1
|
Temaj G, Chichiarelli S, Telkoparan-Akillilar P, Saha S, Nuhii N, Hadziselimovic R, Saso L. P53: A key player in diverse cellular processes including nuclear stress and ribosome biogenesis, highlighting potential therapeutic compounds. Biochem Pharmacol 2024; 226:116332. [PMID: 38830426 DOI: 10.1016/j.bcp.2024.116332] [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: 01/21/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/05/2024]
Abstract
The tumor suppressor proteins are key transcription factors involved in the regulation of various cellular processes, such as apoptosis, DNA repair, cell cycle, senescence, and metabolism. The tumor suppressor protein p53 responds to different type of stress signaling, such as hypoxia, DNA damage, nutrient deprivation, oncogene activation, by activating or repressing the expression of different genes that target processes mentioned earlier. p53 has the ability to modulate the activity of many other proteins and signaling pathway through protein-protein interaction, post-translational modifications, or non-coding RNAs. In many cancers the p53 is found to be mutated or inactivated, resulting in the loss of its tumor suppressor function and acquisition of new oncogenic properties. The tumor suppressor protein p53 also plays a role in the development of other metabolic disorders such as diabetes, obesity, and fatty liver disease. In this review, we will summarize the current data and knowledge on the molecular mechanisms and the functions of p53 in different pathways and processes at the cellular level and discuss the its implications for human health and disease.
Collapse
Affiliation(s)
- Gazmend Temaj
- Faculty of Pharmacy, College UBT, 10000 Prishtina, Kosovo.
| | - Silvia Chichiarelli
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, 00185 Rome, Italy.
| | | | - Sarmistha Saha
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura 00185, Uttar Pradesh, India.
| | - Nexhibe Nuhii
- Department of Pharmacy, Faculty of Medical Sciences, State University of Tetovo, 1200 Tetovo, Macedonia.
| | - Rifat Hadziselimovic
- Faculty of Science, University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina.
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", La Sapienza University, 00185 Rome, Italy.
| |
Collapse
|
2
|
Thakur A, Rana M, Mishra A, Kaur C, Pan CH, Nepali K. Recent advances and future directions on small molecule VEGFR inhibitors in oncological conditions. Eur J Med Chem 2024; 272:116472. [PMID: 38728867 DOI: 10.1016/j.ejmech.2024.116472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/12/2024]
Abstract
"A journey of mixed emotions" is a quote that best describes the progress chart of vascular endothelial growth factor receptor (VEGFR) inhibitors as cancer therapeutics in the last decade. Exhilarated with the Food and Drug Administration (FDA) approvals of numerous VEGFR inhibitors coupled with the annoyance of encountering the complications associated with their use, drug discovery enthusiasts are on their toes with an unswerving determination to enhance the rate of translation of VEGFR inhibitors from preclinical to clinical stage. The recently crafted armory of VEGFR inhibitors is a testament to their growing dominance over other antiangiogenic therapies for cancer treatment. This review perspicuously underscores the earnest attempts of the researchers to extract the antiproliferative potential of VEGFR inhibitors through the design of mechanistically diverse structural assemblages. Moreover, this review encompasses sections on structural/molecular properties and physiological functions of VEGFR, FDA-approved VEGFR inhibitors, and hurdles restricting the activity range/clinical applicability of VEGFR targeting antitumor agents. In addition, tactics to overcome the limitations of VEGFR inhibitors are discussed. A clear-cut viewpoint transmitted through this compilation can provide practical directions to push the cart of VEGFR inhibitors to advanced-stage clinical investigations in diverse malignancies.
Collapse
Affiliation(s)
- Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Mandeep Rana
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Anshul Mishra
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Charanjit Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Chun-Hsu Pan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan.
| |
Collapse
|
3
|
Song B, Yang P, Zhang S. Cell fate regulation governed by p53: Friends or reversible foes in cancer therapy. Cancer Commun (Lond) 2024; 44:297-360. [PMID: 38311377 PMCID: PMC10958678 DOI: 10.1002/cac2.12520] [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: 07/26/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 02/10/2024] Open
Abstract
Cancer is a leading cause of death worldwide. Targeted therapies aimed at key oncogenic driver mutations in combination with chemotherapy and radiotherapy as well as immunotherapy have benefited cancer patients considerably. Tumor protein p53 (TP53), a crucial tumor suppressor gene encoding p53, regulates numerous downstream genes and cellular phenotypes in response to various stressors. The affected genes are involved in diverse processes, including cell cycle arrest, DNA repair, cellular senescence, metabolic homeostasis, apoptosis, and autophagy. However, accumulating recent studies have continued to reveal novel and unexpected functions of p53 in governing the fate of tumors, for example, functions in ferroptosis, immunity, the tumor microenvironment and microbiome metabolism. Among the possibilities, the evolutionary plasticity of p53 is the most controversial, partially due to the dizzying array of biological functions that have been attributed to different regulatory mechanisms of p53 signaling. Nearly 40 years after its discovery, this key tumor suppressor remains somewhat enigmatic. The intricate and diverse functions of p53 in regulating cell fate during cancer treatment are only the tip of the iceberg with respect to its equally complicated structural biology, which has been painstakingly revealed. Additionally, TP53 mutation is one of the most significant genetic alterations in cancer, contributing to rapid cancer cell growth and tumor progression. Here, we summarized recent advances that implicate altered p53 in modulating the response to various cancer therapies, including chemotherapy, radiotherapy, and immunotherapy. Furthermore, we also discussed potential strategies for targeting p53 as a therapeutic option for cancer.
Collapse
Affiliation(s)
- Bin Song
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
| | - Ping Yang
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
| | - Shuyu Zhang
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
- The Second Affiliated Hospital of Chengdu Medical CollegeChina National Nuclear Corporation 416 HospitalChengduSichuanP. R. China
- Laboratory of Radiation MedicineNHC Key Laboratory of Nuclear Technology Medical TransformationWest China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengduSichuanP. R. China
| |
Collapse
|
4
|
Aleksandrova Y, Neganova M. Deciphering the Mysterious Relationship between the Cross-Pathogenetic Mechanisms of Neurodegenerative and Oncological Diseases. Int J Mol Sci 2023; 24:14766. [PMID: 37834214 PMCID: PMC10573395 DOI: 10.3390/ijms241914766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/22/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
The relationship between oncological pathologies and neurodegenerative disorders is extremely complex and is a topic of concern among a growing number of researchers around the world. In recent years, convincing scientific evidence has accumulated that indicates the contribution of a number of etiological factors and pathophysiological processes to the pathogenesis of these two fundamentally different diseases, thus demonstrating an intriguing relationship between oncology and neurodegeneration. In this review, we establish the general links between three intersecting aspects of oncological pathologies and neurodegenerative disorders, i.e., oxidative stress, epigenetic dysregulation, and metabolic dysfunction, examining each process in detail to establish an unusual epidemiological relationship. We also focus on reviewing the current trends in the research and the clinical application of the most promising chemical structures and therapeutic platforms that have a modulating effect on the above processes. Thus, our comprehensive analysis of the set of molecular determinants that have obvious cross-functional pathways in the pathogenesis of oncological and neurodegenerative diseases can help in the creation of advanced diagnostic tools and in the development of innovative pharmacological strategies.
Collapse
Affiliation(s)
- Yulia Aleksandrova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Margarita Neganova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 420088 Kazan, Russia
| |
Collapse
|
5
|
Yueyang M, Yaqin H, Guolian X, Wenjian Z, Yang J, Chen L, Haiyan C, Min C, Jianping D, Penggao D, Hongli Z, Liang W. Glioma angiogenesis is boosted by ELK3 activating the HIF-1
α
/VEGF-A signaling axis. BMC Cancer 2023; 23:662. [PMID: 37452291 PMCID: PMC10347878 DOI: 10.1186/s12885-023-11069-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/14/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND Clinical studies have shown that first-line use of anti-angiogenetic therapy can prolong progression-free survival but little progress has been made in extending the overall survival of the patients. We explored the role of ELK3 in glioma angiogenesis to improve and design more efficacious therapies. METHODS A tissue microarray and immunohistochemistry analysis were used to determine the expression of ELK3 protein in 400 glioma patients. Cell proliferation, metastasis, cell cycle, and apoptosis were monitored in U87 and U251 cells using CCK-8, EdU, transwell assays, and flow cytometry. A tube-formation assay, a rat aorta ring sprouting assay, and a matrigel plug assay were performed to examine the antiangiogenic activity of ELK3. An ELISA, Western blot, and correlation analysis of the CGGA dataset were used to detect the association between ELK3 and VEGF-A or ELK3 and HIF-1α . Besides, orthotopic transplantation in nude mice and histopathological and immunological analysis of in vitro tumors were used to explore the effect of ELK3 on tumor progression and median survival. RESULTS ELK3 was upregulated in glioma tissues and associated with a poor prognosis. In vitro, ELK3 promoted cell proliferation and cell cycle progression, induced metastasis, and suppressed apoptosis. Then, silencing ELK3 inhibited VEGF-A expression and secretion by facilitating HIF-1α degradation via ubiquitination. Finally, knockdown ELK3 inhibited tumor progression and angiogenesis in vitro and in vivo, as well as prolonged nude mice's median survival. CONCLUSIONS Our findings first evidenced that ELK3 is crucial for glioma because it promotes angiogenesis by activating the HIF-1α /VEGF-A signaling axis. Therefore, we suggest that ELK3 is a prognostic marker with a great potential for glioma angiogenesis and ELK3-targeted therapeutic strategies might hold promise in improving the efficacy of anti-angiogenic therapies.
Collapse
Affiliation(s)
- Mou Yueyang
- College of Life Sciences, Northwest University, Xi’an, China
- Departments of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Hu Yaqin
- College of Life Sciences, Northwest University, Xi’an, China
| | - Xue Guolian
- College of Life Sciences, Northwest University, Xi’an, China
| | - Zhao Wenjian
- Departments of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Jiao Yang
- Departments of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Li Chen
- Departments of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Cao Haiyan
- Departments of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Chao Min
- Departments of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Deng Jianping
- Departments of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi’an, China
| | - Dai Penggao
- College of Life Sciences, Northwest University, Xi’an, China
| | - Zhu Hongli
- College of Life Sciences, Northwest University, Xi’an, China
| | - Wang Liang
- Departments of Neurosurgery, Tangdu Hospital, Air Force Medical University, Xi’an, China
| |
Collapse
|
6
|
Wang J, Li X, Zhou J, Qiu D, Zhang M, Sun L, Li SC. Long-term survival with anlotinib as a front-line treatment in an elderly NSCLC patient: A case report. Front Oncol 2023; 13:1043244. [PMID: 37091182 PMCID: PMC10117841 DOI: 10.3389/fonc.2023.1043244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 03/13/2023] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND Half of the population of non-small cell lung cancer (NSCLC) patients are older than 70 years and have limited therapeutic options due to poor tolerance and being excluded in most clinical trials. Anlotinib hydrochloride, a novel oral multi-target tyrosine kinase inhibitor, has been approved for the standard third-line treatment for NSCLC in China. Herein we report an elderly NSCLC patient without any driver gene mutations who was undergoing anlotinib as a front-line treatment and who achieved long-term survival. CASE SUMMARY The 77-year-old male patient was admitted to the hospital for chest tightness after engaging in physical activity for a week. The patient has been diagnosed with stage IIIB driver gene-negative squamous cell lung carcinoma. After that, he was treated with anlotinib for 2 years and 10 months from the first diagnosis until the last disease progression. Briefly, anlotinib combined with platinum-based chemotherapy was performed as the first-line therapy over six cycles. After 6 more cycles of anlotinib monotherapy maintenance, disease progression occurred. Then, anlotinib combined with tegafur was administered as a salvage treatment, and the disease was controlled again. After 29 cycles of anlotinib combined with tegafur regimens, the disease progressed finally. The patient achieved a total of 34 months of progression-free survival after anlotinib was used as the front-line treatment. He is still alive with a good performance status now (performance status score: 1). CONCLUSION This patient achieved long-term survival using anlotinib as a front-line regimen combined with chemotherapy.
