1
|
Wang Y, Lei F, Lin Y, Han Y, Yang L, Tan H. Peroxisome proliferator-activated receptors as therapeutic target for cancer. J Cell Mol Med 2024; 28:e17931. [PMID: 37700501 PMCID: PMC10902584 DOI: 10.1111/jcmm.17931] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 08/05/2023] [Accepted: 08/18/2023] [Indexed: 09/14/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are transcription factors belonging to the nuclear receptor family. There are three subtypes of PPARs, including PPAR-α, PPAR-β/δ and PPAR-γ. They are expressed in different tissues and act by regulating the expression of target genes in the form of binding to ligands. Various subtypes of PPAR have been shown to have significant roles in a wide range of biological processes including lipid metabolism, body energy homeostasis, cell proliferation and differentiation, bone formation, tissue repair and remodelling. Recent studies have found that PPARs are closely related to tumours. They are involved in cancer cell growth, angiogenesis and tumour immune response, and are essential components in tumour progression and metastasis. As such, they have become a target for cancer therapy research. In this review, we discussed the current state of knowledge on the involvement of PPARs in cancer, including their role in tumourigenesis, the impact of PPARs in tumour microenvironment and the potential of using PPARs combinational therapy to treat cancer by targeting essential signal pathways, or as adjuvants to boost the effects of current chemo and immunotherapies. Our review highlights the complexity of PPARs in cancer and the need for a better understanding of the mechanism in order to design effective cancer therapies.
Collapse
Affiliation(s)
- Yuqing Wang
- Department of Internal MedicineMontefiore Medical Center, Wakefield CampusBronxNew YorkUSA
| | - Feifei Lei
- Department of Infectious Disease, Lab of Liver Disease, Renmin HospitalHubei University of MedicineShiyanChina
| | - Yiyun Lin
- Department of Biomedical SciencesUniversity of Texas, MD Anderson Cancer CenterHoustonTexasUSA
| | - Yuru Han
- Qinghai Provincial People's HospitalXiningChina
| | - Lei Yang
- Department of Biomedical SciencesUniversity of Texas, MD Anderson Cancer CenterHoustonTexasUSA
| | - Huabing Tan
- Department of Infectious Disease, Lab of Liver Disease, Renmin HospitalHubei University of MedicineShiyanChina
| |
Collapse
|
2
|
Lavudi K, Nuguri SM, Olverson Z, Dhanabalan AK, Patnaik S, Kokkanti RR. Targeting the retinoic acid signaling pathway as a modern precision therapy against cancers. Front Cell Dev Biol 2023; 11:1254612. [PMID: 37645246 PMCID: PMC10461636 DOI: 10.3389/fcell.2023.1254612] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 07/28/2023] [Indexed: 08/31/2023] Open
Abstract
Retinoic acid (RA) is a vital metabolite derived from vitamin A. RA plays a prominent role during development, which helps in embryological advancement and cellular differentiation. Mechanistically, RA binds to its definite nuclear receptors including the retinoic acid receptor and retinoid X receptor, thus triggering gene transcription and further consequences in gene regulation. This functional heterodimer activation later results in gene activation/inactivation. Several reports have been published related to the detailed embryonic and developmental role of retinoic acids and as an anti-cancer drug for specific cancers, including acute promyelocytic leukemia, breast cancer, and prostate cancer. Nonetheless, the other side of all-trans retinoic acid (ATRA) has not been explored widely yet. In this review, we focused on the role of the RA pathway and its downstream gene activation in relation to cancer progression. Furthermore, we explored the ways of targeting the retinoic acid pathway by focusing on the dual role of aldehyde dehydrogenase (ALDH) family enzymes. Combination strategies by combining RA targets with ALDH-specific targets make the tumor cells sensitive to the treatment and improve the progression-free survival of the patients. In addition to the genomic effects of ATRA, we also highlighted the role of ATRA in non-canonical mechanisms as an immune checkpoint inhibitor, thus targeting the immune oncological perspective of cancer treatments in the current era. The role of ATRA in activating independent mechanisms is also explained in this review. This review also highlights the current clinical trials of ATRA in combination with other chemotherapeutic drugs and explains the future directional insights related to ATRA usage.
Collapse
Affiliation(s)
- Kousalya Lavudi
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, United States
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| | - Shreya Madhav Nuguri
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, United States
| | - Zianne Olverson
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH, United States
| | - Anantha Krishna Dhanabalan
- Centre for Advanced Study in Crystallography and Biophysics, University of Madras, Guindy Campus, Chennai, India
| | - Srinivas Patnaik
- School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| | - Rekha Rani Kokkanti
- Department of Biotechnology, Sri Padmavati Mahila Visvavidyalayam, Tirupati, Andhra Pradesh, India
| |
Collapse
|
3
|
Takeuchi A, Endo M, Kawai A, Nishida Y, Terauchi R, Matsumine A, Aiba H, Nakamura T, Tandai S, Ozaki T, Hoshi M, Kayano D, Okuda M, Yamamoto N, Hayashi K, Miwa S, Igarashi K, Yoshimura K, Nomura A, Murayama T, Tsuchiya H. Randomized placebo-controlled double-blind phase II study of zaltoprofen for patients with diffuse-type and unresectable localized tenosynovial giant cell tumors: The REALIZE study. Front Oncol 2022; 12:900010. [PMID: 36212437 PMCID: PMC9533097 DOI: 10.3389/fonc.2022.900010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Background A tenosynovial giant cell tumor (TGCT) is a locally aggressive benign neoplasm arising from intra- or extra-articular tissue, categorized as localized (L-TGCT, solitary lesion) and diffuse (D-TGCT, multiple lesions) TGCT. Surgical excision is the mainstay of the treatment, and a high local recurrence rate of approximately 50% has been reported. We focused on zaltoprofen, a nonsteroidal anti-inflammatory drug that can activate peroxisome proliferator-activated receptor gamma (PPARγ) and inhibit the proliferation of TGCT stromal cells. Therefore, we conducted a randomized trial to evaluate the safety and effectiveness of zaltoprofen in patients with D-TGCTs or unresectable L-TGCTs. Methods This randomized, placebo-controlled, double-blind, multicenter trial evaluated the safety and efficacy of zaltoprofen. In the treatment group, zaltoprofen (480 mg/day) was administered for 48 weeks; the placebo group received similar dosages without zaltoprofen. The primary outcome was progression-free rate (PFR) 48 weeks after treatment administration. Disease progression was defined as the following conditions requiring surgical intervention: 1) repetitive joint swelling due to hemorrhage, 2) joint range of motion limitation, 3) invasion of the adjacent cartilage or bone, 4) severe joint space narrowing, and 5) increased tumor size (target lesion). Results Forty-one patients were allocated to the zaltoprofen (n=21) or placebo (n=20) groups. The PFR was not significant between the zaltoprofen group and the placebo group at 48 weeks (84.0% and 90.0%, respectively; p=0.619). The mean Japanese Orthopedic Association knee score significantly improved from baseline to week 48 in the zaltoprofen group (85.38 versus 93.75, p=0.027). There was a significant difference between the values at 48 weeks of placebo and zaltoprofen group (p=0.014). One severe adverse event (grade 3 hypertension) was observed in the zaltoprofen group. Discussion This is the first study to evaluate the efficacy and safety of zaltoprofen in patients with TGCT. No significant differences in PFR were observed between the groups at 48 weeks. Physical function significantly improved after zaltoprofen treatment. The safety profile of zaltoprofen was acceptable. This less invasive and safer treatment with zaltoprofen, compared to surgical removal, could be justified as a novel approach to treating TGCT. Further analysis of long-term administration of zaltoprofen should be considered in future studies. Clinical Trial Registration University Hospital Medical Information Network Clinical Trials Registry, identifier (UMIN000025901).
Collapse
Affiliation(s)
- Akihiko Takeuchi
- Department of Orthopaedic Surgery, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Makoto Endo
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akira Kawai
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yoshihiro Nishida
- Department of Orthopaedic Surgery, Nagoya University School of Medicine, Nagoya, Japan
| | - Ryu Terauchi
- Department of Orthopaedic Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akihiko Matsumine
- Department of Orthopaedics and Rehabilitation Medicine, Unit of Surgery, Division of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Hisaki Aiba
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Tomoki Nakamura
- Department of Orthopedic Surgery, Mie University School of Medicine, Mie, Japan
| | - Susumu Tandai
- Department of Orthopaedic Surgery, Asahikawa Medical University, Hokkaido, Japan
| | - Toshifumi Ozaki
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Manabu Hoshi
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Daiki Kayano
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa, Japan
| | - Miho Okuda
- Department of Radiology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Norio Yamamoto
- Department of Orthopaedic Surgery, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Katsuhiro Hayashi
- Department of Orthopaedic Surgery, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Shinji Miwa
- Department of Orthopaedic Surgery, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Kentaro Igarashi
- Department of Orthopaedic Surgery, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Kenichi Yoshimura
- Future Medical Center, Hiroshima University Hospital, Hiroshima, Japan
| | - Akihiro Nomura
- Innovative Clinical Research Center (iCREK), Kanazawa University Hospital, Kanazawa, Japan
| | - Toshinori Murayama
- Innovative Clinical Research Center (iCREK), Kanazawa University Hospital, Kanazawa, Japan
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic Surgery, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
- *Correspondence: Hiroyuki Tsuchiya,
| |
Collapse
|
4
|
Morosi L, Matteo C, Meroni M, Ceruti T, Fuso Nerini I, Bello E, Frapolli R, D'Incalci M, Zucchetti M, Davoli E. Quantitative measurement of pioglitazone in neoplastic and normal tissues by AP-MALDI mass spectrometry imaging. Talanta 2022; 237:122918. [PMID: 34736656 DOI: 10.1016/j.talanta.2021.122918] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/24/2021] [Accepted: 09/29/2021] [Indexed: 11/26/2022]
Abstract
Pioglitazone is a Peroxisome Proliferator-Activated Receptor (PPAR) agonist of the thiazolidinedione class of compounds with promising anticancer activity. An innovative quantitative mass spectrometry imaging (MSI) method and a HPLC-UV method were developed and validated to investigate its distribution in tumor and liver tissues. The MSI method is based on stable isotope normalization and resulted highly specific and sensitive (0.2 pmol/spot). The correct identification of the drug ion signal is confirmed by MS/MS analysis on tissue. The method shows an optimal lateral resolution (25 μm) relying on the ionization efficiency and fine laser diameter of the atmospheric pressure MALDI source. The HPLC-UV method is simple and straightforward involving quick protein precipitation and shows good sensitivity (50ng/sample) using a small starting volume of biological sample. Thus, it is applicable to samples obtained from both preclinical models and clinical surgical procedures. MSI and HPLC-UV assays were validated assessing linearity, intra- and inter-day precision and accuracy, limit of quantification, selectivity and recovery. These are the first methods developed and validated for the analysis of pioglitazone in tissues, and they were applied successfully to myxoid liposarcoma xenograft-bearing mice, which received clinically relevant drug doses. Pioglitazone was measured by either method in sections of tumor and liver 2, 6 and 24 h post-treatment. Drug distribution was relatively homogeneous.
Collapse
Affiliation(s)
- Lavinia Morosi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy
| | - Cristina Matteo
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy
| | - Marina Meroni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy
| | - Tommaso Ceruti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy
| | - Ilaria Fuso Nerini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy
| | - Ezia Bello
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy
| | - Roberta Frapolli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy.
| | - Maurizio D'Incalci
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy
| | - Massimo Zucchetti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Oncology, Via Mario Negri 2, Milan, Italy
| | - Enrico Davoli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Mass Spectrometry Research Center for Health and Environment and Laboratory of Mass Spectrometry, Via Mario Negri 2, Milan, Italy
| |
Collapse
|
5
|
Chi T, Wang M, Wang X, Yang K, Xie F, Liao Z, Wei P. PPAR-γ Modulators as Current and Potential Cancer Treatments. Front Oncol 2021; 11:737776. [PMID: 34631571 PMCID: PMC8495261 DOI: 10.3389/fonc.2021.737776] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/27/2021] [Indexed: 12/12/2022] Open
Abstract
Worldwide, cancer has become one of the leading causes of mortality. Peroxisome Proliferator-Activated Receptors (PPARs) is a family of critical sensors of lipids as well as regulators of diverse metabolic pathways. They are also equipped with the capability to promote eNOS activation, regulate immunity and inflammation response. Aside from the established properties, emerging discoveries are also made in PPAR's functions in the cancer field. All considerations are given, there exists great potential in PPAR modulators which may hold in the management of cancers. In particular, PPAR-γ, the most expressed subtype in adipose tissues with two isoforms of different tissue distribution, has been proven to be able to inhibit cell proliferation, induce cell cycle termination and apoptosis of multiple cancer cells, promote intercellular adhesion, and cripple the inflamed state of tumor microenvironment, both on transcriptional and protein level. However, despite the multi-functionalities, the safety of PPAR-γ modulators is still of clinical concern in terms of dosage, drug interactions, cancer types and stages, etc. This review aims to consolidate the functions of PPAR-γ, the current and potential applications of PPAR-γ modulators, and the challenges in applying PPAR-γ modulators to cancer treatment, in both laboratory and clinical settings. We sincerely hope to provide a comprehensive perspective on the prospect of PPAR-γ applicability in the field of cancer treatment.
