I inhibitor irinotecan is used clinically to treat colorectal cancer (CRC), often in combination with 5-fluorouracil (5FU). AZD0156 in combination with irinotecan and 5FU was evaluated in preclinical models of CRC to determine whether low doses of AZD0156 enhance the cytotoxicity of irinotecan in chemotherapy regimens used in the clinic.
Methods
Anti-proliferative effects of single-agent AZD0156, the active metabolite of irinotecan (SN38), and combination therapy were evaluated in 12 CRC cell lines. Additional assessment with clonogenic assay, cell cycle analysis, and immunoblotting were performed in 4 selected cell lines. Four colorectal cancer patient derived xenograft (PDX) models were treated with AZD0156, irinotecan, or 5FU alone and in combination for assessment of tumor growth inhibition (TGI). Immunofluorescence was performed on tumor tissues. The DDR mutation profile was compared across in vitro and in vivo models.
Results
Enhanced effects on cellular proliferation and regrowth were observed with the combination of AZD0156 and SN38 in select models. In cell cycle analysis of these models, increased G2/M arrest was observed with combination treatment over either single agent. Immunoblotting results suggest an increase in DDR associated with irinotecan therapy, with a reduced effect noted when combined with AZD0156, which is more pronounced in some models. Increased TGI was observed with the combination of AZD0156 and irinotecan as compared to single-agent therapy in some PDX models. The DDR mutation profile was variable across models.
Conclusions
AZD0156 and irinotecan provide a rational and active combination in preclinical colorectal cancer models. Variability across in vivo and in vitro results may be related to the variable DDR mutation profiles of the models evaluated. Further understanding of the implications of individual DDR mutation profiles may help better identify patients more likely to benefit from treatment with the combination of AZD0156 and irinotecan in the clinical setting.
Collapse
|
6
|
Komen J, van Neerven SM, van den Berg A, Vermeulen L, van der Meer AD. Mimicking and surpassing the xenograft model with cancer-on-chip technology. EBioMedicine 2021; 66:103303. [PMID: 33773183 PMCID: PMC8024912 DOI: 10.1016/j.ebiom.2021.103303] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 03/04/2021] [Accepted: 03/10/2021] [Indexed: 01/11/2023] Open
Abstract
Organs-on-chips are in vitro models in which human tissues are cultured in microfluidic compartments with a controlled, dynamic micro-environment. Specific organs-on-chips are being developed to mimic human tumors, but the validation of such 'cancer-on-chip' models for use in drug development is hampered by the complexity and variability of human tumors. An important step towards validation of cancer-on-chip technology could be to first mimic cancer xenograft models, which share multiple characteristics with human cancers but are significantly less complex. Here we review the relevant biological characteristics of a xenograft tumor and show that organ-on-chip technology is capable of mimicking many of these aspects. Actual comparisons between on-chip tumor growth and xenografts are promising but also demonstrate that further development and empirical validation is still needed. Validation of cancer-on-chip models to xenografts would not only represent an important milestone towards acceptance of cancer-on-chip technology, but could also improve drug discovery, personalized cancer medicine, and reduce animal testing.
Collapse
Affiliation(s)
- Job Komen
- BIOS Lab on a Chip group, MESA+ Institute for Nanotechnology, University of Twente, P. O. Box 217, 7500 AE Enschede, the Netherlands.
| | - Sanne M van Neerven
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, 1105 AZ, Amsterdam, the Netherlands
| | - Albert van den Berg
- BIOS Lab on a Chip group, MESA+ Institute for Nanotechnology, University of Twente, P. O. Box 217, 7500 AE Enschede, the Netherlands
| | - Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam and Amsterdam Gastroenterology and Metabolism, Amsterdam University Medical Centers, 1105 AZ, Amsterdam, the Netherlands
| | - Andries D van der Meer
- Applied Stem Cell Technologies, TechMed Centre, University of Twente, P. O. Box 217, 7500 AE Enschede, the Netherlands
| |
Collapse
|
7
|
Oliveira RC, Abrantes AM, Tralhão JG, Botelho MF. The role of mouse models in colorectal cancer research-The need and the importance of the orthotopic models. Animal Model Exp Med 2020; 3:1-8. [PMID: 32318654 PMCID: PMC7167241 DOI: 10.1002/ame2.12102] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/06/2020] [Accepted: 02/21/2020] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer is a worldwide health burden, with high incidence and mortality, especially in the advanced stages of the disease. Preclinical models are very important and valuable to discover and validate early and specific biomarkers as well as new therapeutic targets. In order to accomplish that, the animal models must replicate the clinical evolution of the disease in all of its phases. In this article, we review the existent mouse models, with their strengths and weaknesses in the replication of human cancer disease progression, with major focus on orthotopic models.
Collapse
Affiliation(s)
- Rui C. Oliveira
- Biophysics UnitFaculty of MedicineUniversity of CoimbraCoimbraPortugal
- Pathology DepartmentUniversity Hospital (CHUC)CoimbraPortugal
| | - Ana Margarida Abrantes
- Biophysics UnitFaculty of MedicineUniversity of CoimbraCoimbraPortugal
- Centre of Investigation on Environment, Genetics and Oncobiology (CIMAGO)CoimbraPortugal
| | - José Guilherme Tralhão
- Biophysics UnitFaculty of MedicineUniversity of CoimbraCoimbraPortugal
- Centre of Investigation on Environment, Genetics and Oncobiology (CIMAGO)CoimbraPortugal
- Surgery A DepartmentFaculty of MedicineUniversity Hospital (CHUC)CoimbraPortugal
| | - Maria Filomena Botelho
- Biophysics UnitFaculty of MedicineUniversity of CoimbraCoimbraPortugal
- Centre of Investigation on Environment, Genetics and Oncobiology (CIMAGO)CoimbraPortugal
| |
Collapse
|
8
|
Tian T, Li X, Zhang J. mTOR Signaling in Cancer and mTOR Inhibitors in Solid Tumor Targeting Therapy. Int J Mol Sci 2019; 20:ijms20030755. [PMID: 30754640 PMCID: PMC6387042 DOI: 10.3390/ijms20030755] [Citation(s) in RCA: 421] [Impact Index Per Article: 70.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 01/28/2019] [Accepted: 02/01/2019] [Indexed: 12/12/2022] Open
Abstract
The mammalian or mechanistic target of rapamycin (mTOR) pathway plays a crucial role in regulation of cell survival, metabolism, growth and protein synthesis in response to upstream signals in both normal physiological and pathological conditions, especially in cancer. Aberrant mTOR signaling resulting from genetic alterations from different levels of the signal cascade is commonly observed in various types of cancers. Upon hyperactivation, mTOR signaling promotes cell proliferation and metabolism that contribute to tumor initiation and progression. In addition, mTOR also negatively regulates autophagy via different ways. We discuss mTOR signaling and its key upstream and downstream factors, the specific genetic changes in the mTOR pathway and the inhibitors of mTOR applied as therapeutic strategies in eight solid tumors. Although monotherapy and combination therapy with mTOR inhibitors have been extensively applied in preclinical and clinical trials in various cancer types, innovative therapies with better efficacy and less drug resistance are still in great need, and new biomarkers and deep sequencing technologies will facilitate these mTOR targeting drugs benefit the cancer patients in personalized therapy.
