1
|
Zhou Y, Wang L, Liu M, Jiang H, Wu Y. Oral squamous cell carcinoma: Insights into cellular heterogeneity, drug resistance, and evolutionary trajectories. Cell Biol Toxicol 2025; 41:101. [PMID: 40504271 PMCID: PMC12162747 DOI: 10.1007/s10565-025-10048-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 05/25/2025] [Indexed: 06/16/2025]
Abstract
Oral squamous cell carcinoma (OSCC) can lead to metastasis and high mortality rates known for its aggressive and invasive properties. Currently, primary treatment options of surgical resection, chemotherapy and radiotherapy have many therapeutic limitations for OSCC patients due to its dynamic evolutionary pathways and the development of resistance to conventional therapies. Moreover, previous studies fail to emphasize the roles of cellular heterogeneity, drug resistance, and evolutionary trajectories in OSCC. This review explores the intricate tumor microenvironment landscape of OSCC, focusing on the cellular heterogeneity, drug resistance, and evolutionary trajectories as well as genetic, epigenetic, and environmental risk factors contributing to the OSCC progression. Tumor heterogeneity arises from environmental exposures (e.g., tobacco, HPV infection, dietary carcinogens) that drive clonal evolution, creating subpopulations of cells with distinct mutational profiles and therapeutic vulnerabilities. Recent advances in in the precision medicine and combination therapy of OSCC paves the way for innovative therapeutic strategies, such as targeting molecular subclones through real-time monitoring and leveraging computational models to predict treatment response. By recognizing tumor heterogeneity as both a driver of therapeutic resistance and a therapeutic target, precision medicine frameworks can integrate environmental risk factor data, molecular profiling, and early detection tools to optimize outcomes. This review underscores the necessity for a multidisciplinary approach to understand and combat the complexity of OSCC, proposing directions for future research to enhance diagnosis and treatment efficacy.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Obstetrics and Gynaecology, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Shenyang, 110000, China
| | - Liyin Wang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Minghua Liu
- Departmentof Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, 110000, China
| | - Hongfang Jiang
- Department of Geriatrics, Shengjing Hospital of China Medical University, Shenyang, 110000, China.
| | - Yan Wu
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
2
|
Moomivand S, Nikbakht M, Majd A, Bikhof Torbati M, Mousavi SA. Combining Chemotherapy Agents and Autophagy Modulators for Enhanced Breast Cancer Cell Death. Adv Pharm Bull 2024; 14:908-917. [PMID: 40190668 PMCID: PMC11970493 DOI: 10.34172/apb.42733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 10/18/2024] [Accepted: 10/29/2024] [Indexed: 04/09/2025] Open
Abstract
Purpose Autophagy, governed by genes with dual roles in cell death and survival, plays a crucial role in cancer persistence. Arsenic trioxide (ATO), carboplatin (CP), and cyclophosphamide (CY) are used to treat various cancers. ATO impedes cell proliferation and triggers apoptosis in cancer cells. CP, a platinum-based drug, damages cancer cell DNA, while CY acts as an alkylating agent, disrupting cell proliferation. This study investigates the combined effects of ATO, CP, and CY on inducing apoptosis and modulating autophagy in triple-negative breast cancer (TNBC) cell lines, BT-20 and MDA-MB-231. Methods The cytotoxic effects of ATO, CP, and CY, alone and in combination, were evaluated using the MTT assay on BT-20 and MDA-MB-231 cells. Apoptosis and cell cycle progression were analyzed by annexin-V FITC/PI staining and flow cytometry. Gene expression of autophagy-and apoptosis-related markers, including Beclin 1, LC3, caspase 3, and BCL2, was quantified using RT-PCR. Data were analyzed using GraphPad Prism 4.0 with one-way ANOVA followed by Dunnett's test. Results The combination of ATO, CP, and CY significantly reduced cell viability and enhanced apoptosis, evidenced by increased caspase-3 activity and reduced BCL2 expression. Cell cycle arrest in the G1 phase was observed, alongside elevated autophagy markers Beclin 1 and LC3. Conclusion The combination of ATO, CP, and CY induces synergistic effects in promoting apoptosis and autophagy in TNBC cell lines. These findings suggest that this combination therapy could be a promising approach to enhancing treatment efficacy in aggressive breast cancers, offering new insights into potential therapeutic strategies.
Collapse
Affiliation(s)
- Soraya Moomivand
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
- Research Institute for Oncology, Hematology and Cell Therapy Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Nikbakht
- Research Institute for Oncology, Hematology and Cell Therapy Tehran University of Medical Sciences, Tehran, Iran
- Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran, Iran
| | - Ahmad Majd
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Maryam Bikhof Torbati
- Department of Biology, Yadegar-e-Imam Khomeini (RAH) Shahre rey branch, Islamic Azad University, Tehran, Iran
| | - Seyed Asadoullah Mousavi
- Research Institute for Oncology, Hematology and Cell Therapy Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Li S, Zhang Y, Li H, He Z, Li A, Yan J, Li D. Carboplatin combined with arsenic trioxide versus carboplatin combined with docetaxel treatment for LACC: A randomized, open-label, phase II clinical study. Open Med (Wars) 2024; 19:20241106. [PMID: 39669373 PMCID: PMC11635763 DOI: 10.1515/med-2024-1106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 12/14/2024] Open
Abstract
Purpose This study compared the efficacy and safety of carboplatin combined with arsenic trioxide versus carboplatin combined with docetaxel in treating locally advanced cervical cancer (LACC). Methods A total of 48 patients were enrolled between January 2019 and December 2022 and randomly assigned to the experimental group (carboplatin + arsenic trioxide, n = 24) or control group (carboplatin + docetaxel, n = 24). The clinical efficacy, adverse reactions, and serological markers were analyzed. Results There was no significant difference in baseline characteristics or total effective rates between the two groups (72.22% vs 68.42%, P > 0.05). Both groups showed significant reductions in serum squamous cell carcinoma antigen levels after chemotherapy (P < 0.05), but no significant difference was observed between groups (6.00 ± 11.36 ng/mL vs 8.42 ± 12.17 ng/mL, P > 0.05). Additionally, there was no significant difference in the incidence of adverse reactions (P > 0.05). Conclusion Arsenic trioxide combined with carboplatin as a preoperative neoadjuvant chemotherapy for LACC is not worse than docetaxel combined with carboplatin in terms of short-term efficacy and safety during the treatment of LACC.
Collapse
Affiliation(s)
- Sijin Li
- Medical Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, China
| | - Yawen Zhang
- Gynecology, Baiyun Branch of the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Haiying Li
- Gynecology, Zhuhai Branch of Guangdong Provincial Hospital of Traditional Chinese Medicine, Zhuhai, Guangdong, China
| | - Zeyang He
- Gynecology, Beijing University of Chinese Medicine Shenzhen Hospital (Longgang), Shenzhen, Guangdong, China
| | - An Li
- Medical Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, China
| | - Jiao Yan
- Respiratory Department, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong, China
| | - Daocheng Li
- Medical Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, Guangdong, China
| |
Collapse
|
4
|
Putnová I, Putnová BM, Hurník P, Štembírek J, Buchtová M, Kolísková P. Primary cilia-associated signalling in squamous cell carcinoma of head and neck region. Front Oncol 2024; 14:1413255. [PMID: 39234399 PMCID: PMC11372790 DOI: 10.3389/fonc.2024.1413255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/29/2024] [Indexed: 09/06/2024] Open
Abstract
Squamous cell carcinoma (SCC) of the head and neck originates from the mucosal lining of the upper aerodigestive tract, including the lip, tongue, nasopharynx, oropharynx, larynx and hypopharynx. In this review, we summarise what is currently known about the potential function of primary cilia in the pathogenesis of this disease. As primary cilia represent a key cellular structure for signal transduction and are related to cell proliferation, an understanding of their role in carcinogenesis is necessary for the design of new treatment approaches. Here, we introduce cilia-related signalling in head and neck squamous cell carcinoma (HNSCC) and its possible association with HNSCC tumorigenesis. From this point of view, PDGF, EGF, Wnt and Hh signalling are discussed as all these pathways were found to be dysregulated in HNSCC. Moreover, we review the clinical potential of small molecules affecting primary cilia signalling to target squamous cell carcinoma of the head and neck area.
Collapse
Affiliation(s)
- Iveta Putnová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Anatomy, Histology and Embryology, University of Veterinary Sciences Brno, Brno, Czechia
| | - Barbora Moldovan Putnová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Pathological Morphology and Parasitology, University of Veterinary Sciences Brno, Brno, Czechia
| | - Pavel Hurník
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Institute of Molecular and Clinical Pathology and Medical Genetics, University Hospital Ostrava, Ostrava, Czechia
- Institute of Molecular and Clinical Pathology and Medical Genetics, Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Jan Štembírek
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Maxillofacial Surgery, University Hospital Ostrava, Ostrava, Czechia
| | - Marcela Buchtová
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Petra Kolísková
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| |
Collapse
|
5
|
Troisi R, Tito G, Ferraro G, Sica F, Massai L, Geri A, Cirri D, Messori L, Merlino A. On the mechanism of action of arsenoplatins: arsenoplatin-1 binding to a B-DNA dodecamer. Dalton Trans 2024; 53:3476-3483. [PMID: 38270175 DOI: 10.1039/d3dt04302a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
The reaction of Pt-based anticancer agents with arsenic trioxide affords robust complexes known as arsenoplatins. The prototype of this family of anticancer compounds is arsenoplatin-1 (AP-1) that contains an As(OH)2 fragment linked to a Pt(II) moiety derived from cisplatin. Crystallographic and spectrometric studies of AP-1 binding to a B-DNA double helix dodecamer are presented here, in comparison with cisplatin and transplatin. Results reveal that AP-1, cisplatin and transplatin react differently with the DNA model system. Notably, in the AP-1/DNA systems, the Pt-As bond can break down with time and As-containing fragments can be released. These results have implications for the understanding of the mechanism of action of arsenoplatins.
Collapse
Affiliation(s)
- Romualdo Troisi
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, 80126, Naples, Italy.
- Institute of Biostructures and Bioimaging, CNR, via Pietro Castellino 111, 80131 Naples, Italy
| | - Gabriella Tito
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, 80126, Naples, Italy.
| | - Giarita Ferraro
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, 80126, Naples, Italy.
| | - Filomena Sica
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, 80126, Naples, Italy.
| | - Lara Massai
- Department of Chemistry "U. Schiff", University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Andrea Geri
- Department of Chemistry "U. Schiff", University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Damiano Cirri
- Department of Chemistry and Industrial Chemistry (DCCI), University of Pisa, Via Giuseppe Moruzzi 13, 56124 Pisa, Italy
| | - Luigi Messori
- Department of Chemistry "U. Schiff", University of Florence, via della Lastruccia 3, 50019 Sesto Fiorentino, Italy
| | - Antonello Merlino
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte Sant'Angelo, via Cintia, 80126, Naples, Italy.
| |
Collapse
|
6
|
Ananta, Benerjee S, Tchounwou PB, Kumar S. Mechanistic update of Trisenox in blood cancer. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2023; 5:100166. [PMID: 38074774 PMCID: PMC10701371 DOI: 10.1016/j.crphar.2023.100166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 10/28/2023] [Accepted: 11/14/2023] [Indexed: 02/12/2024] Open
Abstract
Acute promyelocytic leukemia (APL)/blood cancer is M3 type of acute myeloid leukemia (AML) formed inside bone marrow through chromosomal translocation mutation usually between chromosome 15 & 17. It accounts around 10% cases of AML worldwide. Trisenox (TX/ATO) is used in chemotherapy for treatment of all age group of APL patients with highest efficacy and survival rate for longer period. High concentration of TX inhibits growth of APL cells by diverse mechanism however, it cures only PML-RARα fusion gene/oncogene containing APL patients. TX resistant APL patients (different oncogenic make up) have been reported from worldwide. This review summarizes updated mechanism of TX action via PML nuclear bodies formation, proteasomal degradation, autophagy, p53 activation, telomerase activity, heteromerization of pRb & E2F, and regulation of signaling mechanism in APL cells. We have also provided important information of combination therapy of TX with other molecules mechanism of action in acute leukemia cells. It provides updated information of TX action for researcher which may help finding new target for further research in APL pathophysiology or new TX resistant APL patients drug designing.
Collapse
Affiliation(s)
- Ananta
- Department of Life Sciences, School of Earth, Biological, and Environmental Sciences, Central University of South Bihar, Gaya, India
| | - Swati Benerjee
- Department of Life Sciences, School of Earth, Biological, and Environmental Sciences, Central University of South Bihar, Gaya, India
| | - Paul B. Tchounwou
- RCMI Center for Urban Health Disparities Research and Innovation, Morgan State University, Baltimore, MD 21251, USA
| | - Sanjay Kumar
- Department of Life Sciences, School of Earth, Biological, and Environmental Sciences, Central University of South Bihar, Gaya, India
| |
Collapse
|
7
|
Cierpikowski P, Leszczyszyn A, Bar J. The Role of Hedgehog Signaling Pathway in Head and Neck Squamous Cell Carcinoma. Cells 2023; 12:2083. [PMID: 37626893 PMCID: PMC10453169 DOI: 10.3390/cells12162083] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/12/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth leading malignancy worldwide, with a poor prognosis and limited treatment options. Molecularly targeted therapies for HNSCC are still lacking. However, recent reports provide novel insights about many molecular alterations in HNSCC that may be useful in future therapies. Therefore, it is necessary to identify new biomarkers that may provide a better prediction of the disease and promising targets for personalized therapy. The poor response of HNSCC to therapy is attributed to a small population of tumor cells called cancer stem cells (CSCs). Growing evidence indicates that the Hedgehog (HH) signaling pathway plays a crucial role in the development and maintenance of head and neck tissues. The HH pathway is normally involved in embryogenesis, stem cell renewal, and tissue regeneration. However, abnormal activation of the HH pathway is also associated with carcinogenesis and CSC regulation. Overactivation of the HH pathway was observed in several tumors, including basal cell carcinoma, that are successfully treated with HH inhibitors. However, clinical studies about HH pathways in HNSCC are still rare. In this review, we summarize the current knowledge and recent advances regarding the HH pathway in HNSCC and discuss its possible implications for prognosis and future therapy.