Collapse
Affiliation(s)
- Tiange Chi
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,First Clinical Medical School, Beijing University of Chinese Medicine, Beijing, China
| | - Mina Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,Department of Acupuncture and Moxibustion, Beijing Key Laboratory of Acupuncture Neuromodulation, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xu Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ke Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Feiyu Xie
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,Oncology Department, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Zehuan Liao
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.,Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Peng Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
6
|
Loo SY, Syn NL, Koh APF, Teng JCF, Deivasigamani A, Tan TZ, Thike AA, Vali S, Kapoor S, Wang X, Wang JW, Tan PH, Yip GW, Sethi G, Huang RYJ, Hui KM, Wang L, Goh BC, Kumar AP. Epigenetic derepression converts PPARγ into a druggable target in triple-negative and endocrine-resistant breast cancers. Cell Death Discov 2021; 7:265. [PMID: 34580286 PMCID: PMC8476547 DOI: 10.1038/s41420-021-00635-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 08/23/2021] [Accepted: 09/03/2021] [Indexed: 01/04/2023] Open
Abstract
Clinical trials repurposing peroxisome proliferator-activated receptor-gamma (PPARγ) agonists as anticancer agents have exhibited lackluster efficacy across a variety of tumor types. Here, we report that increased PPARG expression is associated with a better prognosis but is anticorrelated with histone deacetylase (HDAC) 1 and 2 expressions. We show that HDAC overexpression blunts anti-proliferative and anti-angiogenic responses to PPARγ agonists via transcriptional and post-translational mechanisms, however, these can be neutralized with clinically approved and experimental HDAC inhibitors. Supporting this notion, concomitant treatment with HDAC inhibitors was required to license the tumor-suppressive effects of PPARγ agonists in triple-negative and endocrine-refractory breast cancer cells, and combination therapy also restrained angiogenesis in a tube formation assay. This combination was also synergistic in estrogen receptor-alpha (ERα)-positive cells because HDAC blockade abrogated ERα interference with PPARγ-regulated transcription. Following a pharmacokinetics optimization study, the combination of rosiglitazone and a potent pan-HDAC inhibitor, LBH589, stalled disease progression in a mouse model of triple-negative breast cancer greater than either of the monotherapies, while exhibiting a favorable safety profile. Our findings account for historical observations of de-novo resistance to PPARγ agonist monotherapy and propound a therapeutically cogent intervention against two aggressive breast cancer subtypes.
Collapse
Affiliation(s)
- Ser Yue Loo
- Cancer Science Institute of Singapore and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Genome Institute of Singapore, A*STAR, Singapore, Singapore
| | - Nicholas L Syn
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Angele Pei-Fern Koh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Janet Cheng-Fei Teng
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Amudha Deivasigamani
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Aye Aye Thike
- Department of Pathology, Singapore General Hospital, Singapore, Singapore
| | - Shireen Vali
- Cellworks Research India Pvt. Ltd., Bengaluru, India
| | - Shweta Kapoor
- Cellworks Research India Pvt. Ltd., Bengaluru, India
| | - Xiaoyuan Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Cardiovascular Research Institute (CVRI), National University Heart Centre, Singapore (NUHCS), National University Health System, Singapore, Singapore
| | - Jiong Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Puay Hoon Tan
- Department of Pathology, Singapore General Hospital, Singapore, Singapore
| | - George W Yip
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ruby Yun-Ju Huang
- School of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Kam Man Hui
- Division of Cellular and Molecular Research, National Cancer Centre Singapore, Singapore, Singapore
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Boon Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,National University Cancer Institute, National University Health System, Singapore, Singapore.,Department of Haematology-Oncology, National University Hospital, National University Health System, Singapore, Singapore
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore. .,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,National University Cancer Institute, National University Health System, Singapore, Singapore.
| |
Collapse
|
7
|
Islam Z, Ali AM, Naik A, Eldaw M, Decock J, Kolatkar PR. Transcription Factors: The Fulcrum Between Cell Development and Carcinogenesis. Front Oncol 2021; 11:681377. [PMID: 34195082 PMCID: PMC8236851 DOI: 10.3389/fonc.2021.681377] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/26/2021] [Indexed: 12/15/2022] Open
Abstract
Higher eukaryotic development is a complex and tightly regulated process, whereby transcription factors (TFs) play a key role in controlling the gene regulatory networks. Dysregulation of these regulatory networks has also been associated with carcinogenesis. Transcription factors are key enablers of cancer stemness, which support the maintenance and function of cancer stem cells that are believed to act as seeds for cancer initiation, progression and metastasis, and treatment resistance. One key area of research is to understand how these factors interact and collaborate to define cellular fate during embryogenesis as well as during tumor development. This review focuses on understanding the role of TFs in cell development and cancer. The molecular mechanisms of cell fate decision are of key importance in efforts towards developing better protocols for directed differentiation of cells in research and medicine. We also discuss the dysregulation of TFs and their role in cancer progression and metastasis, exploring TF networks as direct or indirect targets for therapeutic intervention, as well as specific TFs' potential as biomarkers for predicting and monitoring treatment responses.
Collapse
Affiliation(s)
- Zeyaul Islam
- Diabetes Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Ameena Mohamed Ali
- Diabetes Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Adviti Naik
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Mohamed Eldaw
- Diabetes Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Julie Decock
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Prasanna R. Kolatkar
- Diabetes Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| |
Collapse
|
8
|
Zhou Y, Guo Y, Zhu Y, Sun Y, Li W, Li Z, Wei L. Dual PPARγ/ɑ agonist oroxyloside suppresses cell cycle progression by glycolipid metabolism switch-mediated increase of reactive oxygen species levels. Free Radic Biol Med 2021; 167:205-217. [PMID: 33713839 DOI: 10.1016/j.freeradbiomed.2021.02.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/26/2021] [Accepted: 02/24/2021] [Indexed: 12/15/2022]
Abstract
Cancer cells prefers to rely on aerobic glycolysis than pyruvate oxidation to meet the high demand of energy for rapidly proliferation. Peroxisome proliferator-activated receptors (PPARs) are a kind of important ligand-inducible transcription factors and play crucial roles in glucose and lipid metabolism. Careful designing of novel agonists for PPARs, may show improvement with the side effects and also increase the therapeutic value for cancer and other metabolic disorder diseases. Compared with normal human liver cells, lower expression or acitivity of PPARs is observed in hepatocellular carcinoma (HCC). In this study, we show that oroxyloside (OAG) is a new dual agonist of PPARγ/ɑ, and inhibits cell proliferation of HCC based on metabolic switch. Via both PPAR-dependent and PPAR-independent regulations on glycolipid metabolic enzymes, OAG shuts down the catabolism of glucose and promotes fatty acids oxidation to generate acetyl-CoA for TCA cycle and oxidative phosphorylation. The metabolic switch induced by OAG results in a marked increase of reactive oxygen species (ROS) levels, leading to rapid dephosphorylation of RB and cell-cycle arrest in G1 phase. Pyruvate dehydrogenase kinase 4 (PDK4) and β-Oxidation are required for the suppression of cell cycle progression by OAG. Together, our findings provide a new drug candidate and a viable therapeutic strategy for HCC based on metabolic reprogram.
Collapse
Affiliation(s)
- Yuxin Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, PR China
| | - Yongjian Guo
- School of Biopharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, PR China
| | - Yejin Zhu
- School of Medicine & Holistic Integrative Medcine, Nanjing University of Chinese Medicine, 138 Xianlin Rd, Nanjing, PR China
| | - Yuening Sun
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, PR China
| | - Wei Li
- Research Center of Basic Medical College, Nanjing University of Chinese Medicine, 138 Xianlin Rd, Nanjing, PR China
| | - Zhiyu Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, PR China
| | - Libin Wei
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, PR China.
| |
Collapse
|
9
|
Peng Y, Zhang Q, Zielinski RM, Howells RD, Welsh WJ. Identification of an irreversible PPARγ antagonist with potent anticancer activity. Pharmacol Res Perspect 2020; 8:e00693. [PMID: 33280279 PMCID: PMC7719157 DOI: 10.1002/prp2.693] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/22/2020] [Accepted: 11/02/2020] [Indexed: 12/15/2022] Open
Abstract
Melanoma is responsible for most skin cancer deaths, and its incidence continues to rise year after year. Different treatment options have been developed for melanoma depending on the stage of the disease. Despite recent advances in immuno- and targeted therapies, advanced melanoma remains incurable and thus an urgent need persists for safe and more effective melanoma therapeutics. In this study, we demonstrate that a novel compound MM902 (3-(3-(bromomethyl)-5-(4-(tert-butyl) phenyl)-1H-1,2,4-triazol-1-yl) phenol) exhibited potent efficacies in inhibiting the growth of different cancer cells, and suppressed tumor growth in a mouse xenograft model of malignant melanoma. Beginning with MM902 instead of specific targets, computational similarity- and docking-based approaches were conducted to search for known anticancer drugs whose structural features match MM902 and whose pharmacological target would accommodate an irreversible inhibitor. Peroxisome proliferator-activated receptor (PPAR) was computationally identified as one of the pharmacological targets and confirmed by in vitro biochemical assays. MM902 was shown to bind to PPARγ in an irreversible mode of action and to function as a selective antagonist for PPARγ over PPARα and PPARδ. It is hoped that MM902 will serve as a valuable research probe to study the functions of PPARγ in tumorigenesis and other pathological processes.
Collapse
Affiliation(s)
- Youyi Peng
- Biomedical Informatics Shared ResourceCancer Institute of New JerseyRutgers, The State University of New JerseyNew BrunswickNJUSA
| | - Qiang Zhang
- Department of PharmacologyRobert Wood Johnson Medical SchoolRutgers, The State University of New JerseyPiscatawayNJUSA
- Present address:
Intra‐Cellular Therapies, Inc.430 East 29th StreetNew YorkNY10016USA
| | - Robert M. Zielinski
- Graduate School of Biomedical SciencesNew Jersey Medical SchoolRutgers, The State University of New JerseyNewarkNJUSA
| | - Richard D. Howells
- Department of Biochemistry & Molecular BiologyNew Jersey Medical SchoolRutgers, The State University of New JerseyNewarkNJUSA
| | - William J. Welsh
- Biomedical Informatics Shared ResourceCancer Institute of New JerseyRutgers, The State University of New JerseyNew BrunswickNJUSA
- Department of PharmacologyRobert Wood Johnson Medical SchoolRutgers, The State University of New JerseyPiscatawayNJUSA
| |
Collapse
|
10
|
Villa ALP, Parra RS, Feitosa MR, de Camargo HP, Machado VF, Tirapelli DPDC, da Rocha JJR, Feres O. PPARG expression in colorectal cancer and its association with staging and clinical evolution. Acta Cir Bras 2020; 35:e202000708. [PMID: 32813759 PMCID: PMC7433669 DOI: 10.1590/s0102-865020200070000008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/11/2020] [Accepted: 06/08/2020] [Indexed: 12/14/2022] Open
Abstract
Purpose To evaluate the gene expression of peroxisome proliferator activated receptors gamma (PPARG) in colorectal tumors and to correlate this data with clinical variables of the patients. Methods We analyzed the gene expression of PPARG in 50 samples of colorectal tumors using real-time reverse transcription polymerase chain reaction, and 20 adjacent normal tissue samples as control. The results of these quantifications were correlated with the respective patients' medical records' clinical information. Results PPARG expression was not different in the tumor tissue compared to the control tissue. Patients older than 60 years, histological type with mucinous differentiation, more advanced staging at the time of diagnosis, and patients who evolved with recurrence of the disease or death did not present higher PPARG expression. Conclusion Expression of PPARGD was not associated with worse prognosis.