Collapse
Affiliation(s)
- Jingyi Wang
- Department of Oncology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoqing Li
- Department of Oncology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Juan Zhou
- Department of Oncology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Qiu
- Department of Oncology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Mengyao Zhang
- Department of Oncology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Lan Sun
- Department of Oncology, Bishan Hospital of Chongqing Medical University, Chongqing, China
| | - Shengwen Calvin Li
- Neuro-Oncology and Stem Cell Research Laboratory, Center for Neuroscience Research, CHOC Children’s Research Institute, Children’s Hospital of Orange County (CHOC), Orange, CA, United States
- Department of Neurology, University of California-Irvine School of Medicine, Orange, CA, United States
| |
Collapse
|
7
|
Seneviratne JA, Carter DR, Mittra R, Gifford A, Kim PY, Luo J, Mayoh C, Salib A, Rahmanto AS, Murray J, Cheng NC, Nagy Z, Wang Q, Kleynhans A, Tan O, Sutton SK, Xue C, Chung SA, Zhang Y, Sun C, Zhang L, Haber M, Norris MD, Fletcher JI, Liu T, Dilda PJ, Hogg PJ, Cheung BB, Marshall GM. Inhibition of mitochondrial translocase SLC25A5 and histone deacetylation is an effective combination therapy in neuroblastoma. Int J Cancer 2023; 152:1399-1413. [PMID: 36346110 PMCID: PMC10953412 DOI: 10.1002/ijc.34349] [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: 02/16/2022] [Revised: 08/08/2022] [Accepted: 09/26/2022] [Indexed: 11/11/2022]
Abstract
The mitochondrion is a gatekeeper of apoptotic processes, and mediates drug resistance to several chemotherapy agents used to treat cancer. Neuroblastoma is a common solid cancer in young children with poor clinical outcomes following conventional chemotherapy. We sought druggable mitochondrial protein targets in neuroblastoma cells. Among mitochondria-associated gene targets, we found that high expression of the mitochondrial adenine nucleotide translocase 2 (SLC25A5/ANT2), was a strong predictor of poor neuroblastoma patient prognosis and contributed to a more malignant phenotype in pre-clinical models. Inhibiting this transporter with PENAO reduced cell viability in a panel of neuroblastoma cell lines in a TP53-status-dependant manner. We identified the histone deacetylase inhibitor, suberanilohydroxamic acid (SAHA), as the most effective drug in clinical use against mutant TP53 neuroblastoma cells. SAHA and PENAO synergistically reduced cell viability, and induced apoptosis, in neuroblastoma cells independent of TP53-status. The SAHA and PENAO drug combination significantly delayed tumour progression in pre-clinical neuroblastoma mouse models, suggesting that these clinically advanced inhibitors may be effective in treating the disease.
Collapse
Affiliation(s)
- Janith A. Seneviratne
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
| | - Daniel R. Carter
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
- School of Biomedical EngineeringUniversity of Technology SydneyNew South WalesAustralia
| | - Rituparna Mittra
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Andrew Gifford
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- Kids Cancer CentreSydney Children's HospitalRandwickNew South WalesAustralia
| | - Patrick Y. Kim
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Jie‐Si Luo
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- Department of PaediatricsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Chelsea Mayoh
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
| | - Alice Salib
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
| | - Aldwin S. Rahmanto
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Jayne Murray
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Ngan C. Cheng
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Zsuzsanna Nagy
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Qian Wang
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Ane Kleynhans
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
| | - Owen Tan
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Selina K. Sutton
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Chengyuan Xue
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Sylvia A. Chung
- Adult Cancer Program, Lowy Cancer Research CentreUNSW SydneyNew South WalesAustralia
| | - Yizhuo Zhang
- Department of PaediatricsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
- Department of Paediatric OncologySun Yat‐sen University Cancer CentreGuangzhouGuangdongChina
| | - Chengtao Sun
- Department of PaediatricsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
- Department of Paediatric OncologySun Yat‐sen University Cancer CentreGuangzhouGuangdongChina
| | - Li Zhang
- Department of PaediatricsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
- Department of Paediatric OncologySun Yat‐sen University Cancer CentreGuangzhouGuangdongChina
| | - Michelle Haber
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
| | - Murray D. Norris
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- University of New South WalesCentre for Childhood Cancer ResearchRandwickNew South WalesAustralia
| | - Jamie I. Fletcher
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
| | - Tao Liu
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
| | - Pierre J. Dilda
- Adult Cancer Program, Lowy Cancer Research CentreUNSW SydneyNew South WalesAustralia
| | - Philip J. Hogg
- Australian Cancer Research Foundation (ACRF), Centenary Cancer Research Centre, Charles Perkins CentreUniversity of SydneyNew South WalesAustralia
| | - Belamy B. Cheung
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
- Department of PaediatricsThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Glenn M. Marshall
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research CentreUNSW SydneyKensingtonNew South WalesAustralia
- School of Women's & Children's HealthUNSW SydneyNew South WalesAustralia
- Kids Cancer CentreSydney Children's HospitalRandwickNew South WalesAustralia
| |
Collapse
|
8
|
Nishikawa S, Iwakuma T. Drugs Targeting p53 Mutations with FDA Approval and in Clinical Trials. Cancers (Basel) 2023; 15:429. [PMID: 36672377 PMCID: PMC9856662 DOI: 10.3390/cancers15020429] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/01/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Mutations in the tumor suppressor p53 (p53) promote cancer progression. This is mainly due to loss of function (LOS) as a tumor suppressor, dominant-negative (DN) activities of missense mutant p53 (mutp53) over wild-type p53 (wtp53), and wtp53-independent oncogenic activities of missense mutp53 by interacting with other tumor suppressors or oncogenes (gain of function: GOF). Since p53 mutations occur in ~50% of human cancers and rarely occur in normal tissues, p53 mutations are cancer-specific and ideal therapeutic targets. Approaches to target p53 mutations include (1) restoration or stabilization of wtp53 conformation from missense mutp53, (2) rescue of p53 nonsense mutations, (3) depletion or degradation of mutp53 proteins, and (4) induction of p53 synthetic lethality or targeting of vulnerabilities imposed by p53 mutations (enhanced YAP/TAZ activities) or deletions (hyperactivated retrotransposons). This review article focuses on clinically available FDA-approved drugs and drugs in clinical trials that target p53 mutations and summarizes their mechanisms of action and activities to suppress cancer progression.
Collapse
Affiliation(s)
- Shigeto Nishikawa
- Department of Pediatrics, Division of Hematology & Oncology, Children’s Mercy Research Institute, Kansas City, MO 64108, USA
| | - Tomoo Iwakuma
- Department of Pediatrics, Division of Hematology & Oncology, Children’s Mercy Research Institute, Kansas City, MO 64108, USA
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
9
|
Molina-Cerrillo J, Santoni M, Ruiz Á, Massari F, Pozas J, Ortego I, Gómez V, Grande E, Alonso-Gordoa T. Epigenetics in advanced renal cell carcinoma: Potential new targets. Crit Rev Oncol Hematol 2022; 180:103857. [DOI: 10.1016/j.critrevonc.2022.103857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/06/2022] [Accepted: 10/12/2022] [Indexed: 11/05/2022] Open
|
10
|
Recent progress on vascular endothelial growth factor receptor inhibitors with dual targeting capabilities for tumor therapy. J Hematol Oncol 2022; 15:89. [PMID: 35799213 PMCID: PMC9263050 DOI: 10.1186/s13045-022-01310-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/30/2022] [Indexed: 02/08/2023] Open
Abstract
Vascular endothelial growth factor receptors (VEGFRs) are a family of receptor protein tyrosine kinases that play an important role in the regulation of tumor-induced angiogenesis. Currently, VEGFR inhibitors have been widely used in the treatment of various tumors. However, current VEGFR inhibitors are limited to a certain extent due to limited clinical efficacy and potential toxicity, which hinder their clinical application. Thus, the development of new strategies to improve the clinical outcomes and minimize the toxic effects of VEGFR inhibitors is required. Given the synergistic effect of VEGFR and other therapies in tumor development and progression, VEGFR dual-target inhibitors are becoming an attractive approach due to their favorable pharmacodynamics, low toxicity, and anti-resistant effects. This perspective provides an overview of the development of VEGFR dual-target inhibitors from multiple aspects, including rational target combinations, drug discovery strategies, structure–activity relationships and future directions.
Collapse
|
11
|
Clinical characteristics and outcomes of phase I cancer patients with CCNE1 amplification: MD Anderson experiences. Sci Rep 2022; 12:8701. [PMID: 35610322 PMCID: PMC9130298 DOI: 10.1038/s41598-022-12669-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 05/13/2022] [Indexed: 11/25/2022] Open
Abstract
Cyclin E is frequently encoded by CCNE1 gene amplification in various malignancies. We reviewed the medical records of patients with solid tumors displaying CCNE1 amplification to determine the effect of this amplification for future therapeutic development. We reviewed the medical records of patients with advanced solid tumors harboring CCNE1 amplification who were seen at the phase I clinic between September 1, 2012, and December 31, 2019. Among 79 patients with solid tumors harboring CCNE1 amplification, 56 (71%) received phase 1 clinical trial therapy, 39 (49%) had 3 or more concurrent genomic aberrances, and 52 (66%) had a concurrent TP53 mutation. The median overall survival (OS) after patients’ initial phase I visit was 8.9 months and after their initial metastasis diagnosis was 41.4 months. We identified four factors associated with poor risk: age < 45 years, body mass index ≥ 25 kg/m2, presence of the TP53 mutation, and elevated LDH > upper limit of normal. In patients treated with gene aberration-related therapy, anti-angiogenic therapy led to significantly longer OS after their initial phase I trial therapy than those who did not: 26 months versus 7.4 months, respectively (P = 0.04). This study provided preliminary evidence that CCNE1 amplification was associated with frequent TP53 mutation and aggressive clinical outcomes. Survival benefit was observed in patients who received antiangiogenic therapy and gene aberration-related treatment, supporting the future development of a personalized approach to combine gene aberration-related therapy with antiangiogenesis for the treatment of advanced malignancies harboring CCNE1 amplification.
Collapse
|
12
|
Su R, Wu X, Tao L, Wang C. The role of epigenetic modifications in Colorectal Cancer Metastasis. Clin Exp Metastasis 2022; 39:521-539. [PMID: 35429301 PMCID: PMC9338907 DOI: 10.1007/s10585-022-10163-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/18/2022] [Indexed: 12/19/2022]
Abstract
Distant metastasis is the major contributor to the high mortality rate of colorectal cancer (CRC). To overcome the poor prognosis caused by distant metastasis, the mechanisms of CRC metastasis should be further explored. Epigenetic events are the main mediators of gene regulation and further affect tumor progression. Recent studies have found that some epigenetic enzymes are often dysregulated or mutated in multiple tumor types, which prompted us to study the roles of these enzymes in CRC metastasis. In this review, we summarized the alteration of enzymes related to various modifications, including histone modification, nonhistone modification, DNA methylation, and RNA methylation, and their epigenetic mechanisms during the progression of CRC metastasis. Existing data suggest that targeting epigenetic enzymes is a promising strategy for the treatment of CRC metastasis.
Collapse
Affiliation(s)
- Riya Su
- Department of pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xinlin Wu
- Department of General Surgery, the Affiliated Hospital of Inner Mongolia Medical University, Huhhot, China
| | - Liang Tao
- Department of pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Changshan Wang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China.
| |
Collapse
|
13
|
Lanzi C, Cassinelli G. Combinatorial strategies to potentiate the efficacy of HDAC inhibitors in fusion-positive sarcomas. Biochem Pharmacol 2022; 198:114944. [DOI: 10.1016/j.bcp.2022.114944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/02/2022] [Accepted: 02/02/2022] [Indexed: 12/12/2022]
|
14
|
Zhang DY, Su L, Wang YW. Malignant solitary fibrous tumor in the central nervous system treated with surgery, radiotherapy and anlotinib: A case report. World J Clin Cases 2022; 10:631-642. [PMID: 35097089 PMCID: PMC8771389 DOI: 10.12998/wjcc.v10.i2.631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 11/01/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Solitary fibrous tumor (SFT) of the central nervous system is rare. It is predominantly benign and rarely malignant. There is no established standardized treatment regimen for malignant intracranial SFTs.
CASE SUMMARY We present a rare case of SFT in a 9-year-old girl with a space-occupying effect in the frontal-parietal lobes. She underwent craniotomy, and the mass was resected. Immunohistochemistry examination of the specimen showed that Ki-67 proliferation index staining was highly positive in 80% of tumor cells. Whole exome sequencing of the surgical tissue showed 38 somatic gene mutations and 1 gene amplification such as fibroblast growth factor receptor 4 or TP53. At 1.5 mo after surgery, head magnetic resonance imaging revealed that the tumor had recurred. The patient received 60 Gy and 30 fractions of intensity modulated radiotherapy. The patient then received anlotinib 8 mg po qd for 1-14 d of a 21 d cycle. Following this regimen, the patient achieved stable disease for > 17 mo. Magnetic resonance imaging at 1.5 year after surgery showed that the tumor had not progressed.
CONCLUSION This is the first reported case of SFT of the central nervous system treated with surgery, radiotherapy and anlotinib. This regimen may be an effective treatment option for malignant intracranial SFT patients.
Collapse
Affiliation(s)
- Dong-Yong Zhang
- Department of Neurosurgery, the First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Lan Su
- Department of Genetron Health, Genetron Health, Beijing 110024, China
| | - Yi-Wei Wang
- Department of Anatomy, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| |
Collapse
|
15
|
Xu H, Liang Q, Xu X, Tan S, Wang S, Liu Y, Liu L. Afatinib combined with anlotinib in the treatment of lung adenocarcinoma patient with novel HER2 mutation: a case report and review of the literature. World J Surg Oncol 2021; 19:330. [PMID: 34794435 PMCID: PMC8600784 DOI: 10.1186/s12957-021-02444-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 11/07/2021] [Indexed: 12/25/2022] Open
Abstract
Background HER2 is a member of the ERBB family of receptor tyrosine kinases, and HER2 mutations occur in 1–4% of non-small cell lung cancer (NSCLC) as an oncogenic driver mutation. We found a rare mutation of HER2 p.Asp769Tyr in NSCLC. Case presentation We presented a case of a 68-year-old nonsmoking male patient with brain metastasis from lung adenocarcinoma harboring a rare mutation of HER2 p.Asp769Tyr. After multiple lines of treatment, he obtained a durable response (10 months) to afatinib and anlotinib. Conclusion We reported for the first time that afatinib and anlotinib have successfully treated lung adenocarcinoma with HER2 p.Asp769Tyr mutation. This finding can provide an insight into the optimal treatment of lung adenocarcinoma patients with novel mutations. Additionally, we summarized the efficacy of targeted therapy for HER2 mutant lung cancer in this article. Supplementary Information The online version contains supplementary material available at 10.1186/s12957-021-02444-7.