Collapse
Affiliation(s)
- Tian Tian
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| | - Xiaoyi Li
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| | - Jinhua Zhang
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China.
| |
Collapse
|
9
|
Augustine T, Maitra R, Zhang J, Nayak J, Goel S. Sensitization of colorectal cancer to irinotecan therapy by PARP inhibitor rucaparib. Invest New Drugs 2019; 37:948-960. [PMID: 30612311 DOI: 10.1007/s10637-018-00717-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 12/19/2018] [Indexed: 12/11/2022]
Abstract
Intended to explore synthetic lethality and develop better combinatorial regimens, we screened colorectal cancer (CRC) cells using poly ADP-ribose (PAR) polymerase (PARP) inhibitors and cytotoxic agents. We studied four PARP inhibitors and three DNA-damaging agents, and their combinations using sulforhodamine B assay. Rucaparib demonstrated the greatest synergy with irinotecan, followed by olaparib and PJ34. Rucaparib and irinotecan was further subjected to detailed examination to determine combination index (CI) and underlying mechanism of action. Effectiveness and sequence dependence of this combination were assessed in microsatellite stable (MSS) and unstable (MSI) CRC and HCT116 isogenic cell lines. The degree of cell cycle arrest and apoptosis was determined by FACS. In vivo studies were performed to confirm efficacy of this combination. PAR levels in MSI and PARP expression in MSI and MSS cell lines were diminished upon combinatorial treatment. HCT116 isogenic cells revealed the importance of p21, p53 and PTEN in exerting synergy. In MSI cells, administration of rucaparib prior to irinotecan enhanced cytotoxicity compared to other strategies explored. FACS revealed S-phase arrest and increased late-stage apoptosis in MSS, and G2-M arrest and total and early-stage apoptosis in MSI cells. In in vivo murine xenograft models, a significant reduction in tumor volume and expression of Ki67, pancytokeratin and RPS6KB1, and increase in expression of caspase 3 were observed with the combination. In conclusion, among the various combinations studied, rucaparib plus irinotecan was the most synergistic one. Alterations in cell cycle arrest and apoptosis were dependent on MSI status in CRC cells.
Collapse
Affiliation(s)
- Titto Augustine
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Radhashree Maitra
- Department of Medical Oncology, Montefiore Medical Center, Bronx, NY, 10461, USA
| | - Jinghang Zhang
- Department of Microbiology & Immunology and Flow Cytometry Core Facility, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Jay Nayak
- Department of Medical Oncology, Montefiore Medical Center, Bronx, NY, 10461, USA
| | - Sanjay Goel
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA. .,Department of Medical Oncology, Montefiore Medical Center, Bronx, NY, 10461, USA.
| |
Collapse
|
10
|
Pitts TM, Bradshaw-Pierce EL, Bagby SM, Hyatt SL, Selby HM, Spreafico A, Tentler JJ, McPhillips K, Klauck PJ, Capasso A, Diamond JR, Davis SL, Tan AC, Arcaroli JJ, Purkey A, Messersmith WA, Ecsedy JA, Eckhardt SG. Antitumor activity of the aurora a selective kinase inhibitor, alisertib, against preclinical models of colorectal cancer. Oncotarget 2018; 7:50290-50301. [PMID: 27385211 PMCID: PMC5226583 DOI: 10.18632/oncotarget.10366] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 06/17/2016] [Indexed: 12/19/2022] Open
Abstract
Background The Aurora kinases are a family of serine/threonine kinases comprised of Aurora A, B, and C which execute critical steps in mitotic and meiotic progression. Alisertib (MLN8237) is an investigational Aurora A selective inhibitor that has demonstrated activity against a wide variety of tumor types in vitro and in vivo, including CRC. Results CRC cell lines demonstrated varying sensitivity to alisertib with IC50 values ranging from 0.06 to > 5 umol/L. Following exposure to alisertib we observed a decrease in pAurora A, B and C in four CRC cell lines. We also observed an increase in p53 and p21 in a sensitive p53 wildtype cell line in contrast to the p53 mutant cell line or the resistant cell lines. The addition of alisertib to standard CRC treatments demonstrated improvement over single agent arms; however, the benefit was largely less than additive, but not antagonistic. Methods Forty-seven CRC cell lines were exposed to alisertib and IC50s were calculated. Twenty-one PDX models were treated with alisertib and the Tumor Growth Inhibition Index was assessed. Additionally, 5 KRAS wildtype and mutant PDX models were treated with alisertib as single agent or in combination with cetuximab or irinotecan, respectively. Conclusion Alisertib demonstrated anti-proliferative effects against CRC cell lines and PDX models. Our data suggest that the addition of alisertib to standard therapies in colorectal cancer if pursued clinically, will require further investigation of patient selection strategies and these combinations may facilitate future clinical studies.
Collapse
Affiliation(s)
- Todd M Pitts
- Division of Medical Oncology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Erica L Bradshaw-Pierce
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Takeda California, San Diego, CA, USA
| | - Stacey M Bagby
- Division of Medical Oncology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Stephanie L Hyatt
- Division of Medical Oncology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Heather M Selby
- Division of Medical Oncology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Anna Spreafico
- Division of Medical Oncology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - John J Tentler
- Division of Medical Oncology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kelly McPhillips
- Division of Medical Oncology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Peter J Klauck
- Division of Medical Oncology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Anna Capasso
- Division of Medical Oncology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Jennifer R Diamond
- Division of Medical Oncology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - S Lindsey Davis
- Division of Medical Oncology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Aik Choon Tan
- Division of Medical Oncology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - John J Arcaroli
- Division of Medical Oncology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alicia Purkey
- Division of Medical Oncology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Wells A Messersmith
- Division of Medical Oncology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jeffery A Ecsedy
- Millennium Pharmaceuticals, Inc., a wholly owned subsidiary of Takeda Pharmaceutical Company Limited, Cambridge, MA, USA
| | - S Gail Eckhardt
- Division of Medical Oncology, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.,University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
11
|
Song HN, Lee C, Kim ST, Kim SY, Kim NKD, Jang J, Kang M, Jang H, Ahn S, Kim SH, Park Y, Cho YB, Heo JW, Lee WY, Park JO, Lim HY, Kang WK, Park YS, Park WY, Lee J, Kim HC. Molecular characterization of colorectal cancer patients and concomitant patient-derived tumor cell establishment. Oncotarget 2017; 7:19610-9. [PMID: 26909603 PMCID: PMC4991405 DOI: 10.18632/oncotarget.7526] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/18/2016] [Indexed: 12/31/2022] Open
Abstract
Background We aimed to establish a prospectively enrolled colorectal cancer (CRC) cohort for targeted sequencing of primary tumors from CRC patients. In parallel, we established collateral PDC models from the matched primary tumor tissues, which may be later used as preclinical models for genome-directed targeted therapy experiments. Results In all, we identified 27 SNVs in the 6 genes such as PIK3CA (N = 16), BRAF (N = 6), NRAS (N = 2), and CTNNB1 (N = 1), PTEN (N = 1), and ERBB2 (N = 1). RET-NCOA4 translocation was observed in one out of 105 patients (0.9%). PDC models were successfully established from 62 (55.4%) of the 112 samples. To confirm the genomic features of various tumor cells, we compared variant allele frequency results of the primary tumor and progeny PDCs. The Pearson correlation coefficient between the variants from primary tumor cells and PDCs was 0.881. Methods Between April 2014 and June 2015, 112 patients with CRC who underwent resection of the primary tumor were enrolled in the SMC Oncology Biomarker study. The PDC culture protocol was performed for all eligible patients. All of the primary tumors from the 112 patients who provided written informed consent were genomically sequenced with targeted sequencing. In parallel, PDC establishment was attempted for all sequenced tumors. Conclusions We have prospectively sequenced a CRC cohort of 105 patients and successfully established 62 PDC in parallel. Each genomically characterized PDCs can be used as a preclinical model especially in rare genomic alteration event.