Collapse
Affiliation(s)
- Andre Luiz Prezotto Villa
- MSc, Division of Coloproctology, Department of Anatomy and Surgery , Medical School , Universidade de São Paulo (USP), Ribeirao Preto - SP , Brazil . Conception and design of the study; acquisition, analysis and interpretation of data, manuscript writing
| | - Rogério Serafim Parra
- PhD, Division of Coloproctology, Department of Anatomy and Surgery , Medical School , USP , Ribeirao Preto - SP , Brazil . Manuscript writing, critical revision
| | - Marley Ribeiro Feitosa
- PhD, Department of Anatomy and Surgery , Medical School , USP , Ribeirao Preto - SP , Brazil . Analysis and interpretation of data, statistics analysis
| | - Hugo Parra de Camargo
- MD, Department of Anatomy and Surgery , Medical School , USP , Ribeirao Preto - SP , Brazil . Analysis of data
| | - Vanessa Foresto Machado
- MD, Department of Anatomy and Surgery , Medical School , USP , Ribeirao Preto - SP , Brazil . Analysis and interpretation of data
| | - Daniela Pretti da Cunha Tirapelli
- PhD, Department of Anatomy and Surgery , Medical School , USP , Ribeirao Preto - SP , Brazil . Analysis and interpretation of data, critical revision
| | - José Joaquim Ribeiro da Rocha
- PhD, Associated Professor, Head, Division of Coloproctology, Department of Anatomy and Surgery , Medical School , USP , Ribeirao Preto - SP , Brazil . Critical revision, final approval
| | - Omar Feres
- PhD, Associated Professor, Division of Coloproctology, Department of Anatomy and Surgery , Medical School , USP , Ribeirao Preto - SP , Brazil . Conception and design of the study, analysis and interpretation of data, critical revision, final approval
| |
Collapse
|
11
|
Myotubularin-related protein 7 activates peroxisome proliferator-activated receptor-gamma. Oncogenesis 2020; 9:59. [PMID: 32522977 PMCID: PMC7286916 DOI: 10.1038/s41389-020-0238-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 04/15/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023] Open
Abstract
Peroxisome proliferator-activated receptor-gamma (PPARγ) is a transcription factor drugable by agonists approved for treatment of type 2 diabetes, but also inhibits carcinogenesis and cell proliferation in vivo. Activating mutations in the Kirsten rat sarcoma viral oncogene homologue (KRAS) gene mitigate these beneficial effects by promoting a negative feedback-loop comprising extracellular signal-regulated kinase 1/2 (ERK1/2) and mitogen-activated kinase kinase 1/2 (MEK1/2)-dependent inactivation of PPARγ. To overcome this inhibitory mechanism, we searched for novel post-translational regulators of PPARγ. Phosphoinositide phosphatase Myotubularin-Related-Protein-7 (MTMR7) was identified as cytosolic interaction partner of PPARγ. Synthetic peptides were designed resembling the regulatory coiled-coil (CC) domain of MTMR7, and their activities studied in human cancer cell lines and C57BL6/J mice. MTMR7 formed a complex with PPARγ and increased its transcriptional activity by inhibiting ERK1/2-dependent phosphorylation of PPARγ. MTMR7-CC peptides mimicked PPARγ-activation in vitro and in vivo due to LXXLL motifs in the CC domain. Molecular dynamics simulations and docking predicted that peptides interact with the steroid receptor coactivator 1 (SRC1)-binding site of PPARγ. Thus, MTMR7 is a positive regulator of PPARγ, and its mimicry by synthetic peptides overcomes inhibitory mechanisms active in cancer cells possibly contributing to the failure of clinical studies targeting PPARγ.
Collapse
|
12
|
Elix CC, Salgia MM, Otto-Duessel M, Copeland BT, Yoo C, Lee M, Tew BY, Ann D, Pal SK, Jones JO. Peroxisome proliferator-activated receptor gamma controls prostate cancer cell growth through AR-dependent and independent mechanisms. Prostate 2020; 80:162-172. [PMID: 31769890 PMCID: PMC8985763 DOI: 10.1002/pros.23928] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 10/15/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Prostate cancer (PC) remains a leading cause of cancer mortality and the most successful chemopreventative and treatment strategies for PC come from targeting the androgen receptor (AR). Although AR plays a key role, it is likely that other molecular pathways also contribute to PC, making it essential to identify and develop drugs against novel targets. Recent studies have identified peroxisome proliferator-activated receptor gamma (PPARγ), a nuclear receptor that regulates fatty acid (FA) metabolism, as a novel target in PC, and suggest that inhibitors of PPARγ could be used to treat existing disease. We hypothesized that PPARγ acts through AR-dependent and independent mechanisms to control PC development and growth and that PPARγ inhibition is a viable PC treatment strategy. METHODS Immunohistochemistry was used to determine expression of PPARү in a cohort of patients with PC. Standard molecular techniques were used to investigate the PPARү signaling in PC cells as well a xenograft mouse model to test PPARү inhibition in vivo. Kaplan-Meier curves were created using cBioportal. RESULTS We confirmed the expression of PPARү in human PC. We then showed that small molecule inhibition of PPARγ decreases the growth of AR-positive and -negative PC cells in vitro and that T0070907, a potent PPARγ antagonist, significantly decreased the growth of human PC xenografts in nude mice. We found that PPARγ antagonists or small interfering RNA (siRNA) do not affect mitochondrial activity nor do they cause apoptosis; instead, they arrest the cell cycle. In AR-positive PC cells, antagonists and siRNAs reduce AR transcript and protein levels, which could contribute to growth inhibition. AR-independent effects on growth appear to be mediated by effects on FA metabolism as the specific FASN inhibitor, Fasnall, inhibited PC cell growth but did not have an additive effect when combined with PPARγ antagonists. Patients with increased PPARү target gene expression, but not alterations in PPARү itself, were found to have significantly worse overall survival. CONCLUSIONS Having elucidated the direct cancer cell effects of PPARγ inhibition, our studies have helped to determine the role of PPARγ in PC growth, and support the hypothesis that PPARγ inhibition is an effective strategy for PC treatment.
Collapse
Affiliation(s)
- Catherine C Elix
- Department of Medical Oncology, City of Hope, Duarte, California
| | - Meghan M Salgia
- Department of Medical Oncology, City of Hope, Duarte, California
| | | | - Ben T Copeland
- Department of Medical Oncology, City of Hope, Duarte, California
| | - Christopher Yoo
- Department of Medical Oncology, City of Hope, Duarte, California
| | - Michael Lee
- Department of Diabetes Complications and Metabolism, City of Hope, Duarte, California
| | - Ben Yi Tew
- Department of Medical Oncology, City of Hope, Duarte, California
| | - David Ann
- Department of Diabetes Complications and Metabolism, City of Hope, Duarte, California
| | - Sumanta K Pal
- Department of Medical Oncology, City of Hope, Duarte, California
| | - Jeremy O Jones
- Department of Medical Oncology, City of Hope, Duarte, California
| |
Collapse
|
13
|
Suh S, Kim KW. Diabetes and Cancer: Cancer Should Be Screened in Routine Diabetes Assessment. Diabetes Metab J 2019; 43:733-743. [PMID: 31902143 PMCID: PMC6943263 DOI: 10.4093/dmj.2019.0177] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 11/17/2019] [Indexed: 12/16/2022] Open
Abstract
Cancer incidence appears to be increased in both type 1 and type 2 diabetes mellitus (DM). DM represents a risk factor for cancer, particularly hepatocellular, hepatobiliary, pancreas, breast, ovarian, endometrial, and gastrointestinal cancers. In addition, there is evidence showing that DM is associated with increased cancer mortality. Common risk factors such as age, obesity, physical inactivity and smoking may contribute to increased cancer risk in patients with DM. Although the mechanistic process that may link diabetes to cancer is not completely understood yet, biological mechanisms linking DM and cancer are hyperglycemia, hyperinsulinemia, increased bioactivity of insulin-like growth factor 1, oxidative stress, dysregulations of sex hormones, and chronic inflammation. However, cancer screening rate is significantly lower in people with DM than that in people without diabetes. Evidence from previous studies suggests that some medications used to treat DM are associated with either increased or reduced risk of cancer. However, there is no strong evidence supporting the association between the use of anti-hyperglycemic medication and specific cancer. In conclusion, all patients with DM should be undergo recommended age- and sex appropriate cancer screenings to promote primary prevention and early detection. Furthermore, cancer should be screened in routine diabetes assessment.
Collapse
Affiliation(s)
- Sunghwan Suh
- Department of Internal Medicine, Dong-A University College of Medicine, Busan, Korea
| | - Kwang Won Kim
- Department of Internal Medicine, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, Korea.
| |
Collapse
|
14
|
Frapolli R, Bello E, Ponzo M, Craparotta I, Mannarino L, Ballabio S, Marchini S, Carrassa L, Ubezio P, Porcu L, Brich S, Sanfilippo R, Casali PG, Gronchi A, Pilotti S, D'Incalci M. Combination of PPARγ Agonist Pioglitazone and Trabectedin Induce Adipocyte Differentiation to Overcome Trabectedin Resistance in Myxoid Liposarcomas. Clin Cancer Res 2019; 25:7565-7575. [PMID: 31481505 DOI: 10.1158/1078-0432.ccr-19-0976] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/01/2019] [Accepted: 08/28/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE This study was aimed at investigating whether the PPARγ agonist pioglitazone-given in combination with trabectedin-is able to reactivate adipocytic differentiation in myxoid liposarcoma (MLS) patient-derived xenografts, overcoming resistance to trabectedin. EXPERIMENTAL DESIGN The antitumor and biological effects of trabectedin, pioglitazone, and the combination of the two drugs were investigated in nude mice bearing well-characterized MLS xenografts representative of innate or acquired resistance against trabectedin. Pioglitazone and trabectedin were given by daily oral and weekly i.v. administrations, respectively. Molecular studies were performed by using microarrays approach, real-time PCR, and Western blotting. RESULTS We found that the resistance of MLS against trabectedin is associated with the lack of activation of adipogenesis. The PPARγ agonist pioglitazone reactivated adipogenesis, assessed by histologic and gene pathway analyses. Pioglitazone was well tolerated and did not increase the toxicity of trabectedin. The ability of pioglitazone to reactivate adipocytic differentiation was observed by morphologic examination, and it is consistent with the increased expression of genes such as ADIPOQ implicated in the adipogenesis process. The determination of adiponectin by Western blotting constitutes a good and reliable biomarker related to MLS adipocytic differentiation. CONCLUSIONS The finding that the combination of pioglitazone and trabectedin induces terminal adipocytic differentiation of some MLSs with the complete pathologic response and cure of tumor-bearing mice provides a strong rationale to test the combination of trabectedin and pioglitazone in patients with MLS.
Collapse
Affiliation(s)
- Roberta Frapolli
- Unit of Preclinical Experimental Therapeutics, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Ezia Bello
- Unit of Preclinical Experimental Therapeutics, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marianna Ponzo
- Unit of Preclinical Experimental Therapeutics, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Ilaria Craparotta
- Unit of Translational Genomic, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Laura Mannarino
- Unit of Translational Genomic, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Sara Ballabio
- Unit of Translational Genomic, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Sergio Marchini
- Unit of Translational Genomic, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Laura Carrassa
- Unit of DNA repair, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Paolo Ubezio
- Unit of Biophysics, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Luca Porcu
- Unit of Methodological Research, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Silvia Brich
- Laboratory of Molecular Pathology, Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Roberta Sanfilippo
- Medical Oncology Unit 2, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paolo Giovanni Casali
- Medical Oncology Unit 2, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Silvana Pilotti
- Laboratory of Molecular Pathology, Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maurizio D'Incalci
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| |
Collapse
|
15
|
Sabatino L, Ziccardi P, Cerchia C, Muccillo L, Piemontese L, Loiodice F, Colantuoni V, Lupo A, Lavecchia A. Chiral phenoxyacetic acid analogues inhibit colon cancer cell proliferation acting as PPARγ partial agonists. Sci Rep 2019; 9:5434. [PMID: 30931956 PMCID: PMC6443668 DOI: 10.1038/s41598-019-41765-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 03/07/2019] [Indexed: 12/13/2022] Open
Abstract
Peroxisome Proliferator-Activated Receptor γ (PPARγ) is an important sensor at the crossroad of diabetes, obesity, immunity and cancer as it regulates adipogenesis, metabolism, inflammation and proliferation. PPARγ exerts its pleiotropic functions upon binding of natural or synthetic ligands. The molecular mechanisms through which PPARγ controls cancer initiation/progression depend on the different mode of binding of distinctive ligands. Here, we analyzed a series of chiral phenoxyacetic acid analogues for their ability to inhibit colorectal cancer (CRC) cells growth by binding PPARγ as partial agonists as assessed in transactivation assays of a PPARG-reporter gene. We further investigated compounds (R,S)-3, (S)-3 and (R,S)-7 because they combine the best antiproliferative activity and a limited transactivation potential and found that they induce cell cycle arrest mainly via upregulation of p21waf1/cip1. Interestingly, they also counteract the β-catenin/TCF pathway by repressing c-Myc and cyclin D1, supporting their antiproliferative effect. Docking experiments provided insight into the binding mode of the most active compound (S)-3, suggesting that its partial agonism could be related to a better stabilization of H3 rather than H11 and H12. In conclusion, we identified a series of PPARγ partial agonists affecting distinct pathways all leading to strong antiproliferative effects. These findings may pave the way for novel therapeutic strategies in CRC.
