1
|
Baghel US, Kriplani P, Patel NM, Kaur M, Sharma K, Meghani M, Sharma A, Singh D, Singh B, Setzer WN, Sharifi-Rad J, Calina D. Flavopiridol: a promising cyclin-dependent kinase inhibitor in cancer treatment. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3489-3511. [PMID: 39589530 DOI: 10.1007/s00210-024-03599-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/01/2024] [Indexed: 11/27/2024]
Abstract
Flavopiridol, a synthetic flavonoid derived from rohitukine, stands out as a powerful cyclin-dependent kinase (CDK) inhibitor with significant anticancer properties. Its action mechanisms involve inducing cell cycle arrest, triggering apoptosis, and inhibiting transcription across various cancer types. Despite these promising effects, flavopiridol's clinical use has been hampered by issues related to toxicity and drug resistance. This study aims to comprehensively review flavopiridol's mechanisms of action, structure-activity relationships, synthetic derivatives, pharmacokinetics, and its potential role in clinical applications, with a focus on how combination therapies can enhance its efficacy and address resistance challenges in cancer treatment. A thorough analysis of key studies was performed, examining flavopiridol's anticancer properties, emphasizing its structure-activity relationships, synthetic modifications, and clinical outcomes. The anticancer effects of flavopiridol are primarily driven by its inhibition of CDKs, induction of apoptosis, promotion of oxidative stress, and antiangiogenic activity. Modifications in its chemical structure, especially in the D ring, have shown a significant impact on its CDK inhibitory potency. Several synthetic derivatives have also demonstrated enhanced anticancer activity. While preclinical models highlight flavopiridol's potential in treating cancers such as leukemia and solid tumors, clinical trials have brought attention to its limitations, particularly regarding toxicity and resistance. However, flavopiridol remains a promising candidate for cancer therapy, especially when used in combination with other treatments. Future research efforts should focus on refining its therapeutic profile, minimizing toxicity, and investigating synergistic treatment combinations, including those with immunotherapy. Understanding the mechanisms of resistance and discovering predictive biomarkers will be crucial for its effective integration into clinical practice.
Collapse
Affiliation(s)
| | | | | | - Manpreet Kaur
- Gurukul Pharmacy College, Ranpur, 325003, Kota, India
| | - Kapil Sharma
- Gurukul Pharmacy College, Ranpur, 325003, Kota, India
| | | | - Abhay Sharma
- Department of Pharmacy, University of Kota, Kota, 324005, Rajasthan, India
| | | | - Bhawani Singh
- Deparment of Pure and Applied Chemistry, University of Kota, Kota, 324005, India
| | - William N Setzer
- Aromatic Plant Research Center, 230 N 1200 E, Suite 100, Lehi, UT, 84043, USA
- Department of Chemistry, University of Alabama in Huntsville, Huntsville, AL, 35899, USA
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, Ecuador.
- Centro de Estudios Tecnológicos, Universitarios del Golfo, Veracruz, Mexico.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| |
Collapse
|
2
|
Walker RL, Hornicek FJ, Duan Z. Transcriptional regulation and therapeutic potential of cyclin-dependent kinase 9 (CDK9) in sarcoma. Biochem Pharmacol 2024; 226:116342. [PMID: 38848777 DOI: 10.1016/j.bcp.2024.116342] [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: 04/01/2024] [Revised: 05/17/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Sarcomas include various subtypes comprising two significant groups - soft tissue and bone sarcomas. Although the survival rate for some sarcoma subtypes has improved over time, the current methods of treatment remain efficaciously limited, as recurrent, and metastatic diseases remain a major obstacle. There is a need for better options and therapeutic strategies in treating sarcoma. Cyclin dependent kinase 9 (CDK9) is a transcriptional kinase and has emerged as a promising target for treating various cancers. The aberrant expression and activation of CDK9 have been observed in several sarcoma subtypes, including rhabdomyosarcoma, synovial sarcoma, osteosarcoma, Ewing sarcoma, and chordoma. Enhanced CDK9 expression has also been correlated with poorer prognosis in sarcoma patients. As a master regulator of transcription, CDK9 promotes transcription elongation by phosphorylation and releasing RNA polymerase II (RNAPII) from its promoter proximal pause. Release of RNAPII from this pause induces transcription of critical genes in the tumor cell. Overexpression and activation of CDK9 have been observed to lead to the expression of oncogenes, including MYC and MCL-1, that aid sarcoma development and progression. Inhibition of CDK9 in sarcoma has been proven to reduce these oncogenes' expression and decrease proliferation and growth in different sarcoma cells. Currently, there are several CDK9 inhibitors in preclinical and clinical investigations. This review aims to highlight the recent discovery and results on the transcriptional role and therapeutic potential of CDK9 in sarcoma.