Collapse
Affiliation(s)
- Huanhuan Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Qi Liang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Xian Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Shanyue Tan
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Sumeng Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Yiqian Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China.
| | - Lingxiang Liu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, People's Republic of China.
| |
Collapse
|
16
|
Nassif EF, Auclin E, Bahleda R, Honoré C, Mir O, Dumont S, Mery B, Hodroj K, Brahmi M, Trédan O, Ray-Coquard I, Blay JY, Massard C, Le Cesne A, Dufresne A. TP53 Mutation as a Prognostic and Predictive Marker in Sarcoma: Pooled Analysis of MOSCATO and ProfiLER Precision Medicine Trials. Cancers (Basel) 2021; 13:3362. [PMID: 34282771 PMCID: PMC8268242 DOI: 10.3390/cancers13133362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/23/2021] [Accepted: 07/02/2021] [Indexed: 11/16/2022] Open
Abstract
(1) Background: locally resected high-grade sarcomas relapse in 40% of cases. There is no prognostic or predictive genomic marker for response to peri-operative chemotherapy. (2) Methods: MOSCATO and ProfiLER are pan-tumor prospective precision medicine trials for advanced tumors. Molecular analysis in both trials comprised targeted next-generation sequencing and comparative genomic hybridization array. We investigated if molecular alterations identified in these trials in sarcomas were associated with disease-free survival (DFS) and response to anthracyclines. (3) Results: this analysis included 215 sarcomas, amongst which 53 leiomyosarcomas, 27 rhabdomyosarcomas, 20 undifferentiated pleomorphic sarcomas, and 17 liposarcomas. The most frequently altered gene was TP53 (46 mutations and eight deletions). There were 149 surgically resected localized sarcomas. Median DFS in TP53 wild type (WT), deleted, and mutated sarcomas was 16, 10, and 10 months, respectively (p = 0.028; deletions: HR = 1.55; 95% CI = 0.75-3.19; mutations: HR = 1.70; 95%CI = 1.13-2.64). In multivariate analysis, TP53 mutations remained associated with shorter DFS (p = 0.027; HR = 2.30; 95%CI = 1.10-4.82). There were 161 localized and advanced sarcomas evaluable for response to anthracyclines. Objective response rates were 35% and 55% in TP53 WT and mutated sarcomas, respectively (OR = 2.24; 95%CI = 1.01-5.03; p = 0.05). In multivariate analysis, TP53 mutations remained associated with increased response (OR = 3.24; 95%CI = 1.30-8.45; p = 0.01). (4) Conclusions: TP53 mutations are associated with shorter DFS and increased response to anthracyclines. Post-validation, these findings could assist in decision-making for peri-operative treatments.
Collapse
Affiliation(s)
- Elise F. Nassif
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Edouard Auclin
- Oncology Department, Hopital Européen Georges Pompidou, 75015 Paris, France;
| | - Rastilav Bahleda
- DITEP (Département d’Innovation Therapeutique et d’Essais Précoces), Drug Development Department, Gustave Roussy, 94805 Villejuif, France; (R.B.); (C.M.)
| | - Charles Honoré
- Surgical Oncology Department, Gustave Roussy, 94805 Villejuif, France;
| | - Olivier Mir
- Ambulatory Cancer Care Department, Gustave Roussy, 94805 Villejuif, France;
| | - Sarah Dumont
- Medical Oncology Department, Gustave Roussy, 94805 Villejuif, France;
| | - Benoite Mery
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Khalil Hodroj
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Mehdi Brahmi
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Olivier Trédan
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Isabelle Ray-Coquard
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Jean-Yves Blay
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| | - Christophe Massard
- DITEP (Département d’Innovation Therapeutique et d’Essais Précoces), Drug Development Department, Gustave Roussy, 94805 Villejuif, France; (R.B.); (C.M.)
| | - Axel Le Cesne
- Medical Oncology Department, Gustave Roussy, 94805 Villejuif, France;
| | - Armelle Dufresne
- Centre Léon Bérard, Medical Oncology Department, 69008 Lyon, France; (E.F.N.); (B.M.); (K.H.); (M.B.); (O.T.); (I.R.-C.); (J.-Y.B.); (A.D.)
| |
Collapse
|
17
|
Bordonaro M. Hypothesis: Sam68 and Pygo2 mediate cell type-specific effects of the modulation of CBP-Wnt and p300-Wnt activities in Colorectal Cancer Cells. J Cancer 2021; 12:5046-5052. [PMID: 34234873 PMCID: PMC8247382 DOI: 10.7150/jca.59726] [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: 02/23/2021] [Accepted: 06/07/2021] [Indexed: 11/08/2022] Open
Abstract
The preventive activity of dietary fiber against colorectal cancer (CRC) may be in part mediated by the fermentation product of fiber, butyrate, a histone deacetylase inhibitor (HDACi) that induces CRC cell growth arrest and apoptosis. This action of butyrate, and other HDACis, is in part due to the hyperactivation of the deregulated Wnt activity found in the relevant CRC cell lines. The histone acetylases CBP and p300 interact with beta-catenin; and the relative levels of CBP-Wnt vs. p300-Wnt activity influences CRC cell physiology. It has previously been observed that there are cell type-specific differences in how cotreatment with butyrate and ICG-001, an agent that blocks CBP-Wnt activity allowing for p300-Wnt activity, affects CRC cell physiology. These differences may have clinical significance in dealing with treatment of CRC patients with ICG-001-like agents. Sam68 is a factor differentially expressed in cancer cells, with higher expression in cancer cell lines that have cancer stem cell (CSC)-like properties. Sam68 expression sensitizes cancer cells to ICG-001 treatment, as ICG-001 enhances nuclear localization of Sam68, where binding between Sam68 and CBP diminishes CBP-beta-catenin binding and thus CBP-Wnt activity. Pygo2 is a chromatin effector involved with Wnt signaling that is differentially acetylated by CBP and p300; thus CBP-mediated acetylation localized Pygo2 to the nucleus where it functions in transcriptional activation, while p300-mediated acetylation localizes Pygo2 to the cytoplasm. This paper proposes the hypothesis that Sam68 and Pygo2 are responsible for cell type-specific response of CRC cell lines cotreated with ICG-001 and butyrate as well as other HDACis. Further, experiments are proposed to evaluate this hypothesis and consider possible expected results that could be obtained from such studies.
Collapse
Affiliation(s)
- Michael Bordonaro
- Department of Medical Education, Geisinger Commonwealth School of Medicine, 525 Pine Street, Scranton, PA 18509, USA
| |
Collapse
|
18
|
Li Q, Zhang Z, Fan Y, Zhang Q. Epigenetic Alterations in Renal Cell Cancer With TKIs Resistance: From Mechanisms to Clinical Applications. Front Genet 2021; 11:562868. [PMID: 33510766 PMCID: PMC7835797 DOI: 10.3389/fgene.2020.562868] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
The appearance of tyrosine kinase inhibitors (TKIs) has been a major breakthrough in renal cell carcinoma (RCC) therapy. Unfortunately, a portion of patients with TKIs resistance experience disease progression after TKIs therapy. Epigenetic alterations play an important role in the development of TKIs resistance. Current evidence suggests that epigenetic alterations occur frequently in RCC patients with poor response to TKIs therapy, and modulation of them could enhance the cytotoxic effect of antitumor therapy. In this review, we summarize the currently known epigenetic alterations relating to TKIs resistance in RCC, focusing on DNA methylation, non-coding RNAs (ncRNAs), histone modifications, and their interactions with TKIs treatment. In addition, we discuss application of epigenetic alteration analyses in the clinical setting to predict prognosis of patients with TKIs treatment, and the potential use of epigenetics-based therapies to surmount TKIs resistance.
Collapse
Affiliation(s)
- Qinhan Li
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Zhenan Zhang
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Yu Fan
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| | - Qian Zhang
- Department of Urology, Peking University First Hospital, Institute of Urology, National Research Center for Genitourinary Oncology, Peking University, Beijing, China
| |
Collapse
|
19
|
Halim NA, Sayed RE, Alameh IA, Khoury J, Nakib CE, Zerdan MB, Charafeddine M, Farhat F, Karak FE, Assi HI. Safety and efficacy of pazopanib as a second-line treatment and beyond for soft tissue sarcomas: A real-life tertiary-center experience in the MENA region. Cancer Treat Res Commun 2020; 26:100275. [PMID: 33340905 DOI: 10.1016/j.ctarc.2020.100275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/27/2020] [Accepted: 12/09/2020] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Sarcomas are uncommon malignancies. No advances have been recently achieved despite multiple efforts. Pazopanib is a safe and effective tyrosine kinase inhibitor used in managing soft tissue sarcomas (STS) after chemotherapy failure. However, its use is limited in developing countries and no efficacy data exist from our region. We aimed to study the efficacy of pazopanib in our population, characterized by response rates of patients with chemotherapy-refractory advanced STS receiving pazopanib. Secondary endpoints included progression-free survival (PFS), overall survival (OS) and toxicity profile. MATERIALS AND METHODS 15 patients (age≥18 year) diagnosed with advanced STS, refractory to first-line chemotherapy, receiving pazopanib as ≥second-line therapy in one tertiary center in Lebanon were included between January 1st, 2014 and October 31st, 2018. Patient and disease characteristics, disease evaluation, as well as tolerance to treatment, were extracted from charts retrospectively. Statistical analysis was done using SPSS version 24. RESULTS The mean age was 48.6 [19-66] years. Eleven patients (73.3%) received pazopanib in second-line, whereas four patients (26.7%) received it in third-line. Thirteen patients (86.7%) progressed, and two patients (13.3%) had stable disease. The median PFS was three months [1-19] and the mean OS was 25.4 months [17.2-33.6]. Five patients required dose-reductions due to poor tolerance. CONCLUSION Conclusions cannot be drawn due to small patient numbers. However, given the 3-month PFS, 13% of patients maintaining stable disease, and tolerable safety profile, it is reasonable to incorporate pazopanib in STS treatment. More focused studies with larger patient populations need to be done in Lebanon.
Collapse
Affiliation(s)
- Nour Abdul Halim
- Department of Internal Medicine, Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Rola El Sayed
- Department of Internal Medicine, Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ibrahim A Alameh
- Department of Internal Medicine, Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Jessica Khoury
- Department of Internal Medicine, Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Clara El Nakib
- Department of Internal Medicine, Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Maroun Bou Zerdan
- Department of Internal Medicine, Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Maya Charafeddine
- Department of Internal Medicine, Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Fadi Farhat
- Department of Internal Medicine, Division of Hematology and Oncology, Hammoud Hospital University Medical Center, Saida, Lebanon
| | - Fadi El Karak
- Department of Internal Medicine, Division of Hematology and Oncology, Hotel Dieu de France University Hospital, Beirut, Lebanon
| | - Hazem I Assi
- Department of Internal Medicine, Division of Hematology and Oncology, Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon.
| |
Collapse
|
20
|
Zhu G, Zhao M, Han Q, Tan Y, Sun YU, Bouvet M, Singh SR, Ye J, Hoffman RM. Pazopanib Inhibits Tumor Growth, Lymph-node Metastasis and Lymphangiogenesis of an Orthotopic Mouse of Colorectal Cancer. Cancer Genomics Proteomics 2020; 17:131-139. [PMID: 32108035 DOI: 10.21873/cgp.20173] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 11/27/2019] [Accepted: 12/03/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND/AIM Pazopanib (PAZ) can inhibit tumor progression, but whether PAZ inhibits lymph node metastasis and lymphangiogenesis in colorectal cancer is still unknown. The aim of the present study was to determine the efficacy of PAZ on tumor growth, lymph node metastasis and lymphangiogenesis in an orthotopic nude mouse model in colorectal cancer. MATERIALS AND METHODS CT-26-green fluorescence protein (GFP)-expressing mouse colon cancer cells were injected into nude mice to establish a subcutaneous colorectal cancer model and were treated with saline and PAZ. Additionals subcutaneous tumors were harvested and cut into 5 mm3 fragments, then tumor fragments were implanted orthotopically in the cecum to establish an orthotopic colorectal-cancer nude mouse model. Orthotopic mice were randomized into two groups for the treatment with saline and PAZ, respectively. Tumor width, length and mouse body weight was measured twice a week. The Fluor Vivo imaging system was used to image the GFP. Hematoxylin & eosin staining and immunohistochemical staining was used for histological analysis. RESULTS PAZ inhibited the growth of subcutaneous colorectal cancer, as wells as orthotopic transplanted colorectal cancer tumors. PAZ suppressed lymph node metastasis and lymphangiogenesis in the orthotopic colon cancer model. No significant changes were observed in the body weight between the control and the mice treated with PAZ. CONCLUSION PAZ can inhibit the growth of colorectal cancer and inhibit lymph node metastasis and lymphangiogenesis in orthotopic colon cancer nude mouse models.