Collapse
Affiliation(s)
- Lina Sabatino
- Dipartimento di Scienze e Tecnologie, Università del Sannio, via Port'Arsa 11, 82100, Benevento, Italy
| | - Pamela Ziccardi
- Dipartimento di Scienze e Tecnologie, Università del Sannio, via Port'Arsa 11, 82100, Benevento, Italy
| | - Carmen Cerchia
- Dipartimento di Farmacia, "Drug Discovery" Laboratory, Università degli Studi di Napoli Federico II, via D. Montesano 49, 80131, Napoli, Italy
| | - Livio Muccillo
- Dipartimento di Scienze e Tecnologie, Università del Sannio, via Port'Arsa 11, 82100, Benevento, Italy
| | - Luca Piemontese
- Dipartimento Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via Orabona 4, 70125, Bari, Italy
| | - Fulvio Loiodice
- Dipartimento Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", via Orabona 4, 70125, Bari, Italy
| | - Vittorio Colantuoni
- Dipartimento di Scienze e Tecnologie, Università del Sannio, via Port'Arsa 11, 82100, Benevento, Italy
| | - Angelo Lupo
- Dipartimento di Scienze e Tecnologie, Università del Sannio, via Port'Arsa 11, 82100, Benevento, Italy.
| | - Antonio Lavecchia
- Dipartimento di Farmacia, "Drug Discovery" Laboratory, Università degli Studi di Napoli Federico II, via D. Montesano 49, 80131, Napoli, Italy.
| |
Collapse
|
16
|
Takeuchi A, Nomura A, Yamamoto N, Hayashi K, Igarashi K, Tandai S, Kawai A, Matsumine A, Miwa S, Nishida Y, Nakamura T, Terauchi R, Hoshi M, Kunisada T, Endo M, Yoshimura K, Murayama T, Tsuchiya H. Randomized placebo-controlled double-blind phase II study of zaltoprofen for patients with diffuse-type and unresectable localized tenosynovial giant cell tumors: a study protocol. BMC Musculoskelet Disord 2019; 20:68. [PMID: 30738433 PMCID: PMC6368706 DOI: 10.1186/s12891-019-2453-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 02/01/2019] [Indexed: 02/06/2023] Open
Abstract
Background A tenosynovial giant cell tumor (TGCT) is a locally aggressive benign neoplasm arising from intra- or extra-articular tissue. Diffuse TGCT (D-TGCT) most commonly develops in the knee, followed by the hip, ankle, elbow, and shoulder. Surgical removal is the only effective treatment option for the patients. However, a local recurrence rate as high as 47% has been reported. Recently, we revealed that zaltoprofen, a nonsteroidal anti-inflammatory drug possessing the ability to activate peroxisome proliferator-activated receptor gamma (PPARγ), can inhibit the proliferation of TGCT stromal cells via PPARγ. PPARγ is a ligand-activated transcription factor that belongs to the nuclear hormone receptor superfamily. It plays an important role in the differentiation of adipocytes from precursor cells and exhibits antitumorigenic effects on certain malignancies. Therefore, we are conducting this investigator-initiated clinical trial to evaluate whether zaltoprofen is safe and effective for patients with D-TGCT or unresectable localized TGCT (L-TGCT). Methods This study is a randomized, placebo-controlled, double-blind, multicenter trial to evaluate the safety and efficacy of zaltoprofen for patients with D-TGCT or L-TGCT. For the treatment group, zaltoprofen 480 mg/day will be administered for 48 weeks; the placebo group will receive similar dosages without zaltoprofen. Twenty participants in each group are needed in this trial (40 participants total). The primary outcome is the progression-free rate at 48 weeks after treatment administration. “Progression” is defined as any serious events (1. Repetitive joint swelling due to hemorrhage, 2. Joint range of motion limitation, 3. Invasion of adjacent cartilage or bone, 4. Severe joint space narrowing, 5. Increase in tumor size) requiring surgical interventions. We hypothesize that the zaltoprofen group will have a higher progression-free rate compared to that of the placebo group at 48 weeks. Discussion This is the first study to evaluate the efficacy of zaltoprofen in patients with D-TGCT or unresectable L-TGCT. We believe that the results of this trial will validate a novel treatment option, zaltoprofen, to stabilize disease progression for TGCT patients. Trial registration University Hospital Medical Information Network (UMIN) Clinical Trials Registry (UMIN000025901) registered on 4/01/2017.
Collapse
Affiliation(s)
- Akihiko Takeuchi
- Department of Orthopaedic Surgery, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8641, Japan
| | - Akihiro Nomura
- Innovative Clinical Research Center (iCREK), Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, 920-8641, Japan
| | - Norio Yamamoto
- Department of Orthopaedic Surgery, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8641, Japan
| | - Katsuhiro Hayashi
- Department of Orthopaedic Surgery, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8641, Japan
| | - Kentaro Igarashi
- Department of Orthopaedic Surgery, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8641, Japan
| | - Susumu Tandai
- Department of Orthopaedic Surgery, Asahikawa Medical University, 2-1E Midorigaoka, Asahikawa, Hokkaido, 078-8510, Japan
| | - Akira Kawai
- Department of Musculoskeletal Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Akihiko Matsumine
- Department of Orthopaedics and Rehabilitation Medicine, Unit of Surgery, Division of Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, 910-1104, Japan
| | - Shinji Miwa
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, 1, Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, Aichi, 467-8601, Japan
| | - Yoshihiro Nishida
- Department of Orthopaedic Surgery, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya, 466-8550, Japan
| | - Tomoki Nakamura
- Department of Orthopedic Surgery, Mie University School of Medicine, 2-174 Edobashi, Tsu, Mie, 514-8507, Japan
| | - Ryu Terauchi
- Department of Orthopaedic Surgery, Kyoto Prefectural University of Medicine, 465 Kajii-cho Hirokoji-agaru Kawaramachi-dori Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Manabu Hoshi
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-Machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Toshiyuki Kunisada
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Makoto Endo
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kenichi Yoshimura
- Innovative Clinical Research Center (iCREK), Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, 920-8641, Japan
| | - Toshinori Murayama
- Innovative Clinical Research Center (iCREK), Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, 920-8641, Japan
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic Surgery, Kanazawa University Graduate School of Medical Sciences, 13-1 Takara-machi, Kanazawa, 920-8641, Japan.
| |
Collapse
|
17
|
Bojková B, Kubatka P, Qaradakhi T, Zulli A, Kajo K. Melatonin May Increase Anticancer Potential of Pleiotropic Drugs. Int J Mol Sci 2018; 19:E3910. [PMID: 30563247 PMCID: PMC6320927 DOI: 10.3390/ijms19123910] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/27/2018] [Accepted: 12/03/2018] [Indexed: 12/14/2022] Open
Abstract
Melatonin (N-acetyl-5-methoxytryptamine) is not only a pineal hormone, but also an ubiquitary molecule present in plants and part of our diet. Numerous preclinical and some clinical reports pointed to its multiple beneficial effects including oncostatic properties, and as such, it has become one of the most aspiring goals in cancer prevention/therapy. A link between cancer and inflammation and/or metabolic disorders has been well established and the therapy of these conditions with so-called pleiotropic drugs, which include non-steroidal anti-inflammatory drugs, statins and peroral antidiabetics, modulates a cancer risk too. Adjuvant therapy with melatonin may improve the oncostatic potential of these drugs. Results from preclinical studies are limited though support this hypothesis, which, however, remains to be verified by further research.
Collapse
Affiliation(s)
- Bianka Bojková
- Department of Animal Physiology, Institute of Biology and Ecology, Faculty of Science, Pavol Jozef Šafárik University in Košice, Šrobárová 2, 041 54 Košice, Slovak Republic.
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Malá Hora 4, 036 01 Martin, Slovak Republic.
- Department of Experimental Carcinogenesis, Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Malá Hora 4C, 036 01 Martin, Slovak Republic.
| | - Tawar Qaradakhi
- Institute for Health and Sport (IHES), Victoria University, Melbourne, VIC 3011, Australia.
| | - Anthony Zulli
- Institute for Health and Sport (IHES), Victoria University, Melbourne, VIC 3011, Australia.
| | - Karol Kajo
- St. Elisabeth Oncology Institute, Heydukova 10, 811 08 Bratislava, Slovak Republic.
- Biomedical Research Center, Slovak Academy of Sciences, Dúbravská cesta 9, 845 05 Bratislava, Slovak Republic.
| |
Collapse
|
18
|
Heudobler D, Rechenmacher M, Lüke F, Vogelhuber M, Klobuch S, Thomas S, Pukrop T, Hackl C, Herr W, Ghibelli L, Gerner C, Reichle A. Clinical Efficacy of a Novel Therapeutic Principle, Anakoinosis. Front Pharmacol 2018; 9:1357. [PMID: 30546308 PMCID: PMC6279883 DOI: 10.3389/fphar.2018.01357] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 11/05/2018] [Indexed: 12/18/2022] Open
Abstract
Classic tumor therapy, consisting of cytotoxic agents and/or targeted therapy, has not overcome therapeutic limitations like poor risk genetic parameters, genetic heterogeneity at different metastatic sites or the problem of undruggable targets. Here we summarize data and trials principally following a completely different treatment concept tackling systems biologic processes: the principle of communicative reprogramming of tumor tissues, i.e., anakoinosis (ancient greek for communication), aims at establishing novel communicative behavior of tumor tissue, the hosting organ and organism via re-modeling gene expression, thus recovering differentiation, and apoptosis competence leading to cancer control - in contrast to an immediate, "poisoning" with maximal tolerable doses of targeted or cytotoxic therapies. Therefore, we introduce the term "Master modulators" for drugs or drug combinations promoting evolutionary processes or regulating homeostatic pathways. These "master modulators" comprise a broad diversity of drugs, characterized by the capacity for reprogramming tumor tissues, i.e., transcriptional modulators, metronomic low-dose chemotherapy, epigenetically modifying agents, protein binding pro-anakoinotic drugs, such as COX-2 inhibitors, IMiDs etc., or for example differentiation inducing therapies. Data on 97 anakoinosis inducing schedules indicate a favorable toxicity profile: The combined administration of master modulators, frequently (with poor or no monoactivity) may even induce continuous complete remission in refractory metastatic neoplasia, irrespectively of the tumor type. That means recessive components of the tumor, successively developing during tumor ontogenesis, are accessible by regulatory active drug combinations in a therapeutically meaningful way. Drug selection is now dependent on situative systems characteristics, to less extent histology dependent. To sum up, anakoinosis represents a new substantive therapy principle besides novel targeted therapies.
Collapse
Affiliation(s)
- Daniel Heudobler
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Michael Rechenmacher
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Florian Lüke
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Martin Vogelhuber
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Sebastian Klobuch
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Simone Thomas
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Tobias Pukrop
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Christina Hackl
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| | - Lina Ghibelli
- Department Biology, Universita' di Roma Tor Vergata, Rome, Italy
| | - Christopher Gerner
- Faculty Chemistry, Institut for Analytical Chemistry, University Vienna, Vienna, Austria
| | - Albrecht Reichle
- Department of Internal Medicine III, Hematology and Oncology, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
19
|
Heudobler D, Rechenmacher M, Lüke F, Vogelhuber M, Pukrop T, Herr W, Ghibelli L, Gerner C, Reichle A. Peroxisome Proliferator-Activated Receptors (PPAR)γ Agonists as Master Modulators of Tumor Tissue. Int J Mol Sci 2018; 19:ijms19113540. [PMID: 30424016 PMCID: PMC6274845 DOI: 10.3390/ijms19113540] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/27/2018] [Accepted: 11/06/2018] [Indexed: 02/08/2023] Open
Abstract
In most clinical trials, thiazolidinediones do not show any relevant anti-cancer activity when used as mono-therapy. Clinical inefficacy contrasts ambiguous pre-clinical data either favoring anti-tumor activity or tumor promotion. However, if thiazolidinediones are combined with additional regulatory active drugs, so-called ‘master modulators’ of tumors, i.e., transcriptional modulators, metronomic low-dose chemotherapy, epigenetically modifying agents, protein binding pro-anakoinotic drugs, such as COX-2 inhibitors, IMiDs, etc., the results indicate clinically relevant communicative reprogramming of tumor tissues, i.e., anakoinosis, meaning ‘communication’ in ancient Greek. The concerted activity of master modulators may multifaceted diversify palliative care or even induce continuous complete remission in refractory metastatic tumor disease and hematologic neoplasia by establishing novel communicative behavior of tumor tissue, the hosting organ, and organism. Re-modulation of gene expression, for example, the up-regulation of tumor suppressor genes, may recover differentiation, apoptosis competence, and leads to cancer control—in contrast to an immediate, ‘poisoning’ with maximal tolerable doses of targeted/cytotoxic therapies. The key for uncovering the therapeutic potential of Peroxisome proliferator-activated receptor γ (PPARγ) agonists is selecting the appropriate combination of master modulators for inducing anakoinosis: Now, anakoinosis is trend setting by establishing a novel therapeutic pillar while overcoming classic obstacles of targeted therapies, such as therapy resistance and (molecular-)genetic tumor heterogeneity.
