1
|
Barbe-Richaud JB, Fattori A, Lindner V, Schuster C, Malouf G, Pencreach E, Somme L. EGFR-Mutant Urothelial Carcinoma Harboring an Ala750_Ile759delinsGlyGly Alteration with a Primary Resistance to Polychemotherapy and a Sensitivity to Osimertinib: A Literature Review on EGFR Alterations and Response to EGFR Tyrosine Kinase Inhibitors in Cancers. J Clin Med 2025; 14:3129. [PMID: 40364160 PMCID: PMC12072851 DOI: 10.3390/jcm14093129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/20/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
Urothelial carcinoma is three to four times more common in men than in women, with a 73-year old mean age at diagnosis which is older than the average age at diagnosis of all cancers. Urothelial carcinoma is rare in people under 40 years of age. Smoking, exposure to industrial chemicals, and family history influence the development of bladder cancer, but age remains one of the most important risk factors. It is well established that women are more likely to be diagnosed with an advanced disease, impacting the prognosis and a higher stage-for-stage mortality compared to men. A gender difference is also observed when considering molecular features; for example, there a higher male/female ratio in Fibroblast Growth Factor Receptor 3 (FGFR3)-mutated bladder cancer. Epidermal Growth Factor Receptor (EGFR) amplifications, which are roughly depicted in 25-50% of urothelial carcinoma, have been correlated with a worse prognosis. Genomic alterations of clinical interest are mainly Human Epidermal Growth Factor Receptor 2 mutations and amplifications, as well as FGFR 3 alterations; however, no EGFR mutation has been routinely reported despite the frequency of its amplifications. Recurrently, no targeted inhibitors have demonstrated a benefit compared to platinum-based chemotherapy. We report a rare case of a 35-year-old woman presenting bone, hepatic, and lymph node metastatic urothelial carcinoma, harboring a deletion of 24 nucleotides in exon 19 of the EGFR gene with a 5-month response to osimertinib, a third-generation EGFR tyrosine kinase inhibitor.
Collapse
Affiliation(s)
- Jean-Baptiste Barbe-Richaud
- Oncology Department, Institut de Cancérologie Strasbourg Europe, 17 Rue Albert Calmette, 67200 Strasbourg, France; (J.-B.B.-R.); (C.S.); (G.M.)
| | - Antonin Fattori
- Pathological Department, Hôpital de Hautepierre, 1 Avenue Molière, 67200 Strasbourg, France; (A.F.); (V.L.)
| | - Véronique Lindner
- Pathological Department, Hôpital de Hautepierre, 1 Avenue Molière, 67200 Strasbourg, France; (A.F.); (V.L.)
| | - Caroline Schuster
- Oncology Department, Institut de Cancérologie Strasbourg Europe, 17 Rue Albert Calmette, 67200 Strasbourg, France; (J.-B.B.-R.); (C.S.); (G.M.)
| | - Gabriel Malouf
- Oncology Department, Institut de Cancérologie Strasbourg Europe, 17 Rue Albert Calmette, 67200 Strasbourg, France; (J.-B.B.-R.); (C.S.); (G.M.)
| | - Erwan Pencreach
- Molecular Departement, Hôpital de Hautepierre, 1 Avenue Molière, 67200 Strasbourg, France;
| | - Laura Somme
- Oncology Department, Institut de Cancérologie Strasbourg Europe, 17 Rue Albert Calmette, 67200 Strasbourg, France; (J.-B.B.-R.); (C.S.); (G.M.)
| |
Collapse
|
2
|
Saosamniang P, Matsumura K, Okajima MK, Kaneko T. Directing mineralization of ZnO nanoparticles in cyanobacterial liquid crystalline polysaccharides for cancer therapies. Int J Biol Macromol 2025; 304:140716. [PMID: 39920940 DOI: 10.1016/j.ijbiomac.2025.140716] [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: 08/29/2024] [Revised: 01/20/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Effective cancer therapy faces significant challenges, including non-selective toxicity, limited structural stability, inconsistent nanoparticle (NP) morphology, and instability under varying biological conditions. These issues hindering targeted delivery and therapeutic efficacy. Previous approaches using polysaccharide-based nanomaterials have shown promise; however, problems such as inconsistent NP sizes and shapes, poor mechanical stability, and limited pH resilience restrict their clinical potential. This study hypothesized that sacran, a cyanobacterial liquid crystalline (LC) polysaccharide, can stabilize ZnO NPs, allowing for controlled mineralization, enhanced stability, and selective cytotoxicity. We developed ZnO nanocomposite xerogels in an LC sacran matrix, yielding block-like ZnO NPs (25-70 nm) with high surface-area-to-volume ratios that improve cellular uptake in tumor environments. Incorporating these NPs into chemically crosslinked sacran matrices resulted in a 3-fold increase in mechanical strength and a 10-fold improvement in swelling capacity compared to physically crosslinked systems. Additionally, the sacran-ZnO nanocomposites demonstrated robust stability under various pH conditions, indicating their resilience in diverse biological environments. Cytotoxicity assays revealed that higher concentrations of ZnO NP selectively increased toxicity toward human lung cancer cells (A549), with less impact on human dermal fibroblasts (HDFa). Moreover, HDFa successfully attached to and proliferated on the smooth surfaces of the xerogels, emphasizing their compatibility with normal cells. This highlights the potential of sacran-ZnO nanocomposite xerogels as cancer-selective therapeutic materials, offering stability and effectiveness even under varying biological conditions, while addressing key challenges associated with earlier NP-based therapies.
Collapse
Affiliation(s)
- Pruetsakorn Saosamniang
- Graduate School of Advanced Science and Technology, Energy and Environment Area, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Kazuaki Matsumura
- Graduate School of Advanced Science and Technology, Energy and Environment Area, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Maiko K Okajima
- Key Laboratory of Synthetic and Biological Colloids, School of Chemical and Material Engineering, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, China; Graduate School of Advanced Science and Technology, Energy and Environment Area, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Tatsuo Kaneko
- Key Laboratory of Synthetic and Biological Colloids, School of Chemical and Material Engineering, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu 214122, China; Graduate School of Advanced Science and Technology, Energy and Environment Area, Japan Advanced Institute of Science and Technology (JAIST), 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan.
| |
Collapse
|
3
|
Yu C, Sun J, Lai X, Tan Z, Wang Y, Du H, Pan Z, Chen T, Yang Z, Ye S, Quan J, Zhou Y. Gefitinib induces apoptosis in Caco-2 cells via ER stress-mediated mitochondrial pathways and the IRE1α/JNK/p38 MAPK signaling axis. Med Oncol 2025; 42:71. [PMID: 39924616 DOI: 10.1007/s12032-025-02622-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 01/29/2025] [Indexed: 02/11/2025]
Abstract
Gefitinib, a selective EGFR-tyrosine kinase inhibitor, exhibits potent cytotoxic effects on colorectal cancer cells, though its precise mechanisms are not fully understood. In this study, we demonstrated that gefitinib induces a dose-dependent cytotoxic response in Caco-2 cells, characterized by disrupted microtubule networks, impaired migration, and reduced viability. Gefitinib triggered apoptosis, as indicated by increased levels of cleaved caspase-3, PARP, and elevated late apoptosis rates. Mechanistically, gefitinib-induced endoplasmic reticulum (ER) stress, marked by the upregulation of IRE1α, CHOP, and ATF4. ER stress inhibition by 4-PBA significantly reduced apoptosis and restored mitochondrial membrane potential (MMP). Additionally, gefitinib-induced apoptosis was mediated through the mitochondrial pathway, reflected by the modulation of Bcl-2 family proteins, including the upregulation of Bax and Bim. Inhibition of the IRE1α-mediated JNK/p38 MAPK pathway further mitigated gefitinib-induced apoptosis and restored MMP. These findings highlight the critical role of ER stress and the IRE1α-JNK/p38 MAPK axis in gefitinib-induced mitochondrial apoptosis, offering potential therapeutic targets for colorectal cancer.
Collapse
Affiliation(s)
- Caiyuan Yu
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Jinhui Sun
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Xinyi Lai
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Zhiming Tan
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Yang Wang
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Haiyan Du
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Zhaobin Pan
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Tingyu Chen
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Ziping Yang
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Shicai Ye
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China
| | - Juanhua Quan
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China.
- Laboratory of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China.
| | - Yu Zhou
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China.
- Laboratory of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, Guangdong, China.
| |
Collapse
|
4
|
Cao W, Jin D, Min W, Li H, Wang R, Zhang J, Gou Y. Prognostic values of intracellular cell-related genes in esophageal cancer and their regulatory mechanisms. BMC Cancer 2025; 25:105. [PMID: 39833728 PMCID: PMC11744837 DOI: 10.1186/s12885-025-13483-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
Esophageal cancer is a grave malignant condition. While radiotherapy, often in conjunction with chemotherapy, serves as a cornerstone in the management of locally advanced or metastatic cases, patient tolerance and treatment resistance frequently hinder its efficacy. Cell-in-cell structures, prevalent in various tumors, have been linked to prognosis. Hence, investigating the prognostic significance and regulatory mechanisms of genes related to these intracellular structures in esophageal cancer is imperative. The Cancer Genome Atlas (TCGA) Esophageal Cancer (ESCA) dataset served as the training set for the analysis. Differentially expressed genes (DEGs) in ESCA samples were identified, with those related to intercellular structures designated cell-in-cell-related differential expression genes (CIC-related DEGs). Cox regression analysis was employed to identify prognostic genes, categorizing samples into high- and low-risk groups based on median risk scores. Validation was conducted using the GSE53624 risk model. Established methodologies included morphological mapping, enrichment analysis, immune infiltration analysis, prognostic gene expression validation, molecular docking, and Reverse Transcription Polymerase Chain Reaction (RT-PCR) validation. Thirty-eight intersecting genes were identified between the disease and normal groups in ESCA samples. Stepwise multivariate Cox analysis pinpointed three prognostic genes: androgen receptor (AR), C-X-C motif chemokine ligand 8 (CXCL8), and epidermal growth factor receptor (EGFR). The risk model's applicability was confirmed in the GSE53624 dataset, revealing eight significantly different immune-related gene sets. Prognostic gene expression validation demonstrated significant differences between the disease and normal groups in both datasets. The proteins corresponding to the three prognostic genes interacted with gefitinib and osimertinib. RT-PCR results corroborated the differential expression of prognostic genes in esophageal cancer tissues. This study identified AR, CXCL8, and EGFR as prognostic genes and demonstrated their molecular interactions with gefitinib and osimertinib, providing a foundation for ESCA diagnosis and treatment.
Collapse
Affiliation(s)
- Wei Cao
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Dacheng Jin
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Weirun Min
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Haochi Li
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Rong Wang
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
| | - Jinlong Zhang
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Yunjiu Gou
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China.