Collapse
Affiliation(s)
- Guangwei Zhu
- AntiCancer, Inc., San Diego, CA, U.S.A.,Department of Surgery, University of California, San Diego, CA, U.S.A.,Department of Gastrointestinal Surgery 2 Section, The First Hospital Affiliated to Fujian Medical University, Fuzhou, P.R. China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, P.R. China
| | - Ming Zhao
- AntiCancer, Inc., San Diego, CA, U.S.A
| | | | | | - Y U Sun
- AntiCancer, Inc., San Diego, CA, U.S.A.,Department of Surgery, University of California, San Diego, CA, U.S.A
| | - Michael Bouvet
- Department of Surgery, University of California, San Diego, CA, U.S.A
| | - Shree Ram Singh
- Basic Research Laboratory, National Cancer Institute, Frederick, MD, U.S.A.
| | - Jianxin Ye
- Department of Gastrointestinal Surgery 2 Section, The First Hospital Affiliated to Fujian Medical University, Fuzhou, P.R. China .,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, P.R. China
| | - Robert M Hoffman
- AntiCancer, Inc., San Diego, CA, U.S.A. .,Department of Surgery, University of California, San Diego, CA, U.S.A
| |
Collapse
|
21
|
Fang S, Cheng W, Zhang M, Yang R. Association of TP53 Mutations with Response to Anlotinib Treatment in Advanced Non-Small Cell Lung Cancer. Onco Targets Ther 2020; 13:6645-6650. [PMID: 32753898 PMCID: PMC7354100 DOI: 10.2147/ott.s257052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/15/2020] [Indexed: 12/24/2022] Open
Abstract
Multitargeted antiangiogenic drugs have demonstrated significant antitumor activity against a variety of solid tumors. Anlotinib, a novel oral multitargeted antiangiogenic tyrosine kinase inhibitor, was approved as a third-line treatment for advanced NSCLC in China. However, predictive biomarkers are currently insufficient and are urgently required. Herein, we report three pre-treated cases of advanced NSCLC with TP53 mutations, wherein these patients showed partial response to anlotinib. Moreover, the three patients have achieved a progression-free survival of 8, 6.5, and 5 months, respectively. The main toxicities were hypertension, hand-foot syndrome and fatigue. In conclusion, TP53 mutations may represent a biomarker for predicting salutary effects of anlotinib.
Collapse
Affiliation(s)
- Shencun Fang
- Department of Respiratory Medicine, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Wanwan Cheng
- Department of Respiratory Medicine, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Meiling Zhang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Rusong Yang
- Department of Thoracic Surgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
22
|
Seo J, Park M. Molecular crosstalk between cancer and neurodegenerative diseases. Cell Mol Life Sci 2020; 77:2659-2680. [PMID: 31884567 PMCID: PMC7326806 DOI: 10.1007/s00018-019-03428-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/11/2019] [Accepted: 12/13/2019] [Indexed: 02/07/2023]
Abstract
The progression of cancers and neurodegenerative disorders is largely defined by a set of molecular determinants that are either complementarily deregulated, or share remarkably overlapping functional pathways. A large number of such molecules have been demonstrated to be involved in the progression of both diseases. In this review, we particularly discuss our current knowledge on p53, cyclin D, cyclin E, cyclin F, Pin1 and protein phosphatase 2A, and their implications in the shared or distinct pathways that lead to cancers or neurodegenerative diseases. In addition, we focus on the inter-dependent regulation of brain cancers and neurodegeneration, mediated by intercellular communication between tumor and neuronal cells in the brain through the extracellular microenvironment. Finally, we shed light on the therapeutic perspectives for the treatment of both cancer and neurodegenerative disorders.
Collapse
Affiliation(s)
- Jiyeon Seo
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, South Korea
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, South Korea
| | - Mikyoung Park
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, South Korea.
- Department of Neuroscience, Korea University of Science and Technology, Daejeon, 34113, South Korea.
| |
Collapse
|
23
|
Hou Z, Sun L, Xu F, Hu F, Lan J, Song D, Feng Y, Wang J, Luo X, Hu J, Wang G. Blocking histone methyltransferase SETDB1 inhibits tumorigenesis and enhances cetuximab sensitivity in colorectal cancer. Cancer Lett 2020; 487:63-73. [PMID: 32473242 DOI: 10.1016/j.canlet.2020.05.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/12/2020] [Accepted: 05/20/2020] [Indexed: 12/18/2022]
Abstract
The histone methyltransferase SETDB1 catalyzes the addition of methyl groups to histone H3 at lysine 9, and upregulation of SETDB1 is associated with poor prognosis in cancer patients. Here, we describe how overexpression of SETDB1 contributes to colorectal cancer (CRC) tumorigenesis and drug resistance. We show that SETDB1 is upregulated in CRC, and its level correlates with poor clinical outcome. SETDB1 attenuation inhibits CRC cell proliferation Mechanistically, SETDB1 promotes cell proliferation by upregulating Akt activation. Further, SETDB1 is essential for the tumorigenic activity of Akt. Functional characterization revealed that inhibition of SETDB1 reduces cell growth in CRC resistant to targeted treatments in vitro and in vivo, KRAS-mutated CRC included. Taken together, our results indicate that SETDB1 is a major driver of CRC and may serve as a potential target for the treatment of KRAS-mutated CRC.
Collapse
Affiliation(s)
- Zhenlin Hou
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China; Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Li Sun
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Feng Xu
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fuqing Hu
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jingqin Lan
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Da Song
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yongdong Feng
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jing Wang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xuelai Luo
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Junbo Hu
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Guihua Wang
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
24
|
Temozolomide and Pazopanib Combined with FOLFOX Regressed a Primary Colorectal Cancer in a Patient-derived Orthotopic Xenograft Mouse Model. Transl Oncol 2020; 13:100739. [PMID: 32143177 PMCID: PMC7058405 DOI: 10.1016/j.tranon.2019.12.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 12/19/2019] [Accepted: 12/23/2019] [Indexed: 01/01/2023] Open
Abstract
Purpose: The goal of the present study was to determine the efficacy of temozolomide (TEM) and pazopanib (PAZ) combined with FOLFOX (oxaliplatin, leucovorin and 5-fluorouracil) on a colorectal cancer patient-derived orthotopic xenograft (PDOX) mouse model. Materials and Methods: A colorectal cancer tumor from a patient previously established in non-transgenic nude mice was implanted subcutaneously in transgenic green fluorescence protein (GFP)-expressing nude mice in order to label the tumor stromal cells with GFP. Then labeled tumors were orthotopically implanted into the cecum of nude mice. Mice were randomized into four groups: Group 1, untreated control; group 2, TEM + PAZ; group 3, FOLFOX; group 4, TEM + PAZ plus FOLFOX. Tumor width, length, and mouse body weight were measured weekly. The Fluor Vivo imaging System was used to image the GFP-lableled tumor stromal cells in vivo. H&E staining and immunohistochemical staining were used for histological analysis. Results: All three treatments inhibited tumor growth as compared to the untreated control group. The combination of TEM + PAZ + FOLFOX regressed tumor growth significantly more effectively than TEM + PAZ or FOLFOX. Only the combination of TEM + PAZ + FOLFOX group caused a decrease in body weight. PAZ suppressed lymph vessels density in the colorectal cancer PDOX mouse model suggesting inhibition of lymphangiogenesis. Conclusion: Our results suggest that the combination of TEM + PAZ + FOLFOX has clinical potential for colorectal cancer patient.
Collapse
|
25
|
Phase I studies of vorinostat with ixazomib or pazopanib imply a role of antiangiogenesis-based therapy for TP53 mutant malignancies. Sci Rep 2020; 10:3080. [PMID: 32080210 PMCID: PMC7033174 DOI: 10.1038/s41598-020-58366-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 01/13/2020] [Indexed: 12/19/2022] Open
Abstract
We performed two phase I trials of the histone deacetylase inhibitor vorinostat combined with either the vascular endothelial growth factor inhibitor pazopanib (NCT01339871) or the proteasome inhibitor ixazomib (NCT02042989) in patients with metastatic TP53 mutant solid tumors. Both trials followed a 3 + 3 dose-escalation design allowing for a dose expansion cohort of up to 14 additional patients with a specific tumor type. Patients had to have a confirmed TP53 mutation to be enrolled in NCT02042989. Among patients enrolled in NCT01339871, TP53 mutation status was determined for those for whom tumor specimens were available. The results of NCT01339871 were reported previously. Common treatment-related adverse events in NCT02042989 included anemia, thrombocytopenia, fatigue, nausea, vomiting, and diarrhea. Compared with patients with metastatic TP53 hotspot mutant solid tumors who were treated with ixazomib and vorinostat (n = 59), those who were treated with pazopanib and vorinostat (n = 11) had a significantly higher rate of clinical benefit, defined as stable disease lasting ≥6 months or an objective response (3.4% vs. 45%; p < 0.001), a significantly longer median progression-free survival duration (1.7 months [95% confidence interval (CI), 1.1–2.3] vs. 3.5 months [95% CI, 1.7–5.2]; p = 0.002), and a longer median overall survival duration (7.3 months [95% CI, 4.8–9.8] vs. 12.7 months [95% CI, 7.1–18.3]; p = 0.24). Our two phase I trials provide preliminary evidence supporting the use of antiangiogenisis-based therapy in patients with metastatic TP53 mutant solid tumors, especially in those with metastatic sarcoma or metastatic colorectal cancer.
Collapse
|
26
|
Bordonaro M. Hypothesis: Retinoblastoma protein inactivation mediates effects of histone deacetylase inhibitor-induced Wnt hyperactivation in colorectal cancer cells. J Cancer 2020; 11:668-677. [PMID: 31942190 PMCID: PMC6959039 DOI: 10.7150/jca.37864] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/26/2019] [Indexed: 01/30/2023] Open
Abstract
Butyrate, a product of dietary fiber and a histone deacetylase inhibitor, induces apoptosis of colorectal cancer cells; this effect of butyrate is in part mediated by its ability to hyperactivate Wnt signaling, and may in part explain the preventive action of dietary fiber against colorectal cancer. However, the mechanisms by which Wnt hyperactivation promotes apoptosis are unknown. Inactivation of the retinoblastoma tumor suppressor occurs in some cancers and can lead to context-dependent cell proliferation or cell death/apoptosis. The function of retinoblastoma protein (Rb) in normal cells is modulation of cell cycle; inactivation of Rb allows for cell cycle progression and, hence, cell proliferation. Wnt signaling is upregulated in a variety of cancers, and deregulated Wnt signaling is a key initiating event in most cases of sporadic colorectal cancer. It has been shown that Wnt signaling activated by APC inactivation can synergize with the inactivation of Rb to induce apoptosis in a manner mediated by increased TORC1 activity, leading to induced metabolic and energy stress. Rb is typically not inactivated in colorectal cancer; however, Rb is phosphorylated and deactivated during cell cycle G1/S transition. This manuscript posits that it is during this time that butyrate/histone deacetylase inhibitor-induced Wnt hyperactivation induces apoptosis in colorectal cancer cells. Thus, the inactivation of Rb in cell cycle progression may synergize with Wnt hyperactivation to induce apoptosis in response to histone deacetylase inhibitors. The hypothesis is that hyperactivation of Wnt signaling enhances colorectal cancer cell apoptosis via the interaction between upregulated Wnt signaling and inactivated Rb during cell cycle progression. This paper discusses this hypothesis and offers initial experimental approaches for testing the hypothesis. A better understanding of how histone deacetylase inhibitors induce colorectal cancer cell apoptosis through hyperactivation of Wnt signaling, and of cross-talk between repression of cell cycle and induction of apoptosis that occurs with treatment with histone deacetylase inhibitors, can assist in the development of novel therapies for colorectal cancer.
Collapse
Affiliation(s)
- Michael Bordonaro
- Department of Medical Education, Geisinger Commonwealth School of Medicine, 525 Pine Street, Scranton, PA 18509, USA
| |
Collapse
|
27
|
Lee ATJ, Jones RL, Huang PH. Pazopanib in advanced soft tissue sarcomas. Signal Transduct Target Ther 2019; 4:16. [PMID: 31123606 PMCID: PMC6522548 DOI: 10.1038/s41392-019-0049-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 04/08/2019] [Accepted: 04/16/2019] [Indexed: 12/18/2022] Open
Abstract
Pazopanib is the first and only tyrosine kinase inhibitor currently approved for the treatment of multiple histological subtypes of soft tissue sarcoma (STS). Initially developed as a small molecule inhibitor of vascular endothelial growth factor receptors, preclinical work indicates that pazopanib exerts an anticancer effect through the inhibition of both angiogenic and oncogenic signaling pathways. Following the establishment of optimal dosing and safety profiles in early phase studies and approval for the treatment of advanced renal cell carcinoma, pazopanib was investigated in STS. A landmark phase III randomized study demonstrated improved progression-free survival with pazopanib compared to that with placebo in pretreated patients with STS of various subtypes. The efficacy of pazopanib in specific STS subtypes has been further described in real-world-based case series in both mixed and subtype-specific STS cohorts. At present, there are no clinically validated predictive biomarkers for use in selecting patients with advanced STS for pazopanib therapy, limiting the clinical effectiveness and cost-effectiveness of the drug. In this review, we summarize the preclinical and clinical data for pazopanib, outline the evidence base for its effect in STS and explore reported studies that have investigated putative biomarkers.