Collapse
Affiliation(s)
- Robert L Walker
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 N.W. 10(th) Avenue, Miami, FL 33136. USA
| | - Francis J Hornicek
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 N.W. 10(th) Avenue, Miami, FL 33136. USA
| | - Zhenfeng Duan
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 N.W. 10(th) Avenue, Miami, FL 33136. USA.
| |
Collapse
|
3
|
Honoki K, Tsujiuchi T, Kishi S, Kuniyasu H. Revisiting 'Hallmarks of Cancer' In Sarcomas. J Cancer 2024; 15:1786-1804. [PMID: 38434982 PMCID: PMC10905407 DOI: 10.7150/jca.92844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 01/08/2024] [Indexed: 03/05/2024] Open
Abstract
There is no doubt that anyone who has participated in cancer care or research has once read the 'Hallmarks of Cancer' papers published by Hanahan and Weinberg in 2001 and 2011. They initially defined the six qualities of cancer cells as cancer hallmarks in 2001, but expanded that to 11 as a next generation in 2011. In their papers, they discussed the potential treatment strategies against cancer corresponding to each of the 11 hallmarks, and to date, proposed therapies that target genes and signaling pathways associated with each of these hallmarks have guided a trail that cancer treatments should take, some of which are now used as standard in clinical practice and some of which have yet to progress that far. Along with the recent advances in cancer research such as genomic analysis with next generation sequencing, they can be reconverged to an alternative six categories defined as selective proliferative advantages, altered stress response, deregulated cellular metabolism, immune modulation and inflammation, tumor microenvironment, tissue invasion and metastasis. In this paper, we will overview the current state of these alternative hallmarks and their corresponding treatments in the current sarcoma practice, then discuss the future direction of sarcoma treatment.
Collapse
Affiliation(s)
- Kanya Honoki
- Dept. Of Orthopedic Oncology & Reconstructive Medicine, Nara Medical University, Japan
| | | | - Shingo Kishi
- Dept. of Clinical Pathology, Nozaki Tokushukai Hospital, Japan
| | - Hiroki Kuniyasu
- Dept. of Molecular Pathology, Nara Medical University, Japan
| |
Collapse
|
4
|
Wang Q, Bode AM, Zhang T. Targeting CDK1 in cancer: mechanisms and implications. NPJ Precis Oncol 2023; 7:58. [PMID: 37311884 DOI: 10.1038/s41698-023-00407-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/25/2023] [Indexed: 06/15/2023] Open
Abstract
Cyclin dependent kinases (CDKs) are serine/threonine kinases that are proposed as promising candidate targets for cancer treatment. These proteins complexed with cyclins play a critical role in cell cycle progression. Most CDKs demonstrate substantially higher expression in cancer tissues compared with normal tissues and, according to the TCGA database, correlate with survival rate in multiple cancer types. Deregulation of CDK1 has been shown to be closely associated with tumorigenesis. CDK1 activation plays a critical role in a wide range of cancer types; and CDK1 phosphorylation of its many substrates greatly influences their function in tumorigenesis. Enrichment of CDK1 interacting proteins with Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis was conducted to demonstrate that the associated proteins participate in multiple oncogenic pathways. This abundance of evidence clearly supports CDK1 as a promising target for cancer therapy. A number of small molecules targeting CDK1 or multiple CDKs have been developed and evaluated in preclinical studies. Notably, some of these small molecules have also been subjected to human clinical trials. This review evaluates the mechanisms and implications of targeting CDK1 in tumorigenesis and cancer therapy.