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China.
| |
Collapse
|
5
|
Mandal T, Shukla D, Pattanayak S, Barman R, Ashraf R, Dixit AK, Kumar S, Kumar D, Srivastava AK. Ellagic Acid Induces DNA Damage and Apoptosis in Cancer Stem-like Cells and Overcomes Cisplatin Resistance. ACS OMEGA 2024; 9:48988-49000. [PMID: 39713677 PMCID: PMC11656259 DOI: 10.1021/acsomega.3c08819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/28/2023] [Accepted: 01/03/2024] [Indexed: 12/24/2024]
Abstract
Cancer stem cells (CSCs) are responsible for chemoresistance and tumor relapse in many solid malignancies, including lung and ovarian cancer. Ellagic acid (EA), a natural polyphenol, exhibits anticancer effects on various human malignancies. However, its impact and mechanism of action on cancer stem-like cells (CSLCs) are only partially understood. In this study, we evaluated the therapeutic potential and underlying molecular mechanism of EA isolated from tropical mango against CSLCs. Herein, we observed that EA treatment reduces the stem-like phenotypes in cancer cells, thereby lowering the cell survival and self-renewal potential of ovarian and lung CSLCs. Additionally, EA treatment limits the populations of lung and ovarian CSLCs characterized by CD133+ and CD44+CD117+, respectively. A mechanistic investigation showed that EA treatment induces ROS generation by altering mitochondrial dynamics, causing changes in the levels of Drp1 and Mfn2, which lead to an increased level of accumulation of DNA damage and eventually trigger apoptosis in CSLCs. Moreover, pretreatment with EA sensitizes CSLCs to cisplatin treatment by enhancing DNA damage accumulation and impairing the DNA repair ability of the CSLCs. Furthermore, EA pretreatment significantly reduces cisplatin-induced mutation frequency and improves drug retention in CSLCs, potentially suppressing the development of acquired drug resistance. Taken together, our results demonstrate an unreported finding that EA inhibits CSLCs by targeting mitochondrial function and triggering apoptosis. Thus, EA can be used either alone or in combination with other chemotherepeutic drugs for the management of cancer.
Collapse
Affiliation(s)
- Tanima Mandal
- Cancer
Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Devendra Shukla
- Cancer
Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Subhamoy Pattanayak
- Organic
and Medicinal Chemistry Division, CSIR-Indian
Institute of Chemical Biology, Kolkata, West Bengal 700032, India
| | - Raju Barman
- Organic
and Medicinal Chemistry Division, CSIR-Indian
Institute of Chemical Biology, Kolkata, West Bengal 700032, India
| | - Rahail Ashraf
- Division
of Biology, Indian Institute of Science
Education & Research Tirupati, Tirupati, Andhra Pradesh 517507, India
| | - Amit Kumar Dixit
- CCRAS-Central
Ayurveda Research Institute, Kolkata, West Bengal 700091, India
| | - Sanjay Kumar
- Division
of Biology, Indian Institute of Science
Education & Research Tirupati, Tirupati, Andhra Pradesh 517507, India
| | - Deepak Kumar
- Organic
and Medicinal Chemistry Division, CSIR-Indian
Institute of Chemical Biology, Kolkata, West Bengal 700032, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amit Kumar Srivastava
- Cancer
Biology & Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal 700032, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
6
|
Metwally K, Abo-Dya NE. Pyrrolo[2,3-D]Pyrimidines as EGFR and VEGFR Kinase Inhibitors: A Comprehensive SAR Review. Curr Med Chem 2024; 31:5918-5936. [PMID: 37581522 DOI: 10.2174/0929867331666230815115111] [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: 05/12/2023] [Revised: 06/17/2023] [Accepted: 07/25/2023] [Indexed: 08/16/2023]
Abstract
Tyrosine kinases are implicated in a wide array of cellular physiological processes, including cell signaling. The discovery of the BCR-ABL tyrosine kinase inhibitor imatinib and its FDA approval in 2001 paved the way for the development of small molecule chemical entities of diverse structural backgrounds as tyrosine kinase inhibitors for the treatment of various ailments. Two of the most prominent tyrosine kinases as drug targets are the epidermal growth factor receptor (EGFR) and the vascular endothelial growth factor receptor (VEGFR), as evidenced by the clinical success of their many inhibitors in the drug market. Among several other physiological roles, EGFR regulates epithelial tissue development and homeostasis, while VEGFR regulates tumor-induced angiogenesis. The pyrrolo[2,3-d]pyrimidine nucleus represents a deaza-isostere of adenine, the nitrogenous base of ATP. The recent introduction of many pyrrolo[2,3-d]pyrimidines to the drug market as tyrosine kinase inhibitors makes them a hot topic in the medicinal chemistry research area at the present time. This review article comprehensively sheds light on the structure-activity relationship (SAR) of pyrrolo[2,3-d]pyrimidines as EGFR and VEGFR tyrosine kinase inhibitors, aiming to provide help medicinal chemists in the design of future pyrrolopyrimidine kinase inhibitors.
Collapse
Affiliation(s)
- Kamel Metwally
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk 71491, Saudi Arabia
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Nader E Abo-Dya
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk 71491, Saudi Arabia
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
7
|
Ren T, Yang MZ, Zhang WM, Qin LJ, Zhou SC, Cheng NN, Huang YJ, Sun J, Xu N, Sun HB, Zhang BB. A novel fluorescent dye selectively images and kills cancer stem cells by targeting mitochondria: Evidence from a cell line‑based zebrafish xenograft model. Oncol Lett 2023; 26:472. [PMID: 37809044 PMCID: PMC10551866 DOI: 10.3892/ol.2023.14058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/23/2023] [Indexed: 10/10/2023] Open
Abstract
Numerous agents such as near-infrared dyes that are characterized by specialized cancer imaging and cytotoxicity effects have key roles in cancer diagnosis and therapy via molecularly targeting special biological tissues, organelles and processes. In the present study, a novel fluorescent compound was demonstrated to inhibit cancer cell proliferation in a zebrafish model with slight in vivo toxicity. Further studies demonstrated selective staining of cancer cells and even putative cancer stem cells via accumulation of the dye in the mitochondria of cancer cells, compared with normal cells. Moreover, this compound was also used to image cancer cells in vivo using a zebrafish model. The compound displayed no apparent toxicity to the host animal. Overall, the data indicated that this compound was worthy of further evaluation due to its low toxicity and selective cancer cell imaging and killing effects. It could be a useful tool in cancer research.
Collapse
Affiliation(s)
- Tao Ren
- Department of Clinical Oncology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Meng-Zhe Yang
- Graduate School, Beijing TongRen Hospital, Capital Medical University, Beijing 100069, P.R. China
| | - Wei-Ming Zhang
- Department of Clinical Oncology, Wuming Hospital of Guangxi Medical University, Nanning, Guangxi 530199, P.R. China
| | - Liu-Jie Qin
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Shou-Chang Zhou
- Life Science Institute, Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Nan-Nan Cheng
- Laboratory of Clinical Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, Guangxi 533000, P.R. China
| | - Yuan-Jiao Huang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
- Life Science Institute, Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Jing Sun
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Ning Xu
- Department of Clinical Oncology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530000, P.R. China
| | - Hua-Bing Sun
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Bei-Bei Zhang
- Institute of Biomedical Research, Yunnan University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
8
|
Zhu J, Wang L, Yang Y, Han M, Yang Y, Feng R, Hu Y. Bruceine D and afatinib combination inhibits ovarian cancer cells proliferation and migration through DNA damage repair and EGFR pathway. J Investig Med 2023; 71:511-525. [PMID: 36859802 DOI: 10.1177/10815589231158043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Owing to the high rates of relapse and migration, ovarian cancer (OC) has been recognized as the most lethal gynecological malignancy worldwide. The activity of the epidermal growth factor receptor (EGFR) signaling pathway is frequently associated with OC cell proliferation and migration. Despite this knowledge, inhibition of EGFR signaling in OC patients failed to achieve satisfactory therapeutic effects. In this study, we identified that bruceine D (BD) and EGFR inhibitor, afatinib, combination resulted in synergistic anti-OC effects. The results indicated that compared with one of both drugs alone, the combination of BD and afatinib slowed the DNA replication rate, inhibition of cell viability, and proliferation and clone formation. This resulted in cell cycle arrest and cell apoptosis. In addition, the combination of BD and afatinib possessed a stronger ability to inhibit the OC cell adhesion and migration than treatment with BD or afatinib alone. Mechanistically, the combined treatment triggered intense DNA damage, suppressed DNA damage repair, and enhanced the inhibition of the EGFR pathway. These results demonstrated that compared with each pathway inhibition, combined blocking of both DNA damage repair and the EGFR pathway appears to more effective against OC treatment. The results support the potential of BD and afatinib combination as a therapeutic strategy for OC patients.
Collapse
Affiliation(s)
- Jufan Zhu
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Luo Wang
- Medical College of Zhejiang University, Hangzhou, Zhejiang, China
| | - Yuanjun Yang
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mengfei Han
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yiheng Yang
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Renqian Feng
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yan Hu
- Department of Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
9
|
Li J, Zhu L, Kwok HF. Nanotechnology-based approaches overcome lung cancer drug resistance through diagnosis and treatment. Drug Resist Updat 2023; 66:100904. [PMID: 36462375 DOI: 10.1016/j.drup.2022.100904] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Lung cancer continues to be a malignant tumor with high mortality. Two obstacles interfere with curative therapy of lung cancer: (i) poor diagnosis at the early stages, as symptoms are not specific or asymptomatic; and (ii) invariably emerging drug resistance after treatment. Some factors contributing to drug resistance include preexisting genetic/genomic drug-resistant alteration(s); activation of adaptive drug resistance pathways; remodeling of the tumor microenvironment; and pharmacological mechanisms or activation of drug efflux pumps. Despite the mechanisms explored to better understand drug resistance, a gap remains between molecular understanding and clinical application. Therefore, facilitating the translation of basic science into the clinical setting is a great challenge. Nanomedicine has emerged as a promising tool for cancer treatment. Because of their excellent physicochemical properties and enhanced permeability and retention effects, nanoparticles have great potential to revolutionize conventional lung cancer diagnosis and combat drug resistance. Nanoplatforms can be designed as carriers to improve treatment efficacy and deliver multiple drugs in one system, facilitating combination treatment to overcome drug resistance. In this review, we describe the difficulties in lung cancer treatment and review recent research progress on nanoplatforms aimed at early diagnosis and lung cancer treatment. Finally, future perspectives and challenges of nanomedicine are also discussed.