Collapse
Affiliation(s)
- Alex T. J. Lee
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - Robin L. Jones
- Sarcoma Unit, The Royal Marsden NHS Foundation Trust, London, UK
- Division of Clinical Studies, Institute of Cancer Research, London, UK
| | - Paul H. Huang
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
| |
Collapse
|
28
|
Wu YS, Lee ZY, Chuah LH, Mai CW, Ngai SC. Epigenetics in Metastatic Breast Cancer: Its Regulation and Implications in Diagnosis, Prognosis and Therapeutics. Curr Cancer Drug Targets 2019; 19:82-100. [PMID: 29714144 DOI: 10.2174/1568009618666180430130248] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 02/21/2018] [Accepted: 04/03/2018] [Indexed: 02/06/2023]
Abstract
Despite advances in the treatment regimen, the high incidence rate of breast cancer (BC) deaths is mostly caused by metastasis. Recently, the aberrant epigenetic modifications, which involve DNA methylation, histone modifications and microRNA (miRNA) regulations become attractive targets to treat metastatic breast cancer (MBC). In this review, the epigenetic alterations of DNA methylation, histone modifications and miRNA regulations in regulating MBC are discussed. The preclinical and clinical trials of epigenetic drugs such as the inhibitor of DNA methyltransferase (DNMTi) and the inhibitor of histone deacetylase (HDACi), as a single or combined regimen with other epigenetic drug or standard chemotherapy drug to treat MBCs are discussed. The combined regimen of epigenetic drugs or with standard chemotherapy drugs enhance the therapeutic effect against MBC. Evidences that epigenetic changes could have implications in diagnosis, prognosis and therapeutics for MBC are also presented. Several genes have been identified as potential epigenetic biomarkers for diagnosis and prognosis, as well as therapeutic targets for MBC. Endeavors in clinical trials of epigenetic drugs against MBC should be continued although limited success has been achieved. Future discovery of epigenetic drugs from natural resources would be an attractive natural treatment regimen for MBC. Further research is warranted in translating research into clinical practice with the ultimate goal of treating MBC by epigenetic therapy in the near future.
Collapse
Affiliation(s)
- Yuan Seng Wu
- School of Biosciences, Faculty of Science, University of Nottingham Malaysia Campus, Selangor, Malaysia
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Zhong Yang Lee
- School of Biosciences, Faculty of Science, University of Nottingham Malaysia Campus, Selangor, Malaysia
| | - Lay-Hong Chuah
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
- Advanced Engineering Platform, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
| | - Chun Wai Mai
- Department of Pharmaceutical Chemistry, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Siew Ching Ngai
- School of Biosciences, Faculty of Science, University of Nottingham Malaysia Campus, Selangor, Malaysia
| |
Collapse
|
29
|
Wang Y, Wang Z, Piha-Paul S, Janku F, Subbiah V, Shi N, Hess K, Broaddus R, Shan B, Naing A, Hong D, Tsimberidou AM, Karp D, Lu C, Papadimitrakopoulou V, Heymach J, Meric-Bernstam F, Fu S. Outcome analysis of Phase I trial patients with metastatic KRAS and/or TP53 mutant non-small cell lung cancer. Oncotarget 2018; 9:33258-33270. [PMID: 30279957 PMCID: PMC6161801 DOI: 10.18632/oncotarget.25947] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 07/18/2018] [Indexed: 01/26/2023] Open
Abstract
KRAS and TP53 mutations, which are the most common genetic drivers of tumorigenesis, are still considered undruggable targets. Therefore, we analyzed these genetic aberrations in metastatic non-small cell lung cancer (NSCLC) for the development of potential therapeutics. One hundred eighty-five consecutive patients with metastatic NSCLC in a phase 1 trial center were included. Their genomic aberrations, clinical characteristics, survivals, and phase 1 trial therapies were analyzed. About 10%, 18%, 36%, and 36% of the patients had metastatic KRAS+/TP53+, KRAS+/TP53-,KRAS-/TP53+, and KRAS-/TP53- NSCLC, respectively. The most common concurrent genetic aberrations beside KRAS and/or TP53 (>5%) were KIT, epidermal growth factor receptor, PIK3CA, c-MET, BRAF, STK11, ATM, CDKN2A, and APC. KRAS+/TP53+ NSCLC did not respond well to the phase 1 trial therapy and was associated with markedly worse progression-free (PFS) and overall (OS) survivals than the other three groups together. KRAS hotspot mutations at locations other than codon G12 were associated with considerably worse OS than those at this codon. Gene aberration-matched therapy produced prolonged PFS and so was anti-angiogenesis in patients with TP53 mutations. Introduction of the evolutionary action score system of TP53 missense mutations enabled us to identify a subgroup of NSCLC patients with low-risk mutant p53 proteins having a median OS duration of 64.5 months after initial diagnosis of metastasis. These data suggested that patients with metastatic dual KRAS+/TP53+ hotspot-mutant NSCLC had poor clinical outcomes. Further analysis identified remarkably prolonged survival in patients with low-risk mutant p53 proteins, which warrants confirmatory studies.
Collapse
Affiliation(s)
- Yudong Wang
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, People’s Republic of China
| | - Zhijie Wang
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Sarina Piha-Paul
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Filip Janku
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naiyi Shi
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenneth Hess
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Russell Broaddus
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Baoen Shan
- Department of Cancer Research, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, People’s Republic of China
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Apostolia M. Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel Karp
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Charles Lu
- Department of Thoracic Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vali Papadimitrakopoulou
- Department of Thoracic Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John Heymach
- Department of Thoracic Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
30
|
Chromatin dynamics at the core of kidney fibrosis. Matrix Biol 2018; 68-69:194-229. [DOI: 10.1016/j.matbio.2018.02.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 02/16/2018] [Accepted: 02/17/2018] [Indexed: 02/06/2023]
|
31
|
Zang J, Liang X, Huang Y, Jia Y, Li X, Xu W, Chou CJ, Zhang Y. Discovery of Novel Pazopanib-Based HDAC and VEGFR Dual Inhibitors Targeting Cancer Epigenetics and Angiogenesis Simultaneously. J Med Chem 2018; 61:5304-5322. [PMID: 29787262 DOI: 10.1021/acs.jmedchem.8b00384] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Herein a novel series of pazopanib hybrids as polypharmacological antitumor agents were developed based on the crosstalk between histone deacetylases (HDACs) and vascular endothelial growth factor (VEGF) pathway. Among them, one ortho-aminoanilide 6d and one hydroxamic acid 13f exhibited considerable total HDACs and VEGFR-2 inhibitory activities. The HDAC inhibitory activities endowed 6d and 13f with potent antiproliferative activities, which was not observed in the approved VEGFR inhibitor pazopanib. Compounds 6d and 13f possessed comparable HDAC isoform selectivity profiles to the clinical class I HDAC inhibitor MS-275 and the approved pan-HDAC inhibitor SAHA, respectively. 6d and 13f also exhibited uncompromised multiple tyrosine kinases inhibitory activities relative to pazopanib. The intracellular dual inhibition to HDAC and VEGFR of 6d and 13f was validated by Western blot analysis. In both HUVECs tube formation assay and rat thoracic aorta rings assay, 6d and 13f showed comparable antiangiogenic potencies to pazopanib. What's more, 6d possessed desirable pharmacokinetic profiles with the oral bioavailability of 72% in SD rats and considerable in vivo antitumor efficacy in a human colorectal adenocarcinoma (HT-29) xenograft model.
Collapse
Affiliation(s)
- Jie Zang
- Department of Medicinal Chemistry, School of Pharmaceutical of Science , Shandong University , Ji'nan , Shandong 250012 , P. R. China
| | - Xuewu Liang
- Department of Medicinal Chemistry, School of Pharmaceutical of Science , Shandong University , Ji'nan , Shandong 250012 , P. R. China
| | - Yongxue Huang
- Weifang Bochuang International Biological Medicinal Institute , Weifang , Shandong 261061 , P. R. China
| | - Yuping Jia
- Shandong Academy of Pharmaceutical Sciences , Ji'nan , Shandong 250101 , P. R. China
| | - Xiaoyang Li
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy , Medical University of South Carolina , Charleston , South Carolina 29425 , United States
| | - Wenfang Xu
- Department of Medicinal Chemistry, School of Pharmaceutical of Science , Shandong University , Ji'nan , Shandong 250012 , P. R. China
| | - C James Chou
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy , Medical University of South Carolina , Charleston , South Carolina 29425 , United States
| | - Yingjie Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical of Science , Shandong University , Ji'nan , Shandong 250012 , P. R. China
| |
Collapse
|
32
|
Antiangiogenesis and gene aberration-related therapy may improve overall survival in patients with concurrent KRAS and TP53 hotspot mutant cancer. Oncotarget 2018; 8:33796-33806. [PMID: 28430579 PMCID: PMC5464912 DOI: 10.18632/oncotarget.16840] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 03/16/2017] [Indexed: 02/07/2023] Open
Abstract
Purpose Genetic alterations such as activating KRAS and/or inactivating TP53 are thought to be the most common drivers to tumorigenesis. Therefore, we assessed phase I cancer patients with KRAS+/TP53+ mutations. Results Approximately 8% of patients referred to phase I clinical trials harbored concurrent KRAS and TP53 mutations. Patients who received a phase I trial therapy (n = 57) had a median OS of 12 months, compared with 4.6 months in those who were not treated (n = 106; p = 0.003). KRAS G13 and TP53 R273 mutations were associated with poor overall survival (OS), while antiangiogenesis and gene aberration-related therapies were associated with prolonged OS. A prognostic model using neutrophilia, thrombocytosis, hypoalbuminemia, body mass index <30 kg/m2, and the absence of lung metastasis was established and validated. Phase I cancer patients in the low-risk group had a median OS of 16.6 months compared with 5.4 months in the high-risk group (p < 0.001). Untreated patients in the low-risk group had a median OS of 6.7 months compared with 3.6 months in the high-risk group (p = 0.033). Experimental Design We analyzed 163 consecutive patients with advanced KRAS+/TP53+ mutant cancer who were referred to phase I clinical trials, to identify molecular aberrations, clinical characteristics, survivals, and potentially effective treatment regimens. Conclusions This study provided preliminary evidence that besides modulation of the proinflammatory state, antiangiogensis and concomitant gene aberration-related therapies may improve the treatment of KRAS+/TP53+ mutant cancer.
Collapse
|
33
|
Zhang L, Wang H, Li W, Zhong J, Yu R, Huang X, Wang H, Tan Z, Wang J, Zhang Y. Pazopanib, a novel multi-kinase inhibitor, shows potent antitumor activity in colon cancer through PUMA-mediated apoptosis. Oncotarget 2018; 8:3289-3303. [PMID: 27924057 PMCID: PMC5356882 DOI: 10.18632/oncotarget.13753] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 10/19/2016] [Indexed: 12/22/2022] Open
Abstract
Colon cancer is still the third most common cancer which has a high mortality but low five-year survival rate. Novel tyrosine kinase inhibitors (TKI) such as pazopanib become effective antineoplastic agents that show promising clinical activity in a variety of carcinoma, including colon cancer. However, the precise underlying mechanism against tumor is unclear. Here, we demonstrated that pazopanib promoted colon cancer cell apoptosis through inducing PUMA expression. Pazopanib induced p53-independent PUMA activation by inhibiting PI3K/Akt signaling pathway, thereby activating Foxo3a, which subsequently bound to the promoter of PUMA to activate its transcription. After induction, PUMA activated Bax and triggered the intrinsic mitochondrial apoptosis pathway. Furthermore, administration of pazopanib highly suppressed tumor growth in a xenograft model. PUMA deletion in cells and tumors led to resistance of pazopanib, indicating PUMA-mediated pro-apoptotic and anti-tumor effects in vitro and in vivo. Combing pazopanib with some conventional or novel drugs, produced heightened and synergistic antitumor effects that were associated with potentiated PUMA induction via different pathways. Taken together, these results establish a critical role of PUMA in mediating the anticancer effects of pazopanib in colon cancer cells and provide the rationale for clinical evaluation.