Collapse
Affiliation(s)
- Qiushi Wang
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN, 55912, USA
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN, 55912, USA.
| | - Tianshun Zhang
- The Hormel Institute, University of Minnesota, 801 16th Ave NE, Austin, MN, 55912, USA.
| |
Collapse
|
5
|
Thiel JT, Daigeler A, Kolbenschlag J, Rachunek K, Hoffmann S. The Role of CDK Pathway Dysregulation and Its Therapeutic Potential in Soft Tissue Sarcoma. Cancers (Basel) 2022; 14:3380. [PMID: 35884441 PMCID: PMC9323700 DOI: 10.3390/cancers14143380] [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: 05/17/2022] [Revised: 07/07/2022] [Accepted: 07/09/2022] [Indexed: 02/04/2023] Open
Abstract
Soft tissue sarcomas (STSs) are tumors that are challenging to treat due to their pathologic and molecular heterogeneity and their tumor biology that is not yet fully understood. Recent research indicates that dysregulation of cyclin-dependent kinase (CDK) signaling pathways can be a strong driver of sarcogenesis. CDKs are enzyme forms that play a crucial role in cell-cycle control and transcription. They belong to the protein kinases group and to the serine/threonine kinases subgroup. Recently identified CDK/cyclin complexes and established CDK/cyclin complexes that regulate the cell cycle are involved in the regulation of gene expression through phosphorylation of critical components of transcription and pre-mRNA processing mechanisms. The current and continually growing body of data shows that CDKs play a decisive role in tumor development and are involved in the proliferation and growth of sarcoma cells. Since the abnormal expression or activation of large numbers of CDKs is considered to be characteristic of cancer development and progression, dysregulation of the CDK signaling pathways occurs in many subtypes of STSs. This review discusses how reversal and regulation can be achieved with new therapeutics and summarizes the current evidence from studies regarding CDK modulation for STS treatment.
Collapse
Affiliation(s)
- Johannes Tobias Thiel
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Unfallklinik Tuebingen, University of Tuebingen, 72076 Tuebingen, Germany; (A.D.); (J.K.); (K.R.); (S.H.)
| | | | | | | | | |
Collapse
|
6
|
Tong Z, Mejia A, Veeranki O, Verma A, Correa AM, Dokey R, Patel V, Solis LM, Mino B, Kathkuda R, Rodriguez-Canales J, Lin SH, Krishnan S, Kopetz S, Blum M, Ajani JA, Hofstetter WL, Maru DM. Targeting CDK9 and MCL-1 by a new CDK9/p-TEFb inhibitor with and without 5-fluorouracil in esophageal adenocarcinoma. Ther Adv Med Oncol 2019; 11:1758835919864850. [PMID: 31384313 PMCID: PMC6659187 DOI: 10.1177/1758835919864850] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 06/16/2019] [Indexed: 01/03/2023] Open
Abstract
Background: CDK9 inhibitors are antitumorigenic against solid tumors, including
esophageal adenocarcinoma (EAC). However, efficacy of a CDK9 inhibitor
combined with 5-fluorouracil (5-FU) and target proteins that are targeted by
these agents in EAC are unknown. Methods: The anti-EAC efficacy of a new CDK9 inhibitor, BAY1143572, with and without
5-FU was assessed in vitro and in xenograft models in
athymic nu/nu mice. Synergy between BAY1143572 and 5-FU in inhibiting cell
proliferation was analyzed by calculating the combination index using
CompuSyn software. Potential targets of BAY1143572 and 5-FU were identified
by reverse-phase protein array. The effects of BAY1143572 and 5-FU on MCL-1
in vitro were analyzed by Western blotting,
quantitative real-time polymerase chain reaction, and chromatin
immunoprecipitation assay. MCL-1 protein expression in tumors from patients
with locoregional EAC treated with chemoradiation and surgery was assessed
by immunohistochemistry. Results: BAY1143572 had dose-dependent antiproliferative and proapoptotic effects and
demonstrated synergy with 5-FU against EAC in vitro. The
median volumes of FLO-1 and ESO-26 xenografts treated with 5-FU plus
BAY114352 were significantly smaller than those of xenografts treated with
either agent alone (p < 0.05). BAY1143572 downregulated
MCL-1 by inhibiting HIF-1α binding to the MCL-1 promoter. 5-FU enhanced
BAY1143572-induced MCL-1 downregulation and stable MCL-1 overexpression
reduced the apoptosis induced by BAY1143572 and 5-FU in
vitro. High patients’ tumor MCL-1 expression was correlated
with shorter overall and recurrence-free survival. Conclusions: BAY1143572 and 5-FU have synergistic antitumorigenic effects against EAC.
MCL-1 is a downstream target of CDK9 inhibitors and a predictor of response
to neoadjuvant chemoradiation in EAC.