Collapse
Affiliation(s)
- Haa-Na Song
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Internal Medicine, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Chung Lee
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Seung Tae Kim
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sun Young Kim
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Nayoung K D Kim
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea
| | - Jiryeon Jang
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Mihyun Kang
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hyojin Jang
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soomin Ahn
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seok Hyeong Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yoona Park
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yong Beom Cho
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeong Wook Heo
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Woo Yong Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Joon Oh Park
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ho Yeong Lim
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won Ki Kang
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young Suk Park
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea.,Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Korea
| | - Jeeyun Lee
- Department of Medicine, Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee Cheol Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
12
|
Lee SC, Kim KH, Kim OH, Lee SK, Hong HE, Choi BJ, Jeong W, Kim SJ. Everolimus Plus Ku0063794 Regimen Promotes Anticancer Effects against Hepatocellular Carcinoma Cells through the Paradoxical Inhibition of Autophagy. Cancer Res Treat 2017; 50:1023-1038. [PMID: 29121714 PMCID: PMC6056969 DOI: 10.4143/crt.2017.085] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 10/30/2017] [Indexed: 12/12/2022] Open
Abstract
Purpose Everolimus only inhibits mammalian target of rapamycin complex 1 (mTORC1), whereas Ku0063794 inhibits both mTORC1 and mTORC2. Although they have similar anticancer effects, their combination has a synergistic effect against hepatocellular carcinoma (HCC) cells. We aimed to determine the mechanism underlying the synergistic effects of everolimus and Ku0063794 associated with autophagy in HCC cells. Materials and Methods We compared the effects of everolimus and Ku0063794, individually or in combination, on both the in vitro and in vivo models of HCCs. Results HepG2 cells treated with both agents had significantly lower rates of cell proliferation and higher apoptosis than the individual monotherapies (p < 0.05). Autophagic studies consistently indicated that, unlike the monotherapies, the combination therapy significantly reduced autophagy (p < 0.05). Autophagic blockage directly promoted the pro-apoptotic effects of combination therapy, suggesting autophagy as the survival mechanism of HCC cells. Unlike the monotherapies, combination therapy showed the potential to inhibit sirtuin 1 (SIRT1), the positive regulator of autophagy. SIRT1 overexpression abrogated the autophagy-inhibiting and pro-apoptotic effects of combination therapy. In a nude mouse xenograft model, the shrinkage of tumors was more prominent in mice treated with combination therapy than in mice treated with the respective monotherapies (p < 0.05). The immunohistochemical and immunofluorescence stains of the tumor obtained from the xenograft model showed that combination therapy had the potential of reducing autophagy and promoting apoptosis. Conclusion The combination of everolimus and Ku0063794 potentiates anticancer effects on HCCs through a decrease in autophagy, which is prompted by SIRT1 downregulation.
Collapse
Affiliation(s)
- Sang Chul Lee
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon, Korea
| | - Kee-Hwan Kim
- Department of Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Korea
| | - Ok-Hee Kim
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.,The Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sang Kuon Lee
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon, Korea
| | - Ha-Eun Hong
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.,The Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Byung Jo Choi
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon, Korea
| | - Wonjun Jeong
- Department of Surgery, Daejeon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Daejeon, Korea
| | - Say-June Kim
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.,The Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
13
|
Mitochondrial phosphoenolpyruvate carboxykinase (PEPCK-M) regulates the cell metabolism of pancreatic neuroendocrine tumors (pNET) and de-sensitizes pNET to mTOR inhibitors. Oncotarget 2017; 8:103613-103625. [PMID: 29262588 PMCID: PMC5732754 DOI: 10.18632/oncotarget.21665] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 09/13/2017] [Indexed: 01/01/2023] Open
Abstract
mTOR pathway activation and hypervascularity have been identified as important characteristics of pancreatic neuroendocrine tumors (pNETs). Agents targeting angiogenesis and mTOR, such as sunitinib and everolimus (RAD001), have been shown to result in progression-free survival of approximately 11 months in patients with advanced pNETs. Novel treatment is needed to extend survival. Mitochondrial phosphoenolpyruvate carboxykinase (PEPCK-M), which is encoded by PCK2, catalyzes the conversion of oxaloacetate to phosphoenolpyruvate. PEPCK-M has been demonstrated to potentiate cytoplasmic phosphoenolpyruvate carboxykinase (PEPCK-C)-mediated gluconeogenesis and to play a critical role in the survival program initiated upon stress during metabolism in cancer cells. Elevated expression of PCK2 has been found in various tumors according to the results of The Cancer Genome Atlas project. However, the role of PEPCK-M aberration in cancers is not well understood. In the current study, we observed that 12 of 21 (57%) pNET patients had high expression of PEPCK-M in the tumors, whereas the normal islet cells had weak expression of PEPCK-M. Knockdown of PCK2 inhibited the proliferation of pNET cells and enhanced the sensitivity of pNET cells to mTOR inhibitors. Knockdown of PCK2 promoted glycolysis but reduced mitochondrial oxidative phosphorylation in pNET cells. The combination of mTOR inhibitors and an anti-glycolysis agent, 2-DG, synergistically or additively inhibited the proliferation of pNET cells, particularly for the cells with high expression of PEPCK-M. Therefore, targeting PEPCK-M or glycolysis combined with inhibiting mTOR is a potential therapeutic approach for the treatment of pNETs.