Collapse
Affiliation(s)
- Daniel Heudobler
- Department of Internal Medicine III, University Hospital Regensburg, Hematology and Oncology, 93042 Regensburg, Germany.
| | - Michael Rechenmacher
- Department of Internal Medicine III, University Hospital Regensburg, Hematology and Oncology, 93042 Regensburg, Germany.
| | - Florian Lüke
- Department of Internal Medicine III, University Hospital Regensburg, Hematology and Oncology, 93042 Regensburg, Germany.
| | - Martin Vogelhuber
- Department of Internal Medicine III, University Hospital Regensburg, Hematology and Oncology, 93042 Regensburg, Germany.
| | - Tobias Pukrop
- Department of Internal Medicine III, University Hospital Regensburg, Hematology and Oncology, 93042 Regensburg, Germany.
| | - Wolfgang Herr
- Department of Internal Medicine III, University Hospital Regensburg, Hematology and Oncology, 93042 Regensburg, Germany.
| | - Lina Ghibelli
- Department Biology, Universita' di Roma Tor Vergata, 00173 Rome, Italy.
| | - Christopher Gerner
- Institut for Analytical Chemistry, Faculty Chemistry, University Vienna, Vienna A-1090, Austria.
| | - Albrecht Reichle
- Department of Internal Medicine III, University Hospital Regensburg, Hematology and Oncology, 93042 Regensburg, Germany.
| |
Collapse
|
20
|
Chang SN, Lee JM, Oh H, Kim U, Ryu B, Park JH. Troglitazone inhibits the migration and invasion of PC-3 human prostate cancer cells by upregulating E-cadherin and glutathione peroxidase 3. Oncol Lett 2018; 16:5482-5488. [PMID: 30250621 DOI: 10.3892/ol.2018.9278] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 04/28/2017] [Indexed: 11/06/2022] Open
Abstract
Troglitazone (TGZ) is a synthetic peroxisome proliferator-activated receptor γ (PPARγ) ligand that exhibits potential antitumor effects on a number of cancer subtypes, including prostate cancer. However, little is known about the effect of TGZ on metastasis in prostate cancer. The aim of the present study was to determine the inhibitory effect and mechanism underlying TGZ on cell growth, migration and invasion using the prostate cancer PC-3 cell line. Cellular migration and invasion were evaluated by performing a wound healing assay and Matrigel assay, respectively. The expression levels of mRNA and protein were determined by reverse transcription-quantitative polymerase chain reaction and western blotting. The results demonstrated that TGZ dose-dependently inhibited cell migration and invasion of PC-3 cells. The present study also revealed that TGZ increased the mRNA and protein levels of E-cadherin and glutathione peroxidase 3 (GPx3) in human prostate cancer PC-3 cells. In addition, GW9662, a PPARγ antagonist, attenuated the increased mRNA and protein levels of E-cadherin and GPx3, suggesting that the PPARγ-dependent signaling pathway was involved. Taken together, these results suggested that the anti-migration and anti-invasion effect of TGZ on PC-3 prostate cancer cells is, at least in part, mediated via upregulation of E-cadherin and GPx3. The present study also concluded that PPARγ may be used as a potential remedial target for the prevention and treatment of prostate cancer cell invasion and metastasis.
Collapse
Affiliation(s)
- Seo-Na Chang
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Republic of Korea
| | - Ji Min Lee
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Republic of Korea
| | - Hanseul Oh
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Republic of Korea
| | - Ukjin Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Republic of Korea
| | - Bokyeong Ryu
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Republic of Korea
| | - Jae-Hak Park
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Republic of Korea
| |
Collapse
|
21
|
Galbraith L, Leung HY, Ahmad I. Lipid pathway deregulation in advanced prostate cancer. Pharmacol Res 2018; 131:177-184. [PMID: 29466694 DOI: 10.1016/j.phrs.2018.02.022] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 02/08/2018] [Accepted: 02/14/2018] [Indexed: 01/03/2023]
Abstract
The link between prostate cancer (PC) development and lipid metabolism is well established, with AR intimately involved in a number of lipogenic processes involving SREBP1, PPARG, FASN, ACC, ACLY and SCD1. Recently, there is growing evidence implicating the role of obesity and peri-prostatic adipose tissue (PPAT) in PC aggressiveness and related mortality, suggesting the importance of lipid pathways in both localised and disseminated disease. A number of promising agents are in development to target the lipogenic axis in PC, and the likelihood is that these agents will form part of combination drug strategies, with targeting of multiple metabolic pathways (e.g. FASN and CPT1), or in combination with AR pathway inhibitors (SCD1 and AR).
Collapse
Affiliation(s)
- Laura Galbraith
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Hing Y Leung
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Imran Ahmad
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK.
| |
Collapse
|
22
|
Elix C, Pal SK, Jones JO. The role of peroxisome proliferator-activated receptor gamma in prostate cancer. Asian J Androl 2018; 20:238-243. [PMID: 28597850 PMCID: PMC5952477 DOI: 10.4103/aja.aja_15_17] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 04/11/2017] [Indexed: 12/15/2022] Open
Abstract
Despite great progress in the detection and treatment of prostate cancer, this disease remains an incredible health and economic burden. Although androgen receptor (AR) signaling plays a key role in the development and progression of prostate cancer, aberrations in other molecular pathways also contribute to the disease, making it essential to identify and develop drugs against novel targets, both for the prevention and treatment of prostate cancer. One promising target is the peroxisome proliferator-activated receptor gamma (PPARγ) protein. PPARγ was originally thought to act as a tumor suppressor in prostate cells because agonist ligands inhibited the growth of prostate cancer cells; however, additional studies found that PPARγ agonists inhibit cell growth independent of PPARγ. Furthermore, PPARγ expression increases with cancer grade/stage, which would suggest that it is not a tumor suppressor but instead that PPARγ activity may play a role in prostate cancer development and/or progression. Indeed, two new studies, taking vastly different, unbiased approaches, have identified PPARγ as a target in prostate cancer and suggest that PPARγ inhibition might be useful in prostate cancer prevention and treatment. These findings could lead to a new therapeutic weapon in the fight against prostate cancer.
Collapse
Affiliation(s)
- Catherine Elix
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA91010, USA
| | - Sumanta K Pal
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA91010, USA
| | - Jeremy O Jones
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA91010, USA
| |
Collapse
|
23
|
Vallée A, Lecarpentier Y. Crosstalk Between Peroxisome Proliferator-Activated Receptor Gamma and the Canonical WNT/β-Catenin Pathway in Chronic Inflammation and Oxidative Stress During Carcinogenesis. Front Immunol 2018; 9:745. [PMID: 29706964 PMCID: PMC5908886 DOI: 10.3389/fimmu.2018.00745] [Citation(s) in RCA: 276] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 03/26/2018] [Indexed: 12/19/2022] Open
Abstract
Inflammation and oxidative stress are common and co-substantial pathological processes accompanying, promoting, and even initiating numerous cancers. The canonical WNT/β-catenin pathway and peroxisome proliferator-activated receptor gamma (PPARγ) generally work in opposition. If one of them is upregulated, the other one is downregulated and vice versa. WNT/β-catenin signaling is upregulated in inflammatory processes and oxidative stress and in many cancers, although there are some exceptions for cancers. The opposite is observed with PPARγ, which is generally downregulated during inflammation and oxidative stress and in many cancers. This helps to explain in part the opposite and unidirectional profile of the canonical WNT/β-catenin signaling and PPARγ in these three frequent and morbid processes that potentiate each other and create a vicious circle. Many intracellular pathways commonly involved downstream will help maintain and amplify inflammation, oxidative stress, and cancer. Thus, many WNT/β-catenin target genes such as c-Myc, cyclin D1, and HIF-1α are involved in the development of cancers. Nuclear factor-kappaB (NFκB) can activate many inflammatory factors such as TNF-α, TGF-β, interleukin-6 (IL-6), IL-8, MMP, vascular endothelial growth factor, COX2, Bcl2, and inducible nitric oxide synthase. These factors are often associated with cancerous processes and may even promote them. Reactive oxygen species (ROS), generated by cellular alterations, stimulate the production of inflammatory factors such as NFκB, signal transducer and activator transcription, activator protein-1, and HIF-α. NFκB inhibits glycogen synthase kinase-3β (GSK-3β) and therefore activates the canonical WNT pathway. ROS activates the phosphatidylinositol 3 kinase/protein kinase B (PI3K/Akt) signaling in many cancers. PI3K/Akt also inhibits GSK-3β. Many gene mutations of the canonical WNT/β-catenin pathway giving rise to cancers have been reported (CTNNB1, AXIN, APC). Conversely, a significant reduction in the expression of PPARγ has been observed in many cancers. Moreover, PPARγ agonists promote cell cycle arrest, cell differentiation, and apoptosis and reduce inflammation, angiogenesis, oxidative stress, cell proliferation, invasion, and cell migration. All these complex and opposing interactions between the canonical WNT/β-catenin pathway and PPARγ appear to be fairly common in inflammation, oxidative stress, and cancers.
Collapse
Affiliation(s)
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), Meaux, France
| |
Collapse
|
24
|
Sebolt-Leopold JS. Development of Preclinical Models to Understand and Treat Colorectal Cancer. Clin Colon Rectal Surg 2018; 31:199-204. [PMID: 29720906 DOI: 10.1055/s-0037-1602240] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The establishment and validation of preclinical models that faithfully recapitulate the pathogenesis and treatment response of human colorectal cancer (CRC) is critical to expedient therapeutic advances in the clinical management of this disease. Integral to the application of precision medicine for patients diagnosed with metastatic CRC is the need to understand the molecular determinants of response for a given therapy. Preclinical models of CRC have proven invaluable in answering many of our basic questions relating to the molecular aberrations that drive colorectal tumor progression. This review will address the comparative merits and limitations of the broad spectrum of in vitro and in vivo models available for study of colorectal tumors and their response to experimental therapies.
Collapse
Affiliation(s)
- Judith S Sebolt-Leopold
- Department of Radiology, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
25
|
Ory V, Kietzman WB, Boeckelman J, Kallakury BV, Wellstein A, Furth PA, Riegel AT. The PPARγ agonist efatutazone delays invasive progression and induces differentiation of ductal carcinoma in situ. Breast Cancer Res Treat 2018; 169:47-57. [PMID: 29350308 DOI: 10.1007/s10549-017-4649-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 12/26/2017] [Indexed: 02/06/2023]
Abstract
PURPOSE Ductal carcinoma in situ (DCIS) is a pre-invasive lesion of the breast considered a precursor of invasive ductal carcinoma. This study aimed to determine whether activated PPARγ acts as a tumor suppressor in human DCIS progression. METHODS We utilized the high-affinity PPARγ agonist, efatutazone, to activate endogenous PPARγ in a well-defined model for the progression of basal (triple negative) DCIS, MCFDCIS cells, cultured under 2D and 3D conditions. We studied the effects of activated PPARγ on DCIS progression in MCFDCIS xenograft and C3(1)/Tag transgenic mice treated with 30 mg/kg of efatutazone. RESULTS In vitro, efatutazone did not alter the MCFDCIS cell proliferation but induced phenotypic and gene expression changes, indicating that activated PPARγ is able to differentiate MCFDCIS cells into more luminal and lactational-like cells. In addition, MCFDCIS tumorsphere formation in 3D was reduced by PPARγ activation. In vivo, efatutazone-treated MCFDCIS tumors exhibited fat deposition along with upregulation of PPARγ responsive genes in both epithelial and stromal compartments, suggesting features of milk-producing mammary epithelial cell differentiation. The efatutazone-treated lesions were less invasive with fewer CD44+/p63+ basal progenitor cells. PPARγ activation downregulated Akt phosphorylation in these tumors, although the ERK pathway remained unchanged. Similar trends in gene expression changes consistent with lactational and luminal cell differentiation were observed in the C3(1)/Tag mouse model after efatutazone treatment. CONCLUSIONS Our data suggest that activation of the PPARγ pathway differentiates DCIS lesions and may be a useful approach to delay DCIS progression.
Collapse
Affiliation(s)
- Virginie Ory
- Department of Oncology, Georgetown University, Washington, DC, USA.
| | | | - Jacob Boeckelman
- Department of Oncology, Georgetown University, Washington, DC, USA
| | - Bhaskar V Kallakury
- Department of Pathology, Georgetown University, Washington, DC, USA.,The Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Anton Wellstein
- Department of Oncology, Georgetown University, Washington, DC, USA.,The Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Priscilla A Furth
- Department of Oncology, Georgetown University, Washington, DC, USA.,Department of Medicine, Georgetown University, Washington, DC, USA.,The Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Anna T Riegel
- Department of Oncology, Georgetown University, Washington, DC, USA.,The Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| |
Collapse
|
26
|
De Lellis L, Cimini A, Veschi S, Benedetti E, Amoroso R, Cama A, Ammazzalorso A. The Anticancer Potential of Peroxisome Proliferator-Activated Receptor Antagonists. ChemMedChem 2018; 13:209-219. [PMID: 29276815 DOI: 10.1002/cmdc.201700703] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 12/17/2017] [Indexed: 12/13/2022]
Abstract
The effects on cancer-cell proliferation and differentiation mediated by peroxisome proliferator-activated receptors (PPARs) have been widely studied, and pleiotropic outcomes in different cancer models and under different experimental conditions have been obtained. Interestingly, few studies report and little preclinical evidence supports the potential antitumor activity of PPAR antagonists. This review focuses on recent findings on the antitumor in vitro and in vivo effects observed for compounds able to inhibit the three PPAR subtypes in different tumor models, providing a rationale for the use of PPAR antagonists in the treatment of tumors expressing the corresponding receptors.