Collapse
Affiliation(s)
- Zhimin Tong
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alicia Mejia
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Omkara Veeranki
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anuj Verma
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Arlene M Correa
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rashmi Dokey
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Viren Patel
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luisa Maren Solis
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Barbara Mino
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Riham Kathkuda
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jaime Rodriguez-Canales
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sunil Krishnan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mariela Blum
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wayne L Hofstetter
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dipen M Maru
- Division of Pathology and Laboratory Medicine, Unit 085, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| |
Collapse
|
7
|
Gallego-Lleyda A, De Miguel D, Anel A, Martinez-Lostao L. Lipid Nanoparticles Decorated with TNF-Related Aptosis-Inducing Ligand (TRAIL) Are More Cytotoxic than Soluble Recombinant TRAIL in Sarcoma. Int J Mol Sci 2018; 19:ijms19051449. [PMID: 29757258 PMCID: PMC5983602 DOI: 10.3390/ijms19051449] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 04/29/2018] [Accepted: 05/11/2018] [Indexed: 01/25/2023] Open
Abstract
Sarcomas are rare and heterogeneous cancers classically associated with a poor outcome. Sarcomas are 1% of the cancer but recent estimations indicate that sarcomas account for 2% of the estimated cancer-related deaths. Traditional treatment with surgery, radiotherapy, and chemotherapy has improved the outcome for some types of sarcomas. However, novel therapeutic strategies to treat sarcomas are necessary. TNF-related apoptosis-inducing ligand (TRAIL) is a death ligand initially described as capable of inducing apoptosis on tumor cell while sparing normal cells. Only few clinical trials have used TRAIL-based treatments in sarcoma, but they show only low or moderate efficacy of TRAIL. Consequently, novel TRAIL formulations with an improved TRAIL bioactivity are necessary. Our group has developed a novel TRAIL formulation based on tethering this death ligand on a lipid nanoparticle surface (LUV-TRAIL) resembling the physiological secretion of TRAIL as a trasmembrane protein inserted into the membrane of exosomes. We have already demonstrated that LUV-TRAIL shows an improved cytotoxic activity when compared to soluble recombinant TRAIL both in hematological malignancies and epithelial-derived cancers. In the present study, we have tested LUV-TRAIL in several human sarcoma tumor cell lines with different sensitivity to soluble recombinant TRAIL, finding that LUV-TRAIL was more efficient than soluble recombinant TRAIL. Moreover, combined treatment of LUV-TRAIL with distinct drugs proved to be especially effective, sensitizing even more resistant cell lines to TRAIL.
Collapse
Affiliation(s)
- Ana Gallego-Lleyda
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50009 Zaragoza, Spain.
- Instituto de Investigación Sanitaria de Aragón (ISS), 50009 Zaragoza, Spain.
| | - Diego De Miguel
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50009 Zaragoza, Spain.
- Cell Death, Cancer and Inflammation, University College of London, London WC1E 6BT, UK.
| | - Alberto Anel
- Departamento de Bioquímica, Biología Molecular y Celular, Universidad de Zaragoza, 50009 Zaragoza, Spain.
- Instituto de Investigación Sanitaria de Aragón (ISS), 50009 Zaragoza, Spain.
| | - Luis Martinez-Lostao
- Instituto de Investigación Sanitaria de Aragón (ISS), 50009 Zaragoza, Spain.
- Servicio de Inmunología, Hospital Clínico Universitario Lozano Blesa, 50009 Zaragoza, Spain.
- Departamento de Microbiología, Medicina Preventiva y Salud Pública, Universidad de Zaragoza, 50009 Zaragoza, Spain.
- Instituto de Nanociencia de Aragón, 50009 Zaragoza, Spain.
| |
Collapse
|
8
|
Deep A, Marwaha RK, Marwaha MG, Jyoti J, Nandal R, Sharma AK. Flavopiridol as cyclin dependent kinase (CDK) inhibitor: a review. NEW J CHEM 2018. [DOI: 10.1039/c8nj04306j] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Flavopiridol (alvocidib) is a synthetic flavonoid based on the extract from an Indian plant developed for potential treatment of cancer whose first clinical trials were initiated in 1994 as a frontline combination therapy for leukaemia.