Collapse
Affiliation(s)
- Junnan Li
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR
| | - Lipeng Zhu
- Molecular Biology Research Center & Center for Medical Genetics, School of Life Sciences, Central South University, Changsha 410078, Hunan, China
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR; MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR.
| |
Collapse
|
10
|
Li M, Hu M, Wang Y, Xia Z, Li Z, Li J, Zheng D, Zheng X, Xi M. Loss of RBPMS in ovarian cancer compromises the efficacy of EGFR inhibitor gefitinib through activating HER2/AKT/mTOR/P70S6K signaling. Biochem Biophys Res Commun 2022; 637:348-357. [DOI: 10.1016/j.bbrc.2022.11.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022]
|
11
|
Eptaminitaki GC, Stellas D, Bonavida B, Baritaki S. Long Non-coding RNAs (lncRNAs) signaling in Cancer Chemoresistance: From Prediction to Druggability. Drug Resist Updat 2022; 65:100866. [DOI: 10.1016/j.drup.2022.100866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 11/03/2022]
|
12
|
Li Y, Nie Y, Guo H, Guo H, Ha C, Li Y. Establish of an Initial Platinum-Resistance Predictor in High-Grade Serous Ovarian Cancer Patients Regardless of Homologous Recombination Deficiency Status. Front Oncol 2022; 12:847085. [PMID: 35372049 PMCID: PMC8971787 DOI: 10.3389/fonc.2022.847085] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
Backgrounds Ovarian cancer (OC) is still the leading aggressive and lethal disease of gynecological cancers, and platinum-based regimes are the standard treatments. However, nearly 20%–30% of patients with OC are initial platinum resistant (IPR), and there is a lack of valid tools to predict whether they will be primary platinum resistant or not prior to chemotherapy. Methods Transcriptome data from The Cancer Genome Atlas (TCGA) was downloaded as the training data, and transcriptome data of GSE15622, GSE102073, GSE19829, and GSE26712 were retrieved from Gene Expression Omnibus (GEO) as the validation cohorts. Differentially expressed genes (DEGs) were selected between platinum-sensitive and platinum-resistant patients from the training cohort, and multiple machine-learning algorithms [including random forest, XGboost, and least absolute shrinkage and selection operator (LASSO) regression] were utilized to determine the candidate genes from DEGs. Then, we applied logistic regression to establish the IPR signature based on the expression. Finally, comprehensive clinical, genomic, and survival feature were analyzed to understand the application value of the established IPR signature. Results A total of 532 DEGs were identified between platinum-resistant and platinum-sensitive samples, and 11 of them were shared by these three-machine learning algorithms and utilized to construct an IPR prediction signature. The area under receiver operating characteristic curve (AUC) was 0.841 and 0.796 in the training and validation cohorts, respectively. Notably, the prediction capacity of this signature was stable and robust regardless of the patients’ homologous recombination deficiency (HRD) and mutation burden status. Meanwhile, the genomic feature was concordant between samples with high- or low-IPR signature, except a significantly higher prevalence of gain at Chr19q.12 (regions including CCNE1) in the high-IPR signature samples. The efficacy of prediction of platinum resistance of IPR signature successfully transferred to the precise survival prediction, with the AUC of 0.71, 0.72, and 0.66 to predict 1-, 3-, and 5-year survival, respectively. At last, we found a significantly different tumor-infiltrated lymphocytes feature, including lower abundance of CD4+ naive T cells in the samples with high-IPR signature. A relatively lower tumor immune dysfunction and exclusion (TIDE) value and more sensitivity to multiple therapies including Gefitinib may suggest the potency to transfer from platinum-based therapy to immunotherapy or target therapies in patients with high-IPR signature. Conclusion Our study established an IPR signature based on the expression of 11 genes that could stably and robustly distinguish OC patients with IPR and/or poor outcomes, which may guide therapeutic regimes tailoring.
Collapse
Affiliation(s)
- Yongmei Li
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yufei Nie
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Hongyan Guo
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Hua Guo
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Chunfang Ha
- Department of Gynecology, General Hospital of Ningxia Medical University, Yinchuan, China
- *Correspondence: Chunfang Ha, ; Yuan Li,
| | - Yuan Li
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- *Correspondence: Chunfang Ha, ; Yuan Li,
| |
Collapse
|
13
|
Parveen S, Hagar M, B. Alnoman R, Ahmed HA, El Ashry ESH, Zakaria MA. Synthesis, Docking and Density Functional Theory Approaches on 1,3-Bis-3-(4-Chlorophenyl)-2,3-Dihydroquinazolin-4(1H)-on-2-Thioxopropane toward the Discovery of Dual Kinase Inhibitor. Polycycl Aromat Compd 2021. [DOI: 10.1080/10406638.2021.1871636] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Shazia Parveen
- Faculty of Science, Chemistry Department, Taibah University, Yanbu Branch, Yanbu, Saudi Arabia
| | - Mohamad Hagar
- Faculty of Science, Chemistry Department, Taibah University, Yanbu Branch, Yanbu, Saudi Arabia
- Faculty of Science, Chemistry Department, Alexandria University, Alexandria, Egypt
| | - Rua B. Alnoman
- Faculty of Science, Chemistry Department, Taibah University, Yanbu Branch, Yanbu, Saudi Arabia
| | - Hoda A. Ahmed
- Faculty of Science, Chemistry Department, Taibah University, Yanbu Branch, Yanbu, Saudi Arabia
- Faculty of Science, Department of Chemistry, Cairo University, Cairo, Egypt
| | - El Sayed H. El Ashry
- Faculty of Science, Chemistry Department, Alexandria University, Alexandria, Egypt
| | - Mohamed A. Zakaria
- Faculty of Science, Chemistry Department, Alexandria University, Alexandria, Egypt
| |
Collapse
|
14
|
Gok D. One‐pot three‐component synthesis of novel 2‐(3‐nitro‐phenyl)‐quinazoline‐4‐carboxylic acid derivatives. J Heterocycl Chem 2019. [DOI: 10.1002/jhet.3731] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Dervis Gok
- Department of Chemistry and Chemical Processing Technologies, Kütahya Technical Sciences Vocational SchoolDumlupinar University Kutahya Turkey
| |
Collapse
|
15
|
Li L, Wang D. MicroRNA‑128‑b regulates epidermal growth factor receptor expression in non‑small cell lung cancer. Mol Med Rep 2019; 20:4803-4810. [PMID: 31638205 PMCID: PMC6854541 DOI: 10.3892/mmr.2019.10731] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 06/07/2018] [Indexed: 12/04/2022] Open
Abstract
The expression of epidermal growth factor receptor (EGFR) is regulated by microRNA (miRNA)-128-b in non-small cell lung cancer (NSCLC); however, the association between miRNA-128-b expression and EGFR expression has not been determined in vivo. The expression of miRNA-128-b was detected by reverse transcription (RT)-quantitative polymerase chain reaction (PCR); semi-quantitative RT-PCR was used to detect EGFR mRNA expression; immunostaining was used to detect EGFR protein expression. The results revealed that expression of miR28b in cancer tissues was decreased compared with normal tissues, and the expression of EGFR mRNA in cancer tissues was increased compared with normal tissues. Immunohistochemistry analysis revealed that the normal tissues did not express EGFR protein, and the positive expression rate of EGFR in cancer tissues was 60%. Furthermore, the relative expression levels of miRNA-128-b were demonstrated to be correlated with EGFR mRNA and protein expression levels. In addition, the results revealed that miRNA-128-b regulated EGFR expression in NSCLC cells. In conclusion, the results of the present study suggested that miRNA-128-b may regulate the expression of EGFR in NSCLC cells, and that optimizing targeted therapy is conducive to the development of novel therapeutic strategies for the treatment of patients with lung cancer.
Collapse
Affiliation(s)
- Ling Li
- Department of Oncology, Xintai People's Hospital, Xintai, Shandong 271200, P.R. China
| | - Dongqing Wang
- Department of Radiation Oncology, Shandong Tumor Hospital, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
16
|
Cho ES, Kang HE, Kim NH, Yook JI. Therapeutic implications of cancer epithelial-mesenchymal transition (EMT). Arch Pharm Res 2019; 42:14-24. [PMID: 30649699 DOI: 10.1007/s12272-018-01108-7] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 12/27/2018] [Indexed: 12/19/2022]
Abstract
The epithelial-mesenchymal transition (EMT) comprises an essential biological process involving cancer progression as well as initiation. While the EMT has been regarded as a phenotypic conversion from epithelial to mesenchymal cells, recent evidence indicates that it plays a critical role in stemness, metabolic reprogramming, immune evasion and therapeutic resistance of cancer cells. Interestingly, several transcriptional repressors including Snail (SNAI1), Slug (SNAI2) and the ZEB family constitute key players for EMT in cancer as well as in the developmental process. Note that the dynamic conversion between EMT and epithelial reversion (mesenchymal-epithelial transition, MET) occurs through variable intermediate-hybrid states rather than being a binary process. Given the close connection between oncogenic signaling and EMT repressors, the EMT has emerged as a therapeutic target or goal (in terms of MET reversion) in cancer therapy. Here we review the critical role of EMT in therapeutic resistance and the importance of EMT as a therapeutic target for human cancer.
Collapse
Affiliation(s)
- Eunae Sandra Cho
- Department of Oral Pathology, Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, 03722, Republic of Korea
| | - Hee Eun Kang
- Department of Oral Pathology, Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, 03722, Republic of Korea
| | - Nam Hee Kim
- Department of Oral Pathology, Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, 03722, Republic of Korea.
| | - Jong In Yook
- Department of Oral Pathology, Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, 03722, Republic of Korea.
| |
Collapse
|
17
|
Bonello M, Sims AH, Langdon SP. Human epidermal growth factor receptor targeted inhibitors for the treatment of ovarian cancer. Cancer Biol Med 2018; 15:375-388. [PMID: 30766749 PMCID: PMC6372909 DOI: 10.20892/j.issn.2095-3941.2018.0062] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 10/26/2018] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer is the second most lethal gynecological cancer worldwide and while most patients respond to initial therapy, they often relapse with resistant disease. Human epidermal growth factor receptors (especially HER1/EGFR and HER2/ERBB2) are involved in disease progression; hence, strategies to inhibit their action could prove advantageous in ovarian cancer patients, especially in patients resistant to first line therapy. Monoclonal antibodies and tyrosine kinase inhibitors are two classes of drugs that act on these receptors. They have demonstrated valuable antitumor activity in multiple cancers and their possible use in ovarian cancer continues to be studied. In this review, we discuss the human epidermal growth factor receptor family; review emerging clinical studies on monoclonal antibodies and tyrosine kinase inhibitors targeting these receptors in ovarian cancer patients; and propose future research possibilities in this area.
Collapse
Affiliation(s)
- Maria Bonello
- Cancer Research UK Edinburgh Center and Division of Pathology Laboratory, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Andrew Harvey Sims
- Cancer Research UK Edinburgh Center and Division of Pathology Laboratory, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Simon Peter Langdon
- Cancer Research UK Edinburgh Center and Division of Pathology Laboratory, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| |
Collapse
|
18
|
Sima N, Sun W, Gorshkov K, Shen M, Huang W, Zhu W, Xie X, Zheng W, Cheng X. Small Molecules Identified from a Quantitative Drug Combinational Screen Resensitize Cisplatin's Response in Drug-Resistant Ovarian Cancer Cells. Transl Oncol 2018; 11:1053-1064. [PMID: 29982103 PMCID: PMC6034569 DOI: 10.1016/j.tranon.2018.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 06/10/2018] [Accepted: 06/11/2018] [Indexed: 12/29/2022] Open
Abstract
Drug resistance to chemotherapy occurs in many ovarian cancer patients resulting in failure of treatment. Exploration of drug resistance mechanisms and identification of new therapeutics that overcome the drug resistance can improve patient prognosis. Following a quantitative combination screen of 6060 approved drugs and bioactive compounds in a cisplatin-resistant A2780-cis ovarian cancer cell line, 38 active compounds with IC50s under 1 μM suppressed the growth of cisplatin-resistant ovarian cancer cells. Among these confirmed compounds, CUDC-101, OSU-03012, oligomycin A, VE-821, or Torin2 in a combination with cisplatin restored cisplatin's apoptotic response in the A2780-cis cells, while SR-3306, GSK-923295, SNX-5422, AT-13387, and PF-05212384 directly suppressed the growth of A2780-cis cells. One of the mechanisms for overcoming cisplatin resistance in these cells is mediated by the inhibition of epidermal growth factor receptor (EGFR), though not all the EGFR inhibitors are equally active. The increased levels of total EGFR and phosphorylated-EGFR (p-EGFR) in the A2780-cis cells were reduced after the combined treatment of cisplatin with EGFR inhibitors. In addition, a knockdown of EGFR mRNA reduced cisplatin resistance in the A2780-cis cells. Therefore, the top active compounds identified in this work can be studied further as potential treatments for cisplatin-resistant ovarian cancer. The quantitative combinational screening approach is a useful method for identifying effective compounds and drug combinations against drug-resistant cancer cells.