Collapse
Affiliation(s)
- Lingling Zhang
- College of Biology, Hunan University, Changsha, China.,Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, China.,Department of Internal Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Huanan Wang
- College of Biology, Hunan University, Changsha, China.,Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Wei Li
- Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, China.,Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Juchang Zhong
- College of Biology, Hunan University, Changsha, China
| | - Rongcheng Yu
- College of Biology, Hunan University, Changsha, China
| | - Xinfeng Huang
- College of Biology, Hunan University, Changsha, China
| | - Honghui Wang
- College of Biology, Hunan University, Changsha, China
| | - Zhikai Tan
- College of Biology, Hunan University, Changsha, China
| | - Jiangang Wang
- Department of Internal Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yingjie Zhang
- College of Biology, Hunan University, Changsha, China.,Shenzhen Institute, Hunan University, Shenzhen, China
| |
Collapse
|
34
|
Frequency of Somatic TP53 Mutations in Combination with Known Pathogenic Mutations in Colon Adenocarcinoma, Non-Small Cell Lung Carcinoma, and Gliomas as Identified by Next-Generation Sequencing. Neoplasia 2018; 20:256-262. [PMID: 29454261 PMCID: PMC5849803 DOI: 10.1016/j.neo.2017.12.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 12/20/2022] Open
Abstract
The tumor suppressor gene TP53 is the most frequently mutated gene in human cancer. It encodes p53, a DNA-binding transcription factor that regulates multiple genes involved in DNA repair, metabolism, cell cycle arrest, apoptosis, and senescence. TP53 is associated with human cancer by mutations that lead to a loss of wild-type p53 function as well as mutations that confer alternate oncogenic functions that enable them to promote invasion, metastasis, proliferation, and cell survival. Identifying the discrete TP53 mutations in tumor cells may help direct therapies that are more effective. In this study, we identified the frequency of individual TP53 mutations in patients with colon adenocarcinoma (48%), non–small cell lung carcinoma (NSCLC) (36%), and glioma/glioblastoma (28%) at our institution using next-generation sequencing. We also identified the occurrence of somatic mutations in numerous actionable genes including BRAF, EGFR, KRAS, IDH1, and PIK3CA that occurred concurrently with these TP53 mutations. Of the 480 tumors examined that contained one or more mutations in the TP53 gene, 219 were colon adenocarcinomas, 215 were NSCLCs, and 46 were gliomas/glioblastomas. Among the patients positive for TP53 mutations diagnosed with colon adenocarcinoma, 50% also showed at least one mutation in pathogenic genes of which 14% were BRAF, 33% were KRAS, and 3% were NRAS. Forty-seven percent of NSCLC patients harboring TP53 mutations also had a mutation in at least one actionable pathogenic variant with the following frequencies: BRAF: 4%, EGFR: 10%, KRAS: 28%, and PIK3CA: 4%. Fifty-two percent of patients diagnosed with glioma/glioblastoma with a positive TP53 mutation had at least one concurrent mutation in a known pathogenic gene of which 9% were CDKN2A, 41% were IDH1, and 11% were PIK3CA.
Collapse
|
35
|
Hou MM, Wang Z, Janku F, Piha-Paul S, Naing A, Hong D, Westin S, Coleman RL, Sood AK, Tsimberidou AM, Subbiah V, Wheler J, Zinner R, Lu K, Meric-Bernstam F, Fu S. Continuous anti-angiogenic therapy after tumor progression in patients with recurrent high-grade epithelial ovarian cancer: phase I trial experience. Oncotarget 2018; 7:35132-43. [PMID: 27147567 PMCID: PMC5085215 DOI: 10.18632/oncotarget.9048] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 04/10/2016] [Indexed: 01/04/2023] Open
Abstract
High-grade epithelial ovarian cancer (HG-EOC) is the most lethal gynecologic malignancy worldwide Once patients develop chemoresistance, effective novel strategies are required to improve prognosis We analyzed characteristics and outcomes of 242 consecutive patients with HG-EOC participating in 94 phase I clinical trials at The University of Texas MD Anderson Cancer Center. Baseline lactate dehydrogenase levels, albumin levels, and number of metastatic sites were independent predictors of overall survival (OS). Receiving more than 1 phase I protocol was associated with improved OS (p < 0.001). Regimens including a chemotherapeutic agent plus bevacizumab or Aurora A kinase inhibitor led to a median progression-free survival (PFS) duration of more than 6 months. Although patients receiving bevacizumab-based regimens in the phase I clinical trials had significantly longer PFS than those receiving other anti-angiogenic therapies (p = 0.017), patients treated with vascular endothelial growth factor receptor-tyrosine kinase inhibitors (VEGFR-TKIs) had significantly longer OS (12.2 months) than those not treated with VEGFR-TKIs (8.6 months, p = 0.015). In conclusion, anti-angiogenic therapy is one of the most important strategies for the treatment of HG-EOC, even in those who have already experienced tumor progression. Therefore, eligible patients with HG-EOC should be encouraged to participate in novel phase I studies of anti-angiogenic therapies, even after disease progression.
Collapse
Affiliation(s)
- Ming-Mo Hou
- Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Division of Hematology-Oncology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Zhijie Wang
- Department of Thoracic Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Beijing Institute for Cancer Research, Beijing, China
| | - Filip Janku
- Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sarina Piha-Paul
- Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Aung Naing
- Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David Hong
- Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shannon Westin
- Departments of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Robert L Coleman
- Departments of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anil K Sood
- Departments of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Apostolia M Tsimberidou
- Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Vivek Subbiah
- Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jennifer Wheler
- Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ralph Zinner
- Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Karen Lu
- Departments of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Funda Meric-Bernstam
- Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Siqing Fu
- Departments of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
36
|
Outcomes of patients with sarcoma enrolled in clinical trials of pazopanib combined with histone deacetylase, mTOR, Her2, or MEK inhibitors. Sci Rep 2017; 7:15963. [PMID: 29162825 PMCID: PMC5698336 DOI: 10.1038/s41598-017-13114-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 09/19/2017] [Indexed: 12/29/2022] Open
Abstract
Pazopanib is US FDA approved for the treatment of advanced soft tissue sarcomas. All patients with this disease ultimately develop resistance to therapy. Mechanisms of resistance include activation of the mTOR, histone deacetylase (HDAC), MAPK, and ERBB4 pathways. We hypothesized that combining pazopanib with other targeted agents inhibiting these pathways would increase response rates. We retrospectively evaluated the safety and efficacy of pazopanib plus vorinostat, everolimus, lapatinib or trastuzumab, and MEK inhibitor in patients with advanced sarcoma. The Cancer Geneome Atlas (TCGA) data was analyzed for HDAC, PI3K, HER2, and MAPK/RAS/RAF gene alterations from sarcoma TCGA. Of the 44 advanced sarcoma patients in these trials, 27 (61%) were male; 18 (41%) had bone sarcoma, and 26 (59%) had soft tissue sarcoma. Best response was partial response (PR) in four patients [(overall response rate (ORR) = 9%, 95% confidence interval [CI] 3% to 22%)]. The median progression-free survival (PFS) for all patients was 9.6 weeks (95% CI 8.0 to 15.7 weeks). Analysis of TCGA data revealed HDAC, PI3K, HER2, and MAPK/RAS/RAF gene alterations in 112/243 (46%) of patients predominantly HDAC1-11 (41%) alterations. Pazopanib combinations did demonstrate safety in combination with other agents. TCGA data suggests further evaluation of epigenetic pathway inhibitors in sarcoma.
Collapse
|
37
|
Lewin J, Garg S, Lau BY, Dickson BC, Traub F, Gokgoz N, Griffin AM, Ferguson PC, Andrulis IL, Sim HW, Kamel-Reid S, Stockley TL, Siu LL, Wunder JS, Razak ARA. Identifying actionable variants using next generation sequencing in patients with a historical diagnosis of undifferentiated pleomorphic sarcoma. Int J Cancer 2017; 142:57-65. [PMID: 28891048 DOI: 10.1002/ijc.31039] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 07/19/2017] [Accepted: 08/24/2017] [Indexed: 12/22/2022]
Abstract
There are limited data regarding the molecular characterization of undifferentiated pleomorphic sarcomas (UPS; formerly malignant fibrous histiocytoma). This study aimed to investigate the utility of next generation sequencing (NGS) in UPS to identify subsets of patients who harbour actionable mutations. Patients diagnosed with UPS underwent pathological re-evaluation by a pathologist specializing in sarcoma. Tumor DNA was isolated from archived fresh frozen tissue samples and genotyped using NGS with the Illumina MiSeq TruSeq Amplicon Cancer Panel (48 genes, 212 amplicons). In total, 95 patients initially classified with UPS were identified. Following pathology re-review the histological subtypes were reclassified to include: Myxofibrosarcoma (MFS, N = 44); UPS(N = 18); and Others (N = 27; including undifferentiated spindle cell sarcoma (N = 15) and dedifferentiated liposarcoma (N = 6)). Seven cases were excluded from further analysis for other reasons. Baseline demographics of the finalized cohort (N = 88) showed a median age of 66 years (32-95), primarily with stage I-III disease (92%) and high-grade (86%) lesions. Somatic mutations were identified in 31 cases (35%)(Total mutations = 36: solitary mutation(n = 27); two mutations( =n = 3); three mutations(n = 1)). The most commonly identified mutations were in TP53 (n = 24), ATM (n = 3) and PIK3CA (n = 2). Three of 43 patients with MFS and one of 18 patients with UPS had clinically relevant mutations, mainly related to biomarkers of prediction of response; however few had targetable driver mutations. Somatic mutation status did not influence disease free or overall survival. Based on the small number of clinically relevant mutations, these data do not support the routine use of targeted NGS panels outside of research protocols in UPS.
Collapse
Affiliation(s)
- Jeremy Lewin
- Sarcoma Program, Mount Sinai Hospital, Toronto, Canada.,Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Swati Garg
- Advanced Molecular Diagnostics Laboratory, Princess Margaret Cancer Centre, Toronto, Canada
| | - Beatrice Y Lau
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada
| | - Brendan C Dickson
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Canada
| | - Frank Traub
- Department of Orthopedic surgery, University Hospital Tuebingen, Erberhad Karls University Tuebingen, Germany
| | - Nalan Gokgoz
- Lunenfeld-Tanenbaum Research Institute, Toronto, Canada
| | | | - Peter C Ferguson
- Department of Surgery, Mount Sinai Hospital, University of Toronto, University Musculoskeletal Oncology Unit and Division of Orthopaedic Surgery, Toronto, Canada
| | - Irene L Andrulis
- Lunenfeld-Tanenbaum Research Institute, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Hao-Wen Sim
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Suzanne Kamel-Reid
- Advanced Molecular Diagnostics Laboratory, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Candada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Tracy L Stockley
- Advanced Molecular Diagnostics Laboratory, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Candada
| | - Lillian L Siu
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| | - Jay S Wunder
- Department of Surgery, Mount Sinai Hospital, University of Toronto, University Musculoskeletal Oncology Unit and Division of Orthopaedic Surgery, Toronto, Canada
| | - Albiruni R A Razak
- Sarcoma Program, Mount Sinai Hospital, Toronto, Canada.,Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, Canada
| |
Collapse
|
38
|
In GK, Hu JS, Tseng WW. Treatment of advanced, metastatic soft tissue sarcoma: latest evidence and clinical considerations. Ther Adv Med Oncol 2017; 9:533-550. [PMID: 28794805 PMCID: PMC5524246 DOI: 10.1177/1758834017712963] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 05/04/2017] [Indexed: 12/24/2022] Open
Abstract
Soft tissue sarcoma (STS) is a biologically heterogeneous malignancy with over 50 subtypes. Historically, there have been few systemic treatment options for this relatively rare disease. Traditional cytotoxic agents, such as anthracyclines, alkylating agents, and taxanes have limited clinical benefit beyond the first-line setting; across all high-grade STS subtypes, median overall survival remains approximately 12-18 months for advanced metastatic disease. The development of targeted therapies has led to recent US Food and Drug Administration approval of four new treatments for high-grade STS in the advanced metastatic setting. Among these, olaratumab is most notable for its improvement in overall survival for patients with anthracycline-naïve disease. Further progress in STS management will rely on novel trial design, subtype-specific therapies and validation of biomarkers to tailor therapy. Immunotherapy has shown promise as a new, but yet undiscovered frontier in the management of STS.
Collapse
Affiliation(s)
- Gino K. In
- Division of Oncology, Norris Comprehensive Cancer Center, Los Angeles, CA, USA
- Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - James S. Hu
- Division of Oncology, Norris Comprehensive Cancer Center, Los Angeles, CA, USA
- Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - William W. Tseng
- Department of Surgery, Section of Surgical Oncology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
39
|
Pili R, Liu G, Chintala S, Verheul H, Rehman S, Attwood K, Lodge MA, Wahl R, Martin JI, Miles KM, Paesante S, Adelaiye R, Godoy A, King S, Zwiebel J, Carducci MA. Combination of the histone deacetylase inhibitor vorinostat with bevacizumab in patients with clear-cell renal cell carcinoma: a multicentre, single-arm phase I/II clinical trial. Br J Cancer 2017; 116:874-883. [PMID: 28222071 PMCID: PMC5379145 DOI: 10.1038/bjc.2017.33] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/04/2017] [Accepted: 01/20/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Class II histone deacetylase (HDAC) inhibitors induce hypoxia-inducible factor-1 and -2α degradation and have antitumour effects in combination with vascular endothelial growth factor (VEGF) inhibitors. In this study, we tested the safety and efficacy of the HDAC inhibitor vorinostat and the VEGF blocker bevacizumab in metastatic clear-cell renal cell carcinoma (ccRCC) patients previously treated with different drugs including sunitinib, sorafenib, axitinib, interleukin-2, interferon, and temsirolimus. METHODS Patients with up to two prior regimens were eligible for treatment, consisting of vorinostat 200 mg orally two times daily × 2 weeks, and bevacizumab 15 mg kg-1 intravenously every 3 weeks. The primary end points were safety and tolerability, and the proportion of patients with 6 months of progression-free survival (PFS). Correlative studies included immunohistochemistry, FDG PET/CT scans, and serum analyses for chemokines and microRNAs. RESULTS Thirty-six patients were enrolled, with 33 evaluable for toxicity and efficacy. Eighteen patients had 1 prior treatment, 13 patients had 2 prior treatments, and 2 patients were treatment naïve. Two patients experienced grade 4 thrombocytopenia and three patients had grade 3 thromboembolic events during the course of exposure. We observed six objective responses (18%), including one complete response and five partial responses. The proportion of patients with PFS at 6 months was 48%. The median PFS and overall survival were 5.7 months (confidence interval (CI): 4.1-11.0) and 13.9 months (CI: 9.8-20.7), respectively. Correlative studies showed that modulation of specific chemokines and microRNAs were associated with clinical benefit. CONCLUSIONS The combination of vorinostat with bevacizumab as described is relatively well tolerated. Response rate and median PFS suggest clinical activity for this combination strategy in previously treated ccRCC.