Collapse
|
14
|
Rattray NJW, Charkoftaki G, Rattray Z, Hansen JE, Vasiliou V, Johnson CH. Environmental influences in the etiology of colorectal cancer: the premise of metabolomics. CURRENT PHARMACOLOGY REPORTS 2017; 3:114-125. [PMID: 28642837 PMCID: PMC5475285 DOI: 10.1007/s40495-017-0088-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW In this review we discuss how environmental exposures predominate the etiology of colorectal cancer (CRC). With CRC being a personalized disease influenced by genes and environment, our goal was to explore the role metabolomics can play in identifying exposures, assessing the interplay between co-exposures, and the development of personalized therapeutic interventions. RECENT FINDINGS Approximately 10 % of CRC cases can be explained by germ-line mutations, whereas the prevailing majority are caused by an initiating exposure event occurring decades prior to diagnosis. Recent research has shown that dietary metabolites are linked to a procarcinogenic or protective environment in the colon which is modulated by the microbiome. In addition, excessive alcohol has been shown to increase the risk of CRC and is dependent on diet (folate), the response of microbiome, and genetic polymorphisms within the folate and alcohol metabolic pathways. Metabolomics can not only be used to identify this modulation of host metabolism, which could affect the progression of the tumors but also response to targeted therapeutics. SUMMARY This review highlights the current understanding of the multifaceted etiology and mechanisms of CRC development but also highlights where the field of metabolomics can contribute to a greater understanding of environmental exposure in CRC.
Collapse
Affiliation(s)
- Nicholas J. W. Rattray
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA, 06520
| | - Georgia Charkoftaki
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA, 06520
| | - Zahra Rattray
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Yale University, CT, USA 06520
| | - James E. Hansen
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, Yale University, CT, USA 06520
- Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT, USA 06520
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA, 06520
| | - Caroline H. Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA, 06520
| |
Collapse
|
15
|
HSP90 inhibitor 17-DMAG exerts anticancer effects against gastric cancer cells principally by altering oxidant-antioxidant balance. Oncotarget 2017; 8:56473-56489. [PMID: 28915605 PMCID: PMC5593576 DOI: 10.18632/oncotarget.17007] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 03/07/2017] [Indexed: 01/13/2023] Open
Abstract
Heat shock protein 90 (HSP90) stabilizes numerous oncoproteins and, therefore, its inhibition has emerged as a promising antineoplastic strategy for diverse malignancies. In this study, we determined the therapeutic effects and mechanisms of action of a specific HSP90 inhibitor, 17-dimethylamino-ethylamino-17-demethoxygeldanamycin (17-DMAG), in gastric cancer cell lines (AGS, SNU-1, and KATO-III), patient-derived tissues, and a mouse xenograft model. 17-DMAG exerted anticancer effects against gastric cancer cells, manifested by significantly decreased proliferation rates (P < 0.05) and increased expression of apoptotic markers. Flow cytometry using dichlorofluorescein (DCF) diacetate revealed that 17-DMAG dose-dependently increases reactive oxygen species (ROS) levels in gastric cancer cells. Inhibition of ROS by N-acetyl-L-cysteine (NAC) abrogated the proapoptotic effects of 17-DMAG, as demonstrated by the decreased expression of proapoptotic proteins. In addition, 17-DMAG dose- and time-dependently reduced the expression of antioxidants such as catalase and glutathione peroxidase (GPx). Moreover, 17-DMAG reduced the expression of nuclear respiratory factor (NRF)-1 and NRF-2, and prevented them from migrating from the cytoplasm to the nucleus dose-dependently. Finally, in a nude mouse xenograft model, the shrinkage of tumors was more prominent in mice treated with 17-DMAG than in control mice (P < 0.05). Taken altogether, our results suggest that 17-DMAG exerts potent antineoplastic activity against gastric cancer cells primarily by promoting ROS generation and suppressing antioxidant enzyme activities.
Collapse
|
16
|
Nguyen A, Moussallieh FM, Mackay A, Cicek AE, Coca A, Chenard MP, Weingertner N, Lhermitte B, Letouzé E, Guérin E, Pencreach E, Jannier S, Guenot D, Namer IJ, Jones C, Entz-Werlé N. Characterization of the transcriptional and metabolic responses of pediatric high grade gliomas to mTOR-HIF-1α axis inhibition. Oncotarget 2017; 8:71597-71617. [PMID: 29069732 PMCID: PMC5641075 DOI: 10.18632/oncotarget.16500] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 01/16/2017] [Indexed: 12/12/2022] Open
Abstract
Pediatric high grade glioma (pHGGs), including sus-tentorial and diffuse intrinsic pontine gliomas, are known to have a very dismal prognosis. For instance, even an increased knowledge on molecular biology driving this brain tumor entity, there is no treatment able to cure those patients. Therefore, we were focusing on a translational pathway able to increase the cell resistance to treatment and to reprogram metabolically tumor cells, which are, then, adapting easily to a hypoxic microenvironment. To establish, the crucial role of the hypoxic pathways in pHGGs, we, first, assessed their protein and transcriptomic deregulations in a pediatric cohort of pHGGs and in pHGG's cell lines, cultured in both normoxic and hypoxic conditions. Secondly, based on the concept of a bi-therapy targeting in pHGGs mTORC1 (rapamycin) and HIF-1α (irinotecan), we hypothesized that the balanced expressions between RAS/ERK, PI3K/AKT and HIF-1α/HIF-2α/MYC proteins or genes may provide a modulation of the cell response to this double targeting. Finally, we could evidence three protein, genomic and metabolomic profiles of response to rapamycin combined with irinotecan. The pattern of highly sensitive cells to mTOR/HIF-1α targeting was linked to a MYC/ERK/HIF-1α over-expression and the cell resistance to a major hyper-expression of HIF-2α.