Collapse
Affiliation(s)
- Ni Sima
- Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Wei Sun
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Kirill Gorshkov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Wei Huang
- Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Wenge Zhu
- Department of Biochemistry and Molecular Biology, The George Washington University Medical School, Washington, DC
| | - Xing Xie
- Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA.
| | - Xiaodong Cheng
- Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
19
|
Muinao T, Pal M, Deka Boruah HP. Origins based clinical and molecular complexities of epithelial ovarian cancer. Int J Biol Macromol 2018; 118:1326-1345. [PMID: 29890249 DOI: 10.1016/j.ijbiomac.2018.06.036] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 06/06/2018] [Accepted: 06/07/2018] [Indexed: 12/25/2022]
Abstract
Ovarian cancer is the most lethal of all common gynaecological malignancies in women worldwide. Ovarian cancer comprises of >15 distinct tumor types and subtypes characterized by histopathological features, environmental and genetic risk factors, precursor lesions and molecular events during oncogenesis. Recent studies on gene signature profiling of different subtypes of ovarian cancer have revealed significant genetic heterogeneity between and within each ovarian cancer histological subtype. Thus, an immense interest have shown towards a more personalized medicine for understanding the clinical and molecular complexities of four major types of epithelial ovarian cancer (serous, endometrioid, clear cell, and mucinous). As such, further in depth studies are needed for identification of molecular signalling network complexities associated with effective prognostication and targeted therapies to prevent or treat metastasis. Therefore, understanding the metastatic potential of primary ovarian cancer and therapeutic interventions against lethal ovarian cancer for the development of personalized therapies is very much indispensable. Consequently, in this review we have updated the key dysregulated genes of four major subtypes of epithelial carcinomas. We have also highlighted the recent advances and current challenges in unravelling the complexities of the origin of tumor as well as genetic heterogeneity of ovarian cancer.
Collapse
Affiliation(s)
- Thingreila Muinao
- Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat, Assam 785006, India; Academy of Scientific & Innovative Research, Jorhat Campus, Assam 785006, India
| | - Mintu Pal
- Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat, Assam 785006, India; Academy of Scientific & Innovative Research, Jorhat Campus, Assam 785006, India.
| | - Hari Prasanna Deka Boruah
- Biological Sciences and Technology Division, CSIR-North East Institute of Science and Technology, Jorhat, Assam 785006, India; Academy of Scientific & Innovative Research, Jorhat Campus, Assam 785006, India
| |
Collapse
|
20
|
Chang SJ, Liao EC, Yeo HY, Kuo WH, Chen HY, Tsai YT, Wei YS, Chen YJ, Wang YS, Li JM, Shih CC, Chan CH, Lai ZY, Chou HC, Chuang YJ, Chan HL. Proteomic investigating the cooperative lethal effect of EGFR and MDM2 inhibitors on ovarian carcinoma. Arch Biochem Biophys 2018; 647:10-32. [PMID: 29655550 DOI: 10.1016/j.abb.2018.04.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 03/23/2018] [Accepted: 04/07/2018] [Indexed: 11/17/2022]
Abstract
With the concept of precision medicine, combining multiple molecular-targeting therapies has brought new approaches to current cancer treatments. Malfunction of the tumor suppressor protein, p53 is a universal hallmark in human cancers. Under normal conditions, p53 is degraded through an ubiquitin-proteosome pathway regulated by its negative regulator, MDM2. In contrast, cellular stress such as DNA damage will activate p53 to carry out DNA repair, cell cycle arrest, and apoptosis. In this study, we focused on ovarian carcinoma with high EGFR and MDM2 overexpression rate. We assessed the effects of combined inhibition by MDM2 (JNJ-26854165) and EGFR (gefitinib) inhibitors on various ovarian cell lines to determine the importance of these two molecular targets on cell proliferation. We then used a proteomic strategy to investigate the relationship between MDM2 and EGFR inhibition to explore the underlying mechanisms of how their combined signaling blockades work together to exert cooperative inhibition. Our results demonstrated that all four cell lines were sensitive to both individual and combined, MDM2 and EGFR inhibition. The proteomic analysis also showed that gefitinib/JNJ-treated CAOV3 cells exhibited downregulation of proteins involved in nucleotide biosynthesis such as nucleoside diphosphate kinase B (NME2). In conclusion, our study showed that the combined treatment with JNJ and gefitinib exerted synergistic inhibition on cell proliferation, thereby suggesting the potential application of combining MDM2 inhibitors with EGFR inhibitors for enhancing efficacy in ovarian cancer treatment.
Collapse
Affiliation(s)
- Shing-Jyh Chang
- Department of Obstetrics and Gynecology, Hsinchu MacKay Memorial Hospital, Hsinchu, Taiwan
| | - En-Chi Liao
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsin-Yueh Yeo
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Wen-Hung Kuo
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsin-Yi Chen
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Ting Tsai
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Shan Wei
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Ying-Jen Chen
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Shiuan Wang
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Ji-Min Li
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Chuan-Chi Shih
- Department of Obstetrics and Gynecology, Hsinchu MacKay Memorial Hospital, Hsinchu, Taiwan
| | - Chia-Hao Chan
- Department of Obstetrics and Gynecology, Hsinchu MacKay Memorial Hospital, Hsinchu, Taiwan
| | - Zih-Yin Lai
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsiu-Chuan Chou
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Yung-Jen Chuang
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan.
| | - Hong-Lin Chan
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
21
|
Pang J, Jiang P, Wang Y, Jiang L, Qian H, Tao Y, Shi R, Gao J, Chen Y, Wu Y. Cross-linked hyaluronan gel inhibits the growth and metastasis of ovarian carcinoma. J Ovarian Res 2018; 11:22. [PMID: 29510732 PMCID: PMC5840805 DOI: 10.1186/s13048-018-0394-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/28/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The recurrence, metastasis and poor prognosis are important characteristics of ovarian carcinoma (OC), which are associated with exfoliation of cells from the primary tumor and colonization of the cells in pelvic cavity. On the other hand, the life quality of the patients undergoing surgical resection of OC was influenced by postoperative adhesions. Therefore, preventing postoperative implant tumor and adhesion may be effective methods to improve OC treatment. HyaRegen Gel, a cross-linked hyaluronan gel (CHAG), has been widely used as an anti-adhesive agent following pelvic operation in clinic. However, whether it can affect the implantation and growth of OC cells or not is still not clear. METHODS Migration and invasion assays were applied to detect the effect of CHAG on migration and invasion of OC cells. Western blotting was performed to detect the phosphorylation/activation of EGFR and ERK, and the expression of PCNA and MMP7. Pull down assay was used to analyze the effect of CHAG on the activation of small G protein Rac1. Nude mice implantation tumor model was applied to observe the effect of CHAG on implantation tumor of OC cells. RESULTS The results of in vitro experiments showed that CHAG suppressed both basic and EGF-induced migration and invasion of OC cells, blocked the activation of EGF-initiated EGFR activation, inhibited downstream signal transduction of EGFR, and decreased expression of proliferation and migration/invasion related proteins. Meanwhile, results of in vivo experiments showed that CHAG not only inhibited the formation of implantation tumor of OC cells but also delayed the of the growth of the tumors. CONCLUSIONS CHAG inhibited migration, invasion and proliferation of OC cells in vitro, and suppressed development of implantation tumor of OC in vivo. This made it as both anti-tumor and anti-adhesion agents.
Collapse
Affiliation(s)
- Ji Pang
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, 212013, People's Republic of China
| | - Pengcheng Jiang
- Department of Obstetrics and Gynecology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou City, Jiangsu Province, China
| | - Ying Wang
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, 212013, People's Republic of China
| | - Lu Jiang
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, 212013, People's Republic of China
| | - Hai Qian
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, 212013, People's Republic of China
| | - Yan Tao
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, 212013, People's Republic of China
| | - Ruxia Shi
- Department of Obstetrics and Gynecology, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou City, Jiangsu Province, China
| | - Jizong Gao
- R & D Department, Changzhou BioRegen Biomedical (Changzhou) Co., Ltd., Changzhou City, Jiangsu Province, China
| | - Yongchang Chen
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, 212013, People's Republic of China
| | - Yan Wu
- Department of Physiology, School of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, 212013, People's Republic of China.
| |
Collapse
|
22
|
Carter CA, Zeman K, Day RM, Richard P, Oronsky A, Oronsky N, Lybeck M, Scicinski J, Oronsky B. Addressing the elephant in the room, therapeutic resistance in non-small cell lung cancer, with epigenetic therapies. Oncotarget 2018; 7:40781-40791. [PMID: 27007055 PMCID: PMC5130044 DOI: 10.18632/oncotarget.8205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 03/07/2016] [Indexed: 12/15/2022] Open
Abstract
Like Chinese boxes nesting inside each other, the classification of non-small cell lung cancer (NSCLC) is subdivided into smaller and smaller subtypes on the basis of histological and molecular attributes. The latter characterizes NSCLC by its molecular alterations and the identification of inhibitors that target these cancer-specific "driver" mutations. Despite the initial promise of precision-guided therapies to inhibit a finer and finer array of molecular subcategories, despite even the curative potential of immunotherapeutic checkpoint blockade, in particular, casualties still abound and true clinical success stories are few and far between; the ever-present, if sometimes unmentioned, "elephant in the room", is the acquisition of resistance, which, sooner or later, rears its ugly head to undermine treatment success and shorten survival. Emerging data suggests that epigenetic therapies are able to reprogram the aberrant tumor-associated epigenome and 'tame the beast of resistance', thereby prolonging survival. This article reviews the role of epigenetic dysregulation in NSCLC, explores PFS2 as a possible surrogate endpoint, briefly mentions possible biomarkers and highlights combinatorial treatment epigenetic strategies to "prime" tumors and reverse resistance.
Collapse
Affiliation(s)
- Corey A Carter
- Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Karen Zeman
- National Naval Medical Center, Bethesda, MD, USA
| | - Regina M Day
- Uniformed Services University of The Health Sciences, Bethesda, MD, USA
| | - Patrick Richard
- Uniformed Services University of The Health Sciences, Bethesda, MD, USA
| | | | | | | | | | | |
Collapse
|
23
|
Vera O, Jimenez J, Pernia O, Rodriguez-Antolin C, Rodriguez C, Sanchez Cabo F, Soto J, Rosas R, Lopez-Magallon S, Esteban Rodriguez I, Dopazo A, Rojo F, Belda C, Alvarez R, Valentin J, Benitez J, Perona R, De Castro J, Ibanez de Caceres I. DNA Methylation of miR-7 is a Mechanism Involved in Platinum Response through MAFG Overexpression in Cancer Cells. Am J Cancer Res 2017; 7:4118-4134. [PMID: 29158814 PMCID: PMC5695001 DOI: 10.7150/thno.20112] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/04/2017] [Indexed: 11/16/2022] Open
Abstract
One of the major limitations associated with platinum use is the resistance that almost invariably develops in different tumor types. In the current study, we sought to identify epigenetically regulated microRNAs as novel biomarkers of platinum resistance in lung and ovarian cancers, the ones with highest ratios of associated chemo-resistance. Methods: We combined transcriptomic data from microRNA and mRNA under the influence of an epigenetic reactivation treatment in a panel of four paired cisplatin -sensitive and -resistant cell lines, followed by real-time expression and epigenetic validations for accurate candidate selection in 19 human cancer cell lines. To identify specific candidate genes under miRNA regulation, we assembled “in silico” miRNAs and mRNAs sequences by using ten different algorithms followed by qRT-PCR validation. Functional assays of site-directed mutagenesis and luciferase activity, miRNAs precursor overexpression, silencing by antago-miR and cell viability were performed to confirm their specificity in gene regulation. Results were further explored in 187 primary samples obtained from ovarian tumors and controls. Results: We identified 4 candidates, miR-7, miR-132, miR-335 and miR-148a, which deregulation seems to be a common event in the development of resistance to cisplatin in both tumor types. miR-7 presented specific methylation in resistant cell lines, and was associated with poorer prognosis in ovarian cancer patients. Our experimental results strongly support the direct regulation of MAFG through miR-7 and their involvement in the development of CDDP resistance in human tumor cells. Conclusion: The basal methylation status of miR-7 before treatment may be a potential clinical epigenetic biomarker, predictor of the chemotherapy outcome to CDDP in ovarian cancer patients. To the best of our knowledge, this is the first report linking the regulation of MAFG by miRNA-7 and its role in chemotherapy response to CDDP. Furthermore, this data highlights the possible role of MAFG as a novel therapeutic target for platinum resistant tumors.