Collapse
Affiliation(s)
- Piotr Cierpikowski
- Department of Maxillofacial Surgery, The Ludwik Rydygier Specialist Hospital, Osiedle Zlotej Jesieni 1, 31-826 Krakow, Poland
| | - Anna Leszczyszyn
- Dental Surgery Outpatient Clinic, 4th Military Clinical Hospital, Weigla 5, 53-114 Wroclaw, Poland;
| | - Julia Bar
- Department of Immunopathology and Molecular Biology, Wroclaw Medical University, Bujwida 44, 50-345 Wroclaw, Poland
| |
Collapse
|
8
|
Onda M, Ota A, Ito K, Ono T, Karnan S, Kato M, Kondo S, Furuhashi A, Hayashi T, Hosokawa Y, Kazaoka Y. Inhibition of VEGFR2 and EGFR signaling cooperatively suppresses the proliferation of oral squamous cell carcinoma. Cancer Med 2023; 12:16416-16430. [PMID: 37341071 PMCID: PMC10469792 DOI: 10.1002/cam4.6282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 06/05/2023] [Accepted: 06/12/2023] [Indexed: 06/22/2023] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) is frequently overexpressed in oral squamous cell carcinoma (OSCC), and EGFR-targeting therapeutics have been widely employed to treat patients with a variety of carcinomas including OSCC. Here, we aimed to investigate alternative signaling for OSCC survival under the disruption of EGFR signaling. METHODS OSCC cell lines, namely HSC-3 and SAS, were utilized to investigate how EGFR disruption affects cell proliferation. Gene set enrichment analysis was performed to examine how EGFR disruption affects oncogenic signaling in OSCC cells. Disruption of KDR gene was performed using CRISPR/Cas9 techniques. A VEGFR inhibitor, vatalanib was used to research the impact of VEGFR inhibition on OSCC survival. RESULTS EGFR disruption significantly decreased the proliferation and oncogenic signaling including Myc and PI3K-Akt, in OSCC cells. Chemical library screening assays revealed that VEGFR inhibitors continued to inhibit the proliferation of EGFR-deficient OSCC cells. In addition, CRISPR-mediated disruption of KDR/VEGFR2 retarded OSCC cell proliferation. Furthermore, combined erlotinib-vatalanib treatment exhibited a more potent anti-proliferative effect on OSCC cells, compared to either monotherapy. The combined therapy effectively suppressed the phosphorylation levels of Akt but not p44/42. CONCLUSION VEGFR-mediated signaling would be an alternative signaling pathway for the survival of OSCC cells under the disruption of EGFR signaling. These results highlight the clinical application of VEGFR inhibitors in the development of multi-molecular-targeted therapeutics against OSCC.
Collapse
Affiliation(s)
- Maho Onda
- Department of Oral and Maxillofacial SurgeryAichi Medical University HospitalNagakuteJapan
| | - Akinobu Ota
- Department of BiochemistryAichi Medical University School of MedicineNagakuteJapan
- Department of Food and Nutritional EnvironmentCollege of Human Life and EnvironmentKinjo Gakuin UniversityNagoyaJapan
| | - Kunihiro Ito
- Department of Oral and Maxillofacial SurgeryAichi Medical University HospitalNagakuteJapan
| | - Takayuki Ono
- Department of Oral and Maxillofacial SurgeryAichi Medical University HospitalNagakuteJapan
| | - Sivasundaram Karnan
- Department of BiochemistryAichi Medical University School of MedicineNagakuteJapan
| | - Mikako Kato
- Department of Oral and Maxillofacial SurgeryAichi Medical University HospitalNagakuteJapan
| | - Sayuri Kondo
- Department of Oral and Maxillofacial SurgeryAichi Medical University HospitalNagakuteJapan
| | - Akifumi Furuhashi
- Department of Oral and Maxillofacial SurgeryAichi Medical University HospitalNagakuteJapan
| | - Tomio Hayashi
- Department of Oral and Maxillofacial SurgeryAichi Medical University HospitalNagakuteJapan
| | - Yoshitaka Hosokawa
- Department of BiochemistryAichi Medical University School of MedicineNagakuteJapan
| | - Yoshiaki Kazaoka
- Department of Oral and Maxillofacial SurgeryAichi Medical University HospitalNagakuteJapan
| |
Collapse
|
9
|
Silva JPN, Pinto B, Monteiro L, Silva PMA, Bousbaa H. Combination Therapy as a Promising Way to Fight Oral Cancer. Pharmaceutics 2023; 15:1653. [PMID: 37376101 PMCID: PMC10301495 DOI: 10.3390/pharmaceutics15061653] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Oral cancer is a highly aggressive tumor with invasive properties that can lead to metastasis and high mortality rates. Conventional treatment strategies, such as surgery, chemotherapy, and radiation therapy, alone or in combination, are associated with significant side effects. Currently, combination therapy has become the standard practice for the treatment of locally advanced oral cancer, emerging as an effective approach in improving outcomes. In this review, we present an in-depth analysis of the current advancements in combination therapies for oral cancer. The review explores the current therapeutic options and highlights the limitations of monotherapy approaches. It then focuses on combinatorial approaches that target microtubules, as well as various signaling pathway components implicated in oral cancer progression, namely, DNA repair players, the epidermal growth factor receptor, cyclin-dependent kinases, epigenetic readers, and immune checkpoint proteins. The review discusses the rationale behind combining different agents and examines the preclinical and clinical evidence supporting the effectiveness of these combinations, emphasizing their ability to enhance treatment response and overcome drug resistance. Challenges and limitations associated with combination therapy are discussed, including potential toxicity and the need for personalized treatment approaches. A future perspective is also provided to highlight the existing challenges and possible resolutions toward the clinical translation of current oral cancer therapies.
Collapse
Affiliation(s)
- João P. N. Silva
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal; (J.P.N.S.); (B.P.); (L.M.)
| | - Bárbara Pinto
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal; (J.P.N.S.); (B.P.); (L.M.)
| | - Luís Monteiro
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal; (J.P.N.S.); (B.P.); (L.M.)
| | - Patrícia M. A. Silva
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal; (J.P.N.S.); (B.P.); (L.M.)
- TOXRUN—Toxicology Research Unit, University Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal
| | - Hassan Bousbaa
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS), Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra, Portugal; (J.P.N.S.); (B.P.); (L.M.)
| |
Collapse
|
10
|
Nogueira RLR, de Araújo TBS, Valverde LF, Silva VAO, Cavalcante BRR, Rossi EA, Allahdadi KJ, dos Reis MG, Pereira TA, Coletta RD, Bezerra DP, de Freitas Souza BS, Dias RB, Rocha CAG. Arsenic Trioxide Triggers Apoptosis of Metastatic Oral Squamous Cells Carcinoma with Concomitant Downregulation of GLI1 in Hedgehog Signaling. Biomedicines 2022; 10:biomedicines10123293. [PMID: 36552049 PMCID: PMC9775978 DOI: 10.3390/biomedicines10123293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/03/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Given the lack of advances in Oral Squamous Cell Carcinoma (OSCC) therapy in recent years, pharmacological strategies to block OSCC-related signaling pathways have gained prominence. The present study aimed to evaluate the therapeutic potential of Arsenic Trioxide (ATO) concerning its antitumoral effects and the inhibition of the Hedgehog (HH) pathway in OSCC. Initially, ATO cytotoxicity was assessed in a panel of cell lines. Cell viability, cell cycle, death patterns, and cell morphology were analyzed, as well as the effect of ATO on the expression of HH pathway components. After the cytotoxic assay, HSC3 cells were chosen for all in vitro assays. ATO increased apoptotic cell death and nuclear fragmentation in the sub-G1 cell cycle phase and promoted changes in cell morphology. In addition, the reduced expression of GLI1 indicated that ATO inhibits HH activity. The present study provides evidence of ATO as an effective cytotoxic drug for oral cancer treatment in vitro.
Collapse
Affiliation(s)
- Raphael Luís Rocha Nogueira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil
| | - Taís Bacelar Sacramento de Araújo
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador 40100-150, Bahia, Brazil
| | - Ludmila Faro Valverde
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador 40100-150, Bahia, Brazil
| | - Viviane Aline Oliveira Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil
| | - Bruno Raphael Ribeiro Cavalcante
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil
| | - Erik Aranha Rossi
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil
- Center for Biotechnology and Cell Therapy, D’Or Institute for Research and Education (IDOR), São Rafael Hospital, Salvador 41253-190, Bahia, Brazil
| | - Kyan James Allahdadi
- Center for Biotechnology and Cell Therapy, D’Or Institute for Research and Education (IDOR), São Rafael Hospital, Salvador 41253-190, Bahia, Brazil
| | - Mitermayer Galvão dos Reis
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil
| | - Thiago Almeida Pereira
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Ricardo D. Coletta
- Department of Oral Diagnosis, School of Dentistry University of Campinas, Piracicaba 13414-903, São Paulo, Brazil
- Graduate Program in Oral Biology, School of Dentistry University of Campinas, Piracicaba 13414-903, São Paulo, Brazil
| | - Daniel Pereira Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
| | - Bruno Solano de Freitas Souza
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil
- Center for Biotechnology and Cell Therapy, D’Or Institute for Research and Education (IDOR), São Rafael Hospital, Salvador 41253-190, Bahia, Brazil
| | - Rosane Borges Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador 40100-150, Bahia, Brazil
| | - Clarissa A. Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil
- Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador 40100-150, Bahia, Brazil
- Center for Biotechnology and Cell Therapy, D’Or Institute for Research and Education (IDOR), São Rafael Hospital, Salvador 41253-190, Bahia, Brazil
- Correspondence: ; Tel.: +55-71-3176-2209
| |
Collapse
|
11
|
Hsieh CY, Lin CC, Huang YW, Chen JH, Tsou YA, Chang LC, Fan CC, Lin CY, Chang WC. Macrophage secretory IL-1β promotes docetaxel resistance in head and neck squamous carcinoma via SOD2/CAT-ICAM1 signaling. JCI Insight 2022; 7:157285. [PMID: 36264639 PMCID: PMC9746909 DOI: 10.1172/jci.insight.157285] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 10/18/2022] [Indexed: 01/12/2023] Open
Abstract
Docetaxel (DTX) combined with cisplatin and 5-fluorouracil has been used as induction chemotherapy for head and neck squamous cell carcinoma (HNSCC). However, the development of acquired resistance remains a major obstacle to treatment response. Tumor-associated macrophages are associated with chemotherapeutic resistance. In the present study, increased infiltration of macrophages into the tumor microenvironment (TME) was significantly associated with shorter overall survival and increased resistance to chemotherapeutic drugs, particularly DTX, in patients with HNSCC. Macrophage coculture induced expression of intercellular adhesion molecule 1 (ICAM1), which promotes stemness and the formation of polyploid giant cancer cells, thereby reducing the efficacy of DTX. Both genetic silencing and pharmacological inhibition of ICAM1 sensitized HNSCC to DTX. Macrophage secretion of IL-1β was found to induce tumor expression of ICAM1. IL-1β neutralization and IL-1 receptor blockade reversed DTX resistance induced by macrophage coculture. IL-1β activated superoxide dismutase 2 and inhibited catalase, thereby modulating intracellular levels of ROS and inducing ICAM1 expression. Arsenic trioxide (ATO) reduced macrophage infiltration into the TME and impaired IL-1β secretion by macrophages. The combinatorial use of ATO enhanced the in vivo efficacy of DTX in a mouse model, which may provide a revolutionary approach to overcoming acquired therapeutic resistance in HNSCC.