Collapse
Affiliation(s)
- Laura De Lellis
- Department of Pharmacy, University of Chieti, Via dei Vestini 31, 66100, Chieti, Italy.,Unit of General Pathology, CeSI-MeT, University of Chieti, Chieti, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,National Institute for Nuclear Physics (INFN), Gran Sasso National Laboratory (LNGS), Assergi (Aq), Italy.,Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, 1900 N. 12th Street, Philadelphia, PA, 19122, USA
| | - Serena Veschi
- Department of Pharmacy, University of Chieti, Via dei Vestini 31, 66100, Chieti, Italy.,Unit of General Pathology, CeSI-MeT, University of Chieti, Chieti, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Rosa Amoroso
- Department of Pharmacy, University of Chieti, Via dei Vestini 31, 66100, Chieti, Italy
| | - Alessandro Cama
- Department of Pharmacy, University of Chieti, Via dei Vestini 31, 66100, Chieti, Italy.,Unit of General Pathology, CeSI-MeT, University of Chieti, Chieti, Italy
| | | |
Collapse
|
27
|
Alpha-Tocopherol prevents esophageal squamous cell carcinoma by modulating PPARγ-Akt signaling pathway at the early stage of carcinogenesis. Oncotarget 2017; 8:95914-95930. [PMID: 29221176 PMCID: PMC5707070 DOI: 10.18632/oncotarget.21437] [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: 06/09/2017] [Accepted: 08/17/2017] [Indexed: 02/07/2023] Open
Abstract
The poor prognosis of esophageal squamous cell carcinoma (ESCC) emphasizes the urgent need to better understand the carcinogenesis and develop prevention strategies. Previous studies have highlighted the potential of using Vitamin E (tocopherols) for cancer chemoprevention, but the preventive activity of α-Tocopherol against ESCC remains to be elucidated. Our data showed that early-stage supplementation with α-Tocopherol significantly prevented esophageal carcinogenesis induced by N-nitrosomethylbenzylamine (NMBA) in ESCC rat model. In the Het-1A cell model, α-Tocopherol markedly suppressed cell proliferation, promoted cell cycle G2-phase arrest and increased apoptosis. Gene microarray and proteins array analysis indicated that Akt signaling was a potential target for α-Tocopherol. We further demonstrated that α-Tocopherol increased the expression of PPARγ and its downstream tumor suppressor PTEN. Knockdown of PPARγ activated Akt signaling transduction, whereas this process was attenuated by the presence of α-Tocopherol and PPARγ agonist Rosiglitazone. In contrast, the effect of α-Tocopherol on Akt inhibition was not observed in established tumors, neither in cancerous cell lines which constitutively expressed higher levels of PPARγ. These results were closely correlated with the ineffectiveness of α-Tocopherol in the late stage of ESCC carcinogenesis. Taken together, our study suggested that α-Tocopherol may serve as a PPARγ agonist for the chemoprevention of esophageal cancer.
Collapse
|
28
|
Behera R, Kaur A, Webster MR, Kim S, Ndoye A, Kugel CH, Alicea GM, Wang J, Ghosh K, Cheng P, Lisanti S, Marchbank K, Dang V, Levesque M, Dummer R, Xu X, Herlyn M, Aplin AE, Roesch A, Caino C, Altieri DC, Weeraratna AT. Inhibition of Age-Related Therapy Resistance in Melanoma by Rosiglitazone-Mediated Induction of Klotho. Clin Cancer Res 2017; 23:3181-3190. [PMID: 28232477 PMCID: PMC5474161 DOI: 10.1158/1078-0432.ccr-17-0201] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 02/08/2017] [Accepted: 02/10/2017] [Indexed: 01/21/2023]
Abstract
Purpose: Aging is a poor prognostic factor for melanoma. We have shown that melanoma cells in an aged microenvironment are more resistant to targeted therapy than identical cells in a young microenvironment. This is dependent on age-related secreted factors. Klotho is an age-related protein whose serum levels decrease dramatically by age 40. Most studies on klotho in cancer have focused on the expression of klotho in the tumor cell. We have shown that exogenous klotho inhibits internalization and signaling of Wnt5A, which drives melanoma metastasis and resistance to targeted therapy. We investigate here whether increasing klotho in the aged microenvironment could be an effective strategy for the treatment of melanoma.Experimental Design: PPARγ increases klotho levels and is increased by glitazones. Using rosiglitazone, we queried the effects of rosiglitazone on Klotho/Wnt5A cross-talk, in vitro and in vivo, and the implications of that for targeted therapy in young versus aged animals.Results: We show that rosiglitazone increases klotho and decreases Wnt5A in tumor cells, reducing the burden of both BRAF inhibitor-sensitive and BRAF inhibitor-resistant tumors in aged, but not young mice. However, when used in combination with PLX4720, tumor burden was reduced in both young and aged mice, even in resistant tumors.Conclusions: Using glitazones as adjuvant therapy for melanoma may provide a new treatment strategy for older melanoma patients who have developed resistance to vemurafenib. As klotho has been shown to play a role in other cancers too, our results may have wide relevance for multiple tumor types. Clin Cancer Res; 23(12); 3181-90. ©2017 AACR.
Collapse
Affiliation(s)
- Reeti Behera
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Amanpreet Kaur
- The Wistar Institute, Philadelphia, Pennsylvania
- University of the Sciences, Philadelphia, Pennsylvania
| | | | - Suyeon Kim
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Abibatou Ndoye
- The Wistar Institute, Philadelphia, Pennsylvania
- University of the Sciences, Philadelphia, Pennsylvania
| | | | - Gretchen M Alicea
- The Wistar Institute, Philadelphia, Pennsylvania
- University of the Sciences, Philadelphia, Pennsylvania
| | - Joshua Wang
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Kanad Ghosh
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Phil Cheng
- University of Zurich, Zurich, Switzerland
| | | | | | - Vanessa Dang
- The Wistar Institute, Philadelphia, Pennsylvania
| | | | | | - Xiaowei Xu
- Department of Pathology, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Andrew E Aplin
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Alexander Roesch
- Department of Dermatology, University Hospital, West German Cancer Center, University Duesburg-Essen, Essen, Germany
| | | | | | | |
Collapse
|
29
|
Thermodynamics in cancers: opposing interactions between PPAR gamma and the canonical WNT/beta-catenin pathway. Clin Transl Med 2017; 6:14. [PMID: 28405929 PMCID: PMC5389954 DOI: 10.1186/s40169-017-0144-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/20/2017] [Indexed: 01/03/2023] Open
Abstract
Cancer cells are the site of numerous metabolic and thermodynamic abnormalities. We focus this review on the interactions between the canonical WNT/beta-catenin pathway and peroxisome proliferator-activated receptor gamma (PPAR gamma) in cancers and their implications from an energetic and metabolic point of view. In numerous tissues, PPAR gamma activation induces inhibition of beta-catenin pathway, while the activation of the canonical WNT/beta-catenin pathway inactivates PPAR gamma. In most cancers but not all, PPAR gamma is downregulated while the WNT/beta-catenin pathway is upregulated. In cancer cells, upregulation of the WNT/beta-catenin signaling induces dramatic changes in key metabolic enzymes that modify their thermodynamic behavior. This leads to activation of pyruvate dehydrogenase kinase1 (PDK-1) and monocarboxylate lactate transporter. Consequently, phosphorylation of PDK-1 inhibits the pyruvate dehydrogenase complex (PDH). Thus, a large part of pyruvate cannot be converted into acetyl-coenzyme A (acetyl-CoA) in mitochondria and only a part of acetyl-CoA can enter the tricarboxylic acid cycle. This leads to aerobic glycolysis in spite of the availability of oxygen. This phenomenon is referred to as the Warburg effect. Cytoplasmic pyruvate is converted into lactate. The WNT/beta-catenin pathway induces the transcription of genes involved in cell proliferation, i.e., MYC and CYCLIN D1. This ultimately promotes the nucleotide, protein and lipid synthesis necessary for cell growth and multiplication. In cancer, activation of the PI3K-AKT pathway induces an increase of the aerobic glycolysis. Moreover, prostaglandin E2 by activating the canonical WNT pathway plays also a role in cancer. In addition in many cancer cells, PPAR gamma is downregulated. Moreover, PPAR gamma contributes to regulate some key circadian genes. In cancers, abnormalities in the regulation of circadian rhythms (CRs) are observed. CRs are dissipative structures which play a key-role in far-from-equilibrium thermodynamics. In cancers, metabolism, thermodynamics and CRs are intimately interrelated.
Collapse
|
30
|
Commonalities in the Association between PPARG and Vitamin D Related with Obesity and Carcinogenesis. PPAR Res 2016; 2016:2308249. [PMID: 27579030 PMCID: PMC4992792 DOI: 10.1155/2016/2308249] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 05/15/2016] [Indexed: 02/07/2023] Open
Abstract
The PPAR nuclear receptor family has acquired great relevance in the last decade, which is formed by three different isoforms (PPARα, PPARβ/δ, and PPAR ϒ). Those nuclear receptors are members of the steroid receptor superfamily which take part in essential metabolic and life-sustaining actions. Specifically, PPARG has been implicated in the regulation of processes concerning metabolism, inflammation, atherosclerosis, cell differentiation, and proliferation. Thus, a considerable amount of literature has emerged in the last ten years linking PPARG signalling with metabolic conditions such as obesity and diabetes, cardiovascular disease, and, more recently, cancer. This review paper, at crossroads of basic sciences, preclinical, and clinical data, intends to analyse the last research concerning PPARG signalling in obesity and cancer. Afterwards, possible links between four interrelated actors will be established: PPARG, the vitamin D/VDR system, obesity, and cancer, opening up the door to further investigation and new hypothesis in this fascinating area of research.
Collapse
|
31
|
Abstract
Adiponectin (APN), an adipokine produced by adipocytes, has been shown to have a critical role in the pathogenesis of obesity-associated malignancies. Through its receptor interactions, APN may exert its anti-carcinogenic effects including regulating cell survival, apoptosis and metastasis via a plethora of signalling pathways. Despite the strong evidence supporting this notion, some work may indicate otherwise. Our review addresses all controversies critically. On the whole, hypoadiponectinaemia is associated with increased risk of several malignancies and poor prognosis. In addition, various genetic polymorphisms may predispose individuals to increased risk of obesity-associated malignancies. We also provide an updated summary on therapeutic interventions to increase APN levels that are of key interest in this field. To date efforts to manipulate APN levels have been promising, but much work remains to be done.
Collapse
Affiliation(s)
- Arnav Katira
- UCL Medical School, UCL Faculty of Medical Science, University College London, London WC1E 6BT, UK
| | - Peng H Tan
- UCL Medical School, UCL Faculty of Medical Science, University College London, London WC1E 6BT, UK; Breast Unit, Whittington Health, London N19 5NF, UK
| |
Collapse
|
32
|
Gallagher EJ, LeRoith D. Obesity and Diabetes: The Increased Risk of Cancer and Cancer-Related Mortality. Physiol Rev 2015; 95:727-48. [PMID: 26084689 DOI: 10.1152/physrev.00030.2014] [Citation(s) in RCA: 534] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Obesity and type 2 diabetes are becoming increasingly prevalent worldwide, and both are associated with an increased incidence and mortality from many cancers. The metabolic abnormalities associated with type 2 diabetes develop many years before the onset of diabetes and, therefore, may be contributing to cancer risk before individuals are aware that they are at risk. Multiple factors potentially contribute to the progression of cancer in obesity and type 2 diabetes, including hyperinsulinemia and insulin-like growth factor I, hyperglycemia, dyslipidemia, adipokines and cytokines, and the gut microbiome. These metabolic changes may contribute directly or indirectly to cancer progression. Intentional weight loss may protect against cancer development, and therapies for diabetes may prove to be effective adjuvant agents in reducing cancer progression. In this review we discuss the current epidemiology, basic science, and clinical data that link obesity, diabetes, and cancer and how treating obesity and type 2 diabetes could also reduce cancer risk and improve outcomes.
Collapse
Affiliation(s)
| | - Derek LeRoith
- Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
33
|
Chemotherapy and chemoprevention by thiazolidinediones. BIOMED RESEARCH INTERNATIONAL 2015; 2015:845340. [PMID: 25866814 PMCID: PMC4383438 DOI: 10.1155/2015/845340] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 07/29/2014] [Accepted: 08/27/2014] [Indexed: 12/13/2022]
Abstract
Thiazolidinediones (TZDs) are synthetic ligands of Peroxisome-Proliferator-Activated Receptor gamma (PPARγ). Troglitazone, rosiglitazone, and pioglitazone have been approved for treatment of diabetes mellitus type II. All three compounds, together with the first TZD ciglitazone, also showed an antitumor effect in preclinical studies and a beneficial effect in some clinical trials. This review summarizes hypotheses on the role of PPARγ in tumors, on cellular targets of TZDs, antitumor effects of monotherapy and of TZDs in combination with other compounds, with a focus on their role in the treatment of differentiated thyroid carcinoma. The results of chemopreventive effects of TZDs are also considered. Existing data suggest that the action of TZDs is highly complex and that actions do not correlate with cellular PPARγ expression status. Effects are cell-, species-, and compound-specific and concentration-dependent. Data from human trials suggest the efficacy of TZDs as monotherapy in prostate cancer and glioma and as chemopreventive agent in colon, lung, and breast cancer. TZDs in combination with other therapies might increase antitumor effects in thyroid cancer, soft tissue sarcoma, and melanoma.