Collapse
Affiliation(s)
- Aurélia Nguyen
- Laboratory EA 3430, Progression Tumorale et Micro-Environnement, Approches Translationnelles et Epidémiologie, University of Strasbourg, Strasbourg, France
| | | | - Alan Mackay
- Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | - A Ercument Cicek
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA, USA.,Computer Engineering Department, Bilkent University, Cankaya, Ankara, Turkey
| | - Andres Coca
- Department of Neurosurgery, University Hospital of Strasbourg, Strasbourg, France
| | - Marie Pierre Chenard
- Department of Pathology, University Hospital of Strasbourg, Strasbourg, France.,Centre de Ressources Biologiques, University Hospital of Strasbourg, Strasbourg, France
| | - Noelle Weingertner
- Department of Pathology, University Hospital of Strasbourg, Strasbourg, France
| | - Benoit Lhermitte
- Department of Pathology, University Hospital of Strasbourg, Strasbourg, France
| | - Eric Letouzé
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, Paris, France
| | - Eric Guérin
- Laboratory EA 3430, Progression Tumorale et Micro-Environnement, Approches Translationnelles et Epidémiologie, University of Strasbourg, Strasbourg, France
| | - Erwan Pencreach
- Laboratory EA 3430, Progression Tumorale et Micro-Environnement, Approches Translationnelles et Epidémiologie, University of Strasbourg, Strasbourg, France
| | - Sarah Jannier
- Laboratory EA 3430, Progression Tumorale et Micro-Environnement, Approches Translationnelles et Epidémiologie, University of Strasbourg, Strasbourg, France.,Department of Pediatric Onco-hematology, University Hospital of Strasbourg, Strasbourg, France
| | - Dominique Guenot
- Laboratory EA 3430, Progression Tumorale et Micro-Environnement, Approches Translationnelles et Epidémiologie, University of Strasbourg, Strasbourg, France
| | - Izzie Jacques Namer
- Department of Nuclear Medicine, University Hospital of Strasbourg, Strasbourg, France
| | - Chris Jones
- Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | - Natacha Entz-Werlé
- Laboratory EA 3430, Progression Tumorale et Micro-Environnement, Approches Translationnelles et Epidémiologie, University of Strasbourg, Strasbourg, France.,Department of Pediatric Onco-hematology, University Hospital of Strasbourg, Strasbourg, France
| |
Collapse
|
17
|
Lamego I, Marques MPM, Duarte IF, Martins AS, Oliveira H, Gil AM. Impact of the Pd 2Spermine Chelate on Osteosarcoma Metabolism: An NMR Metabolomics Study. J Proteome Res 2017; 16:1773-1783. [PMID: 28244322 DOI: 10.1021/acs.jproteome.7b00035] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
A metabolomics study of Pd2Spermine(Spm) on osteosarcoma MG-63 and osteoblastic HOb cells is presented to assess the impact of the potential palladium drug on cell metabolism compared with cisplatin (cDDP). Despite its higher cytotoxicity, Pd2Spm induced lower (and reversible) metabolic impact on MG-63 cells and the absence of apoptosis; conversely, it induced significant deviations in osteoblastic amino acid metabolism. However, when in combination with doxorubicin and methotrexate, Pd2Spm induced strong metabolic deviations on lipids, choline compounds, amino acids, nucleotides, and compounds related to antioxidative mechanisms (e.g., glutathione, inositol, hypoxanthine), similarly to the cDDP cocktail. Synergetic effects included triggering of lipid biosynthesis by Pd2Spm in the presence of doxorubicin (and reinforced by methotrexate) and changes in the glycosylation substrate uridine diphosphate acetylgalactosamine and methionine and serine metabolisms. This work provides promising results related to the impact of Pd2Spm on osteosarcoma cellular metabolism, particularly in drug combination protocols. Lipid metabolism, glycosylation, and amino acid metabolisms emerge as relevant features for targeted studies to further understand a potential anticancer mechanism of combined Pd2Spm.
Collapse
Affiliation(s)
- Inês Lamego
- Department of Chemistry and CICECO-Aveiro Institute of Materials, (CICECO/UA), University of Aveiro , 3810 Aveiro, Portugal.,R&D Unit "Molecular Physical-Chemistry", University of Coimbra , 3000-213 Coimbra, Portugal
| | - M Paula M Marques
- R&D Unit "Molecular Physical-Chemistry", University of Coimbra , 3000-213 Coimbra, Portugal.,Department of Life Sciences, Faculty of Science and Technology, University of Coimbra , 3000-213 Coimbra, Portugal
| | - Iola F Duarte
- Department of Chemistry and CICECO-Aveiro Institute of Materials, (CICECO/UA), University of Aveiro , 3810 Aveiro, Portugal
| | - Ana S Martins
- Department of Chemistry and CICECO-Aveiro Institute of Materials, (CICECO/UA), University of Aveiro , 3810 Aveiro, Portugal
| | - Helena Oliveira
- Department of Biology & CESAM, University of Aveiro , 3810 Aveiro, Portugal
| | - Ana M Gil
- Department of Chemistry and CICECO-Aveiro Institute of Materials, (CICECO/UA), University of Aveiro , 3810 Aveiro, Portugal
| |
Collapse
|
18
|
Assessment of non-linear combination effect terms for drug-drug interactions. J Pharmacokinet Pharmacodyn 2016; 43:461-79. [PMID: 27638639 DOI: 10.1007/s10928-016-9490-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 08/31/2016] [Indexed: 12/20/2022]
Abstract
Drugs interact with their targets in different ways. A diversity of modeling approaches exists to describe the combination effects of two drugs. We investigate several combination effect terms (CET) regarding their underlying mechanism based on drug-receptor binding kinetics, empirical and statistical summation principles and indirect response models. A list with properties is provided and the interrelationship of the CETs is analyzed. A method is presented to calculate the optimal drug concentration pair to produce the half-maximal combination effect. This work provides a comprehensive overview of typically applied CETs and should shed light into the question as to which CET is appropriate for application in pharmacokinetic/pharmacodynamic models to describe a specific drug-drug interaction mechanism.
Collapse
|
19
|
Hosoi H. Current status of treatment for pediatric rhabdomyosarcoma in the USA and Japan. Pediatr Int 2016; 58:81-7. [PMID: 26646016 DOI: 10.1111/ped.12867] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 11/25/2015] [Indexed: 02/05/2023]
Abstract
This article reviews the current status of treatment for children with rhabdomyosarcoma, according to the four risk groups. Low-risk subgroup A: the Children's Oncology Group in the USA recently performed a clinical trial consisting of a chemotherapy regimen with a shortened treatment period and a reduced drug dosage. Patients in this group received only four cycles of vincristine and actinomycin D (VA) after four cycles of vincristine, actinomycin D, and cyclophosphamide (VAC) with cyclophosphamide (CPM) 1.2 g/m(2) and their outcome was no worse than that obtained with previous regimens. Low-risk subgroup B: although marked improvement in survival was seen with an intensive VAC regimen with CPM 2.2 g/m(2) /cycle (Intergroup Rhabdomyosarcoma Study [IRS]-V, 1997-2004), the total dose of CPM in this regimen caused serious and fatal hepatic veno-occlusive disease during treatment and probably cannot avoid infertility or possible secondary cancer as a late effect. Thereafter, a reduced-dose regimen consisting of four cycles of VAC with CPM 1.2 g/m(2) followed by 12 cycles of VA was investigated in the next study, but the outcome appeared to be worse than in IRS-V. Intermediate-risk group: no significant difference was found between VAC/vincristine, topotecan and cyclophispahamide (VTC) and intensive VAC in IRS-V. The results of a subsequent regimen of VAC with CPM 1.2 g/m(2) alternating with vincristine and irinotecan are awaited. High-risk group: overall survival is approximately 30% and has not improved over the last 25 years. Although 18 month failure-free survival (FFS) was improved with an intensive combination therapy regimen, 36 month FFS dropped to 32% and thus better novel approaches or additive treatments are needed.