Collapse
Affiliation(s)
- Ching-Yun Hsieh
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Ching-Chan Lin
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Yu-Wen Huang
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Jong-Hang Chen
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Yung-An Tsou
- Department of Otolaryngology-Head and Neck Surgery and
| | - Ling-Chu Chang
- Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan.,Chinese Medicinal Research and Development Center, China Medical University Hospital, and,Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chi-Chen Fan
- Department of Research and Development, Marker Exploration Corporation, Taipei, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu, Taiwan
| | - Chen-Yuan Lin
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Wei-Chao Chang
- Center for Molecular Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| |
Collapse
|
12
|
Kumar S, Tchounwou PB. p53 as a unique target of action of cisplatin in acute leukaemia cells. J Cell Mol Med 2022; 26:4727-4739. [PMID: 35946055 PMCID: PMC9443951 DOI: 10.1111/jcmm.17502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/18/2022] [Accepted: 06/26/2022] [Indexed: 12/03/2022] Open
Abstract
Acute promyelocytic leukaemia (APL) occurs in approximately 10% of acute myeloid leukaemia patients. Arsenic trioxide (ATO) has been for APL chemotherapy, but recently several ATO-resistant cases have been reported worldwide. Cisplatin (CDDP) enhances the toxicity of ATO in ovarian, lung cancer, chronic myelogenous leukaemia, and HL-60 cells. Hence, the goal of this study was to investigate a novel target of CDDP action in APL cells, as an alternate option for the treatment of ATO-resistant APL patients. We applied biochemical, molecular, confocal microscopy and advanced gene editing (CRISPR-Cas9) techniques to elucidate the novel target of CDDP action and its functional mechanism in APL cells. Our main findings revealed that CDDP activated p53 in APL cells through stress signals catalysed by ATM and ATR protein kinases, CHK1 and CHK2 phosphorylation at Ser 345 and Thr68 residues, and downregulation and dissociation of MDM2-DAXX-HAUSP complex. Our functional studies confirmed that CDDP-induced repression of MDM2-DAXX-HAUSP complex was significantly reversed in both nutilin-3-treated KG1a and p53-knockdown NB4 cells. Our findings also showed that CDDP stimulated an increased number of promyelocytes with dense granules, activated p53 expression, and downregulated MDM2 in liver and bone marrow of APL mice. Principal conclusion of our study highlights a novel mode of action of CDDP targeting p53 expression which may provide a basis for designing new anti-leukaemic compounds for treatment of APL patients.
Collapse
Affiliation(s)
- Sanjay Kumar
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD‐RCMI Center for Health Disparities ResearchJackson State UniversityJacksonMississippiUSA
- Department of life Sciences, School of Earth, Biological, and Environmental SciencesCentral University South BiharGayaIndia
| | - Paul B. Tchounwou
- Cellomics and Toxicogenomics Research Laboratory, NIH/NIMHD‐RCMI Center for Health Disparities ResearchJackson State UniversityJacksonMississippiUSA
| |
Collapse
|
13
|
Chen Q, Qiu Y, Chen L, Lin J, Yan LJ, Bao XD, Lin LS, Pan LZ, Shi B, Zheng XY, Chen F, He BC, Wang J, Liu FQ. Association between serum arsenic and oral cancer risk: A case-control study in southeast China. Community Dent Oral Epidemiol 2022; 50:83-90. [PMID: 33748987 DOI: 10.1111/cdoe.12633] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 01/14/2023]
Abstract
OBJECTIVES Evidence on serum arsenic and oral cancer risk was limited. We aimed to evaluate the association between serum arsenic and the risk of oral cancer in a southeast China population. METHODS Serum arsenic was determined for 325 oral cancer patients and 648 controls using inductively coupled plasma-mass spectrometry (ICP-MS). Logistic regression and restricted cubic spline were analysed the association between serum arsenic level and oral cancer risk, and crude and adjusted odds ratios (aOR) with 95% confidence interval (95% CI) were calculated. Factors adjusted for included age, gender, BMI, smoking, drinking, education, residence, marital status and dietary factors. Stratification analysis was further performed according to drinking, smoking and dietary characteristics. RESULTS Serum arsenic level was lower in the case group (P50 = 19.2μg/L, IQR = 11.6 ~ 26.4μg/L) than in the control group (P50 = 30.2 μg/L, IQR = 25.0 ~ 36.4 μg/L). An inverse but nonlinear association was observed between arsenic level and oral cancer risk by restricted cubic spline. These with moderate serum arsenic levels had a lower risk of oral cancer than those with low levels (OR = 0.11; 95%CI: 0.07-0.18), after adjusting for demographic and dietary intake factors. We also kept serum arsenic as a continuous variable in a regression model, where a similar inverse association between arsenic and oral cancer was observed, with OR = 0.86 (95%CI: 0.84-0.88). Stratification analysis revealed no significant multiplicative interactions between serum arsenic and smoking, drinking or dietary intake. CONCLUSION Serum arsenic is inversely related to oral cancer risk. Relative to those with low levels of arsenic, people with moderate serum arsenic levels had a lower risk of oral cancer. If confirmed, serum arsenic level may be a useful predictive marker for oral cancer risk.
Collapse
Affiliation(s)
- Qing Chen
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Yu Qiu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Lin Chen
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jing Lin
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Ling-Jun Yan
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Xiao-Dan Bao
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Li-Song Lin
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Li-Zhen Pan
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Bin Shi
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiao-Yan Zheng
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Fa Chen
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Bao-Chang He
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| | - Jing Wang
- Laboratory Center, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Feng-Qiong Liu
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, China
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
| |
Collapse
|
14
|
Agrawal Y, Nadkarni K, Gupta NA, Manne RK, Santra MK. F-box protein FBXO41 plays vital role in arsenic trioxide-mediated autophagic death of cancer cells. Toxicol Appl Pharmacol 2022; 441:115973. [PMID: 35278439 DOI: 10.1016/j.taap.2022.115973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 11/29/2022]
Abstract
Arsenic trioxide (ATO), a potent anti-neoplastic drug, is known to prevent cancer cell growth through induction of autophagic cell death. However, importance of cellular factors in ATO-mediated autophagic cell death is poorly understood. In this study, using biochemical and immunofluorescence techniques, we show that F-box protein FBXO41 plays a critical role in anti-proliferative activity of ATO. Our study reveals the importance of FBXO41 in induction of autophagic death of cancer cells by ATO. Further, we show that the autophagic cell death induced by FBXO41 is distinct and independent of apoptosis and necrosis, showing that FBXO41 may play vital role in inducing autophagic death of apoptosis resistant cancer cells. Overall, our study elucidates the importance of FBXO41 in ATO induced autophagic cell death to prevent cancer progression, which could be explored to develop promising cancer therapeutic strategy.
Collapse
Affiliation(s)
- Yashika Agrawal
- Molecular Oncology Laboratory, National Centre for Cell Science, Ganeshkhind Road, Pune, Maharashtra 411007, India; Department of Biotechnology, S.P. Pune University, Ganeshkhind Road, Pune, Maharashtra 411007, India
| | - Kaustubh Nadkarni
- Molecular Oncology Laboratory, National Centre for Cell Science, Ganeshkhind Road, Pune, Maharashtra 411007, India; Department of Biotechnology, S.P. Pune University, Ganeshkhind Road, Pune, Maharashtra 411007, India
| | - Neha A Gupta
- Molecular Oncology Laboratory, National Centre for Cell Science, Ganeshkhind Road, Pune, Maharashtra 411007, India
| | - Rajesh Kumar Manne
- Molecular Oncology Laboratory, National Centre for Cell Science, Ganeshkhind Road, Pune, Maharashtra 411007, India
| | - Manas Kumar Santra
- Molecular Oncology Laboratory, National Centre for Cell Science, Ganeshkhind Road, Pune, Maharashtra 411007, India.
| |
Collapse
|
15
|
Zhao Y, Yao H, Yang K, Han S, Chen S, Li Y, Chen S, Huang K, Lian G, Li J. Arsenic Trioxide-loaded nanoparticles Enhance the Chemosensitivity of Gemcitabine in Pancreatic Cancer via Reversal of Pancreatic Stellate Cells Desmoplasia through Targeting AP4/Galectin-1 Pathway. Biomater Sci 2022; 10:5989-6002. [DOI: 10.1039/d2bm01039a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Pancreatic stellate cell (PSCs) constitutes the fibrotic tumor microenvironment composed of the stroma matrix, which blocks the penetration of Gemcitabine (GEM) in pancreatic adenocarcinoma (PDAC) and results in chemoresistance. We...
Collapse
|
16
|
Cheng Y, Li S, Gao L, Zhi K, Ren W. The Molecular Basis and Therapeutic Aspects of Cisplatin Resistance in Oral Squamous Cell Carcinoma. Front Oncol 2021; 11:761379. [PMID: 34746001 PMCID: PMC8569522 DOI: 10.3389/fonc.2021.761379] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/04/2021] [Indexed: 02/06/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a kind of malignant tumors with low survival rate and prone to have early metastasis and recurrence. Cisplatin is an alkylating agent which induces DNA damage through the formation of cisplatin-DNA adducts, leading to cell cycle arrest and apoptosis. In the management of advanced OSCC, cisplatin-based chemotherapy or chemoradiotherapy has been considered as the first-line treatment. Unfortunately, only a portion of OSCC patients can benefit from cisplatin treatment, both inherent resistance and acquired resistance greatly limit the efficacy of cisplatin and even cause treatment failure. Herein, this review outline the underlying mechanisms of cisplatin resistance in OSCC from the aspects of DNA damage and repair, epigenetic regulation, transport processes, programmed cell death and tumor microenvironment. In addition, this review summarizes the strategies applicable to overcome cisplatin resistance, which can provide new ideas to improve the clinical therapeutic outcome of OSCC.
Collapse
Affiliation(s)
- Yali Cheng
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China
| | - Shaoming Li
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.,School of Stomatology of Qingdao University, Qingdao, China
| | - Ling Gao
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.,Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Keqian Zhi
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.,Key Lab of Oral Clinical Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenhao Ren
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
17
|
Wahiduzzaman M, Ota A, Hosokawa Y. Novel Mechanistic Insights into the Anti-cancer Mode of Arsenic Trioxide. Curr Cancer Drug Targets 2021; 20:115-129. [PMID: 31736446 DOI: 10.2174/1568009619666191021122006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/23/2019] [Accepted: 09/19/2019] [Indexed: 12/19/2022]
Abstract
Arsenic, a naturally-occurring toxic element, and a traditionally-used drug, has received a great deal of attention worldwide due to its curative anti-cancer properties in patients with acute promyelocytic leukemia. Among the arsenicals, arsenic trioxide has been most widely used as an anti-cancer drug. Recent advances in cancer therapeutics have led to a paradigm shift away from traditional cytotoxic drugs towards the targeting of proteins closely associated with driving the cancer phenotype. Due to the diverse anti-cancer effects of ATO on different types of malignancies, numerous studies have made efforts to uncover the mechanisms of ATO-induced tumor suppression. From in vitro cellular models to studies in clinical settings, ATO has been extensively studied. The outcomes of these studies have opened doors to establishing improved molecular-targeted therapies for cancer treatment. The efficacy of ATO has been augmented by combination with other drugs. In this review, we discuss recent arsenic-based cancer therapies and summarize the novel underlying molecular mechanisms of the anti-cancer effects of ATO.
Collapse
Affiliation(s)
- Md Wahiduzzaman
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| | - Akinobu Ota
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| | - Yoshitaka Hosokawa
- Department of Biochemistry, School of Medicine, Aichi Medical University, Nagakute, Aichi 480-1195, Japan
| |
Collapse
|
18
|
The effect of aflibercept and arsenic trioxide on the proliferation, migration and apoptosis of oral squamous cell carcinoma in vitro. Mol Biol Rep 2021; 48:3223-3235. [PMID: 33929648 DOI: 10.1007/s11033-021-06341-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/07/2021] [Indexed: 10/21/2022]
Abstract
Aflibercept and arsenic trioxide drugs apply a cytotoxic effect on some human cancer cell lines. However, no more study has followed the effects of both drugs, especially arsenic trioxide, on oral squamous cell carcinoma (OCC). We used three OCC lines as a model to show the effect of these drugs on the genetically complex disease and investigate its targeted therapy. In this study, three human OCC cell lines were used from different patients. We treated cell lines with both medications to detect the effect and relevant molecular basis. First, methyl thiazolyl tetrazolium (MTT) assay was performed to detect the cytotoxicity effect and cell growth. Second, flow cytometry, gene and protein expression were performed to evaluate the anti-angiogenic effect on OCC lines. Next apoptosis was analyzed by flow cytometry. Finally, clonogenesis capacity and cell migration were assessed by colony formation assay and wound healing, respectively. Aflibercept had no cytotoxic effect on the three OCC cell lines but decreased cell growth rate. Arsenic trioxide had a significant cytotoxic effect on three cell lines. Our results demonstrated that both drugs significantly decreased endoglin, VEGFA, and VEGFB expression. In addition, Migration and colony formation assays confirmed that these drugs have significant anti-proliferative and anti-migration effect on oral carcinoma cells. These results revealed that both medications might be a potential drug for the management of oral cancer patients.
Collapse
|
19
|
Huang GZ, Wu QQ, Zheng ZN, Shao TR, Li F, Lu XY, Ye HY, Chen GX, Song YX, Zeng WS, Ai YL, Lv XZ. Bioinformatics Analyses Indicate That Cathepsin G (CTSG) is a Potential Immune-Related Biomarker in Oral Squamous Cell Carcinoma (OSCC). Onco Targets Ther 2021; 14:1275-1289. [PMID: 33658795 PMCID: PMC7920606 DOI: 10.2147/ott.s293148] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose Plenty of studies showed that the immune system was associated with cancer initiation and progression. This study aimed to explore the prognostic biomarkers from immune-related genes (IRGs) in oral squamous cell carcinoma (OSCC). Materials and Methods RNA-seq data were downloaded from The Cancer Genome Atlas (TCGA) and IRGs and transcription factors (TFs) were extracted. Then, the co-expression network between IRGs and TFs was constructed using the "WGCNA" package in R software. Furthermore, a gene expression signature according to IRGs was constructed to predict OSCC prognosis and its accuracy was validated by survival analysis. Subsequently, correlation analyses between risk-score and immune cells level and clinical parameters were performed. Finally, immune-related biomarkers were selected and further investigated using gain-of-function assays in vitro. Results A total of 32 normal cases and 317 OSCC cases were selected in our study. Differentially-expressed analysis indicated that there were 381 differentially-expressed IRGs and 62 TFs in OSCC. Among them, 25 TFs and 21 IRGs were enrolled in the co-expression network. Furthermore, we found that gene expression signature on the basis of 10 IRGs could predict the prognosis accurately and a high-risk score based on gene expression signature meant a high T classification, terminal clinical stage, and low immune cells level in OSCC. Finally, cathepsin G (CTSG) was identified as a potential immune-related biomarker and therapeutic target in OSCC. Conclusion In conclusion, IRGs were directly involved in the development and progression of OSCC. Furthermore, CTSG was identified as a potential independent biomarker and might be an immunotherapeutic target in OSCC treatment.