Collapse
|
34
|
Griffin F. Farmed deer: A veterinary model for chronic mycobacterial diseases that is accessible, appropriate and cost-effective. J Pharm Bioallied Sci 2014; 6:10-5. [PMID: 24459398 PMCID: PMC3895287 DOI: 10.4103/0975-7406.124302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Revised: 11/20/2013] [Accepted: 11/20/2013] [Indexed: 12/15/2022] Open
Abstract
Although most studies in immunology have used inbred mice as the experimental model to study fundamental immune mechanisms they have been proven to be limited in their ability to chart complex functional immune pathways, such as are seen in outbred populations of humans or animals. Translation of the findings from inbred mouse studies into practical solutions in therapeutics or the clinic has been remarkably unproductive compared with many other areas of clinical practice in human and veterinary medicine. Access to an unlimited array of mouse strains and an increasing number of genetically modified strains continues to sustain their paramount position in immunology research. Since the mouse studies have provided little more than the dictionary and glossary of immunology, another approach will be required to write the classic exposition of functional immunity. Domestic animals such as ruminants and swine present worthwhile alternatives as models for immunological research into infectious diseases, which may be more informative and cost effective. The original constraint on large animal research through a lack of reagents has been superseded by new molecular technologies and robotics that allow research to progress from gene discovery to systems biology, seamlessly. The current review attempts to highlight how exotic animals such as deer can leverage off the knowledge of ruminant genomics to provide cost-effective models for research into complex, chronic infections. The unique opportunity they provide relates to their diversity and polymorphic genotypes and the integrity of their phenotype for a range of infectious diseases.
Collapse
Affiliation(s)
- Frank Griffin
- Department of Microbiology and Immunology, Disease Research Laboratory, University of Otago, Dunedin, New Zealand
| |
Collapse
|
35
|
Positive and negative effects of glitazones in carcinogenesis: experimental models vs. clinical practice. Pathol Res Pract 2014; 210:465-72. [PMID: 25023882 DOI: 10.1016/j.prp.2014.06.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 10/31/2013] [Accepted: 06/03/2014] [Indexed: 01/30/2023]
Abstract
Diabetes increases cancer risk, which may be modulated by careful choice of treatment. Experimental reports showed efficacy of glitazones in various in vitro and in vivo models of carcinogenesis, but procarcinogenic effects in some models were reported too, and, similarly, data on cancer incidence in glitazone users are inconsistent. This review summarizes oncostatic effects of glitazones in preclinical and clinical studies and brings a brief summary of their impact on cancer risk in diabetic patients, with a focus on the association between pioglitazone use and bladder cancer.
Collapse
|
36
|
Qin L, Gong C, Chen AM, Guo FJ, Xu F, Ren Y, Liao H. Peroxisome proliferator‑activated receptor γ agonist rosiglitazone inhibits migration and invasion of prostate cancer cells through inhibition of the CXCR4/CXCL12 axis. Mol Med Rep 2014; 10:695-700. [PMID: 24842333 DOI: 10.3892/mmr.2014.2232] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 03/07/2014] [Indexed: 11/06/2022] Open
Abstract
It has been indicated that the C‑X‑C chemokine receptor type 4/C‑X‑C chemokine ligand 12 (CXCR4/CXCL12) axis is involved in promoting invasion and metastasis in tumors. Therefore, novel drugs capable of downregulating the CXCR4/CXCL12 axis may demonstrate potential for the treatment of metastatic prostate cancer (PCa). Rosiglitazone (RSG), a thiazolidinedione ligand of the peroxisome proliferator‑activated receptor (PPAR) γ, has been found to inhibit proliferation, induce apoptosis, suppress angiogenesis and inhibit metastasis. However, the precise mechanisms by which RSG regulates CXCR4 gene expression and the consequent effects on prostate cell migration and invasion are not fully understood. In this study, it was observed that RSG is capable of downregulating the expression of CXCR4 in PCa cells in a dose‑, time‑ and PPARγ‑dependent manner. Furthermore, it was observed that the downregulation of CXCR4 expression occurred at a transcriptional level, as indicated by a reduction in CXCR4 mRNA expression. Suppression of CXCR4 expression by RSG further correlated with the inhibition of CXCL12‑induced migration and invasion in PCa cells. Analysis of the predominant intracellular signaling pathways that act downstream of the activated CXCR4/CXCL12 axis, namely the phosphatidyl inositol 3‑kinase (PI3K)‑protein kinase B (Akt) cascades, revealed that RSG rapidly interferes with the phosphorylation/activation of Akt, which mediates CXCL12‑stimulated migration and invasion. Overall, the findings of this study suggest that RSG represents a novel inhibitor of CXCR4 expression and, thus, has significant potential as a powerful therapeutic agent for the treatment of metastatic PCa.
Collapse
Affiliation(s)
- Liang Qin
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Chen Gong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - An-Min Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Feng-Jing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Fei Xu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ye Ren
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Hui Liao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
37
|
Wang ZY, Quan Y, Zhang HY. Medical genetic inspirations for anticancer drug repurposing. Trends Pharmacol Sci 2013; 35:1-3. [PMID: 24315157 DOI: 10.1016/j.tips.2013.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 11/14/2013] [Indexed: 10/25/2022]
Affiliation(s)
- Zhong-Yi Wang
- National Key Laboratory of Crop Genetic Improvement, Center for Bioinformatics, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, P. R. China
| | - Yuan Quan
- National Key Laboratory of Crop Genetic Improvement, Center for Bioinformatics, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, P. R. China
| | - Hong-Yu Zhang
- National Key Laboratory of Crop Genetic Improvement, Center for Bioinformatics, College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, P. R. China.
| |
Collapse
|
38
|
Koyama M, Sowa Y, Horinaka M, Goda AE, Fujiwara J, Sakai T. Peroxisome proliferator-activated receptor γ ligand troglitazone and TRAIL synergistically induce apoptosis. Oncol Rep 2013; 31:947-54. [PMID: 24276615 DOI: 10.3892/or.2013.2868] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 10/25/2013] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is known to cause apoptosis in several types of malignant tumor cells through its interaction with the death domain-containing receptor, death receptor 5 (DR5). In the present study, we showed that co-treatment with troglitazone (TGZ), a synthetic ligand of peroxisome proliferator-activated receptor γ (PPARγ), and TRAIL synergistically induced apoptosis through DR5 upregulation in human colon cancer DLD-1 cells. TGZ elevated DR5 expression at the promoter level through the CCAAT/enhancer-binding protein homologous protein (CHOP) binding site. These results suggest that combined treatment with TGZ and TRAIL may be promising as a new therapy against malignant tumors.
Collapse
Affiliation(s)
- Makoto Koyama
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yoshihiro Sowa
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Mano Horinaka
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Ahmed E Goda
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Jun Fujiwara
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Toshiyuki Sakai
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
39
|
Sawayama H, Ishimoto T, Watanabe M, Yoshida N, Sugihara H, Kurashige J, Hirashima K, Iwatsuki M, Baba Y, Oki E, Morita M, Shiose Y, Baba H. Small molecule agonists of PPAR-γ exert therapeutic effects in esophageal cancer. Cancer Res 2013; 74:575-85. [PMID: 24272485 DOI: 10.1158/0008-5472.can-13-1836] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The transcription factor PPAR-γ plays various roles in lipid metabolism, inflammation, cellular differentiation, and apoptosis. PPAR-γ agonists used to treat diabetes may have utility in cancer treatment. Efatutazone is a novel later generation PPAR-γ agonist that selectively activates PPAR-γ target genes and has antiproliferative effects in a range of malignancies. In this study, we investigated PPAR-γ status in esophageal squamous cell carcinoma (ESCC) and investigated the antiproliferative effects of efatutazone. PPAR-γ was expressed heterogeneously in ESCC, in which it exhibited an inverse relationship with Ki-67 expression. PPAR-γ expression was associated independently with good prognosis in ESCC. Efatutazone, but not the conventional PPAR-γ agonist troglitazone, inhibited ESCC cell proliferation in vitro and in vivo. Mechanistic investigations suggested that efatutazone acted by upregulating p21Cip1 protein in the nucleus through inactivation of the Akt pathway and dephosphorylation of p21Cip1 at Thr145 without affecting the transcriptional activity of p21Cip1. We also found that treatment with efatutazone led to phosphorylation of the EGF receptor and activation of the mitogen-activated protein kinase (MAPK) pathway. Accordingly, the combination of efatutazone with the antiepithelial growth factor receptor antibody cetuximab synergized to negatively regulate the phosphoinositide 3-kinase-Akt and MAPK pathways. Together, our results suggest that efatutazone, alone or in combination with cetuximab, may offer therapeutic effects in ESCC.
Collapse
Affiliation(s)
- Hiroshi Sawayama
- Authors' Affiliations: Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto; Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka; and Research and Development Division, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Rousseaux C, El-Jamal N, Fumery M, Dubuquoy C, Romano O, Chatelain D, Langlois A, Bertin B, Buob D, Colombel JF, Cortot A, Desreumaux P, Dubuquoy L. The 5-aminosalicylic acid antineoplastic effect in the intestine is mediated by PPARγ. Carcinogenesis 2013; 34:2580-6. [PMID: 23843037 PMCID: PMC3810841 DOI: 10.1093/carcin/bgt245] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Epidemiological evidences suggested that 5-aminosalicylic acid (5-ASA) therapy may prevent the development of colorectal cancer in inflammatory bowel disease patients. Our aim is to investigate whether peroxisome proliferator-activated receptor-γ (PPARγ) mediates the antineoplastic effects of 5-ASA. HT-29 and Caco-2 cells were treated by 5-ASA, rosiglitazone (PPARγ ligand) or etoposide (anticarcinogenic drug). Epithelial cell growth, proliferation and apoptosis were assessed by cell count, Ki-67 staining and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay, respectively. The antineoplastic effect of 5-ASA was evaluated in a xenograft tumor model in SCID mice and in azoxymethane (AOM)-induced colon carcinogenesis in A/JOlaHsd mice. The role of PPARγ was examined by administration of PPARγ antagonist, GW9662 and in PPAR knockdown cells. Compared with untreated cells, treatment of HT-29 cells by 5-ASA inhibited significantly cell growth and cell proliferation (respectively, 60% and 63%) and induced apoptosis in 75% of cells. These effects were abolished by co-treatment with GW9662 and blunted in PPAR knockdown cells. Contrarily to etoposide, similar inhibitory effects of GW9662 were obtained in HT-29 cells treated with rosiglitazone. In the xenograft model, GW9662 abolished the therapeutic effect of 5-ASA, which decreased tumor weight and volume by 80% in SCID mice compared with untreated mice. In A/JOlaHsd mice, 5-ASA suppressed colon carcinogenesis by decreasing the number of aberrant crypt foci (75%) and aberrant crypts (22%) induced by AOM treatment with an absence of 5-ASA response after GW9662 administration. In conclusion, 5-ASA exerts potent antineoplastic effects that are mediated through PPARγ. These data provide new rational for designing more effective and safe antineoplastic PPARγ ligands with topical effects.
Collapse
Affiliation(s)
- Christel Rousseaux
- Department of Project Management, Intestinal Biotech Development, 59045 Lille, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Park J, Morley TS, Scherer PE. Inhibition of endotrophin, a cleavage product of collagen VI, confers cisplatin sensitivity to tumours. EMBO Mol Med 2013; 5:935-48. [PMID: 23629957 PMCID: PMC3779453 DOI: 10.1002/emmm.201202006] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2012] [Revised: 03/17/2013] [Accepted: 03/18/2013] [Indexed: 01/24/2023] Open
Abstract
Endotrophin is a cleavage product of collagenVIα3 (COL6A3). Here, we explore the relationship between thiazolidinediones (TZDs), endotrophin and cisplatin resistance in the context of a mammary tumour model. COL6A3 levels are increased in response to cisplatin exposure in tumours. Endotrophin, in turn, causes cisplatin resistance. The effects of endotrophin can be bypassed, either through use of COL6 null (COL6−/−) mice or by administering TZDs in wild-type mice (leading to a downregulation of endotrophin). Both approaches sensitize tumours to cisplatin through the suppression of endotrophin-induced epithelial–mesenchymal transition. The beneficial effects of TZDs on cisplatin sensitivity are diminished in COL6−/− mice, whereas endotrophin+ tumours are sensitive to the TZD/cisplatin combination. Therefore, the chemosensitization obtained with TZDs is achieved through a downregulation of endotrophin. Treatment with an endotrophin neutralizing antibody in combination with cisplatin completely inhibits tumour growth of tumour allografts. Combined, our data suggest that endotrophin levels are a strong prognostic marker for the effectiveness of the combination therapy of TZDs with cisplatin, and neutralization of endotrophin activity dramatically improves the therapeutic response to combination therapy.
Collapse
Affiliation(s)
- Jiyoung Park
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | |
Collapse
|
42
|
Chen SW, Tsan YT, Chen JD, Hsieh HI, Lee CH, Lin HH, Wang JD, Chen PC. Use of thiazolidinediones and the risk of colorectal cancer in patients with diabetes: a nationwide, population-based, case-control study. Diabetes Care 2013; 36:369-75. [PMID: 23043163 PMCID: PMC3554275 DOI: 10.2337/dc11-2197] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Preclinical data suggest that peroxisome proliferator-activated receptor γ (PPARγ) agonists have antineoplastic effects in colorectal cancer. We aimed to assess the association between the use of synthetic PPARγ agonists, represented by thiazolidinediones (TZDs), and the risk of developing colorectal cancer. RESEARCH DESIGN AND METHODS We conducted a nationwide, population-based, case-control study using the Taiwan National Health Insurance Research Database. Case subjects were defined as patients who were diagnosed with diabetes at least 365 days prior to a new diagnosis of colorectal cancer between 2000 and 2008. We randomly selected diabetic control subjects for each case subject, which were matched by sex, age, and the duration of diabetes. Among the 24,496 eligible case subjects and control subjects, we used conditional logistic regression to assess the risk of colorectal cancer in association with the use of TZDs. An additional analysis was conducted to assess the effects of concomitant use of TZDs and low-dose aspirin or nonsteroidal anti-inflammatory drugs (NSAIDs) on the risk of colorectal cancer. RESULTS A decreased risk of colorectal cancer was observed in patients who had used TZDs compared with those who had never used TZDs (adjusted odds ratio 0.86 [95% CI 0.79-0.94]). Furthermore, the benefit of a decreased colorectal cancer risk was also found with concomitant use of TZDs and low-dose aspirin or NSAIDs. CONCLUSIONS The use of TZDs may be associated with a decreased risk of colorectal cancer in patients with diabetes. Further studies are warranted to confirm our findings.