Collapse
Affiliation(s)
- Hajime Hosoi
- Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
20
|
Mallick P, Shah P, Gandhi A, Ghose R. Impact of obesity on accumulation of the toxic irinotecan metabolite, SN-38, in mice. Life Sci 2015; 139:132-8. [PMID: 26334566 DOI: 10.1016/j.lfs.2015.08.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 08/19/2015] [Accepted: 08/22/2015] [Indexed: 01/21/2023]
Abstract
AIM Our aim is to investigate the impact of high fat diet-induced obesity on plasma concentrations of the toxic irinotecan metabolite, SN-38, in mice. MAIN METHODS Diet-induced obese (DIO, 60% kcal fed) and lean mice (10% kcal fed) were treated orally with a single dose of 10mg/kg irinotecan to determine pharmacokinetic (PK) parameters. Feces and livers were collected for quantification of irinotecan and its metabolites (SN-38 & SN-38G). SN-38G formation by Ugt1a1 enzyme was analyzed in liver S9 fractions. Expression of the pro-inflammatory cytokine, TNF-α was determined in liver and plasma. Hepatic β-glucuronidase and carboxylesterase enzymes (CES) were also determined. KEY FINDINGS AUC0-8 and Cmax of SN-38 increased by 2-fold in DIO mice compared to their lean controls. This was accompanied by a~2-fold reduction in AUC0-8 and Cmax of SN-38G in DIO mice. There were no differences in the PK parameters of irinotecan in DIO or lean mice. Conversion of SN-38 to SN-38G by Ugt1a1 enzyme was reduced by ~2-fold in liver S9 fractions in DIO mice. Furthermore, in DIO mice, β-glucuronidase activity increased by 2-fold, whereas there was no change in CES activity. TNF-α mRNA expression was 3 fold higher in DIO mice. SIGNIFICANCE Our study demonstrates that reduced hepatic Ugt1a activity during obesity likely contributes to reduced glucuronidation, and results in higher levels of the toxic metabolite, SN-38. Thus, irinotecan dosage should be closely monitored for effective and safe chemotherapy in obese cancer patients who are at a higher risk of developing liver toxicity.
Collapse
Affiliation(s)
- Pankajini Mallick
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA
| | - Pranav Shah
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences, Rockville, MD, USA
| | - Adarsh Gandhi
- Department of Bioanalysis and Physiology, Lundbeck Research Inc. USA, Paramus, NJ, USA
| | - Romi Ghose
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA.
| |
Collapse
|
21
|
Zheng JF, Lu J, Wang XZ, Guo WH, Zhang JX. Comparative Metabolomic Profiling of Hepatocellular Carcinoma Cells Treated with Sorafenib Monotherapy vs. Sorafenib-Everolimus Combination Therapy. Med Sci Monit 2015; 21:1781-91. [PMID: 26092946 PMCID: PMC4479262 DOI: 10.12659/msm.894669] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Sorafenib-everolimus combination therapy may be more effective than sorafenib monotherapy for hepatocellular carcinoma (HCC). To better understand this effect, we comparatively profiled the metabolite composition of HepG2 cells treated with sorafenib, everolimus, and sorafenib-everolimus combination therapy. Material/Methods A 2D HRMAS 1H-NMR metabolomic approach was applied to identify the key differential metabolites in 3 experimental groups: sorafenib (5 μM), everolimus (5 μM), and combination therapy (5 μM sorafenib +5 μM everolimus). MetaboAnalyst 3.0 was used to perform pathway analysis. Results All OPLS-DA models displayed good separation between experimental groups, high-quality goodness of fit (R2), and high-quality goodness of predication (Q2). Sorafenib and everolimus have differential effects with respect to amino acid, methane, pyruvate, pyrimidine, aminoacyl-tRNA biosynthesis, and glycerophospholipid metabolism. The addition of everolimus to sorafenib resulted in differential effects with respect to pyruvate, amino acid, methane, glyoxylate and dicarboxylate, glycolysis or gluconeogenesis, glycerophospholipid, and purine metabolism. Conclusions Sorafenib and everolimus have differential effects on HepG2 cells. Sorafenib preferentially affects glycerophospholipid and purine metabolism, while the addition of everolimus preferentially affects pyruvate, amino acid, and glucose metabolism. This phenomenon may explain (in part) the synergistic effects of sorafenib-everolimus combination therapy observed in vivo.
Collapse
Affiliation(s)
- Jian-Feng Zheng
- The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Juan Lu
- Jiangxi Children's Hospital, Nanchang, Jiangxi, China (mainland)
| | - Xiao-Zhong Wang
- The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Wu-Hua Guo
- The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Ji-Xiang Zhang
- The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| |
Collapse
|
22
|
Davis SL, Robertson KM, Pitts TM, Tentler JJ, Bradshaw-Pierce EL, Klauck PJ, Bagby SM, Hyatt SL, Selby HM, Spreafico A, Ecsedy JA, Arcaroli JJ, Messersmith WA, Tan AC, Eckhardt SG. Combined inhibition of MEK and Aurora A kinase in KRAS/PIK3CA double-mutant colorectal cancer models. Front Pharmacol 2015; 6:120. [PMID: 26136684 PMCID: PMC4468631 DOI: 10.3389/fphar.2015.00120] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 05/21/2015] [Indexed: 12/22/2022] Open
Abstract
Aurora A kinase and MEK inhibitors induce different, and potentially complementary, effects on the cell cycle of malignant cells, suggesting a rational basis for utilizing these agents in combination. In this work, the combination of an Aurora A kinase and MEK inhibitor was evaluated in pre-clinical colorectal cancer models, with a focus on identifying a subpopulation in which it might be most effective. Increased synergistic activity of the drug combination was identified in colorectal cancer cell lines with concomitant KRAS and PIK3CA mutations. Anti-proliferative effects were observed upon treatment of these double-mutant cell lines with the drug combination, and tumor growth inhibition was observed in double-mutant human tumor xenografts, though effects were variable within this subset. Additional evaluation suggests that degree of G2/M delay and p53 mutation status affect apoptotic activity induced by combination therapy with an Aurora A kinase and MEK inhibitor in KRAS and PIK3CA mutant colorectal cancer. Overall, in vitro and in vivo testing was unable to identify a subset of colorectal cancer that was consistently responsive to the combination of a MEK and Aurora A kinase inhibitor.