Collapse
Affiliation(s)
- Guang-Zhao Huang
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Qing-Qing Wu
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Ze-Nan Zheng
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Ting-Ru Shao
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Fei Li
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Xin-Yan Lu
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Heng-Yu Ye
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Gao-Xiang Chen
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Yu-Xing Song
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Wei-Sen Zeng
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Yi-Long Ai
- Foshan Stomatological Hospital, School of Stomatology and Medicine, Foshan University, Foshan, Guangdong Province, People's Republic of China
| | - Xiao-Zhi Lv
- Department of Oral & Maxillofacial Surgery, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong Province, People's Republic of China
| |
Collapse
|
20
|
Parise A, Russo N, Marino T. The platination mechanism of RNase A by arsenoplatin: insight from the theoretical study. Inorg Chem Front 2021. [DOI: 10.1039/d0qi01165g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A detailed metalation process of the bovine pancreatic ribonuclease (RNase A) by a novel multitarget anti-cancer agent arsenoplatin-1, ([Pt(μ-NHC(CH3)O)2ClAs(OH)2]), performed at DFT level and using different models size is provided.
Collapse
Affiliation(s)
- A. Parise
- Dipartimento di Chimica e Tecnologie Chimiche
- Università della Calabria
- 87036 Arcavacata di Rende
- Italy
- Université Paris-Saclay
| | - N. Russo
- Dipartimento di Chimica e Tecnologie Chimiche
- Università della Calabria
- 87036 Arcavacata di Rende
- Italy
| | - T. Marino
- Dipartimento di Chimica e Tecnologie Chimiche
- Università della Calabria
- 87036 Arcavacata di Rende
- Italy
| |
Collapse
|
21
|
Hu WC, Teo WH, Huang TF, Lee TC, Lo JF. Combinatorial Low Dose Arsenic Trioxide and Cisplatin Exacerbates Autophagy via AMPK/STAT3 Signaling on Targeting Head and Neck Cancer Initiating Cells. Front Oncol 2020; 10:463. [PMID: 32351887 PMCID: PMC7174769 DOI: 10.3389/fonc.2020.00463] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/16/2020] [Indexed: 12/11/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a highly lethal disease with high-level of epidemic both in the world and Taiwan. Previous studies support that head and neck cancer-initiating cells (HN-CICs), a subpopulation of cancer cells with enhanced stemness properties, contribute to therapy resistance and tumor recurrence. Arsenic trioxide (As2O3; ATO) has shown to be an effective anti-cancer drug targeting acute promyelocytic leukemia (APL). Combinatorial treatment with high dose of ATO and cisplatin (CDDP) exert synergistic apoptotic effects in cancer cell lines of various solid tumors, however, it may cause of significant side effect to the patients. Nevertheless, none has reported the anti-cancerous effect of ATO/CDDP targeting HN-CICs. In this study, we aim to evaluate the low dose combination of ATO with conventional chemo-drugs CDDP treatment on targeting HN-CICs. We first analyzed the inhibitory tumorigenicity of co-treatment with ATO and chemo-drugs on HN-CICs which are enriched from HNSCC cells. We observed that ATO/CDDP therapeutic regimen successfully synergized the cell death on HN-CICs with a Combination Index (CI) <1 by Chou-Talalay's analysis in vitro. Interestingly, the ATO/CDDP regimen also induced exaggerated autophagy on HN-CICs. Additionally, this drug combination strategy also empowered both preventive and therapeutic effect by in vivo xenograft assays. Finally, we provide the underlying molecular mechanisms of ATO-based therapeutic regimen on HN-CICs. Together, low dose of combinatorial ATO/CDDP regimen induced cell death as well as exacerbated autophagy via AMPK-STAT3 mediated pathway in HN-CICs.
Collapse
Affiliation(s)
- Wei-Chun Hu
- Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan
| | - Wan-Huai Teo
- Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan
| | - Tung-Fu Huang
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Orthopedics and Traumatology, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Te-Chang Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jeng-Fan Lo
- Institute of Oral Biology, National Yang-Ming University, Taipei, Taiwan.,Department of Dentistry, School of Dentistry, National Yang-Ming University, Taipei, Taiwan.,Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan.,Department of Dentistry, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
22
|
Kondo S, Ota A, Ono T, Karnan S, Wahiduzzaman M, Hyodo T, Lutfur Rahman M, Ito K, Furuhashi A, Hayashi T, Konishi H, Tsuzuki S, Hosokawa Y, Kazaoka Y. Discovery of novel molecular characteristics and cellular biological properties in ameloblastoma. Cancer Med 2020; 9:2904-2917. [PMID: 32096304 PMCID: PMC7163100 DOI: 10.1002/cam4.2931] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/09/2020] [Accepted: 02/04/2020] [Indexed: 12/15/2022] Open
Abstract
Ameloblastoma is a rare odontogenic benign tumor accounting for less than 1% of head and neck tumors. Advanced next generation sequencing (NGS) analyses identified high frequency of BRAF V600E and SMO L412F mutations in ameloblastoma. Despite the existence of whole genomic sequence information from patients with ameloblastoma, entire molecular signature of and the characteristics of ameloblastoma cells are still obscure. In this study, we sought to uncover the molecular basis of ameloblastoma and to determine the cellular phenotype of ameloblastoma cells with BRAF mutations. Our comparative cDNA microarray analysis and gene set enrichment analysis (GSEA) showed that ameloblastoma exhibited a distinct gene expression pattern from the normal tissues: KRAS-responsive gene set is significantly activated in ameloblastoma. Importantly, insulin like growth factor 2 (IGF2), a member of KRAS-responsive genes, enhances the proliferation of an ameloblastoma cell line AMU-AM1 with BRAF mutation. In addition, Toll-like receptor 2 (TLR2) knockdown readily inactivated KRAS-responsive gene sets as well as increases caspase activities, suggesting that TLR2 signaling may mediate cell survival signaling in ameloblastoma cells. Collectively, the findings may help to further clarify the pathophysiology of ameloblastoma and lead to the development of precision medicine for patients with ameloblastoma.
Collapse
Affiliation(s)
- Sayuri Kondo
- Department of Oral and Maxillofacial Surgery, Aichi Medical University Hospital, Nagakute, Japan
| | - Akinobu Ota
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Takayuki Ono
- Department of Oral and Maxillofacial Surgery, Aichi Medical University Hospital, Nagakute, Japan
| | - Sivasundaram Karnan
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Md Wahiduzzaman
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Toshinori Hyodo
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Md Lutfur Rahman
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kunihiro Ito
- Department of Oral and Maxillofacial Surgery, Aichi Medical University Hospital, Nagakute, Japan
| | - Akifumi Furuhashi
- Department of Oral and Maxillofacial Surgery, Aichi Medical University Hospital, Nagakute, Japan
| | - Tomio Hayashi
- Department of Oral and Maxillofacial Surgery, Aichi Medical University Hospital, Nagakute, Japan
| | - Hiroyuki Konishi
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Shinobu Tsuzuki
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Yoshitaka Hosokawa
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Yoshiaki Kazaoka
- Department of Oral and Maxillofacial Surgery, Aichi Medical University Hospital, Nagakute, Japan
| |
Collapse
|
23
|
Inhibition of PI3K pathway using BKM120 intensified the chemo-sensitivity of breast cancer cells to arsenic trioxide (ATO). Int J Biochem Cell Biol 2019; 116:105615. [DOI: 10.1016/j.biocel.2019.105615] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 09/05/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022]
|
24
|
Miodragović Ð, Swindell EP, Waxali ZS, Bogachkov A, O'Halloran TV. Beyond Cisplatin: Combination Therapy with Arsenic Trioxide. Inorganica Chim Acta 2019; 496:119030. [PMID: 32863421 PMCID: PMC7453736 DOI: 10.1016/j.ica.2019.119030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Platinum drugs (cisplatin, oxaliplatin, and carboplatin) and arsenic trioxide are the only commercial inorganic non-radioactive anticancer drugs approved by the US Food and Drug Administration. Numerous efforts are underway to take advantage of the synergy between the anticancer activity of cisplatin and arsenic trioxide - two drugs with strikingly different mechanisms of action. These include co-encapsulation of the two drugs in novel nanoscale delivery systems as well as the development of small molecule agents that combine the activity of these two inorganic materials. Several of these new molecular entities containing Pt-As bonds have broad anticancer activity, are robust in physiological buffer solutions, and form stable complexes with biopolymers. This review summarizes results from a number of preclinical studies involving the combination of cisplatin and As2O3, co-encapsulation and nanoformulation efforts, and the chemistry and cytotoxicity of the first member of platinum anticancer agents with an arsenous acid moiety bound to the platinum(II) center: arsenoplatins.
Collapse
Affiliation(s)
- Ðenana Miodragović
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
- Northeastern Illinois University, 5500 North St Louis Avenue, Chicago, Illinois 60625, United States
| | - Elden P Swindell
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Zohra Sattar Waxali
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Abraham Bogachkov
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Thomas V O'Halloran
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| |
Collapse
|
25
|
Miodragović Đ, Merlino A, Swindell EP, Bogachkov A, Ahn RW, Abuhadba S, Ferraro G, Marzo T, Mazar AP, Messori L, O’Halloran TV. Arsenoplatin-1 Is a Dual Pharmacophore Anticancer Agent. J Am Chem Soc 2019; 141:6453-6457. [PMID: 30943017 PMCID: PMC6830503 DOI: 10.1021/jacs.8b13681] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Arsenoplatins are adducts of two chemically important anticancer drugs, cisplatin and arsenic trioxide, that have a Pt(II) bond to an As(III) hydroxide center. Screens of the NCI-60 human tumor cell lines reveal that arsenoplatin-1 (AP-1), [Pt(μ-NHC(CH3)O)2ClAs(OH)2], the first representative of this novel class of anticancer agents, displays a superior activity profile relative to the parent drugs As2O3 or cisplatin in a majority of cancer cell lines tested. These activity profiles are important because the success of arsenic trioxide in blood cancers (such as APL) has not been seen in solid tumors due to the rapid clearance of arsenous acid from the body. To understand the biological chemistry of these compounds, we evaluated interactions of AP-1 with the two important classes of biomolecules-proteins and DNA. The first structural studies of AP-1 bound to model proteins reveal that platinum(II) binds the Nε of His in a manner that preserves the Pt-As bond. We find that AP-1 readily enters cells and binds to DNA with an intact Pt-As bond (Pt:As ratio of 1). At longer incubation times, however, the Pt:As ratio in DNA samples increases, suggesting that the Pt-As bond breaks and releases the As(OH)2 moiety. We conclude that arsenoplatin-1 has the potential to deliver both Pt and As species to a variety of hematological and solid cancers.
Collapse
Affiliation(s)
- Đenana Miodragović
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois
60208, United States
- Northeastern Illinois University, 5500 North St Louis Avenue, Chicago, Illinois 60625, United
States
| | - Antonello Merlino
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario di Monte
Sant’Angelo, Via Cintia, I-80126 Napoli, Italy
| | - Elden P. Swindell
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois
60208, United States
| | - Abraham Bogachkov
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois
60208, United States
| | - Richard W. Ahn
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois
60208, United States
| | - Sara Abuhadba
- Northeastern Illinois University, 5500 North St Louis Avenue, Chicago, Illinois 60625, United
States
| | - Giarita Ferraro
- Department of Chemistry “Ugo Schiff”, Università degli Studi Firenze, via della
Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| | - Tiziano Marzo
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano 6, 56126 Pisa, Italy
| | - Andrew P. Mazar
- Pharmacology, Feinberg School of Medicine, Northwestern University, 2145 Sheridan Road, Evanston,
Illinois 60208, United States
| | - Luigi Messori
- Department of Chemistry “Ugo Schiff”, Università degli Studi Firenze, via della
Lastruccia 3-13, 50019 Sesto Fiorentino, Italy
| | - Thomas V. O’Halloran
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Road, Evanston, Illinois
60208, United States
| |
Collapse
|
26
|
Chen Z, Zhang Z, Chen M, Xie S, Wang T, Li X. Synergistic antitumor efficacy of hybrid micelles with mitochondrial targeting and stimuli-responsive drug release behavior. J Mater Chem B 2019; 7:1415-1426. [PMID: 32255012 DOI: 10.1039/c8tb02843e] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The term synergism means that the overall therapeutic benefits should be greater than the sum of the effects of individual agents and that the optimal therapeutic efficacy can be achieved at reduced doses. Micellar systems usually fail to deliver multiple drugs to target sites at synergistic doses and thus are not able to maximize the antitumor efficacy. In the current study, we demonstrate a strategy to coordinate the release of camptothecin (CPT) and α-tocopheryl succinate (TOS) from hybrid micelles for nucleus and mitochondrion interferences. TOS is decorated with cationic triphenylphosphonium (TPP) to promote the targeting capability of TOS-TPP to mitochondria. The combination of CPT and TOS-TPP shows strong synergistism with a combination index of 0.186. Hyaluronic acid (HA) is conjugated with CPT or TOS-TPP via disulfide linkages for tumor cell targeting and intracellular reduction-triggered release. Both conjugates either separately self-assemble into MC and MT micelles, or are blended at different ratios to form MC-T hybrid micelles. In response to elevated intracellular glutathione levels, the coordinated release of CPT and TOS-TPP from MC-T results in a combination index of 0.26 and the dose-reduction indexes of CPT and TOS are 7.7 and 3.4, respectively. Compared with MC and MT, MC-T micelles with 5 fold lower doses exhibit higher intracellular reactive oxygen species (ROS) levels, comparable tumor growth inhibition and animal survival, indicating no hematologic and intestinal toxicities. Moreover, the HA conjugates of MC-T are linked to polylactide via acid-labile linkages and electrospun into short fibers (MC-T@SF) as an injectable depot to release MC-T in response to the acidic tumor microenvironment. At a predetermined synergistic ratio, MC-T@SF with 5 fold lower doses achieves antitumor profiles comparable to those of individual micelle-loaded short fibers. Therefore, the hybrid micelles and micelle-releasing short fibers represent a feasible strategy to synergistically enhance the therapeutic efficacy and enable significant reduction in effective doses of chemotherapeutic agents.