Collapse
|
24
|
Increased EGFR expression induced by a novel oncogene, CUG2, confers resistance to doxorubicin through Stat1-HDAC4 signaling. Cell Oncol (Dordr) 2017; 40:549-561. [DOI: 10.1007/s13402-017-0343-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2017] [Indexed: 12/22/2022] Open
|
25
|
Mehner C, Oberg AL, Goergen KM, Kalli KR, Maurer MJ, Nassar A, Goode EL, Keeney GL, Jatoi A, Radisky DC, Radisky ES. EGFR as a prognostic biomarker and therapeutic target in ovarian cancer: evaluation of patient cohort and literature review. Genes Cancer 2017; 8:589-599. [PMID: 28740577 PMCID: PMC5511892 DOI: 10.18632/genesandcancer.142] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Background Limited effectiveness of therapeutic agents targeting epidermal growth factor receptor (EGFR) in clinical trials using unselected ovarian cancer patients has prompted efforts to more effectively stratify patients who might best benefit from these therapies. A series of studies that have evaluated immunohistochemical (IHC) staining of EGFR in ovarian cancer biopsies has produced unclear results as to the utility of this measure as a prognostic biomarker. Here, we used one of the largest, single institution cohorts to date to determine possible associations of EGFR expression with patient outcome. Methods We performed IHC staining of EGFR in tissue microarrays including nearly 500 patient tumor samples. Staining was classified by subcellular localization (membranous, cytoplasmic) or by automated image analysis algorithms. We also performed a literature review to place these results in the context of previous studies. Results No significant associations were found between EGFR subcellular localization or expression and histology, stage, grade, or outcome. These results were broadly consistent with the consensus of the reviewed literature. Conclusions These results suggest that IHC staining for EGFR may not be a useful prognostic biomarker for ovarian cancer patients. Future studies should pursue other staining methods or analysis in combination with other pathway mediators.
Collapse
Affiliation(s)
- Christine Mehner
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Ann L Oberg
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Krista M Goergen
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | | | - Matthew J Maurer
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Aziza Nassar
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, USA
| | - Ellen L Goode
- Department of Health Sciences Research, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Gary L Keeney
- Department of Laboratory Medicine and Pathology, Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA
| | - Aminah Jatoi
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Evette S Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
26
|
Liu S, Yang H, Chen Y, He B, Chen Q. Krüppel-Like Factor 4 Enhances Sensitivity of Cisplatin to Lung Cancer Cells and Inhibits Regulating Epithelial-to-Mesenchymal Transition. Oncol Res 2017; 24:81-7. [PMID: 27296948 PMCID: PMC7838665 DOI: 10.3727/096504016x14597766487717] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In order to improve therapeutic efficacy, it is a current emergency to better know the mechanisms underlying cisplatin resistance in lung cancer cells. In this study, we aim to investigate the role of Krüppel-like factor 4 (KLF4) in cisplatin-resistant lung cancer cells. We developed cisplatin-resistant lung cancer cell line A549/DDP, and then a battery of experiments was used to analyze the effects of KLF4 in cisplatin resistance of lung cancer. We found that KLF4 was significantly downregulated in cisplatin-resistant A549 cells and forced KLF4 expression inhibited cell growth and induced apoptosis. Further, we found that overexpression of KLF4 was able to inhibit cell migration and invasion, to inhibit the expression of Slug, Twist, and vimentin, and to increase the expression of E-cadherin and subsequent inhibition of the EMT process. Thus, overexpression of KLF4 may be a potential strategy for lung cancer treatment, especially for cisplatin-resistant cases.
Collapse
Affiliation(s)
- Shenggang Liu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | | | | | | | | |
Collapse
|
27
|
Li B, Li Q, Mo J, Dai H. Drug-Loaded Polymeric Nanoparticles for Cancer Stem Cell Targeting. Front Pharmacol 2017; 8:51. [PMID: 28261093 PMCID: PMC5306366 DOI: 10.3389/fphar.2017.00051] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 01/24/2017] [Indexed: 12/15/2022] Open
Abstract
Cancer stem cells (CSCs) have been reported to play critical roles in tumor initiation, propagation, and regeneration of cancer. Nano-size vehicles are employed to deliver drugs to target the CSCs for cancer therapy. Polymeric nanoparticles have been considered as the most efficient vehicles for drug delivery due to their excellent pharmacokinetic properties. The CSCs specific antibodies or ligands can be conjugated onto the surface or interior of nanoparticles to successfully target and finally eliminate CSCs. In this review, we focus on the approaches of polymeric nanoparticles design for loading drug, and their potential application for CSCs targeting in cancer therapy.
Collapse
Affiliation(s)
- Binbin Li
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of TechnologyWuhan, China
- Biomedical Materials and Engineering Research Center of Hubei ProvinceWuhan, China
| | - Qinghua Li
- Department of Neurology, Affiliated Hospital of Guilin Medical UniversityGuilin, China
| | - Jingxin Mo
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of EducationGuangzhou, China
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen UniversityGuangzhou, China
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of TechnologyWuhan, China
- Biomedical Materials and Engineering Research Center of Hubei ProvinceWuhan, China
| |
Collapse
|
28
|
Diagnostic Value of Serum Angiogenesis Markers in Ovarian Cancer Using Multiplex Immunoassay. Int J Mol Sci 2017; 18:ijms18010123. [PMID: 28075407 PMCID: PMC5297757 DOI: 10.3390/ijms18010123] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 12/14/2016] [Accepted: 12/20/2016] [Indexed: 12/14/2022] Open
Abstract
As cancer development involves pathological vessel formation, 16 angiogenesis markers were evaluated as potential ovarian cancer (OC) biomarkers. Blood samples collected from 172 patients were divided based on histopathological result: OC (n = 38), borderline ovarian tumours (n = 6), non-malignant ovarian tumours (n = 62), healthy controls (n = 50) and 16 patients were excluded. Sixteen angiogenesis markers were measured using BioPlex Pro Human Cancer Biomarker Panel 1 immunoassay. Additionally, concentrations of cancer antigen 125 (CA125) and human epididymis protein 4 (HE4) were measured in patients with adnexal masses using electrochemiluminescence immunoassay. In the comparison between OC vs. non-OC, osteopontin achieved the highest area under the curve (AUC) of 0.79 (sensitivity 69%, specificity 78%). Multimarker models based on four to six markers (basic fibroblast growth factor-FGF-basic, follistatin, hepatocyte growth factor-HGF, osteopontin, platelet-derived growth factor AB/BB-PDGF-AB/BB, leptin) demonstrated higher discriminatory ability (AUC 0.80-0.81) than a single marker (AUC 0.79). When comparing OC with benign ovarian tumours, six markers had statistically different expression (osteopontin, leptin, follistatin, PDGF-AB/BB, HGF, FGF-basic). Osteopontin was the best single angiogenesis marker (AUC 0.825, sensitivity 72%, specificity 82%). A three-marker panel consisting of osteopontin, CA125 and HE4 better discriminated the groups (AUC 0.958) than HE4 or CA125 alone (AUC 0.941 and 0.932, respectively). Osteopontin should be further investigated as a potential biomarker in OC screening and differential diagnosis of ovarian tumours. Adding osteopontin to a panel of already used biomarkers (CA125 and HE4) significantly improves differential diagnosis between malignant and benign ovarian tumours.
Collapse
|
29
|
Targeted in vivo delivery of EGFR siRNA inhibits ovarian cancer growth and enhances drug sensitivity. Sci Rep 2016; 6:36518. [PMID: 27819259 PMCID: PMC5098171 DOI: 10.1038/srep36518] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/18/2016] [Indexed: 01/05/2023] Open
Abstract
A functionalized nanohydrogel siRNA delivery system and a mouse model of serous ovarian cancer were used to test predictions from previous cell line studies that knockdown of EGFR (epidermal growth factor receptor) may be of clinical significance in the treatment of epithelial tumors especially with respect to the enhancement of platinum based therapies. Our results support these predictions and suggest that targeted delivery of EGFR siRNA may be an effective strategy for the treatment of ovarian and other epithelial tumors associated with elevated levels of EGFR and especially those demonstrating resistance to platinum-based therapies.
Collapse
|
30
|
Synthesis of chalcone incorporated quinazoline derivatives as anticancer agents. Saudi Pharm J 2016; 25:275-279. [PMID: 28344479 PMCID: PMC5355550 DOI: 10.1016/j.jsps.2016.06.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 06/19/2016] [Indexed: 11/12/2022] Open
Abstract
A series of ten novel chalcone incorporated quinazoline derivatives (11a–11j) were designed and synthesized. All the synthesized compounds were evaluated for their anticancer activities against four human cancer cell lines (A549, HT-29, MCF-7 and A375). Among them, four compounds, 11f, 11g, 11i and 11j showed more potent anticancer activity than the control drug, Combretastatin – A4.
Collapse
|
31
|
Granados ML, Hudson LG, Samudio-Ruiz SL. Contributions of the Epidermal Growth Factor Receptor to Acquisition of Platinum Resistance in Ovarian Cancer Cells. PLoS One 2015; 10:e0136893. [PMID: 26351843 PMCID: PMC4564275 DOI: 10.1371/journal.pone.0136893] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 08/09/2015] [Indexed: 01/12/2023] Open
Abstract
Acquisition of platinum resistance following first line platinum/taxane therapy is commonly observed in ovarian cancer patients and prevents clinical effectiveness. There are few options to prevent platinum resistance; however, demethylating agents have been shown to resensitize patients to platinum therapy thereby demonstrating that DNA methylation is a critical contributor to the development of platinum resistance. We previously reported the Epidermal Growth Factor Receptor (EGFR) is a novel regulator of DNA methyltransferase (DNMT) activity and DNA methylation. Others have shown that EGFR activation is linked to cisplatin treatment and platinum resistance. We hypothesized that cisplatin induced activation of the EGFR mediates changes in DNA methylation associated with the development of platinum resistance. To investigate this, we evaluated EGFR signaling and DNMT activity after acute cisplatin exposure. We also developed an in vitro model of platinum resistance to examine the effects of EGFR inhibition on acquisition of cisplatin resistance. Acute cisplatin treatment activates the EGFR and downstream signaling pathways, and induces an EGFR mediated increase in DNMT activity. Cisplatin resistant cells also showed increased DNMT activity and global methylation. EGFR inhibition during repeated cisplatin treatments generated cells that were more sensitive to cisplatin and did not develop increases in DNA methylation or DNMT activity compared to controls. These findings suggest that activation of EGFR during platinum treatment contributes to the development of platinum resistance. Furthermore, EGFR inhibition may be an effective strategy at attenuating the development of platinum resistance thereby enhancing the effectiveness of chemotherapeutic treatment in ovarian cancer.