Collapse
Affiliation(s)
- Roberto Pili
- Genitourinary Program, Indiana University-Simon Cancer Center, Indianapolis, IN, USA
| | - Glenn Liu
- University of Wisconsin Carbone Cancer Center, Wisconsin, WI, USA
| | - Sreenivasulu Chintala
- Genitourinary Program, Indiana University-Simon Cancer Center, Indianapolis, IN, USA
| | - Hendrick Verheul
- Vrije Universiteit Amsterdam, De Boelelaan 1105, 1081 HV Amsterdam, The Netherlands
| | | | | | | | - Richard Wahl
- Johns Hopkins Kimmel Cancer Center, Baltimore, MD, USA
| | | | | | | | - Remi Adelaiye
- Genitourinary Program, Indiana University-Simon Cancer Center, Indianapolis, IN, USA
| | | | - Serina King
- Johns Hopkins Kimmel Cancer Center, Baltimore, MD, USA
| | | | | |
Collapse
|
40
|
Tiwari A, Gupta VG, Bakhshi S. Newer medical therapies for metastatic soft tissue sarcoma. Expert Rev Anticancer Ther 2017; 17:257-270. [PMID: 28103739 DOI: 10.1080/14737140.2017.1285229] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 01/18/2017] [Indexed: 12/20/2022]
Abstract
Metastatic/advanced soft tissue sarcoma has a poor prognosis conventionally, treatment options have been limited. In recent years, this area has been a rich ground for research with many new drugs being approved and several more in the pipeline. With multiple new treatment options available, it is vital to keep up pace with this rapidly changing field. Areas covered: Recent data regarding use of novel agents in advanced soft tissue sarcoma is reviewed with a focus on clinical applicability. The goal is to guide the clinician into choosing appropriate lines of therapy for the individual patient in light of recent availability of multiple new treatment options. Expert commentary: Patients with advanced soft tissue sarcoma can expect to receive several lines of therapy in the modern era. Tumor histology should ideally guide the choice of therapy. The new FDA approved second line drugs viz, trabectedin, pazopanib and eribulin should be considered first after failure of doxorubicin-based chemotherapy. Additional options have become available, such as antiangiogenic agents, mTOR inhibitors, and several new molecules targeting specific oncogenic pathways. All these agents have a role in treating soft tissue sarcoma, and careful individualization of therapy can help achieve optimal outcomes in these challenging patients.
Collapse
Affiliation(s)
- Akash Tiwari
- a Department of Medical Oncology , Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences , New Delhi , India
| | - Vineet Govinda Gupta
- a Department of Medical Oncology , Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences , New Delhi , India
| | - Sameer Bakhshi
- a Department of Medical Oncology , Dr. B.R.A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences , New Delhi , India
| |
Collapse
|
41
|
Wang Z, Shi N, Naing A, Janku F, Subbiah V, Araujo DM, Patel SR, Ludwig JA, Ramondetta LM, Levenback CF, Ramirez PT, Piha‐Paul SA, Hong D, Karp DD, Tsimberidou AM, Meric‐Bernstam F, Fu S. Survival of patients with metastatic leiomyosarcoma: the MD Anderson Clinical Center for targeted therapy experience. Cancer Med 2016; 5:3437-3444. [PMID: 27882721 PMCID: PMC5224847 DOI: 10.1002/cam4.956] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 10/11/2016] [Accepted: 10/12/2016] [Indexed: 12/13/2022] Open
Abstract
Advanced stage leiomyosarcoma (LMS) is incurable with current systemic antitumor therapies. Therefore, there is clinical interest in exploring novel therapeutic regimens to treat LMS. We reviewed the medical records of 75 consecutive patients with histologically confirmed metastatic LMS, who had been referred to the Clinical Center for Targeted Therapy at MD Anderson Cancer Center. To lay the foundation for potential phase I trials for the treatment of advanced LMS, we analyzed tumor response and survival outcome data. The frequent hotspot gene aberrations that we observed were the TP53 mutation (65%) and RB1 loss/mutation (45%) detected by Sequenom or next-generation sequencing. Among patients treated with gene aberration-related phase I trial therapy, the median progression-free survival was 5.8 months and the median overall survival was 15.9 months, significantly better than in patients without therapy (1.9 months, P = 0.001; and 8.7 months, P = 0.013, respectively). Independent risk factors that predicted shorter overall survival included hemoglobin <10 g/dL, body mass index <30 kg/m2 , serum albumin <3.5 g/dL, and neutrophil above upper limit of normal. The median survivals were 19.9, 7.6, and 0.9 months for patients with 0, 1 or 2, and ≥3 of the above risk factors, respectively (P < 0.001). A prognostic scoring system that included four independent risk factors might predict survival in patients with metastatic LMS who were treated in a phase I trial. Gene aberration-related therapies led to significantly better clinical benefits, supporting that further exploration with novel mechanism-driven therapeutic regimens is warranted.
Collapse
Affiliation(s)
- Zhijie Wang
- Department of Investigational Cancer TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexas
- Department of Medical OncologyCancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Naiyi Shi
- Department of Investigational Cancer TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Aung Naing
- Department of Investigational Cancer TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Filip Janku
- Department of Investigational Cancer TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Vivek Subbiah
- Department of Investigational Cancer TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Dejka M. Araujo
- Department of Sarcoma Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Shreyaskumar R. Patel
- Department of Sarcoma Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Joseph A. Ludwig
- Department of Sarcoma Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Lois M. Ramondetta
- Department of Gynecologic Oncology and Reproductive MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Charles F. Levenback
- Department of Gynecologic Oncology and Reproductive MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Pedro T. Ramirez
- Department of Gynecologic Oncology and Reproductive MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Sarina A. Piha‐Paul
- Department of Investigational Cancer TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - David Hong
- Department of Investigational Cancer TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Daniel D. Karp
- Department of Investigational Cancer TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Apostolia M. Tsimberidou
- Department of Investigational Cancer TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Funda Meric‐Bernstam
- Department of Investigational Cancer TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexas
| | - Siqing Fu
- Department of Investigational Cancer TherapeuticsThe University of Texas MD Anderson Cancer CenterHoustonTexas
| |
Collapse
|
42
|
Histology-Specific Uses of Tyrosine Kinase Inhibitors in Non-gastrointestinal Stromal Tumor Sarcomas. Curr Treat Options Oncol 2016; 17:11. [PMID: 26931561 DOI: 10.1007/s11864-015-0382-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OPINION STATEMENT Adult sarcomas, especially those with metastatic or unresectable disease, have limited treatment options. Traditional chemotherapeutic options have been limited by poor response rates in patients with advanced sarcomas. The important clinical question is whether the success of targeted therapy in GIST can be extended to other sarcomas and also if preclinical data describing targets across this heterogeneous group of cancers can be translated to clinical efficacy of known and upcoming target specific agents. Multi-targeted tyrosine kinase inhibitors (TKI) such as pazopanib, sorafenib, sunutinib, cediranib have shown benefits across various histologies of soft tissue sarcoma as well as bone sarcomas. The efficacy of imatinib in Dermatofibrosarcoma Protruberans; sunitinib and cediranib in alveolar soft part sarcoma; and sorafenib and imatinib in chordomas have provided a treatment option of these rare tumors where no effective options existed. TKIs are being tested in combination with chemotherapy as well as radiation to improve response. Although traditional RECIST criteria may not adequately reflect response to these targeted agents, the studies have shown promise for the efficacy of TKIs across the spectrum of sarcomas. The integration of biomarker studies with clinical trials may help further identify responders beyond that defined by histology. With the current data, TKIs are being used both as first-line treatment and beyond in non-GIST sarcomas.
Collapse
|
43
|
Chen S, Cavazza E, Barlier C, Salleron J, Filhine-Tresarrieu P, Gavoilles C, Merlin JL, Harlé A. Beside P53 and PTEN: Identification of molecular alterations of the RAS/MAPK and PI3K/AKT signaling pathways in high-grade serous ovarian carcinomas to determine potential novel therapeutic targets. Oncol Lett 2016; 12:3264-3272. [PMID: 27899992 PMCID: PMC5103928 DOI: 10.3892/ol.2016.5083] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 07/01/2016] [Indexed: 01/16/2023] Open
Abstract
Despite great histological and molecular heterogeneity, the clinical management of high-grade ovarian carcinomas remains unspecialized. As a major subgroup, high-grade serous ovarian carcinomas (HGSOCs) require novel therapies. In addition to utilizing conventional histological prognostic markers and performing oncogenetic investigations, the molecular diagnostic method of next generation sequencing (NGS) was performed to identify ‘druggable’ targets that could provide access to innovative therapy. The present study was performed in 45 HGSOC patients (mean age, 59.1 years; range, 25–87 years) with histologically proven HGSOC. Breast cancer 1/2 (BRCA1/2) germline mutations were screened in 17 patients with a familial or personal history of cancer, which was justified by oncogenetic investigations. Tumor protein 53 (P53) and phosphatase and tensin homolog (PTEN) expression were assessed in formalin-fixed paraffin-embedded tissues using immunohistochemistry. Somatic mutations of Kirsten rat sarcoma viral oncogene homolog, neuroblastoma RAS viral oncogene homolog (NRAS), B-Raf proto-oncogene, serine/threonine kinase, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit α (PIK3CA) and MET proto-oncogene, receptor tyrosine kinase (MET) were screened using NGS on DNA extracts from frozen tumor specimens obtained at diagnosis. With a median follow-up of 38 months (range, 6–93 months), 20 patients are alive, 10 patients are disease-free and 14 patients progressed within 6 months following platinum-based therapy. P53 overexpression was detected in 67% of patients and PTEN loss was detected in 38% of the patients. The overexpression of mutant P53 was found to be associated with a longer progression-free and overall survival. In total, 2 NRAS (exon 3), 3 PIK3CA (exon 5 and 10) and 5 MET mutations (exons 14 and 18) were detected. In HGSOCs, in addition to P53 and PTEN alterations, somatic genetic abnormalities can be detected using NGS and provide molecular rationale for targeted therapies, potentially offering novel therapeutic opportunities to patients.
Collapse
Affiliation(s)
- Shuhui Chen
- Faculty of Pharmacy, Université de Lorraine, 54001 Nancy, France; CNRS UMR 7039 CRAN, Université de Lorraine, 54506 Vandoeuvre-les-Nancy, France; Department of Biopathology, Institut de Cancérologie de Lorraine, 54519 Vandoeuvre-les-Nancy, France; Department of Gynecological Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Elisa Cavazza
- Faculty of Pharmacy, Université de Lorraine, 54001 Nancy, France
| | | | - Julia Salleron
- Department of Data Biostatistics, Institut de Cancérologie de Lorraine, 54519 Vandoeuvre-les-Nancy, France
| | | | - Céline Gavoilles
- Department of Medical Oncology, Institut de Cancérologie de Lorraine, 54519 Vandoeuvre-les-Nancy, France
| | - Jean-Louis Merlin
- Faculty of Pharmacy, Université de Lorraine, 54001 Nancy, France; CNRS UMR 7039 CRAN, Université de Lorraine, 54506 Vandoeuvre-les-Nancy, France; Department of Biopathology, Institut de Cancérologie de Lorraine, 54519 Vandoeuvre-les-Nancy, France
| | - Alexandre Harlé
- Faculty of Pharmacy, Université de Lorraine, 54001 Nancy, France; CNRS UMR 7039 CRAN, Université de Lorraine, 54506 Vandoeuvre-les-Nancy, France; Department of Biopathology, Institut de Cancérologie de Lorraine, 54519 Vandoeuvre-les-Nancy, France
| |
Collapse
|
44
|
Wheler JJ, Janku F, Naing A, Li Y, Stephen B, Zinner R, Subbiah V, Fu S, Karp D, Falchook GS, Tsimberidou AM, Piha-Paul S, Anderson R, Ke D, Miller V, Yelensky R, Lee JJ, Hong D, Kurzrock R. TP53 Alterations Correlate with Response to VEGF/VEGFR Inhibitors: Implications for Targeted Therapeutics. Mol Cancer Ther 2016; 15:2475-2485. [PMID: 27466356 DOI: 10.1158/1535-7163.mct-16-0196] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 07/10/2016] [Indexed: 11/16/2022]
Abstract
TP53 tumor-suppressor gene mutations are among the most frequent abnormalities in cancer, affecting approximately 40% of patients. Yet, there is no accepted way to target these alterations in the clinic. At the same time, antagonists of VEGFR or its ligand are best-selling oncology drugs, with multiple, expensive compounds approved. Although only a subset of patients benefit from these antiangiogenesis agents, no relevant biomarker has been identified. Interestingly, TP53 mutations upregulate VEGF-A and VEGFR2. We prospectively enrolled 500 patients, to be interrogated by comprehensive genomic profiling (CGP) (next-generation sequencing, 236 genes), and to be matched, whenever possible, with targeted agents. Herein, we analyze outcomes based on VEGF/VEGFR inhibitor treatment and presence of TP53 mutations. Of the 500 patients, 188 (37.6%; with ≥1 alteration) were treated; 106 (56% of 188) had tumors that harbored TP53 mutations. VEGF/VEGFR inhibitor therapy was independently associated with improvement in all outcome parameters [rate of stable disease (SD) ≥6 months/partial and complete remission (PR/CR); (31% versus 7%; TP53-mutant patients (who received no other molecular-matched agents) treated with versus without VEGF/VEGFR inhibitors), time-to-treatment failure, and overall survival (multivariate analysis: all P ≤ 0.01)] for the patients harboring TP53-mutant cancers, but improvement was not seen in any of these parameters for patients with TP53 wild-type neoplasms. We conclude that TP53 mutations predict sensitivity to VEGF/VEGFR inhibitors in the clinic. TP53 alterations may therefore be a ready biomarker for treatment with antiangiogenesis agents, a finding of seminal importance across the cancer field. Mol Cancer Ther; 15(10); 2475-85. ©2016 AACR.