Collapse
Affiliation(s)
- Zhoujiang Chen
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China.
| | | | | | | | | | | |
Collapse
|
27
|
Su W, Tang J, Wang Y, Sun S, Shen Y, Yang H. Long non-coding RNA highly up-regulated in liver cancer promotes epithelial-to-mesenchymal transition process in oral squamous cell carcinoma. J Cell Mol Med 2019; 23:2645-2655. [PMID: 30677230 PMCID: PMC6433680 DOI: 10.1111/jcmm.14160] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 12/24/2018] [Accepted: 12/27/2018] [Indexed: 12/17/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is an oral and maxillofacial malignancy that exhibits high incidence worldwide. In diverse human cancers, the long non-coding RNA (lncRNA) highly up-regulated in liver cancer (HULC) is aberrantly expressed, but how HULC affects OSCC development and progression has remained mostly unknown. We report that HULC was abnormally up-regulated in oral cancer tissues and OSCC cell lines, and that suppression of HULC expression in OSCC cells not only inhibited the proliferation, drug tolerance, migration and invasion of the cancer cells, but also increased their apoptosis rate. Notably, in a mouse xenograft model, HULC depletion reduced tumorigenicity and inhibited the epithelial-to-mesenchymal transition process. Collectively, our findings reveal a crucial role of the lncRNA HULC in regulating oral cancer carcinogenesis and tumour progression, and thus suggest that HULC could serve as a novel therapeutic target for OSCC.
Collapse
Affiliation(s)
- Wen Su
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China.,Peking University Shenzhen Hospital Clinical College, Anhui Medical University, Hefei, Anhui, China
| | - Jing Tang
- Jingzhou Central Hospital, Jingzhou, Hubei
| | - Yufan Wang
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Shuai Sun
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yuehong Shen
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Hongyu Yang
- Department of Oral and Maxillofacial Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
28
|
Wahiduzzaman M, Ota A, Karnan S, Hanamura I, Mizuno S, Kanasugi J, Rahman ML, Hyodo T, Konishi H, Tsuzuki S, Takami A, Hosokawa Y. Novel combined Ato-C treatment synergistically suppresses proliferation of Bcr-Abl-positive leukemic cells in vitro and in vivo. Cancer Lett 2018; 433:117-130. [PMID: 29944906 DOI: 10.1016/j.canlet.2018.06.027] [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: 03/04/2018] [Revised: 05/31/2018] [Accepted: 06/18/2018] [Indexed: 12/28/2022]
Abstract
Chronic myelogenous leukemia (CML) accounts for 15-20% of all leukemias affecting adults. Despite recent advances in the development of specific Bcr-Abl tyrosine kinase inhibitors (TKIs), some CML patients suffer from relapse due to TKI resistance. Here, we assessed the efficacy of a novel combinatorial arsenic trioxide (ATO) and cisplatin (CDDP) treatment (Ato-C) in human Bcr-Abl-positive leukemic cells. Combination index analyses revealed that a synergistic interaction of ATO and CDDP elicits a wide range of effects in K562, KU-812, MEG-A2, and KCL-22 cells. Notably, Ato-C synergistically enhanced apoptosis and decreased the survival of both acquired TKI-resistant CML cells and the cells expressing mutant Bcr-AblT315I. In addition, Ato-C dramatically decreased the phosphorylation level of forkhead transcription factor FOXO1/3a and STAT5 as well as c-Myc protein level. Interestingly, results of gene set enrichment analysis showed that Ato-C significantly downregulates the expression of MYC- and/or E2F1-target genes. Furthermore, Ato-C significantly suppressed the proliferation of MEG-A2-derived tumor when compared with that following monotherapy in vivo. Collectively, these results suggest that combined Ato-C treatment could be a promising alternative to the current therapeutic regime in CML.
Collapse
Affiliation(s)
- Md Wahiduzzaman
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| | - Akinobu Ota
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan.
| | - Sivasundaram Karnan
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| | - Ichiro Hanamura
- Division of Hematology, Department of Internal Medicine, Aichi Medical University, Aichi, Japan
| | - Shohei Mizuno
- Division of Hematology, Department of Internal Medicine, Aichi Medical University, Aichi, Japan
| | - Jo Kanasugi
- Division of Hematology, Department of Internal Medicine, Aichi Medical University, Aichi, Japan
| | - Md Lutfur Rahman
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| | - Toshinori Hyodo
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| | - Hiroyuki Konishi
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| | - Shinobu Tsuzuki
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| | - Akiyoshi Takami
- Division of Hematology, Department of Internal Medicine, Aichi Medical University, Aichi, Japan
| | - Yoshitaka Hosokawa
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| |
Collapse
|
29
|
Xiong X, Li Y, Liu L, Qi K, Zhang C, Chen Y, Fang J. Arsenic trioxide induces cell cycle arrest and affects Trk receptor expression in human neuroblastoma SK-N-SH cells. Biol Res 2018; 51:18. [PMID: 29898774 PMCID: PMC5998579 DOI: 10.1186/s40659-018-0167-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 06/06/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Arsenic trioxide (As2O3), a drug that has been used in China for approximately two thousand years, induces cell death in a variety of cancer cell types, including neuroblastoma (NB). The tyrosine kinase receptor (Trk) family comprises three members, namely TrkA, TrkB and TrkC. Various studies have confirmed that TrkA and TrkC expression is associated with a good prognosis in NB, while TrkB overexpression can lead to tumor cell growth and invasive metastasis. Previous studies have shown that As2O3 can inhibit the growth and proliferation of a human NB cell line and can also affect the N-Myc mRNA expression. It remains unclear whether As2O3 regulates Trks for the purposes of treating NB. METHODS The aim of the present study was to investigate the effect of As2O3 on Trk expression in NB cell lines and its potential therapeutic efficacy. SK-N-SH cells were grown with increasing doses of As2O3 at different time points. We cultured SK-N-SH cells, which were treated with increasing doses of As2O3 at different time points. Trk expression in the NB samples was quantified by immunohistochemistry, and the cell cycle was analyzed by flow cytometry. TrkA, TrkB and TrkC mRNA expression was evaluated by real-time PCR analysis. RESULTS Immunohistochemical and real-time PCR analyses indicated that TrkA and TrkC were over-expressed in NB, and specifically during stages 1, 2 and 4S of the disease progression. TrkB expression was increased in stage 3 and 4 NB. As2O3 significantly arrested SK-N-SH cells in the G2/M phase. In addition, TrkA, TrkB and TrkC expression levels were significantly upregulated by higher concentrations of As2O3 treatment, notably in the 48-h treatment period. Our findings suggested that to achieve the maximum effect and appropriate regulation of Trk expression in NB stages 1, 2 and 4S, As2O3 treatment should be at relatively higher concentrations for longer delivery times;however, for NB stages 3 and 4, an appropriate concentration and infusion time for As2O3 must be carefully determined. CONCLUSION The present findings suggested that As2O3 induced Trk expression in SK-N-SH cells to varying degrees and may be a promising adjuvant to current treatments for NB due to its apoptotic effects.
Collapse
Affiliation(s)
- Xilin Xiong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
- Pediatric Hematology/Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
| | - Yang Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
- Pediatric Hematology/Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
| | - Ling Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
- Department of Pediatric Hematology/Oncology, Affiliated Hospital of Guangdong Medical College, Zhanjiang, 524000 Guangdong China
| | - Kai Qi
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
- Pediatric Hematology/Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
| | - Chi Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
- Pediatric Hematology/Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
| | - Yueqin Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
- Department of Life Sciences, Sun Yat-Sen University, Guangzhou, 510120 Guangdong China
| | - Jianpei Fang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
- Pediatric Hematology/Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120 China
| |
Collapse
|
30
|
Yamaji M, Ota A, Wahiduzzaman M, Karnan S, Hyodo T, Konishi H, Tsuzuki S, Hosokawa Y, Haniuda M. Novel ATP-competitive Akt inhibitor afuresertib suppresses the proliferation of malignant pleural mesothelioma cells. Cancer Med 2017; 6:2646-2659. [PMID: 28960945 PMCID: PMC5673922 DOI: 10.1002/cam4.1179] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 07/26/2017] [Indexed: 12/12/2022] Open
Abstract
Malignant pleural mesothelioma (MPM), an asbestos-related occupational disease, is an aggressive and incurable tumor of the thoracic cavity. Despite recent advances in MPM treatment, overall survival of patients with MPM is very low. Recent studies have implicated that PI3K/Akt signaling is involved in MPM cell survival and development. To investigate the effects of Akt inhibitors on MPM cell survival, we examined the effects of nine selective Akt inhibitors, namely, afuresertib, Akti-1/2, AZD5363, GSK690693, ipatasertib, MK-2206, perifosine, PHT-427, and TIC10, on six MPM cell lines, namely, ACC-MESO-4, Y-MESO-8A, MSTO-211H, NCI-H28, NCI-H290, and NCI-H2052, and a normal mesothelial cell line MeT-5A. Comparison of IC50 values of the Akt inhibitors showed that afuresertib, an ATP-competitive specific Akt inhibitor, exerted tumor-specific effects on MPM cells. Afuresertib significantly increased caspase-3 and caspase-7 activities and apoptotic cell number among ACC-MESO-4 and MSTO-211H cells. Moreover, afuresertib strongly arrested the cell cycle in the G1 phase. Western blotting analysis showed that afuresertib increased the expression of p21WAF1/CIP1 and decreased the phosphorylation of Akt substrates, including GSK-3β and FOXO family proteins. These results suggest that afuresertib-induced p21 expression promotes G1 phase arrest by inducing FOXO activity. Furthermore, afuresertib significantly enhanced cisplatin-induced cytotoxicity. Interestingly, results of gene set enrichment analysis showed that afuresertib modulated the expression E2F1 and MYC, which are associated with fibroblast core serum response. Together, these results suggest that afuresertib is a useful anticancer drug for treating patients with MPM.
Collapse
Affiliation(s)
- Masayuki Yamaji
- Division of Chest Surgery, Department of Surgery, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan.,Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| | - Akinobu Ota
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| | - Md Wahiduzzaman
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| | - Sivasundaram Karnan
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| | - Toshinori Hyodo
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| | - Hiroyuki Konishi
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| | - Shinobu Tsuzuki
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| | - Yoshitaka Hosokawa
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| | - Masayuki Haniuda
- Division of Chest Surgery, Department of Surgery, Aichi Medical University School of Medicine, Nagakute, Aichi, 480-1195, Japan
| |
Collapse
|
31
|
Detection of Glutathione in Oral Squamous Cell Carcinoma Cells With a Fluorescent Probe During the Course of Oxidative Stress and Apoptosis. J Oral Maxillofac Surg 2017; 75:223.e1-223.e10. [DOI: 10.1016/j.joms.2016.08.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 08/01/2016] [Accepted: 08/08/2016] [Indexed: 11/20/2022]
|
32
|
Mohapatra S, Saha A, Mondal P, Jana B, Ghosh S, Biswas A, Ghosh S. Synergistic Anticancer Effect of Peptide-Docetaxel Nanoassembly Targeted to Tubulin: Toward Development of Dual Warhead Containing Nanomedicine. Adv Healthc Mater 2017; 6. [PMID: 27782376 DOI: 10.1002/adhm.201600718] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/12/2016] [Indexed: 01/09/2023]
Abstract
Microtubule dynamics play a crucial role in cancer cell division. Various drugs are developed to target microtubule. Although a few of them show potential in treatment of cancer, but success rate is limited due to their poor bioavailability and lack of specificity. Thus, development of highly bioavailable and target specific anticancer drug is extremely necessary. To address these key issues, here, a combination of approaches such as development of a dodecapeptide-docetaxel nanoassembly targeted to tubulin and MUC1 (mucin 1, cell surface associated glycoprotein) targeting oligonucleotide aptamer conjugated liposome for delivering peptide-docetaxel nanoassembly into the breast cancer cell have been demonstrated. These studies reveal that the peptide forms nanoassembly and entraps docetaxel drug. Further, the liposomal formulation of peptide-docetaxel exerts synergistic anticancer effect, activates key mitotic check point proteins, and inhibits bipolar spindle formation, metastatic cancer cell migration, and growth of tumor mimicking 3D multicellular spheroid.