Collapse
Affiliation(s)
- Michaela L. Granados
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, United States of America
| | - Laurie G. Hudson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, United States of America
| | - Sabrina L. Samudio-Ruiz
- Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, United States of America
| |
Collapse
|
32
|
Hyun DS, Kim JD, Park JH, Park SJ, Song CH, Ku SK. Effects of Bojungikkitang (a Polyherbal Formula), on Gefitinib Pharmacokinetics in Rats. INT J PHARMACOL 2015. [DOI: 10.3923/ijp.2015.604.610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
33
|
Du P, Zhang X, Liu H, Chen L. Lentivirus-Mediated RNAi silencing targeting ERCC1 reverses cisplatin resistance in cisplatin-resistant ovarian carcinoma cell line. DNA Cell Biol 2015; 34:497-502. [PMID: 25941922 DOI: 10.1089/dna.2015.2805] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The aim of the study was to investigate the potential mechanisms that interferencing of excision repair cross-complementing gene 1 (ERCC1) mediated by lentiviral vector in cisplatin-resistant ovarian cancer SKOV3/DDP cells. The human platinum-resistant ovarian carcinoma cell line SKOV3/DDP was transfected by pLVX-shRNA lentivirus. Interference efficiency for ERCC1 by lentiviruses transfection was detected by real-time polymerase chain reaction and western blot assay. CCK-8 assay was used for cell proliferation on cell resistance after transfection with ERCC1. Effects of cell apoptosis and cell cycles were detected by flow cytometry. The expression levels of ERCC1 were significantly suppressed in SKOV3/DDP cells after stably transfecting with shERCC1-recombinant plasmid. The results of cell viability assay demonstrated that interference with ERCC1 gene increased the sensitivity of SKOV3/DDP cells to cisplatin (p<0.01). ERCC1 gene-specific silencing promoted cell apoptosis of SKOV3/DDP cells (p<0.01) detected by flow cytometry. Cell cycle analysis results showed that the proportion of cells in G1 and S phase decreased, while the proportion of G2 phase cells increased in ERCC1-silencing cells. The differences were statistically significant (p<0.01), which demonstrated that stable interferencing with ERCC1 induced the cells arrest in G2 phase after being treated by DDP and silencing the expression of ERCC1 inhibited cell proliferation by preventing the progression of cell mitosis. ERCC1 gene silencing effectively reversed SKOV3/DDP cell resistance to cisplatin and increased sensitivity to cisplatin resistance in cisplatin-resistant ovarian cancer cells. Interference of ERCC1 promoted the apoptosis of SKOV3/DDP cells and prevented cell mitosis by inducing G0/G1 phase arrest. Thus, ERCC1 could be a potential therapeutic target for the therapy of cisplatin-resistant ovarian cancer and it would provide new ideas for epigenetic therapy of drug-resistant epithelial ovarian cancer.
Collapse
Affiliation(s)
- Pei Du
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| | - Xiaowei Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| | - Hualin Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| | - Liquan Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| |
Collapse
|
34
|
Fernandes J. The study of homology between tumor progression genes and members of retroviridae as a tool to predict target-directed therapy failure. Front Pharmacol 2015; 6:92. [PMID: 25983693 PMCID: PMC4416442 DOI: 10.3389/fphar.2015.00092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 04/16/2015] [Indexed: 11/30/2022] Open
Abstract
Oncogenes are the primary candidates for target-directed therapy, given that they are involved directly in the progression and resistance of tumors. However, the appearance of point mutations can hinder the treatment of patients with these new molecules, raising costs and the need to development new analogs that target the novel mutations. Based on an analysis of homologies, the present study discusses the possibility of predicting the failure of a protein as a pharmacological target, due to its similarities with retrovirus sequences, which have extremely high mutation rates. This analysis was based on the molecular evidence available in the literature, and widely-used and well-established PSI-BLAST, with two iterations and maximum of 500 aligned sequences. The possibility of predicting which newly-discovered genes involved in tumor progression would likely result in the failure of targeted therapy, using free, simple and automated bioinformatics tools, could provide substantial savings in the time and financial resources needed for long-term drug development.
Collapse
Affiliation(s)
- Janaina Fernandes
- NUMPEX-BIO, Federal University of Rio de Janeiro, Duque de Caxias , Rio de Janeiro, Brazil ; Institute for Translational Research on Health and Environment in the Amazon Region - INPeTAm, Federal University of Rio de Janeiro , Rio de Janeiro, Brazil
| |
Collapse
|
35
|
Synergistic anti-tumor effect of combined inhibition of EGFR and JAK/STAT3 pathways in human ovarian cancer. Mol Cancer 2015; 14:100. [PMID: 25928246 PMCID: PMC4437681 DOI: 10.1186/s12943-015-0366-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 04/10/2015] [Indexed: 01/05/2023] Open
Abstract
Background The EGFR signaling pathway is frequently activated in human ovarian cancer and associated with poor prognosis. However, inhibition of EGFR signaling in patients with recurrent ovarian cancer has been disappointing. It remains to be addressed whether ovarian cancer patients could benefit from targeting EGFR signaling. Here we investigated the mechanisms underlying the resistance to EGFR inhibition in ovarian cancer and developed a strategy to overcome it. Results We found that treatment of human ovarian cancer cells with an EGFR inhibitor, gefitinib, resulted in increased STAT3 phosphorylation in a dose- and time-dependent manner. Inhibiting STAT3 activation with a small molecule inhibitor of JAK, an upstream kinase that phosphorylates and activates STAT3, synergistically increased the anti-tumor activity of gefitinib in vitro. Similar results were obtained when STAT3 or JAK1 expression was knocked down. In contrast, inhibiting other signaling pathways, such as AKT/mTOR, MEK or SRC, was relatively less effective. The combined treatment resulted in simultaneous attenuation of multiple survival pathways and increased inhibition of ERK pathway. In addition, the dual inhibition showed a stronger suppression of xenograft tumor growth than either single inhibition. Conclusions Our findings demonstrate that feedback activation of STAT3 pathway might contribute to the resistance to EGFR inhibition. Combined blockade of both pathways appears to be more effective against human ovarian cancer than inhibition of each pathway alone both in vitro and in vivo. This study may provide a strategy to improve clinical benefit of targeting EGFR pathway in ovarian cancer patients.
Collapse
|
36
|
An antibody to amphiregulin, an abundant growth factor in patients’ fluids, inhibits ovarian tumors. Oncogene 2015; 35:438-47. [DOI: 10.1038/onc.2015.93] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 02/03/2023]
|
37
|
Milagre CS, Gopinathan G, Everitt G, Thompson RG, Kulbe H, Zhong H, Hollingsworth RE, Grose R, Bowtell DDL, Hochhauser D, Balkwill FR. Adaptive Upregulation of EGFR Limits Attenuation of Tumor Growth by Neutralizing IL6 Antibodies, with Implications for Combined Therapy in Ovarian Cancer. Cancer Res 2015; 75:1255-64. [PMID: 25670170 PMCID: PMC4384986 DOI: 10.1158/0008-5472.can-14-1801] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 01/16/2015] [Indexed: 01/05/2023]
Abstract
Excess production of the proinflammatory IL6 has both local and systemic tumor-promoting activity in many cancers, including ovarian cancer. However, treatment of advanced ovarian cancer patients with a neutralizing IL6 antibody yielded little efficacy in a previous phase II clinical trial. Here, we report results that may explain this outcome, based on the finding that neutralizing antibodies to IL6 and STAT3 inhibition are sufficient to upregulate the EGFR pathway in high-grade serous and other ovarian cancer cells. Cell treatment with the EGFR inhibitor gefitinib abolished upregulation of the EGFR pathway. Combining neutralizing IL6 antibodies and gefitinib inhibited malignant cell growth in 2D and 3D culture. We found that ErbB-1 was localized predominantly in the nucleus of ovarian cancer cells examined, contrasting with plasma membrane localization in lung cancer cells. Treatment with anti-IL6, gefitinib, or their combination all led to partial restoration of ErbB-1 on the plasma membrane. In vivo experiments confirmed the effects of IL6 inhibition on the EGFR pathway and the enhanced activity of a combination of anti-IL6 antibodies and gefitinib on malignant cell growth. Taken together, our results offer a preclinical rationale to combine anti-IL6 and gefitinib to treat patients with advanced stage ovarian cancer.
Collapse
Affiliation(s)
- Carla S Milagre
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Ganga Gopinathan
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Gemma Everitt
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Richard G Thompson
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Hagen Kulbe
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Haihong Zhong
- MedImmune, One MedImmune Way, Gaithersburg, Maryland
| | | | - Richard Grose
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - David D L Bowtell
- Cancer Genomics and Genetics Program, Peter MacCallum Cancer Centre, Research Division, Peter MacCallum Cancer Centre, Melbourne, Australia
| | | | - Frances R Balkwill
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, United Kingdom.
| |
Collapse
|
38
|
Burotto M, Manasanch EE, Wilkerson J, Fojo T. Gefitinib and erlotinib in metastatic non-small cell lung cancer: a meta-analysis of toxicity and efficacy of randomized clinical trials. Oncologist 2015; 20:400-10. [PMID: 25795635 DOI: 10.1634/theoncologist.2014-0154] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 02/16/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Tyrosine kinase inhibitors (TKIs) targeting the epidermal growth factor receptor (EGFR) have been evaluated in patients with metastatic and advanced non-small cell lung cancer (NSCLC). The U.S. Food and Drug Administration initially granted accelerated approval to gefitinib but subsequently rescinded the authorization. Erlotinib and afatinib are similar compounds approved for the treatment of metastatic NSCLC. The objective of this study was to compare the efficacy and toxicity of erlotinib, gefitinib, and afatinib in NSCLC. METHODS We tabulated efficacy variables including overall response rate (ORR), progression-free survival (PFS), and overall survival (OS) and quantitated toxicities and rates of dose reductions and discontinuation. Summary odds ratios were calculated using random and fixed-effects models. An odds ratio was the summary measure used for pooling of studies. RESULTS We examined 28 studies including three randomized trials with afatinib. Clinical toxicities, including pruritus, rash, anorexia, diarrhea, nausea, fatigue, mucositis, paronychia, and anemia, were similar between erlotinib and gefitinib, although some statistical differences were observed. Afatinib treatment resulted in more diarrhea, rash, and paronychia compared with erlotinib and gefitinib. Regarding efficacy, similar outcomes were recorded for ORR, PFS, or OS in the total population and in specific subgroups of patients between erlotinib and gefitinib. All three TKIs demonstrated higher ORRs in first line in tumors harboring EGFR mutations. CONCLUSION Gefitinib has similar activity and toxicity compared with erlotinib and offers a valuable alternative to patients with NSCLC. Afatinib has similar efficacy compared with erlotinib and gefitinib in first-line treatment of tumors harboring EGFR mutations but may be associated with more toxicity, although further studies are needed. Gefitinib deserves consideration for U.S. marketing as a primary treatment for EGFR-mutant NSCLC.