Collapse
Affiliation(s)
- S Lindsey Davis
- Division of Medical Oncology, Department of Internal Medicine, University of Colorado Anschutz Medical Campus Aurora, CO, USA ; University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus Aurora, CO, USA
| | - Kelli M Robertson
- Division of Medical Oncology, Department of Internal Medicine, University of Colorado Anschutz Medical Campus Aurora, CO, USA
| | - Todd M Pitts
- Division of Medical Oncology, Department of Internal Medicine, University of Colorado Anschutz Medical Campus Aurora, CO, USA ; University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus Aurora, CO, USA
| | - John J Tentler
- Division of Medical Oncology, Department of Internal Medicine, University of Colorado Anschutz Medical Campus Aurora, CO, USA ; University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus Aurora, CO, USA
| | - Erica L Bradshaw-Pierce
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus Aurora, CO, USA ; Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus Aurora, CO, USA ; Department of Drug Metabolism and Pharmacokinetics, Takeda California, Inc. San Diego, CA, USA
| | - Peter J Klauck
- Division of Medical Oncology, Department of Internal Medicine, University of Colorado Anschutz Medical Campus Aurora, CO, USA
| | - Stacey M Bagby
- Division of Medical Oncology, Department of Internal Medicine, University of Colorado Anschutz Medical Campus Aurora, CO, USA
| | - Stephanie L Hyatt
- Division of Medical Oncology, Department of Internal Medicine, University of Colorado Anschutz Medical Campus Aurora, CO, USA
| | - Heather M Selby
- Division of Medical Oncology, Department of Internal Medicine, University of Colorado Anschutz Medical Campus Aurora, CO, USA
| | - Anna Spreafico
- Division of Medical Oncology, Department of Internal Medicine, University of Colorado Anschutz Medical Campus Aurora, CO, USA
| | - Jeffrey A Ecsedy
- Department of Translational Medicine, Millenium Pharmaceuticals, Inc., A wholly owned Subsidiary of a Takeda Pharmaceutical Company Limited Cambridge, MA, USA
| | - John J Arcaroli
- Division of Medical Oncology, Department of Internal Medicine, University of Colorado Anschutz Medical Campus Aurora, CO, USA ; University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus Aurora, CO, USA
| | - Wells A Messersmith
- Division of Medical Oncology, Department of Internal Medicine, University of Colorado Anschutz Medical Campus Aurora, CO, USA ; University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus Aurora, CO, USA
| | - Aik Choon Tan
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus Aurora, CO, USA
| | - S Gail Eckhardt
- Division of Medical Oncology, Department of Internal Medicine, University of Colorado Anschutz Medical Campus Aurora, CO, USA ; University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus Aurora, CO, USA
| |
Collapse
|
23
|
Afonso J, Longatto-Filho A, DA Silva VM, Amaro T, Santos LL. Phospho-mTOR in non-tumour and tumour bladder urothelium: Pattern of expression and impact on urothelial bladder cancer patients. Oncol Lett 2014; 8:1447-1454. [PMID: 25202348 PMCID: PMC4156165 DOI: 10.3892/ol.2014.2392] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 06/24/2014] [Indexed: 12/28/2022] Open
Abstract
Urothelial bladder carcinoma (UBC) is heterogeneous in its pathology and clinical behaviour. Evaluation of prognostic and predictive biomarkers is necessary, in order to produce personalised treatment options. The present study used immunohistochemistry to evaluate UBC sections containing tumour and non-tumour areas from 76 patients, for the detection of p-mTOR, CD31 and D2-40 (blood and lymphatic vessels identification, respectively). Of the non-tumour and tumour sections, 36 and 20% were scored positive for p-mTOR expression, respectively. Immunoexpression was observed in umbrella cells from non-tumour urothelium, in all cell layers from non-muscle-invasive (NMI) tumours (including expression in superficial cells), and in spots of cells from muscle-invasive (MI) tumours. Positive expression decreased from non-tumour to tumour urothelium, and from pT1/pTis to pT3/pT4 tumours; however, the few pT3/pT4 positive cases had worse survival rates, with 5-year disease-free survival being significantly lower. Angiogenesis occurrence was impaired in pT3/pT4 tumours that did not express p-mTOR. In conclusion, p-mTOR expression in non-tumour umbrella cells is likely a reflection of their metabolic plasticity, and extension to the inner layers of the urothelium in NMI tumours is consistent with an enhanced malignant potential. The expression in cell spots in a few MI tumours and absence of expression in the remaining tumours is intriguing and requires further research. Additional studies regarding the up- and downstream effectors of the mTOR pathway should be conducted.
Collapse
Affiliation(s)
- Julieta Afonso
- Life and Health Sciences Research Institute (ICVS), School of Healh Sciences (ECS) University of Minho, Braga 4710-057, Portugal ; ICVS/3B's, PT Government Associate Laboratory, Braga 4710-057/Guimarães 4806-909, Portugal
| | - Adhemar Longatto-Filho
- Life and Health Sciences Research Institute (ICVS), School of Healh Sciences (ECS) University of Minho, Braga 4710-057, Portugal ; ICVS/3B's, PT Government Associate Laboratory, Braga 4710-057/Guimarães 4806-909, Portugal ; Laboratory of Medical Investigation (LIM 14), Faculty of Medicine, São Paulo State University, São Paulo 01246-000, Brazil ; Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo 14784-400, Brazil
| | | | - Teresina Amaro
- Experimental Pathology and Therapeutics Research Center, Portuguese Institute of Oncology (IPO), Porto 4200-072, Portugal
| | - Lúcio L Santos
- Department of Surgical Oncology, Portuguese Institute of Oncology (IPO), Porto 4200-072, Portugal ; Faculty of Health Sciences, University Fernando Pessoa, Porto 4200-150, Portugal
| |
Collapse
|
24
|
A phase I trial of everolimus in combination with 5-FU/LV, mFOLFOX6 and mFOLFOX6 plus panitumumab in patients with refractory solid tumors. Cancer Chemother Pharmacol 2014; 74:117-23. [PMID: 24819684 DOI: 10.1007/s00280-014-2474-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 04/23/2014] [Indexed: 12/22/2022]
Abstract
PURPOSE This phase I study investigated the safety, dose-limiting toxicity, and efficacy in three cohorts all treated with the mTOR inhibitor everolimus that was delivered (1) in combination with 5-fluorouracil with leucovorin (5-FU/LV), (2) with mFOLFOX6 (5-FU/LV + oxaliplatin), and (3) with mFOLFOX6 + panitumumab in patients with refractory solid tumors. METHODS Patients were accrued using a 3-patient cohort design consisting of two sub-trials in which the maximum tolerated combination (MTC) and dose-limiting toxicity (DLT) of everolimus and 5-FU/LV was established in Sub-trial A and of everolimus in combination with mFOLFOX6 and mFOLFOX6 plus panitumumab in Sub-trial B. RESULTS Thirty-six patients were evaluable for toxicity, 21 on Sub-trial A and 15 on Sub-trial B. In Sub-trial A, DLT was observed in 1/6 patients enrolled on dose level 1A and 2/3 patients in level 6A. In Sub-trial B, 2/3 patients experienced DLT on level 1B and subsequent patients were enrolled on level 1B-1 without DLT. Three of six patients in cohort 2B-1 experienced grade 3 mucositis, and further study of the combination of everolimus, mFOLFOX6 and panitumumab was aborted. Among the 24 patients enrolled with refractory metastatic colorectal cancer, the median time on treatment was 2.7 months with 45 % of patients remaining on treatment with stable disease for at least 3 months. CONCLUSIONS While a regimen of everolimus in addition to 5-FU/LV and mFOLFOX6 appears safe and tolerable, the further addition of panitumumab resulted in an unacceptable level of toxicity that cannot be recommended for further study. Further investigation is warranted to better elucidate the role which mTOR inhibitors play in patients with refractory solid tumors, with a specific focus on mCRC as a potential for the combination of this targeted and cytotoxic therapy in future studies.