Collapse
Affiliation(s)
- Saswat Mohapatra
- Organic and Medicinal Chemistry Division; Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology (IICB); 4, Raja S. C. Mullick Road, Jadavpur Kolkata 700032 West Bengal India
- Academy of Scientific and Innovative Research (AcSIR); CSIR-Indian Institute of Chemical Biology Campus; 4, Raja S. C. Mullick Road Kolkata 700 032 West Bengal India
| | - Abhijit Saha
- Organic and Medicinal Chemistry Division; Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology (IICB); 4, Raja S. C. Mullick Road, Jadavpur Kolkata 700032 West Bengal India
| | - Prasenjit Mondal
- Organic and Medicinal Chemistry Division; Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology (IICB); 4, Raja S. C. Mullick Road, Jadavpur Kolkata 700032 West Bengal India
- Academy of Scientific and Innovative Research (AcSIR); CSIR-Indian Institute of Chemical Biology Campus; 4, Raja S. C. Mullick Road Kolkata 700 032 West Bengal India
| | - Batakrishna Jana
- Organic and Medicinal Chemistry Division; Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology (IICB); 4, Raja S. C. Mullick Road, Jadavpur Kolkata 700032 West Bengal India
| | - Subhajit Ghosh
- Organic and Medicinal Chemistry Division; Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology (IICB); 4, Raja S. C. Mullick Road, Jadavpur Kolkata 700032 West Bengal India
| | - Atanu Biswas
- Organic and Medicinal Chemistry Division; Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology (IICB); 4, Raja S. C. Mullick Road, Jadavpur Kolkata 700032 West Bengal India
| | - Surajit Ghosh
- Organic and Medicinal Chemistry Division; Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology (IICB); 4, Raja S. C. Mullick Road, Jadavpur Kolkata 700032 West Bengal India
- Academy of Scientific and Innovative Research (AcSIR); CSIR-Indian Institute of Chemical Biology Campus; 4, Raja S. C. Mullick Road Kolkata 700 032 West Bengal India
| |
Collapse
|
33
|
Ito K, Ota A, Ono T, Nakaoka T, Wahiduzzaman M, Karnan S, Konishi H, Furuhashi A, Hayashi T, Yamada Y, Hosokawa Y, Kazaoka Y. Inhibition of Nox1 induces apoptosis by attenuating the AKT signaling pathway in oral squamous cell carcinoma cell lines. Oncol Rep 2016; 36:2991-2998. [PMID: 27600098 DOI: 10.3892/or.2016.5068] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/08/2016] [Indexed: 11/06/2022] Open
Abstract
NADPH oxidases, also known as the Nox family, are major sources of reactive oxygen species generation that regulate redox-sensitive signaling pathways. Recent studies have implicated the Nox family in cancer development and progression. However, the involvement of its members in the development of oral squamous cell carcinoma (OSCC) remains to be elucidated. To clarify this issue, we first analyzed mRNA expression of Nox/Duox family members (Nox1, Nox2, Nox3, Nox4, Nox5, Duox1 and Duox2) in five OSCC cell lines. Nox1 and Nox4 mRNAs were highly expressed in four OSCC cell lines. Western blot analysis revealed that the protein expression level of Nox1 was higher than that of Nox4 in the OSCC cell lines. In addition, knockdown of Nox1, but not Nox4, significantly suppressed cell viability and induced apoptosis in the HSC-2 and HSC-3 cells. We also found that a specific AKT inhibitor, perifosine, dose-dependently suppressed OSCC cell growth. Notably, Nox1 knockdown significantly attenuated the phosphorylation level of AKT. Furthermore, both Nox1 knockdown and perifosine treatment markedly enhanced the cisplatin-induced cytotoxic effect. Taken together, our results highlight that the Nox1/AKT signaling pathway plays an important role in cell survival in OSCC cells.
Collapse
Affiliation(s)
- Kunihiro Ito
- Department of Oral and Maxillofacial Surgery, Aichi Medical University Hospital, Aichi 480-1195, Japan
| | - Akinobu Ota
- Department of Biochemistry, Aichi Medical University School of Medicine, Aichi 480-1195, Japan
| | - Takayuki Ono
- Department of Oral and Maxillofacial Surgery, Aichi Medical University Hospital, Aichi 480-1195, Japan
| | - Toshiki Nakaoka
- Department of Oral and Maxillofacial Surgery, Aichi Medical University Hospital, Aichi 480-1195, Japan
| | - Md Wahiduzzaman
- Department of Biochemistry, Aichi Medical University School of Medicine, Aichi 480-1195, Japan
| | - Sivasundaram Karnan
- Department of Biochemistry, Aichi Medical University School of Medicine, Aichi 480-1195, Japan
| | - Hiroyuki Konishi
- Department of Biochemistry, Aichi Medical University School of Medicine, Aichi 480-1195, Japan
| | - Akifumi Furuhashi
- Department of Oral and Maxillofacial Surgery, Aichi Medical University Hospital, Aichi 480-1195, Japan
| | - Tomio Hayashi
- Department of Oral and Maxillofacial Surgery, Aichi Medical University Hospital, Aichi 480-1195, Japan
| | - Yoichi Yamada
- Department of Oral and Maxillofacial Surgery, Aichi Medical University Hospital, Aichi 480-1195, Japan
| | - Yoshitaka Hosokawa
- Department of Biochemistry, Aichi Medical University School of Medicine, Aichi 480-1195, Japan
| | - Yoshiaki Kazaoka
- Department of Oral and Maxillofacial Surgery, Aichi Medical University Hospital, Aichi 480-1195, Japan
| |
Collapse
|
34
|
Ribeiro IP, Caramelo F, Marques F, Domingues A, Mesquita M, Barroso L, Prazeres H, Julião MJ, Baptista IP, Ferreira A, Melo JB, Carreira IM. WT1, MSH6, GATA5 and PAX5 as epigenetic oral squamous cell carcinoma biomarkers - a short report. Cell Oncol (Dordr) 2016; 39:573-582. [PMID: 27491556 DOI: 10.1007/s13402-016-0293-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2016] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Oral squamous cell carcinoma (OSCC) is a frequently occurring aggressive malignancy with a heterogeneous clinical behavior. Based on the paucity of specific early diagnostic and prognostic biomarkers, which hampers the appropriate treatment and, ultimately the development of novel targeted therapies, we aimed at identifying such biomarkers through a genetic and epigenetic analysis of these tumors. METHODS 93 primary OSCCs were subjected to DNA copy number alteration (CNA) and methylation status analyses using methylation-specific multiplex ligation-dependent probe amplification (MS-MPLA). The genetic and epigenetic OSCC profiles obtained were associated with the patients' clinic-pathological features. RESULTS We found that WT1 gene promoter methylation is a predictor of a better prognosis and that MSH6 and GATA5 gene promoter methylation serve as predictors of a worse prognosis. GATA5 gene promoter methylation was found to be significantly associated with a shorter survival rate. In addition, we found that PAX5 gene promoter methylation was significantly associated with tongue tumors. To the best of our knowledge, this is the first study that highlights this specific set of genes as epigenetic diagnostic and prognostic biomarkers in OSCC. CONCLUSIONS Our data highlight the importance of epigenetically assessing OSCCs to identify key genes that may serve as diagnostic and prognostic biomarkers and, potentially, as candidate therapeutic targets.
Collapse
Affiliation(s)
- Ilda Patrícia Ribeiro
- Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra, Polo Ciências da Saúde, 3000-354, Coimbra, Portugal.,CIMAGO - Center of Investigation on Environment Genetics and Oncobiology - Faculty of Medicine, University of Coimbra, 3000-354, Coimbra, Portugal
| | - Francisco Caramelo
- Laboratory of Biostatistics and Medical Informatics, IBILI - Faculty of Medicine, University of Coimbra, 3000-354, Coimbra, Portugal
| | - Francisco Marques
- CIMAGO - Center of Investigation on Environment Genetics and Oncobiology - Faculty of Medicine, University of Coimbra, 3000-354, Coimbra, Portugal.,Department of Dentistry, Faculty of Medicine, University of Coimbra, 3000-075, Coimbra, Portugal.,Stomatology Unit, Coimbra Hospital and University Centre (CHUC), EPE, 3000-075, Coimbra, Portugal
| | - Ana Domingues
- Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra, Polo Ciências da Saúde, 3000-354, Coimbra, Portugal
| | - Margarida Mesquita
- Maxillofacial Surgery Department, Coimbra Hospital and University Centre (CHUC), EPE, 3000-075, Coimbra, Portugal
| | - Leonor Barroso
- Maxillofacial Surgery Department, Coimbra Hospital and University Centre (CHUC), EPE, 3000-075, Coimbra, Portugal
| | - Hugo Prazeres
- Molecular Pathology Laboratory, Portuguese Institute of Oncology of Coimbra FG, EPE, 3000-075, Coimbra, Portugal
| | - Maria José Julião
- Department of Pathology, Coimbra Hospital and University Centre (CHUC), EPE, 3000-075, Coimbra, Portugal
| | - Isabel Poiares Baptista
- CIMAGO - Center of Investigation on Environment Genetics and Oncobiology - Faculty of Medicine, University of Coimbra, 3000-354, Coimbra, Portugal.,Department of Dentistry, Faculty of Medicine, University of Coimbra, 3000-075, Coimbra, Portugal
| | - Artur Ferreira
- Maxillofacial Surgery Department, Coimbra Hospital and University Centre (CHUC), EPE, 3000-075, Coimbra, Portugal
| | - Joana Barbosa Melo
- Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra, Polo Ciências da Saúde, 3000-354, Coimbra, Portugal.,CIMAGO - Center of Investigation on Environment Genetics and Oncobiology - Faculty of Medicine, University of Coimbra, 3000-354, Coimbra, Portugal
| | - Isabel Marques Carreira
- Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra, Polo Ciências da Saúde, 3000-354, Coimbra, Portugal. .,CIMAGO - Center of Investigation on Environment Genetics and Oncobiology - Faculty of Medicine, University of Coimbra, 3000-354, Coimbra, Portugal.
| |
Collapse
|
35
|
Cidlinsky N, Dogliotti G, Pukrop T, Jung R, Weber F, Krahn MP. Inactivation of the LKB1-AMPK signaling pathway does not contribute to salivary gland tumor development - a short report. Cell Oncol (Dordr) 2016; 39:389-96. [PMID: 27480082 DOI: 10.1007/s13402-016-0290-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/15/2016] [Indexed: 01/05/2023] Open
Abstract
PURPOSE Activation of AMPK by the tumor suppressor LKB1 represents an essential gatekeeping step for cells under energetic stress to prevent their growth and proliferation by inhibiting mTOR activation, until the energy supply normalizes. The LKB1/AMPK pathway is frequently downregulated in various types of cancer, thereby uncoupling tumor cell growth and proliferation from energy supply. As yet, little information is available on the role of the LKB1/AMPK pathway in tumors derived from salivary gland tissues. METHODS We performed LKB1 protein expression and AMPK and mTOR activation analyses in several salivary gland tumor types and their respective healthy control tissues using immunohistochemistry. RESULTS No significant downregulation of LKB1 expression or decreased activation of AMPK or mTOR were observed in any of the salivary gland tumors tested. In contrast, we found that the salivary gland tumors exhibited an increased rather than a decreased AMPK activation. Although the PI3K/Akt pathway was found to be activated in most of the analyzed tumor samples, the unchanged robust activity of LKB1/AMPK likely prevents (over)activation of mTOR. CONCLUSION In contrast to many other types of cancer, inactivation or downregulation of the LKB1/AMPK pathway does not substantially contribute to the pathogenesis of salivary gland tumors.