Collapse
Affiliation(s)
- Mauricio Burotto
- Medical Oncology and Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Elisabet E Manasanch
- Medical Oncology and Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Julia Wilkerson
- Medical Oncology and Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tito Fojo
- Medical Oncology and Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA; Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
39
|
Koo DH, Kim KP, Choi CM, Lee DH, Lee JC, Lee JS, Jang SJ, Kim SW. EGFR-TKI is effective regardless of treatment timing in pulmonary adenocarcinoma with EGFR mutation. Cancer Chemother Pharmacol 2014; 75:197-206. [PMID: 25422153 DOI: 10.1007/s00280-014-2631-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 11/14/2014] [Indexed: 02/01/2023]
Abstract
PURPOSE Although epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) have become key therapeutic agents for non-small cell lung cancer (NSCLC) patients with EGFR mutation, little is known about the efficacy of EGFR-TKIs according to different treatment timings. METHODS A total of 1,250 patients with NSCLC were screened for EGFR mutations at a single institution between March 2006 and May 2010. The efficacy of EGFR-TKIs in terms of response rate (RR), progression-free survival (PFS), and overall survival (OS) were compared according to the treatment timing. RESULTS Among the 437 patients (36.1 %) with EGFR mutation, we analyzed 222 patients who received EGFR-TKI treatment. With a median follow-up duration of 27.5 months (range 8.3-69.2), EGFR-TKI was given to 97 (43.7 %), 109 (49.1 %), and 16 (7.2 %) patients as first-line, second-line, and third-line therapy, respectively. All three groups showed similar RR (71.1, 72.5, and 75.0 %, respectively) to EGFR-TKI (p = 0.802). No significant difference was observed according to treatment timing of EGFR-TKI in terms of PFS (median 10.6, 13.0, and 10.4 months; p = 0.670) and OS (median 20.5, 26.2, and 17.1 months; p = 0.142). The treatment timing of EGFR-TKI still showed no association with PFS or OS after adjusting significant prognostic factors including performance, disease status, and EGFR mutation types. CONCLUSIONS EGFR-TKIs showed similar efficacy in patients with EGFR mutation-positive adenocarcinoma in terms of RR, PFS, and OS irrespective of treatment timing. Although EGFR-TKIs are currently the treatment of choice of first-line treatment in patients with EGFR-positive tumors, the sequential treatment with EGFR-TKI could be a reasonable option when EGFR mutation status cannot be obtained in a short time.
Collapse
Affiliation(s)
- Dong-Hoe Koo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 86, Asanbyeongwon-gil, Songpa-gu, Seoul, 138-736, Korea
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Lawlor D, Martin P, Busschots S, Thery J, O'Leary JJ, Hennessy BT, Stordal B. PARP Inhibitors as P-glyoprotein Substrates. J Pharm Sci 2014; 103:1913-20. [PMID: 24700236 DOI: 10.1002/jps.23952] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 02/27/2014] [Accepted: 02/28/2014] [Indexed: 01/23/2023]
Abstract
The cytotoxicity of PARP inhibitors olaparib, veliparib, and CEP-8983 were investigated in two P-glycoprotein (P-gp) overexpressing drug-resistant cell models (IGROVCDDP and KB-8-5-11). IGROVCDDP and KB-8-5-11 were both resistant to olaparib and resistance was reversible with the P-gp inhibitors elacridar, zosuquidar, and valspodar. In contrast, the P-gp overexpressing models were not resistant to veliparib or CEP-8983. Olaparib and veliparib did not induce protein expression of P-gp in IGROVCDDP or KB-8-5-11 at doses that successfully inhibit PARP. Olaparib therefore appears to be a P-gp substrate. Veliparib and CEP-8983 do not appear to be substrates. Veliparib and CEP-8983 may therefore be more useful in combined chemotherapy regimens with P-gp substrates and may be active in platinum and taxane-resistant ovarian cancer.
Collapse
Affiliation(s)
- Denise Lawlor
- Department of Histopathology, St James' Hospital and Trinity College, Dublin, Dublin 8, Ireland
| | | | | | | | | | | | | |
Collapse
|
41
|
Niu Q, Wang W, Li Q, Li Y, Ruden DM, He B. Percutaneous Fine-Needle 5% Ethanol-Cisplatin Intratumoral Injection Combined with Second-Line Chemotherapy Improves On the Standard of Care in Patients with Platinum-Pretreated Stage IV Non-Small Cell Lung Cancer. Transl Oncol 2014; 7:303-8. [PMID: 24726235 PMCID: PMC4101294 DOI: 10.1016/j.tranon.2014.02.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 09/23/2013] [Accepted: 02/11/2014] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND: Efficacy of second-line chemotherapy in platinum-pretreated non–small cell lung cancer (NSCLC) is poor. This study investigated efficacy of computed tomography–guided percutaneous fine-needle 5% ethanol-cisplatin intratumoral injection (CT-PFNECII) combined with second-line chemotherapy in patients with platinum-pretreated stage IV NSCLC. PATIENTS: Between October 2011 and July 2013, 34 eligible patients were randomly assigned to receive either CT-PFNECII combined with second-line chemotherapy (combination group, n = 17) or second-line chemotherapy alone (chemotherapy group, n = 17). The primary end points were the proportions of patients who achieved an overall response rate (ORR) and disease control rate (DCR). Secondary end points were median survival and progression-free survival (PFS). RESULTS: The ORR and DCR in the combination group were significantly higher than in the chemotherapy group (23.53% vs 11.76% for ORR, P < .01; and 58.82% vs 35.29% for DCR, P < .01). Compared with patients in the chemotherapy group, patients in the combination group had significantly longer PFS (5.4 months vs 3.0 months, P < .01) and median survival (9.5 months vs 5.3 months, P < .01). CONCLUSIONS: CT-PFNECII combined with second-line chemotherapy provided a higher response rate and improved survival than second-line chemotherapy for patients with platinum-pretreated stage IV NSCLC.
Collapse
Affiliation(s)
- Qi Niu
- Department of Medical Oncology, No. 309 People's Liberation Army Hospital, Beijing, People's Republic of China.
| | - Wei Wang
- Department of Medical Oncology, No. 309 People's Liberation Army Hospital, Beijing, People's Republic of China
| | - Qian Li
- Department of Internal Medicine, Beijing Language and Culture University Hospital, Beijing, People's Republic of China
| | - Yong Li
- Department of Medical Oncology, No. 309 People's Liberation Army Hospital, Beijing, People's Republic of China
| | - Douglas M Ruden
- Department of Obstetrics and Gynecology, Institute of Environmental Health Sciences, CS Mott Center for Human Health and Development, Wayne State University, Detroit, MI, USA
| | - Baoming He
- Department of Radiology, No. 309 People's Liberation Army Hospital, Beijing, People's Republic of China
| |
Collapse
|
42
|
Synthetic lethal therapy for KRAS mutant non-small-cell lung carcinoma with nanoparticle-mediated CDK4 siRNA delivery. Mol Ther 2014; 22:964-73. [PMID: 24496383 DOI: 10.1038/mt.2014.18] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 12/21/2013] [Indexed: 12/19/2022] Open
Abstract
The KRAS mutation is present in ~20% of lung cancers and has not yet been effectively targeted for therapy. This mutation is associated with a poor prognosis in non-small-cell lung carcinomas (NSCLCs) and confers resistance to standard anticancer treatment drugs, including epidermal growth factor receptor tyrosine kinase inhibitors. In this study, we exploited a new therapeutic strategy based on the synthetic lethal interaction between cyclin-dependent kinase 4 (CDK4) downregulation and the KRAS mutation to deliver micellar nanoparticles (MNPs) containing small interfering RNA targeting CDK4 (MNPsiCDK4) for treatment in NSCLCs harboring the oncogenic KRAS mutation. Following MNPsiCDK4 administration, CDK4 expression was decreased, accompanied by inhibited cell proliferation, specifically in KRAS mutant NSCLCs. However, this intervention was harmless to normal KRAS wild-type cells, confirming the proposed mechanism of synthetic lethality. Moreover, systemic delivery of MNPsiCDK4 significantly inhibited tumor growth in an A549 NSCLC xenograft murine model, with depressed expression of CDK4 and mutational KRAS status, suggesting the therapeutic promise of MNPsiCDK4 delivery in KRAS mutant NSCLCs via a synthetic lethal interaction between KRAS and CDK4.
Collapse
|
43
|
Lum CT, Wai-Yin Sun R, Zou T, Che CM. Gold(iii) complexes inhibit growth of cisplatin-resistant ovarian cancer in association with upregulation of proapoptotic PMS2 gene. Chem Sci 2014. [DOI: 10.1039/c3sc53203h] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
44
|
Ge XH, Lin Q, Ren XC, Liu YE, Chen XJ, Wang DY, Wang YQ, Cao B, Li ZG, Liu ML. Phase II clinical trial of whole-brain irradiation plus three-dimensional conformal boost with concurrent topotecan for brain metastases from lung cancer. Radiat Oncol 2013; 8:238. [PMID: 24125485 PMCID: PMC3853318 DOI: 10.1186/1748-717x-8-238] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Accepted: 09/22/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Patients with brain metastases from lung cancer have poor prognoses and short survival time, and they are often excluded from clinical trials. Whole-cranial irradiation is considered to be the standard treatment, but its efficacy is not satisfactory. The purpose of this phase II clinical trial was to evaluate the preliminary efficacy and safety of the treatment of whole-brain irradiation plus three-dimensional conformal boost combined with concurrent topotecan for the patients with brain metastases from lung cancer. METHODS Patients with brain metastasis from lung cancer received concurrent chemotherapy and radiotherapy: conventional fractionated whole-brain irradiation, 2 fields/time, 1 fraction/day, 2 Gy/fraction, 5 times/week, and DT 40 Gy/20 fractions; for the patients with ≤ 3 lesions with diameter ≥ 2 cm, a three-dimensional (3-D) conformal localised boost was given to increase the dosage to 56-60 Gy; and during radiotherapy, concurrent chemotherapy with topotecan was given (the chemoradiotherapy group, CRT). The patients with brain metastasis from lung cancer during the same period who received radiotherapy only were selected as the controls (the radiotherapy-alone group, RT). RESULTS From March 2009 to March 2012, both 38 patients were enrolled into two groups. The median progression-free survival(PFS) time , the 1- and 2-year PFS rates of CRT group and RT group were 6 months, 42.8%, 21.6% and 3 months, 11.6%, 8.7% (χ2 = 6.02, p = 0.014), respectively. The 1- and 2-year intracranial lesion control rates of CRT and RT were 75.9% , 65.2% and 41.6% , 31.2% (χ2 = 3.892, p = 0.049), respectively. The 1- and 2-year overall survival rates (OS) of CRT and RT were 50.8% , 37.9% and 40.4% , 16.5% (χ2 = 1.811, p = 0.178), respectively. The major side effects were myelosuppression and digestive toxicities, but no differences were observed between the two groups. CONCLUSION Compared with radiotherapy alone, whole-brain irradiation plus 3-D conformal boost irradiation and concurrent topotecan chemotherapy significantly improved the PFS rate and the intracranial lesion control rate of patients with brain metastases from lung cancer, and no significant increases in side effects were observed. Based on these results, this treatment method is recommended for phase III clinical trial.