Collapse
|
25
|
Wang XW, Zhang YJ. Targeting mTOR network in colorectal cancer therapy. World J Gastroenterol 2014; 20:4178-88. [PMID: 24764656 PMCID: PMC3989954 DOI: 10.3748/wjg.v20.i15.4178] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 12/28/2013] [Accepted: 01/20/2014] [Indexed: 02/06/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) integrates growth factor signals with cellular nutrient and energy levels and coordinates cell growth, proliferation and survival. A regulatory network with multiple feedback loops has evolved to ensure the exquisite regulation of cell growth and division. Colorectal cancer is the most intensively studied cancer because of its high incidence and mortality rate. Multiple genetic alterations are involved in colorectal carcinogenesis, including oncogenic Ras activation, phosphatidylinositol 3-kinase pathway hyperactivation, p53 mutation, and dysregulation of wnt pathway. Many oncogenic pathways activate the mTOR pathway. mTOR has emerged as an effective target for colorectal cancer therapy. In vitro and preclinical studies targeting the mTOR pathway for colorectal cancer chemotherapy have provided promising perspectives. However, the overall objective response rates in major solid tumors achieved with single-agent rapalog therapy have been modest, especially in advanced metastatic colorectal cancer. Combination regimens of mTOR inhibitor with agents such as cytotoxic chemotherapy, inhibitors of vascular endothelial growth factor, epidermal growth factor receptor and Mitogen-activated protein kinase kinase (MEK) inhibitors are being intensively studied and appear to be promising. Further understanding of the molecular mechanism in mTOR signaling network is needed to develop optimized therapeutic regimens. In this paper, oncogenic gene alterations in colorectal cancer, as well as their interaction with the mTOR pathway, are systematically summarized. The most recent preclinical and clinical anticancer therapeutic endeavors are reviewed. New players in mTOR signaling pathway, such as non-steroidal anti-inflammatory drug and metformin with therapeutic potentials are also discussed here.
Collapse
|
26
|
Bezabeh T, Ijare OB, Nikulin AE, Somorjai RL, Smith IC. MRS-based Metabolomics in Cancer Research. MAGNETIC RESONANCE INSIGHTS 2014; 7:1-14. [PMID: 25114549 PMCID: PMC4122556 DOI: 10.4137/mri.s13755] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 12/30/2013] [Accepted: 12/30/2013] [Indexed: 12/18/2022]
Abstract
Metabolomics is a relatively new technique that is gaining importance very rapidly. MRS-based metabolomics, in particular, is becoming a useful tool in the study of body fluids, tissue biopsies and whole organisms. Advances in analytical techniques and data analysis methods have opened a new opportunity for such technology to contribute in the field of diagnostics. In the MRS approach to the diagnosis of disease, it is important that the analysis utilizes all the essential information in the spectra, is robust, and is non-subjective. Although some of the data analytic methods widely used in chemical and biological sciences are sketched, a more extensive discussion is given of a 5-stage Statistical Classification Strategy. This proposes powerful feature selection methods, based on, for example, genetic algorithms and novel projection techniques. The applications of MRS-based metabolomics in breast cancer, prostate cancer, colorectal cancer, pancreatic cancer, hepatobiliary cancers, gastric cancer, and brain cancer have been reviewed. While the majority of these applications relate to body fluids and tissue biopsies, some in vivo applications have also been included. It should be emphasized that the number of subjects studied must be sufficiently large to ensure a robust diagnostic classification. Before MRS-based metabolomics can become a widely used clinical tool, however, certain challenges need to be overcome. These include manufacturing user-friendly commercial instruments with all the essential features, and educating physicians and medical technologists in the acquisition, analysis, and interpretation of metabolomics data.
Collapse
Affiliation(s)
- Tedros Bezabeh
- Department of Chemistry, University of Winnipeg, Winnipeg, Manitoba, Canada. ; Human Nutritional Sciences, University of Manitoba, Winnipeg, Manitoba, Canada. ; Innovative Biodiagnostics Inc, Winnipeg, Manitoba, Canada
| | - Omkar B Ijare
- Department of Chemistry, University of Winnipeg, Winnipeg, Manitoba, Canada. ; Innovative Biodiagnostics Inc, Winnipeg, Manitoba, Canada
| | | | | | - Ian Cp Smith
- Department of Chemistry, University of Winnipeg, Winnipeg, Manitoba, Canada. ; Departments of Anatomy and Human Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada. ; Innovative Biodiagnostics Inc, Winnipeg, Manitoba, Canada
| |
Collapse
|
27
|
Investigation of the Material Basis Underlying the Correlation between Presbycusis and Kidney Deficiency in Traditional Chinese Medicine via GC/MS Metabolomics. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:762092. [PMID: 24371466 PMCID: PMC3858872 DOI: 10.1155/2013/762092] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 10/24/2013] [Accepted: 10/24/2013] [Indexed: 02/06/2023]
Abstract
Objective. To investigate the correlation between presbycusis and kidney deficiency as defined by traditional Chinese medicine (TCM) and its material basis from the perspective of metabolism. Methods. Pure-tone audiometry was used to test auditory function. A kidney deficiency symptom scoring table was used to measure the kidney deficiency accumulated scores of the research subjects. Gas chromatography/mass spectrometry (GC/MS) was used to measure the metabolites in the urine samples from 11 presbycusis patients and 9 elderly people with normal hearing. Results. Hearing loss in the elderly was positively correlated with kidney deficiency score in TCM. There were significant differences in urine metabolite profile between the presbycusis patients and the controls. A total of 23 differentially expressed metabolites were found. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that these metabolites were related to glutathione metabolism, amino acid metabolism, glucose metabolism, the N-methyl-D-aspartic acid (NMDA) receptor pathway, and the γ -aminobutyric acid (GABA) receptor pathway. Conclusion. Glutathione metabolism, amino acid metabolism, glucose metabolism, NMDA receptors, and GABA receptors may be related to the pathogenesis of presbycusis and may be the material basis underlying the correlation between presbycusis and kidney deficiency in TCM.
Collapse
|