Collapse
Affiliation(s)
- Natascha Cidlinsky
- Molecular and Cellular Anatomy, University of Regensburg, Universitätsstr. 31, 93053, Regensburg, Germany
| | - Giada Dogliotti
- Molecular and Cellular Anatomy, University of Regensburg, Universitätsstr. 31, 93053, Regensburg, Germany
| | - Tobias Pukrop
- Department of Internal Medicine III, Hematology and Medical Oncology, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Rudolf Jung
- Institute for Pathology, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Florian Weber
- Institute for Pathology, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Michael P Krahn
- Molecular and Cellular Anatomy, University of Regensburg, Universitätsstr. 31, 93053, Regensburg, Germany.
| |
Collapse
|
36
|
Lang M, Wang X, Wang H, Dong J, Lan C, Hao J, Huang C, Li X, Yu M, Yang Y, Yang S, Ren H. Arsenic trioxide plus PX-478 achieves effective treatment in pancreatic ductal adenocarcinoma. Cancer Lett 2016; 378:87-96. [PMID: 27212442 DOI: 10.1016/j.canlet.2016.05.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 05/12/2016] [Accepted: 05/17/2016] [Indexed: 02/06/2023]
Abstract
Arsenic trioxide (ATO) has been selected as a promising treatment not only in leukemia but also in solid tumors. Previous studies showed that the cytotoxicity of ATO mainly depends on the induction of reactive oxygen species. However, ATO has only achieved a modest effect in pancreatic ductal adenocarcinoma, suggesting that the existing radical scavenging proteins, such as hypoxia inducible factor-1, attenuate the effect. The goal of this study is to investigate the effect of combination treatment of ATO plus PX-478 (hypoxia-inducible factor-1 inhibitor) and its underlying mechanism. Here, we showed that PX-478 robustly strengthened the anti-growth and pro-apoptosis effect of ATO on Panc-1 and BxPC-3 pancreatic cancer cells in vitro. Meanwhile, in vivo mouse xenograft models also showed the synergistic effect of ATO plus PX-478 compared with any single agent. Further studies showed that the anti-tumor effect of ATO plus PX-478 was derived from the reactive oxygen species-induced apoptosis. We next confirmed that Hypoxia-inducible factor-1 cleared reactive oxygen species by its downstream target, forkhead box O transcription factors, and this effect may justify the strategy of ATO plus PX-478 in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Mingxiao Lang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiuchao Wang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hongwei Wang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jie Dong
- School of Public Health, Tianjin Medical University, Tianjin, China
| | - Chungen Lan
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jihui Hao
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Chongbiao Huang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xin Li
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ming Yu
- School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yanhui Yang
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Shengyu Yang
- Department of Tumor Biology and Comprehensive Melanoma Research Center, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - He Ren
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
| |
Collapse
|
37
|
Owonikoko TK, Zhang G, Kim HS, Stinson RM, Bechara R, Zhang C, Chen Z, Saba NF, Pakkala S, Pillai R, Deng X, Sun SY, Rossi MR, Sica GL, Ramalingam SS, Khuri FR. Patient-derived xenografts faithfully replicated clinical outcome in a phase II co-clinical trial of arsenic trioxide in relapsed small cell lung cancer. J Transl Med 2016; 14:111. [PMID: 27142472 PMCID: PMC4855771 DOI: 10.1186/s12967-016-0861-5] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 04/12/2016] [Indexed: 02/03/2023] Open
Abstract
Background SCLC has limited treatment options and inadequate preclinical models. Promising activity of arsenic trioxide (ASO) recorded in conventional preclinical models of SCLC supported the clinical evaluation of ASO in patients. We assessed the efficacy of ASO in relapsed SCLC patients and in corresponding patient-derived xenografts (PDX). Methods Single arm, Simon 2-stage, phase II trial to enroll patients with relapsed SCLC who have failed at least one line of therapy. ASO was administered as an intravenous infusion over 1–2 h daily for 4 days in week 1 and for 2 days in weeks 2–6 of an 8-week cycle. Treatment continued until disease progression. Pretreatment tumor biopsy was employed for PDX generation through direct implantation into subcutaneous pockets of SCID mice without in vitro manipulation and serially propagated for five generations. Ex vivo efficacy of cisplatin (3 mg/kg i.p. weekly) and ASO (3.75 mg/kg i.p. every other day) was tested in PDX representative of platinum sensitive and platinum refractory SCLC. Results The best response in 17 evaluable patients was stable disease in 2 (12 %), progressive disease in 15 (88 %) patients and median time-to-progression of seven (range 1–7) weeks. PDX was successfully grown in 5 of 9 (56 %) transplanted biopsy samples. Serially-propagated PDXs preserved characteristic small cell histology and genomic stability confirmed by immunohistochemistry, short tandem repeat (STR) profiling and targeted sequencing. ASO showed in vitro cytotoxicity but lacked in vivo efficacy against SCLC PDX tumor growth. Conclusions Cisplatin inhibited growth of PDX derived from platinum-sensitive SCLC but was ineffective against PDX from platinum-refractory SCLC. Strong concordance between clinical and ex vivo effects of ASO and cisplatin in SCLC supports the use of PDX models to prescreen promising anticancer agents prior to clinical testing in SCLC patients. Trial Registration The study was registered at http://www.clinicaltrials.gov (NCT01470248) Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-0861-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Taofeek K Owonikoko
- Department of Hematology & Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, 1365C Clifton Road, NE, Suite C3080, Atlanta, GA, 30322, USA.
| | - Guojing Zhang
- Department of Hematology & Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, 1365C Clifton Road, NE, Suite C3080, Atlanta, GA, 30322, USA
| | - Hyun S Kim
- Department of Radiology, Division of Interventional Radiology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA, 30322, USA
| | | | - Rabih Bechara
- Department of Medicine, Division of Interventional Pulmonology, Winship Cancer Institute, Atlanta, GA, 30322, USA
| | - Chao Zhang
- Department of Biostatistics, Rollins School of Public Health and Biostatistics Shared Resource, Winship Cancer Institute, Atlanta, GA, 30322, USA
| | - Zhengjia Chen
- Department of Biostatistics, Rollins School of Public Health and Biostatistics Shared Resource, Winship Cancer Institute, Atlanta, GA, 30322, USA
| | - Nabil F Saba
- Department of Hematology & Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, 1365C Clifton Road, NE, Suite C3080, Atlanta, GA, 30322, USA
| | - Suchita Pakkala
- Department of Hematology & Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, 1365C Clifton Road, NE, Suite C3080, Atlanta, GA, 30322, USA
| | - Rathi Pillai
- Department of Hematology & Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, 1365C Clifton Road, NE, Suite C3080, Atlanta, GA, 30322, USA
| | - Xingming Deng
- Department of Radiation Oncology, Winship Cancer Institute, Atlanta, GA, 30322, USA
| | - Shi-Yong Sun
- Department of Hematology & Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, 1365C Clifton Road, NE, Suite C3080, Atlanta, GA, 30322, USA
| | - Michael R Rossi
- Department of Radiation Oncology, Winship Cancer Institute, Atlanta, GA, 30322, USA.,Department of Pathology, Winship Cancer Institute, Atlanta, GA, 30322, USA
| | - Gabriel L Sica
- Department of Pathology, Winship Cancer Institute, Atlanta, GA, 30322, USA
| | - Suresh S Ramalingam
- Department of Hematology & Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, 1365C Clifton Road, NE, Suite C3080, Atlanta, GA, 30322, USA
| | - Fadlo R Khuri
- Department of Hematology & Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, 1365C Clifton Road, NE, Suite C3080, Atlanta, GA, 30322, USA
| |
Collapse
|
38
|
Liu J, Wei X, Wu Y, Wang Y, Qiu Y, Shi J, Zhou H, Lu Z, Shao M, Yu L, Tong L. Giganteaside D induces ROS-mediated apoptosis in human hepatocellular carcinoma cells through the MAPK pathway. Cell Oncol (Dordr) 2016; 39:333-42. [PMID: 27016209 DOI: 10.1007/s13402-016-0273-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2016] [Indexed: 10/22/2022] Open
Abstract
PURPOSE Every year, almost one million individuals are diagnosed with hepatocellular carcinoma (HCC) worldwide and more than 690,000 patients die of it. At present, most therapeutic anti-HCC agents are not effective, which is due to the appearance of chemo-resistance and/or toxic side effects. Therefore, it is imperative to find novel more effective anti-HCC agents. Here, we evaluated the effect of giganteaside D (GD), an oleanolic acid saponin from P. scabiosaefolia, on the growth and apoptosis of HCC cells. METHODS AND RESULTS Using MTT and clonogenic assays, we found that GD exhibited a significant growth inhibitory effect on the HCC-derived cell lines HepG2 and Bel-7402. In addition, we found that GD induced mitochondria-mediated apoptosis in these HCC-derived cells, as indicated by a decreased mitochondrial potential, activation of Caspase-9 and Caspase-3, cleavage of PARP and release of Cytochrome C from the mitochondria. Besides, we found that GD stimulated the generation of reactive oxygen species (ROS) and that blockage of ROS attenuated the GD-induced mitochondria-mediated apoptosis. Additionally, we found that GD treatment led to a decrease in phosphorylated Erk (p-Erk) and triggered the generation of p-JNK, both components of the mitogen-activated protein kinase (MAPK) signaling pathway. Inhibition of Erk or JNK by specific inhibitors or siRNAs augmented or attenuated the cytotoxic and apoptotic effects of GD. CONCLUSIONS From our results we conclude that GD can induce ROS-mediated apoptosis in HCC-derived cells through the MAPK pathway. This observation may open up avenues to explore the future use of GD as a HCC chemotherapeutic agent.
Collapse
Affiliation(s)
- Junshan Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Xiduan Wei
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Yafeng Wu
- Inpatient Department, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, People's Republic of China
| | - Yanni Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Yuwen Qiu
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Junmin Shi
- Southern Institute of Pharmaceutical Research, South China Normal University, Guangzhou, 510631, People's Republic of China
| | - Hongling Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Zibin Lu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Meng Shao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| | - Linzhong Yu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| | - Li Tong
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
39
|
Robles-Escajeda E, Das U, Ortega NM, Parra K, Francia G, Dimmock JR, Varela-Ramirez A, Aguilera RJ. A novel curcumin-like dienone induces apoptosis in triple-negative breast cancer cells. Cell Oncol (Dordr) 2016; 39:265-77. [PMID: 26920032 DOI: 10.1007/s13402-016-0272-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2016] [Indexed: 12/31/2022] Open
Abstract
PURPOSE According to the World Health Organization (WHO), breast cancer is the most common cancer affecting women worldwide. In the USA ~12.3 % of all women are expected to be diagnosed with various types of breast cancer, exhibiting varying degrees of therapeutic response rates. Therefore, the identification of novel anti-breast cancer drugs is of paramount importance. METHODS The 1,5-diaryl-3-oxo-1,4-pentadienyl pharmacophore was incorporated into a number of cytotoxins. Three of the resulting dienones, 2a, 2b and 2c, were tested for their anti-neoplastic potencies in a variety of human breast cancer-derived cell lines, including the triple negative MDA-MB-231 cell line and its metastatic variant, using a live-cell bio-imaging method. Special emphasis was put on dienone 2c, since its anti-cancer activity and its mode of inflicting cell death have so far not been reported. RESULTS We found that all three dienones exhibited potent cytotoxicities towards the breast cancer-derived cell lines tested, whereas significantly lower toxicities were observed towards the non-cancerous human breast cell line MCF-10A. The dienones 2b and 2c exhibited the greatest selective cytotoxicity at submicromolar concentration levels. We found that these two dienones induced phosphatidylserine externalization in MDA-MB-231 cells in a concentration-dependent manner, suggesting that their cytotoxic effect might be mediated by apoptosis. This possibility was confirmed by our observation that the dienone 2c can induce mitochondrial depolarization, caspase-3 activation, cell cycle disruption and DNA fragmentation in MDA-MB-231 cells. CONCLUSION Our findings indicate that dienone 2c uses the mitochondrial/intrinsic pathway to inflict apoptosis in triple negative MDA-MB-231 breast cancer-derived cells. This observation warrants further assessment of dienone 2c as a potential anti-breast cancer drug.
Collapse
Affiliation(s)
- Elisa Robles-Escajeda
- Cytometry, Screening and Imaging Core Facility, Border Biomedical Research Center, Department of Biological Sciences, the University of Texas at El Paso, 500 West University Avenue, El Paso, TX, 79968-0519, USA
| | - Umashankar Das
- Drug Discovery & Development Research Group, College of Pharmacy & Nutrition, University of Saskatchewan, 110 Science Place, Saskatoon, SK, S7N 5C9, Canada
| | - Nora M Ortega
- Cytometry, Screening and Imaging Core Facility, Border Biomedical Research Center, Department of Biological Sciences, the University of Texas at El Paso, 500 West University Avenue, El Paso, TX, 79968-0519, USA
| | - Karla Parra
- Cytometry, Screening and Imaging Core Facility, Border Biomedical Research Center, Department of Biological Sciences, the University of Texas at El Paso, 500 West University Avenue, El Paso, TX, 79968-0519, USA
| | - Giulio Francia
- Cytometry, Screening and Imaging Core Facility, Border Biomedical Research Center, Department of Biological Sciences, the University of Texas at El Paso, 500 West University Avenue, El Paso, TX, 79968-0519, USA
| | - Jonathan R Dimmock
- Drug Discovery & Development Research Group, College of Pharmacy & Nutrition, University of Saskatchewan, 110 Science Place, Saskatoon, SK, S7N 5C9, Canada
| | - Armando Varela-Ramirez
- Cytometry, Screening and Imaging Core Facility, Border Biomedical Research Center, Department of Biological Sciences, the University of Texas at El Paso, 500 West University Avenue, El Paso, TX, 79968-0519, USA.
| | - Renato J Aguilera
- Cytometry, Screening and Imaging Core Facility, Border Biomedical Research Center, Department of Biological Sciences, the University of Texas at El Paso, 500 West University Avenue, El Paso, TX, 79968-0519, USA.
| |
Collapse
|
40
|
Duz MB, Karatas OF, Guzel E, Turgut NF, Yilmaz M, Creighton CJ, Ozen M. Identification of miR-139-5p as a saliva biomarker for tongue squamous cell carcinoma: a pilot study. Cell Oncol (Dordr) 2015; 39:187-93. [PMID: 26650483 DOI: 10.1007/s13402-015-0259-z] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2015] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND Of all human oral carcinomas, 41 % are localized to the tongue. Despite considerable improvements in both diagnosis and treatment, tongue squamous cell carcinoma (TSCC) has remained one of the most lethal types of cancer. Here, we aimed at identifying a salivary microRNA (miRNA) expression signature specific for TSCC patients. METHODS To identify putative diagnostic biomarkers, we compared the miRNA expression profiles of saliva samples from three TSCC patients and four healthy control individuals using an Agilent miRNA microarray platform (V19). Three of the differentially expressed miRNAs identified were selected for further validation using quantitative reverse-transcription PCR (qRT-PCR) in saliva samples from 25 TSCC patients and 25 healthy control individuals. RESULTS Through microarray-based expression profiling, we found that 419 miRNAs were deregulated in the saliva samples from the TSCC patients compared to those from the healthy control individuals tested. Subsequent qRT-PCR analysis revealed that the expression level of miR-139-5p was significantly reduced in the TSCC validation samples compared to the controls. Further analysis of post-operative saliva samples derived from TSCC patients revealed that the miR-139-5p expression levels had turned back to normal again. In addition, we found that miR-139-5p exhibited enough power to discriminate pre-operative TSCC patients from both normal individuals (AUC: 0.805) and post-operative TSCC patients (AUC: 0.713), thereby underscoring its diagnostic potential. CONCLUSIONS From our results we conclude that saliva can be used as a feasible source for routine TSCC diagnostics and that miR-139-5p may serve as a potential biomarker for early TSCC detection.