Collapse
Affiliation(s)
- Shih-Wei Chen
- Institute of Occupational Medicine and Industrial Hygiene, National Taiwan University College of Public Health, Taipei, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Targeting PPARγ Signaling Cascade for the Prevention and Treatment of Prostate Cancer. PPAR Res 2012; 2012:968040. [PMID: 23213321 PMCID: PMC3504464 DOI: 10.1155/2012/968040] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 10/08/2012] [Accepted: 10/18/2012] [Indexed: 12/21/2022] Open
Abstract
The peroxisome proliferator-activated receptor-gamma (PPARγ) is a member of the hormone-activated nuclear receptor superfamily. PPARγ can be activated by a diverse group of agents, such as endogenous polyunsaturated fatty acids, 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2), and thiazolidinedione (TZD) drugs. PPARγ induces antiproliferative, antiangiogenic, and prodifferentiation pathways in several tissue types, thus making it a highly useful target for downregulation of carcinogenesis. These TZD-derived novel therapeutic agents, alone or in combination with other anticancer drugs, have translational relevance in fostering effective strategies for cancer treatment. TZDs have been proven for antitumor activity in a wide variety of experimental cancer models, both in vitro and in vivo, by affecting the cell cycle, inducing cell differentiation and apoptosis, as well as by inhibiting tumor angiogenesis. Angiogenesis inhibition mechanisms of TZDs include direct inhibition of endothelial cell proliferation and migration, as well as reduction in tumor cell vascular endothelial growth factor production. In prostate cancer, PPARγ ligands such as troglitazone and 15d-PGJ2 have also shown to inhibit tumor growth. This paper will focus on current discoveries in PPARγ activation, targeting prostate carcinogenesis as well as the role of PPARγ as a possible anticancer therapeutic option. Here, we review PPARγ as an antitumor agent and summarize the antineoplastic effects of PPARγ agonists in prostate cancer.
Collapse
|
44
|
Roberts PJ, Usary JE, Darr DB, Dillon PM, Pfefferle AD, Whittle MC, Duncan JS, Johnson SM, Combest AJ, Jin J, Zamboni WC, Johnson GL, Perou CM, Sharpless NE. Combined PI3K/mTOR and MEK inhibition provides broad antitumor activity in faithful murine cancer models. Clin Cancer Res 2012; 18:5290-303. [PMID: 22872574 DOI: 10.1158/1078-0432.ccr-12-0563] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Anticancer drug development is inefficient, but genetically engineered murine models (GEMM) and orthotopic, syngeneic transplants (OST) of cancer may offer advantages to in vitro and xenograft systems. EXPERIMENTAL DESIGN We assessed the activity of 16 treatment regimens in a RAS-driven, Ink4a/Arf-deficient melanoma GEMM. In addition, we tested a subset of treatment regimens in three breast cancer models representing distinct breast cancer subtypes: claudin-low (T11 OST), basal-like (C3-TAg GEMM), and luminal B (MMTV-Neu GEMM). RESULTS Like human RAS-mutant melanoma, the melanoma GEMM was refractory to chemotherapy and single-agent small molecule therapies. Combined treatment with AZD6244 [mitogen-activated protein-extracellular signal-regulated kinase kinase (MEK) inhibitor] and BEZ235 [dual phosphoinositide-3 kinase (PI3K)/mammalian target of rapamycin (mTOR) inhibitor] was the only treatment regimen to exhibit significant antitumor activity, showed by marked tumor regression and improved survival. Given the surprising activity of the "AZD/BEZ" combination in the melanoma GEMM, we next tested this regimen in the "claudin-low" breast cancer model that shares gene expression features with melanoma. The AZD/BEZ regimen also exhibited significant activity in this model, leading us to testing in even more diverse GEMMs of basal-like and luminal breast cancer. The AZD/BEZ combination was highly active in these distinct breast cancer models, showing equal or greater efficacy compared with any other regimen tested in studies of over 700 tumor-bearing mice. This regimen even exhibited activity in lapatinib-resistant HER2(+) tumors. CONCLUSION These results show the use of credentialed murine models for large-scale efficacy testing of diverse anticancer regimens and predict that combinations of PI3K/mTOR and MEK inhibitors will show antitumor activity in a wide range of human malignancies.
Collapse
Affiliation(s)
- Patrick J Roberts
- Department of Genetics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Roper J, Hung KE. Priceless GEMMs: genetically engineered mouse models for colorectal cancer drug development. Trends Pharmacol Sci 2012; 33:449-55. [DOI: 10.1016/j.tips.2012.05.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 04/11/2012] [Accepted: 05/02/2012] [Indexed: 12/13/2022]
|
46
|
Skelhorne-Gross G, Nicol CJB. The Key to Unlocking the Chemotherapeutic Potential of PPARγ Ligands: Having the Right Combination. PPAR Res 2012; 2012:946943. [PMID: 22966225 PMCID: PMC3395155 DOI: 10.1155/2012/946943] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 03/14/2012] [Indexed: 01/03/2023] Open
Abstract
Despite extensive preclinical evidence that peroxisome proliferator-activated receptor (PPAR)γ activation protects against tumourigenesis, results from a few clinical trials using PPARγ ligands as monotherapy show modest success. In spite of this, several groups reported exciting results with therapeutic regimens that combine PPARγ ligands with other compounds: chemotherapeutic agents, retinoid x receptor (RXR)α agonists, statins, or cell-to-cell signaling molecules in preclinical cancer models and human trials. Here we have compiled an extensive review, consolidating the existing literature, which overwhelmingly supports a beneficial effect of treating with PPARγ ligands in combination with existing chemotherapies versus their monotherapy in cancer. There are many examples in which combination therapy resulted in synergistic/additive effects on apoptosis, differentiation, and the ability to reduce cell growth and tumour burden. There are also studies that indicate that PPARγ ligand pretreatment overcomes resistance and reduces toxicities. Several mechanisms are explored to explain these protective effects. This paper highlights each of these studies that, collectively, make a very strong case for the use of PPARγ ligands in combination with other agents in the treatment and management of several cancers.
Collapse
Affiliation(s)
- Graham Skelhorne-Gross
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada K7L 3N6
- Cancer Biology and Genetics Division, Cancer Research Institute, Queen's University, Kingston, ON, Canada K7L 3N6
| | - Christopher J. B. Nicol
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada K7L 3N6
- Cancer Biology and Genetics Division, Cancer Research Institute, Queen's University, Kingston, ON, Canada K7L 3N6
- Department of Biomedical and Molecular Sciences (Pharmacology and Toxicology), Queen's University, Kingston, ON, Canada K7L 3N6
| |
Collapse
|
47
|
Vamecq J, Colet JM, Vanden Eynde JJ, Briand G, Porchet N, Rocchi S. PPARs: Interference with Warburg' Effect and Clinical Anticancer Trials. PPAR Res 2012; 2012:304760. [PMID: 22654896 PMCID: PMC3357561 DOI: 10.1155/2012/304760] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 02/15/2012] [Accepted: 02/19/2012] [Indexed: 02/07/2023] Open
Abstract
The metabolic/cell signaling basis of Warburg's effect ("aerobic glycolysis") and the general metabolic phenotype adopted by cancer cells are first reviewed. Several bypasses are adopted to provide a panoramic integrated view of tumoral metabolism, by attributing a central signaling role to hypoxia-induced factor (HIF-1) in the expression of aerobic glycolysis. The cancer metabolic phenotype also results from alterations of other routes involving ras, myc, p53, and Akt signaling and the propensity of cancer cells to develop signaling aberrances (notably aberrant surface receptor expression) which, when present, offer unique opportunities for therapeutic interventions. The rationale for various emerging strategies for cancer treatment is presented along with mechanisms by which PPAR ligands might interfere directly with tumoral metabolism and promote anticancer activity. Clinical trials using PPAR ligands are reviewed and followed by concluding remarks and perspectives for future studies. A therapeutic need to associate PPAR ligands with other anticancer agents is perhaps an important lesson to be learned from the results of the clinical trials conducted to date.
Collapse
Affiliation(s)
- Joseph Vamecq
- Inserm, HMNO, CBP, CHRU Lille, 59037 Lille, France
- Biochemistry and Molecular Biology, HMNO, CBP, CHRU Lille, 59037 Lille, France
| | - Jean-Marie Colet
- Department of Human Biology and Toxicology, Faculty of Medicine and Pharmacy, UMons, 7000 Mons, Belgium
| | | | - Gilbert Briand
- Biochemistry and Molecular Biology, HMNO, CBP, CHRU Lille, 59037 Lille, France
| | - Nicole Porchet
- Biochemistry and Molecular Biology, HMNO, CBP, CHRU Lille, 59037 Lille, France
| | - Stéphane Rocchi
- Inserm U1065, IFR 50, Mediterranean Center of Molecular Medicine, 06204 Nice, France
| |
Collapse
|
48
|
Allstadt Frazier S, McKemie DS, Guerrero TA, LaChapelle H, Skorupski KA, Kass PH, Rodriguez CO. Phase I clinical trial of oral rosiglitazone in combination with intravenous carboplatin in cancer-bearing dogs. Vet Comp Oncol 2012; 12:1-9. [DOI: 10.1111/j.1476-5829.2012.00322.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- S. Allstadt Frazier
- Veterinary Medical Teaching Hospital; University of California; Davis CA USA
- Department of Veterinary Surgical and Radiological Sciences; University of California; Davis CA USA
| | - D. S. McKemie
- Department of Molecular Biosciences; University of California; Davis CA USA
| | - T. A. Guerrero
- Department of Veterinary Surgical and Radiological Sciences; University of California; Davis CA USA
| | - H. LaChapelle
- Veterinary Medical Teaching Hospital; University of California; Davis CA USA
| | - K. A. Skorupski
- Veterinary Medical Teaching Hospital; University of California; Davis CA USA
- Department of Veterinary Surgical and Radiological Sciences; University of California; Davis CA USA
| | - P. H. Kass
- Department of Population, Health & Reproduction; University of California; Davis CA USA
| | - C. O. Rodriguez
- Veterinary Medical Teaching Hospital; University of California; Davis CA USA
- Department of Veterinary Surgical and Radiological Sciences; University of California; Davis CA USA
| |
Collapse
|
49
|
Peters JM, Shah YM, Gonzalez FJ. The role of peroxisome proliferator-activated receptors in carcinogenesis and chemoprevention. Nat Rev Cancer 2012; 12:181-95. [PMID: 22318237 PMCID: PMC3322353 DOI: 10.1038/nrc3214] [Citation(s) in RCA: 375] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that are involved in regulating glucose and lipid homeostasis, inflammation, proliferation and differentiation. Although all of these functions might contribute to the influence of PPARs in carcinogenesis, there is a distinct need for a review of the literature and additional experimentation to determine the potential for targeting PPARs for cancer therapy and cancer chemoprevention. As PPAR agonists include drugs that are used for the treatment of metabolic diseases, a more complete understanding of the roles of PPARs in cancer will aid in determining any increased cancer risk for patients undergoing therapy with PPAR agonists.
Collapse
Affiliation(s)
- Jeffrey M Peters
- Department of Veterinary and Biomedical Sciences and The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, Pennsylvania 16802, USA.
| | | | | |
Collapse
|
50
|
Youssef J, Badr M. Peroxisome proliferator-activated receptors and cancer: challenges and opportunities. Br J Pharmacol 2012; 164:68-82. [PMID: 21449912 DOI: 10.1111/j.1476-5381.2011.01383.x] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs), members of the nuclear hormone receptor superfamily, function as transcription factors and modulators of gene expression. These actions allow PPARs to regulate a variety of biological processes and to play a significant role in several diseases and conditions. The current literature describes frequently opposing and paradoxical roles for the three PPAR isotypes, PPARα, PPARβ/δ and PPARγ, in cancer. While some studies have implicated PPARs in the promotion and development of cancer, others, in contrast, have presented evidence for a protective role for these receptors against cancer. In some tissues, the expression level of these receptors and/or their activation correlates with a positive outcome against cancer, while, in other tissue types, their expression and activation have the opposite effect. These disparate findings raise the possibility of (i) PPAR receptor-independent effects, including effects on receptors other than PPARs by the utilized ligands; (ii) cancer stage-specific effect; and/or (iii) differences in essential ligand-related pharmacokinetic considerations. In this review, we highlight the latest available studies on the role of the various PPAR isotypes in cancer in several major organs and present challenges as well as promising opportunities in the field.
Collapse
Affiliation(s)
- Jihan Youssef
- University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | | |
Collapse
|