Collapse
Affiliation(s)
- Xiao-hui Ge
- Department of Radiation Oncology, Affiliated Hospital of Hebei University, 212 East Yuhua Avenue, Baoding, Hebei Province 071000, PR China
| | - Qiang Lin
- Department of Oncology, North China Petroleum Bureau General Hospital of Hebei Medical University, 8 Huizhan Avenue, Renqiu, Hebei Province 062552, PR China
| | - Xiao-cang Ren
- Department of Oncology, North China Petroleum Bureau General Hospital of Hebei Medical University, 8 Huizhan Avenue, Renqiu, Hebei Province 062552, PR China
| | - Yue-e Liu
- Department of Oncology, North China Petroleum Bureau General Hospital of Hebei Medical University, 8 Huizhan Avenue, Renqiu, Hebei Province 062552, PR China
| | - Xue-ji Chen
- Department of Oncology, North China Petroleum Bureau General Hospital of Hebei Medical University, 8 Huizhan Avenue, Renqiu, Hebei Province 062552, PR China
| | - Dong-ying Wang
- Department of Oncology, North China Petroleum Bureau General Hospital of Hebei Medical University, 8 Huizhan Avenue, Renqiu, Hebei Province 062552, PR China
| | - Yong-qiang Wang
- Department of Radiation Oncology, Affiliated Hospital of Hebei University, 212 East Yuhua Avenue, Baoding, Hebei Province 071000, PR China
| | - Bin Cao
- Department of Oncology, North China Petroleum Bureau General Hospital of Hebei Medical University, 8 Huizhan Avenue, Renqiu, Hebei Province 062552, PR China
| | - Zhi-gang Li
- Department of Radiation Oncology, Affiliated Hospital of Hebei University, 212 East Yuhua Avenue, Baoding, Hebei Province 071000, PR China
| | - Miao-ling Liu
- Department of Radiation Oncology, Affiliated Hospital of Hebei University, 212 East Yuhua Avenue, Baoding, Hebei Province 071000, PR China
| |
Collapse
|
45
|
Glaysher S, Bolton LM, Johnson P, Atkey N, Dyson M, Torrance C, Cree IA. Targeting EGFR and PI3K pathways in ovarian cancer. Br J Cancer 2013; 109:1786-94. [PMID: 24022196 PMCID: PMC3790180 DOI: 10.1038/bjc.2013.529] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 08/09/2013] [Accepted: 08/13/2013] [Indexed: 12/02/2022] Open
Abstract
Background: The epidermal growth factor receptor (EGFR) is expressed in ovarian cancer, but agents targeting this pathway have shown little effect as single agents. This may be due to the presence of alternative pathways, particularly activation of the PI3K/Akt/MTOR pathway. Methods: We have therefore examined the effect of inhibitors of this pathway (ZSTK474 and sirolimus) in combination with the EGFR inhibitors erlotinib and gefitinib in ovarian cancer primary cell cultures. Results: The single-agent EGFR inhibitors showed little activity, although some activity was seen with the single-agent PI3K inhibitor, ZSTK474. Combinations of ZSTK474 with EGFR inhibitors showed enhanced activity with some evidence of synergy, whereas sirolimus combinations were less active. The results were not explicable on the basis of PIK3CA mutation or amplification, or PTEN loss, although one tumour with a KRAS mutation showed resistance to EGFR inhibitors. However, there was correlation of the EGFR expression with sensitivity to EGFR and resistance to PI3K active agents, and inverse correlation in the sensitivity of individual tumours to agents active against these pathways, suggesting a mechanism of action for the combination. Conclusion: Phase I/II clinical trials with these agents should include further pharmacodynamic endpoints and molecular characterisation to identify patients most likely to benefit from this strategy.
Collapse
Affiliation(s)
- S Glaysher
- Translational Oncology Research Centre, Queen Alexandra Hospital, Portsmouth PO6 3LY, UK
| | | | | | | | | | | | | |
Collapse
|
46
|
Chen K, Huang YH, Chen JL. Understanding and targeting cancer stem cells: therapeutic implications and challenges. Acta Pharmacol Sin 2013; 34:732-40. [PMID: 23685952 PMCID: PMC3674516 DOI: 10.1038/aps.2013.27] [Citation(s) in RCA: 441] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 03/06/2013] [Indexed: 02/07/2023]
Abstract
Cancer stem cells (CSCs) have been identified as rare cell populations in many cancers, including leukemia and solid tumors. Accumulating evidence has suggested that CSCs are capable of self-renewal and differentiation into various types of cancer cells. Aberrant regulation of gene expression and some signaling pathways has been observed in CSCs compared to other tumor cells. CSCs are thought to be responsible for cancer initiation, progression, metastasis, recurrence and drug resistance. The CSC hypothesis has recently attracted much attention due to the potential for discovery and development of CSC-related therapies and the identification of key molecules involved in controlling the unique properties of CSC populations. Over the past several years, a tremendous amount of effort has been invested in the development of new drugs, such as nanomedicines, that can take advantage of the "Achilles' heel" of CSCs by targeting cell-surface molecular markers or various signaling pathways. Novel compounds and therapeutic strategies that selectively target CSCs have been identified, some of which have been evaluated in preclinical and clinical studies. In this article, we review new findings related to the investigation of the CSC hypothesis, and discuss the crucial pathways involved in regulating the development of CSC populations and the advances in studies of drug resistance. In addition, we review new CSC-targeted therapeutic strategies aiming to eradicate malignancies.
Collapse
Affiliation(s)
- Ke Chen
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing 100101, China
| | - Ying-hui Huang
- China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Ji-long Chen
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing 100101, China
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| |
Collapse
|
47
|
Marzinke MA, Choi CH, Chen L, Shih IM, Chan DW, Zhang H. Proteomic analysis of temporally stimulated ovarian cancer cells for biomarker discovery. Mol Cell Proteomics 2013; 12:356-68. [PMID: 23172893 PMCID: PMC3567859 DOI: 10.1074/mcp.m112.019521] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 11/03/2012] [Indexed: 11/06/2022] Open
Abstract
While ovarian cancer remains the most lethal gynecological malignancy in the United States, there are no biomarkers available that are able to predict therapeutic responses to ovarian malignancies. One major hurdle in the identification of useful biomarkers has been the ability to obtain enough ovarian cancer cells from primary tissues diagnosed in the early stages of serous carcinomas, the most deadly subtype of ovarian tumor. In order to detect ovarian cancer in a state of hyperproliferation, we analyzed the implications of molecular signaling cascades in the ovarian cancer cell line OVCAR3 in a temporal manner, using a mass-spectrometry-based proteomics approach. OVCAR3 cells were treated with EGF(1), and the time course of cell progression was monitored based on Akt phosphorylation and growth dynamics. EGF-stimulated Akt phosphorylation was detected at 12 h post-treatment, but an effect on proliferation was not observed until 48 h post-exposure. Growth-stimulated cellular lysates were analyzed for protein profiles between treatment groups and across time points using iTRAQ labeling and mass spectrometry. The protein response to EGF treatment was identified via iTRAQ analysis in EGF-stimulated lysates relative to vehicle-treated specimens across the treatment time course. Validation studies were performed on one of the differentially regulated proteins, lysosomal-associated membrane protein 1 (LAMP-1), in human tissue lysates and ovarian tumor tissue sections. Further, tissue microarray analysis was performed to demarcate LAMP-1 expression across different stages of epithelial ovarian cancers. These data support the use of this approach for the efficient identification of tissue-based markers in tumor development related to specific signaling pathways. LAMP-1 is a promising biomarker for studies of the progression of EGF-stimulated ovarian cancers and might be useful in predicting treatment responses involving tyrosine kinase inhibitors or EGF receptor monoclonal antibodies.
Collapse
Affiliation(s)
- Mark A. Marzinke
- From the ‡Department of Pathology, Johns Hopkins University, Baltimore, MD 21231
| | - Caitlin H. Choi
- From the ‡Department of Pathology, Johns Hopkins University, Baltimore, MD 21231
| | - Li Chen
- From the ‡Department of Pathology, Johns Hopkins University, Baltimore, MD 21231
| | - Ie-Ming Shih
- From the ‡Department of Pathology, Johns Hopkins University, Baltimore, MD 21231
| | - Daniel W. Chan
- From the ‡Department of Pathology, Johns Hopkins University, Baltimore, MD 21231
| | - Hui Zhang
- From the ‡Department of Pathology, Johns Hopkins University, Baltimore, MD 21231
| |
Collapse
|
48
|
Liu L, Shao X, Gao W, Zhang Z, Liu P, Wang R, Huang P, Yin Y, Shu Y. MicroRNA-133b inhibits the growth of non-small-cell lung cancer by targeting the epidermal growth factor receptor. FEBS J 2012; 279:3800-12. [PMID: 22883469 DOI: 10.1111/j.1742-4658.2012.08741.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 07/26/2012] [Accepted: 08/07/2012] [Indexed: 12/15/2022]
Abstract
Both the deregulation of microRNAs and epidermal growth factor receptor (EGFR) are emerging as important factors in non-small-cell lung cancer (NSCLC). Here, miR-133b was found to be associated with tumor stage, the extent of regional lymph node involvement, stage, visceral pleura or vessel invasion and EGFR mRNA expression in Chinese patients with NSCLC. Bioinformatic analysis and luciferase reporter assay revealed that miR-133b can interact specifically with the 3'-UTR of EGFR mRNA. Functionally, miR-133b transfection showed regulatory activity in translationally repressing EGFR mRNA. Moreover, miR-133b transfection may modulate apoptosis, invasion and sensitivity to EGFR-TKI through the EGFR signaling pathways, especially in EGFR-addicted NSCLC cells. Taken together, our findings show that miR-133b can inhibit cell growth of NSCLC through targeting EGFR and regulating its downstream signaling pathway. This finding has important implications for the development of targeted therapeutics for a number of EGFR-addicted cancers.
Collapse
Affiliation(s)
- Lingxiang Liu
- Cancer Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
With about 22,000 new cases estimated in 2012 in the US and 15,500 related deaths, ovarian cancer is a heterogeneous and aggressive disease. Even though most of patients are sensitive to chemotherapy treatment following surgery, recurring disease is almost always lethal, and only about 30% of the women affected will be cured. Thanks to a better understanding of the molecular mechanisms underlying ovarian cancer malignancy, new therapeutic options with molecular-targeted agents have become available. This review discusses the rationale behind molecular-targeted therapies and examines how newly identified molecular targets may enhance personalized therapies for ovarian cancer patients.
Collapse
|
50
|
Shigetomi H, Higashiura Y, Kajihara H, Kobayashi H. Targeted molecular therapies for ovarian cancer: an update and future perspectives (Review). Oncol Rep 2012; 28:395-408. [PMID: 22641286 DOI: 10.3892/or.2012.1833] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 05/03/2012] [Indexed: 11/06/2022] Open
Abstract
Identification of the potential gene expression profiles of epithelial ovarian cancer and the arrival of newly targeted therapies have advanced the strategies used for treatment of this disease. This review focuses on the design of ongoing and planned clinical trials and offers a synopsis of the English-language literature for preclinical and clinical targeted therapies for epithelial ovarian cancer. Among many targeted agents, a promising, novel class of targeted drugs for special patient populations expected to improve the effectiveness of current therapy include inhibitors of angiogenesis, poly (ADP ribose) polymerase (PARP) and DNA repair mechanisms. Inhibition of PARP or homologous recombination (HR) repair mediated by Chk1 (checkpoint kinase 1) would selectively sensitize p53 mutation, BRCAness phenotype (serous type ovarian cancer) or HNF (hepatocyte nuclear factor)-1β-overexpressing tumor cells (clear cell type ovarian cancer) to chemotherapeutic agents. The therapeutic response is likely to be limited to a targeted patient, but not to the broad population. This review discusses some of the key current developments and existing challenges.
Collapse
Affiliation(s)
- Hiroshi Shigetomi
- Department of Obstetrics and Gynecology, Nara Medical University, Nara 634-8522, Japan
| | | | | | | |
Collapse
|