Collapse
Affiliation(s)
- Mehmet Bugrahan Duz
- Department of Medical Genetics, Istanbul University Cerrahpasa Medical School, Istanbul, Turkey
| | - Omer Faruk Karatas
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, Turkey
| | - Esra Guzel
- Department of Medical Genetics, Istanbul University Cerrahpasa Medical School, Istanbul, Turkey.,Departments of Medical Genetics and Molecular Biology and Genetics, Biruni University, 10. Yil Caddesi Protokol Yolu No: 45, 34010, Topkapi, Istanbul, Turkey
| | - Nesrettin Fatih Turgut
- Department of Otorhinolaryngology, Cerrahpasa Medical School, Istanbul University, Istanbul, Turkey
| | - Mehmet Yilmaz
- Department of Otorhinolaryngology, Cerrahpasa Medical School, Istanbul University, Istanbul, Turkey
| | - Chad J Creighton
- Department of Medicine and Dan L. Duncan Cancer Center Division of Biostatistics, Baylor College of Medicine, Houston, TX, USA
| | - Mustafa Ozen
- Department of Medical Genetics, Istanbul University Cerrahpasa Medical School, Istanbul, Turkey. .,Departments of Medical Genetics and Molecular Biology and Genetics, Biruni University, 10. Yil Caddesi Protokol Yolu No: 45, 34010, Topkapi, Istanbul, Turkey. .,Department of Pathology & Immunology Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
41
|
Patel S, Kumar L, Singh N. Metformin and epithelial ovarian cancer therapeutics. Cell Oncol (Dordr) 2015; 38:365-75. [DOI: 10.1007/s13402-015-0235-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2015] [Indexed: 12/13/2022] Open
|
42
|
Abstract
Arsenic is an enigmatic xenobiotic that causes a multitude of chronic diseases including cancer and also is a therapeutic with promise in cancer treatment. Arsenic causes mitotic delay and induces aneuploidy in diploid human cells. In contrast, arsenic causes mitotic arrest followed by an apoptotic death in a multitude of virally transformed cells and cancer cells. We have explored the hypothesis that these differential effects of arsenic exposure are related by arsenic disruption of mitosis and are differentiated by the target cell's ability to regulate or modify cell cycle checkpoints. Functional p53/CDKN1A axis has been shown to mitigate the mitotic block and to be essential to induction of aneuploidy. More recent preliminary data suggest that microRNA modulation of chromatid cohesion also may play a role in escape from mitotic block and in generation of chromosomal instability. Other recent studies suggest that arsenic may be useful in treatment of solid tumors when used in combination with other cytotoxic agents such as cisplatin.
Collapse
Affiliation(s)
- J Christopher States
- Department of Pharmacology and Toxicology, University of Louisville, 505 S. Hancock St, Louisville, KY, 40202, USA,
| |
Collapse
|
43
|
Suzuki T, Ishibashi K, Yumoto A, Nishio K, Ogasawara Y. Utilization of arsenic trioxide as a treatment of cisplatin-resistant non-small cell lung cancer PC-9/CDDP and PC-14/CDDP cells. Oncol Lett 2015; 10:805-809. [PMID: 26622574 DOI: 10.3892/ol.2015.3352] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 05/20/2015] [Indexed: 11/06/2022] Open
Abstract
Cisplatin is a commonly used drug in combination chemotherapy. However, various malignant tumors frequently acquire resistance to cisplatin. Arsenic trioxide (ATO) has been approved as a chemotherapeutic drug for the treatment of acute promyelocytic leukemia, and the combination of ATO and cisplatin has been revealed to demonstrate synergistic effects in ovarian and small cell lung cancer cells. Thus, it was hypothesized that ATO may also be active against cisplatin-resistant non-small cell lung cancer (NSCLC) PC-9/CDDP and PC-14/CDDP cells. The present study also evaluated the effects of ATO on the cisplatin-sensitive NSCLC PC-9 and PC-14 cell lines. Notably, ATO demonstrated a markedly decreased IC50 in the cisplatin-resistant PC-9/CDDP and PC-14/CDDP cells compared with the IC50 in the cisplatin-sensitive parental PC-9 and PC-14 cells. Additionally, it was found that arsenite accumulation in the PC-9 cell line was affected through the downregulation of GS-X pump systems. Although it is likely that cisplatin resistance in PC-9 cells does not depend on the GS-X pump systems, ATO was effective against cisplatin-resistant NSCLC PC-9/CDDP and PC-14/CDDP cells in combination chemotherapy.
Collapse
Affiliation(s)
- Toshihiro Suzuki
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, Kiyose, Tokyo 204-8588, Japan
| | - Kenichi Ishibashi
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, Kiyose, Tokyo 204-8588, Japan
| | - Atsushi Yumoto
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, Kiyose, Tokyo 204-8588, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kinki University School of Medicine, Osaka-sayama, Osaka 589-8511, Japan
| | - Yuki Ogasawara
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, Kiyose, Tokyo 204-8588, Japan
| |
Collapse
|
44
|
Ono T, Ota A, Ito K, Nakaoka T, Karnan S, Konishi H, Furuhashi A, Hayashi T, Yamada Y, Hosokawa Y, Kazaoka Y. Plumbagin suppresses tumor cell growth in oral squamous cell carcinoma cell lines. Oral Dis 2015; 21:501-11. [PMID: 25580997 DOI: 10.1111/odi.12310] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 12/13/2014] [Accepted: 12/22/2014] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Plumbagin (PL), a naturally occurring quinoid, exerts antitumoral effects in diverse types of cancer cells. However, the effect of PL on tumor cell proliferation in oral squamous cell carcinoma (OSCC) remains poorly understood. In this study, we assessed the efficacy of PL, in human OSCC cells. METHODS The effect of PL on the cell growth and apoptosis of OSCC cell lines was evaluated using MTT and Annexin V assays, respectively. The effect of PL on mitochondrial membrane potential loss and reactive oxygen species (ROS) generation was evaluated using flow cytometry analysis. RESULTS MTT assay showed that PL dose-dependently suppressed OSCC cell growth, with IC50 values ranging from 3.87 to 14.6 μM. Flow cytometry analysis revealed that PL treatment resulted in a significant decrease in mitochondrial membrane potential and an increase in the number of apoptotic cells. Notably, ROS generation was significantly elevated after PL treatment. Furthermore, a ROS scavenger, N-acetylcysteine (NAC), clearly suppressed the decrease in mitochondrial membrane potential, increase of caspase-3/7 activity, and apoptosis after PL treatment. CONCLUSION This study provides the considerable evidence of the tumor-suppressive effect of PL, thereby highlighting its therapeutic potential for OSCC treatment.
Collapse
Affiliation(s)
- T Ono
- Department of Oral and Maxillofacial Surgery, Aichi Medical University Hospital, Nagakute, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Matsuo Y, Sakagami H, Mimaki Y. A rare type of sesquiterpene and β-santalol derivatives from Santalum album and their cytotoxic activities. Chem Pharm Bull (Tokyo) 2014; 62:1192-9. [PMID: 25450628 DOI: 10.1248/cpb.c14-00457] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A rare type of sesquiterpene with a spiro bicyclic system (1) and seven new (2-8) and four known (9-12) β-santalol derivatives were isolated from the heartwood of Santalum album (Santalaceae). The structures of these new compounds were determined by analysis of extensive spectroscopic data. The isolated compounds and derivatives were evaluated for their cytotoxic activity against HL-60 human promyelocytic leukemia cells, A549 human lung adenocarcinoma cells, HSC-2 and HSC-4 human oral squamous cell carcinoma cell lines, and TIG-3 normal human diploid fibroblasts. cis-β-Santalol (9) and β-santaldiol (10) induced apoptotic cell death in HL-60 cells.
Collapse
Affiliation(s)
- Yukiko Matsuo
- Tokyo University of Pharmacy and Life Sciences, School of Pharmacy
| | | | | |
Collapse
|
46
|
Apoptosis induction by an analog of curcumin (BDMC-A) in human laryngeal carcinoma cells through intrinsic and extrinsic pathways. Cell Oncol (Dordr) 2014; 37:439-54. [DOI: 10.1007/s13402-014-0207-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2014] [Indexed: 01/08/2023] Open
|
47
|
Cotylenin A and arsenic trioxide cooperatively suppress cell proliferation and cell invasion activity in human breast cancer cells. Int J Oncol 2014; 46:841-8. [PMID: 25405645 DOI: 10.3892/ijo.2014.2760] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 11/04/2014] [Indexed: 11/05/2022] Open
Abstract
Arsenic trioxide (ATO) is an approved treatment for acute promyelocytic leukemia (APL). It has also shown potential for treatment of multiple myeloma and various solid tumors including breast cancer. The requirement of high, toxic concentrations for the induction of apoptosis in non-APL and solid tumor cells is a major limitation for its use in other hematological malignancies and solid tumors. We have examined whether inducers of differentiation of leukemia cells can control the growth of solid tumor cells. In the present study, we found that cotylenin A, a plant growth regulator and a potent inducer of differentiation in myeloid leukemia cells, significantly potentiated both ATO-induced inhibition of cell growth in a liquid culture, and ATO-induced inhibition of anchorage-independent growth in a semi-solid culture in human breast cancer MCF-7 and MDA-MB-231 cells. ISIR-005 (a synthetic cotylenin A-derivative) was also able to enhance ATO-induced growth inhibition. The combined treatment with cotylenin A and ATO induced cleaved caspase-7 in MCF-7 cells at the concentrations which ATO alone scarcely induced and cotylenin A alone only weakly induced. Expression of survivin in MCF-7 cells was markedly decreased with the presence of both cotylenin A and ATO, although the expression of survivin was only slightly decreased by cotylenin A or ATO alone. The pretreatment with N-acetylcysteine significantly reduced the combination treatment-induced cell growth inhibition. These data suggest that induction of cleaved caspase-7, inhibition of survivin and oxidative responses are important events in the corporative inhibition in the growth of MCF-7 cells induced by both cotylenin A and ATO. Furthermore, we found that the combined treatment with cotylenin A and ATO also could be effective in suppressing the invasive capacity of MDA-MB-231 cells determined with the impedance-based xCELLigence Real-Time Cell Analysis technology. These results suggest that cotylenin A is an attractive enhancer for the ATO-induced anticancer activities in human breast cancer.
Collapse
|
48
|
A novel saliva-based microRNA biomarker panel to detect head and neck cancers. Cell Oncol (Dordr) 2014; 37:331-8. [PMID: 25156495 DOI: 10.1007/s13402-014-0188-2] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2014] [Indexed: 10/24/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are known to play an important role in cancer development by post-transcriptionally affecting the expression of critical genes. The aims of this study were two-fold: (i) to develop a robust method to isolate miRNAs from small volumes of saliva and (ii) to develop a panel of saliva-based diagnostic biomarkers for the detection of head and neck squamous cell carcinoma (HNSCC). METHODS Five differentially expressed miRNAs were selected from miScript™ miRNA microarray data generated using saliva from five HNSCC patients and five healthy controls. Their differential expression was subsequently confirmed by RT-qPCR using saliva samples from healthy controls (n = 56) and HNSCC patients (n = 56). These samples were divided into two different cohorts, i.e., a first confirmatory cohort (n = 21) and a second independent validation cohort (n = 35), to narrow down the miRNA diagnostic panel to three miRNAs: miR-9, miR-134 and miR-191. This diagnostic panel was independently validated using HNSCC miRNA expression data from The Cancer Genome Atlas (TCGA), encompassing 334 tumours and 39 adjacent normal tissues. Receiver operating characteristic (ROC) curve analysis was performed to assess the diagnostic capacity of the panel. RESULTS On average 60 ng/μL miRNA was isolated from 200 μL of saliva. Overall a good correlation was observed between the microarray data and the RT-qPCR data. We found that miR-9 (P <0.0001), miR-134 (P <0.0001) and miR-191 (P <0.001) were differentially expressed between saliva from HNSCC patients and healthy controls, and that these miRNAs provided a good discriminative capacity with area under the curve (AUC) values of 0.85 (P <0.0001), 0.74 (P < 0.001) and 0.98 (P < 0.0001), respectively. In addition, we found that the salivary miRNA data showed a good correlation with the TCGA miRNA data, thereby providing an independent validation. CONCLUSIONS We show that we have developed a reliable method to isolate miRNAs from small volumes of saliva, and that the saliva-derived miRNAs miR-9, miR-134 and miR-191 may serve as novel biomarkers to reliably detect HNSCC.
Collapse
|