Collapse
Affiliation(s)
- Jennifer J Wheler
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Filip Janku
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yali Li
- Foundation Medicine, Cambridge, Massachusetts
| | - Bettzy Stephen
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ralph Zinner
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel Karp
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sarina Piha-Paul
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Roosevelt Anderson
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Danxia Ke
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | | - J Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy, Moores Cancer Center, The University of California, San Diego, La Jolla, California.
| |
Collapse
|
45
|
Vorinostat in refractory soft tissue sarcomas - Results of a multi-centre phase II trial of the German Soft Tissue Sarcoma and Bone Tumour Working Group (AIO). Eur J Cancer 2016; 64:74-82. [PMID: 27367154 DOI: 10.1016/j.ejca.2016.05.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 05/15/2016] [Accepted: 05/17/2016] [Indexed: 11/22/2022]
Abstract
INTRODUCTION New treatment options for patients with metastatic Soft Tissue Sarcoma are urgently needed. Preclinical studies suggested activity of vorinostat, a histone deacetylase inhibitor. METHODS A multi-centre, open-label, non-randomised phase II trial to investigate the efficacy and safety of vorinostat in patients with locally advanced or metastatic Soft Tissue Sarcoma failing 1st-line anthracycline-based chemotherapy was initiated. Patients were treated with vorinostat 400 mg po qd for 28 d followed by a treatment-free period of 7 d, representing a treatment cycle of 5 weeks. Restaging was performed every three cycles or at clinical progression. RESULTS Between 06/10 and 09/13, 40 Soft Tissue Sarcoma patients were treated with vorinostat at seven participating centres. Patients had received 1 (n=8, 20%), 2 (n=10, 25%) or ≥3 (n=22, 55%) previous lines of chemotherapy. Best response after three cycles of treatment was stable disease (n=9, 23%). Median progression-free survival and overall survival were 3.2 and 12.3 months, respectively. Six patients showed long-lasting disease stabilisation for up to ten cycles. Statistical analyses failed to identify baseline predictive markers in this subgroup. Major toxicities (grade ≥III) included haematological toxicity (n=6, 15%) gastrointestinal disorders (n=5, 13%), fatigue (n=4, 10%), musculoskeletal pain (n=4, 10%), and pneumonia (n=2, 5%). CONCLUSION In a heavily pre-treated patient population, objective response to vorinostat was low. However, a small subgroup of patients had long-lasting disease stabilisation. Further studies aiming to identify predictive markers for treatment response as well as exploration of combination regimens are warranted. TRIAL REGISTRATION NCT00918489 (ClinicalTrials.gov) EudraCT-number: 2008-008513-19.
Collapse
|
46
|
Cranmer LD, Loggers ET, Pollack SM. Pazopanib in the management of advanced soft tissue sarcomas. Ther Clin Risk Manag 2016; 12:941-55. [PMID: 27354810 PMCID: PMC4907704 DOI: 10.2147/tcrm.s84792] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Therapy of soft tissue sarcomas represents an area of significant unmet need in oncology. Angiogenesis has been explored as a potential target both preclinically and clinically, with suggestions of activity. Pazopanib is a multitargeted tyrosine kinase inhibitor with prominent antiangiogenic effects. In a Phase II study, pazopanib demonstrated activity in strata enrolling patients with leiomyosarcomas, synovial sarcomas, or other sarcomas but not those enrolling adipocytic sarcomas. PALETTE, the pivotal Phase III trial, demonstrated improved progression-free survival versus placebo in pazopanib-treated patients previously treated for advanced soft tissue sarcomas. No survival benefit was observed, and adipocytic sarcomas were excluded. Health-related quality-of-life assessments indicated significant decrements in several areas affected by pazopanib toxicities, but no global deterioration. Cost-effectiveness analyses indicate that pazopanib therapy may or may not be cost-effective in different geographic settings. Pazopanib provides important proof-of-concept for antiangiogenic therapy in soft tissue sarcomas. Its use can be improved by further biological studies of its activity profile in sarcomas, studies of biological rational combinations, and clinicopathologic/biological correlative studies of activity to allow better drug targeting.
Collapse
Affiliation(s)
- Lee D Cranmer
- Division of Medical Oncology, University of Washington, Seattle, WA, USA
| | - Elizabeth T Loggers
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Seth M Pollack
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
47
|
Nervi C, De Marinis E, Codacci-Pisanelli G. Epigenetic treatment of solid tumours: a review of clinical trials. Clin Epigenetics 2015; 7:127. [PMID: 26692909 PMCID: PMC4676165 DOI: 10.1186/s13148-015-0157-2] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 11/10/2015] [Indexed: 12/12/2022] Open
Abstract
Epigenetic treatment has been approved by regulatory agencies for haematological malignancies. The success observed in cutaneous lymphomas represents a proof of principle that similar results may be obtained in solid tumours. Several agents that interfere with DNA methylation-demethylation and histones acetylation/deacetylation have been studied, and some (such as azacytidine, decitabine, valproic acid and vorinostat) are already in clinical use. The aim of this review is to provide a brief overview of the molecular events underlying the antitumour effects of epigenetic treatments and to summarise data available on clinical trials that tested the use of epigenetic agents against solid tumours. We not only list results but also try to indicate how the proper evaluation of this treatment might result in a better selection of effective agents and in a more rapid development. We divided compounds in demethylating agents and HDAC inhibitors. For each class, we report the antitumour activity and the toxic side effects. When available, we describe plasma pharmacokinetics and pharmacodynamic evaluation in tumours and in surrogate tissues (generally white blood cells). Epigenetic treatment is a reality in haematological malignancies and deserves adequate attention in solid tumours. A careful consideration of available clinical data however is required for faster drug development and possibly to re-evaluate some molecules that were perhaps discarded too early.
Collapse
Affiliation(s)
- Clara Nervi
- Department of Medical and Surgical Sciences and Biotechnology, University of Rome "la Sapienza", Corso della Repubblica, 97, 04100 Latina, Italy
| | - Elisabetta De Marinis
- Department of Medical and Surgical Sciences and Biotechnology, University of Rome "la Sapienza", Corso della Repubblica, 97, 04100 Latina, Italy
| | - Giovanni Codacci-Pisanelli
- Department of Medical and Surgical Sciences and Biotechnology, University of Rome "la Sapienza", Corso della Repubblica, 97, 04100 Latina, Italy
| |
Collapse
|
48
|
Koehler K, Liebner D, Chen JL. TP53 mutational status is predictive of pazopanib response in advanced sarcomas. Ann Oncol 2015; 27:539-43. [PMID: 26646755 DOI: 10.1093/annonc/mdv598] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 11/20/2015] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND To investigate whether TP53 DNA mutational status impacts progression-free survival (PFS) in patients with advanced sarcomas (soft tissue sarcoma) treated with vascular endothelial growth factor receptors (VEGFR) inhibition. PATIENTS AND METHODS We retrospectively reviewed 19 cases of patients treated at the Ohio State James Comprehensive Cancer Center with advanced sarcoma treated with VEGFR inhibition who also had next-generation sequencing of their tumors (via FoundationOne Heme panel). We evaluated TP53 as well as mutations that were observed in at least 20% of patients and evaluated its contribution to PFS using the Kaplan-Meier survival analysis of available radiology end points. RESULTS Mutations that were observed in at least 20% of patients included TP53 and Rb1. Only TP53 was predictive of PFS in the context of VEGFR inhibition. The PFS of patients with TP53 mutations was significantly greater than TP53 wild-type tumors with the median PFS of 208 versus 136 days, respectively [P = 0.036, hazards ratio 0.38 (95% confidence interval 0.09-0.83)]. CONCLUSIONS Mutations in TP53 may serve as a predictive biomarker of response to VEGFR inhibition in patients with advanced sarcoma. Larger, prospective studies are necessary to confirm these findings.
Collapse
Affiliation(s)
- K Koehler
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus
| | - D Liebner
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus Division of Bioinformatics, Department of Biomedical Informatics, The Ohio State University, Columbus, USA
| | - J L Chen
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus Division of Bioinformatics, Department of Biomedical Informatics, The Ohio State University, Columbus, USA
| |
Collapse
|
49
|
Wang Z, Fu S. An overview of tyrosine kinase inhibitors for the treatment of epithelial ovarian cancer. Expert Opin Investig Drugs 2015; 25:15-30. [PMID: 26560712 DOI: 10.1517/13543784.2016.1117071] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy and the fifth most common cause of cancer-related deaths in women. Initial treatment with surgery and chemotherapy has improved survival significantly. However, the disease progresses or recurs in most patients. Thus, there is an urgent need to develop more effective treatment strategies. AREAS COVERED This article provides an overview of tyrosine kinase inhibitors (TKIs) for the treatment of EOC, which is based on English peer-reviewed articles on MEDLINE and related abstracts presented at major conferences. The authors highlight the data from the published clinical trials in EOC patients who were treated with TKIs or TKI-based regimens. EXPERT OPINION EOC is responsive to most chemotherapeutic drugs and/or biological agents and represents an ideal disease model for investigating novel anti-cancer agents. Numerous small-molecule TKIs targeting the VEGFR, PARP, PI3K-AKT-mTOR, MAPK, Src, PKC, Wee1 and HER1/2 signaling pathways are currently being tested in clinical trials. Research is needed for devising regimens combining TKIs with other agents in an optimal timing schedule and for identifying potential biomarkers predictive of response and survival.
Collapse
Affiliation(s)
- Zhijie Wang
- a Department of Investigational Cancer Therapeutics , The University of Texas MD Anderson Cancer Center , 1515 Holcombe Boulevard, Houston , TX 77030 , USA.,b Department of Thoracic Medical Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education) , Peking University Cancer Hospital & Beijing Institute for Cancer Research , Beijing , China
| | - Siqing Fu
- a Department of Investigational Cancer Therapeutics , The University of Texas MD Anderson Cancer Center , 1515 Holcombe Boulevard, Houston , TX 77030 , USA
| |
Collapse
|
50
|
Ali SR, Humphreys KJ, McKinnon RA, Michael MZ. Impact of Histone Deacetylase Inhibitors on microRNA Expression and Cancer Therapy: A Review. Drug Dev Res 2015; 76:296-317. [PMID: 26303212 DOI: 10.1002/ddr.21268] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chromatin-modifying drugs, such as histone deacetylase inhibitors (HDACi), have shown potential as cancer therapeutics, either alone or in combination with other therapies. HDACi have the ability to reverse aberrant epigenetic modifications associated with cancer, namely dysregulated histone acetylation. There are currently three FDA approved HDACi; vorinostat, romidepsin, and panobinostat. Epigenetic modifications can regulate the expression of protein coding genes, and in addition can alter expression of microRNA (miRNA) genes. Many miRNAs play key roles in cell proliferation and apoptosis, and are commonly dysregulated in cancer states. A number of in vitro and in vivo studies have demonstrated the ability of chromatin-modifying drugs to alter miRNA expression, which may provide the basis for further investigation of miRNAs as therapeutic targets or as biomarkers of drug response. This review summarises findings from studies investigating the effects of HDACi on miRNA expression, as well as key clinical trials involving HDACi. Understanding how chromatin-modifying drugs epigenetically modulate miRNA genes provides further insight into the cellular mechanisms that deliver therapeutic responses, and may assist in refining treatment strategies.
Collapse
Affiliation(s)
- Saira R Ali
- Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, Adelaide, South Australia, Australia
| | - Karen J Humphreys
- Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, Adelaide, South Australia, Australia
| | - Ross A McKinnon
- Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, Adelaide, South Australia, Australia
| | - Michael Z Michael
- Flinders Centre for Innovation in Cancer, School of Medicine, Flinders University, Adelaide, South Australia, Australia.,Department of Gastroenterology and Hepatology, Flinders Medical Centre, Adelaide, South Australia, Australia
| |
Collapse
|