Collapse
Affiliation(s)
- Aakash Deep
- Department of Pharmaceutical Sciences
- Chaudhary Bansi Lal University
- Bhiwani-127021
- India
| | - Rakesh Kumar Marwaha
- Department of Pharmaceutical Sciences
- Maharshi Dayanand University
- Rohtak-124001
- India
| | | | - Jyoti Jyoti
- Department of Pharmaceutical Sciences
- Maharshi Dayanand University
- Rohtak-124001
- India
| | - Rimmy Nandal
- Department of Pharmaceutical Sciences
- Chaudhary Bansi Lal University
- Bhiwani-127021
- India
| | - Arun Kumar Sharma
- Department of Pharmacology
- Amity Institute of Pharmacy
- Amity University
- Gurugram
- India
| |
Collapse
|
9
|
Bleloch JS, Ballim RD, Kimani S, Parkes J, Panieri E, Willmer T, Prince S. Managing sarcoma: where have we come from and where are we going? Ther Adv Med Oncol 2017; 9:637-659. [PMID: 28974986 PMCID: PMC5613860 DOI: 10.1177/1758834017728927] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 07/26/2017] [Indexed: 02/06/2023] Open
Abstract
Sarcomas are a heterogeneous group of neoplasms of mesenchymal origin. Approximately 80% arise from soft tissue and 20% originate from bone. To date more than 100 sarcoma subtypes have been identified and they vary in molecular characteristics, pathology, clinical presentation and response to treatment. While sarcomas represent <1% of adult cancers, they account for approximately 21% of paediatric malignancies and thus pose some of the greatest risks of mortality and morbidity in children and young adults. Metastases occur in one-third of all patients and approximately 10-20% of sarcomas recur locally. Surgery in combination with preoperative and postoperative therapies is the primary treatment for localized sarcoma tumours and is the most promising curative possibility. Metastasized sarcomas, on the other hand, are treated primarily with single-agent or combination chemotherapy, but this rarely leads to a complete and robust response and often becomes a palliative form of treatment. The heterogeneity of sarcomas results in variable responses to current generalized treatment strategies. In light of this and the lack of curative strategies for metastatic and unresectable sarcomas, there is a need for novel subtype-specific treatment strategies. With the more recent understanding of the molecular mechanisms underlying the pathogenesis of some of these tumours, the treatment of sarcoma subtypes with targeted therapies is a rapidly evolving field. This review discusses the current management of sarcomas as well as promising new therapies that are currently underway in clinical trials.
Collapse
Affiliation(s)
- Jenna S Bleloch
- Department of Human Biology, University of Cape Town, South Africa
| | - Reyna D Ballim
- Department of Human Biology, University of Cape Town, South Africa
| | - Serah Kimani
- Department of Human Biology, University of Cape Town, South Africa
| | - Jeannette Parkes
- Department of Radiation Oncology, University of Cape Town, South Africa
| | - Eugenio Panieri
- Department of Surgery, University of Cape Town, South Africa
| | - Tarryn Willmer
- Department of Human Biology, University of Cape Town, South Africa
| | - Sharon Prince
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, 7925, South Africa
| |
Collapse
|
10
|
The antineoplastic drug flavopiridol reverses memory impairment induced by Amyloid-ß1-42 oligomers in mice. Pharmacol Res 2016; 106:10-20. [PMID: 26875816 DOI: 10.1016/j.phrs.2016.02.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 02/03/2016] [Accepted: 02/05/2016] [Indexed: 01/06/2023]
Abstract
The ectopic re-activation of cell cycle in neurons is an early event in the pathogenesis of Alzheimer's disease (AD), which could lead to synaptic failure and ensuing cognitive deficits before frank neuronal death. Cytostatic drugs that act as cyclin-dependent kinase (CDK) inhibitors have been poorly investigated in animal models of AD. In the present study, we examined the effects of flavopiridol, an inhibitor of CDKs currently used as antineoplastic drug, against cell cycle reactivation and memory loss induced by intracerebroventricular injection of Aß1-42 oligomers in CD1 mice. Cycling neurons, scored as NeuN-positive cells expressing cyclin A, were found both in the frontal cortex and in the hippocampus of Aβ-injected mice, paralleling memory deficits. Starting from three days after Aβ injection, flavopiridol (0.5, 1 and 3mg/kg) was intraperitoneally injected daily, for eleven days. Here we show that a treatment with flavopiridol (0.5 and 1mg/kg) was able to rescue the loss of memory induced by Aβ1-42, and to prevent the occurrence of ectopic cell-cycle events in the mouse frontal cortex and hippocampus. This is the first evidence that a cytostatic drug can prevent cognitive deficits in a non-transgenic animal model of AD.
Collapse
|
11
|
Sborov D, Chen JL. Targeted therapy in sarcomas other than GIST tumors. J Surg Oncol 2015; 111:632-40. [PMID: 25330750 PMCID: PMC4436975 DOI: 10.1002/jso.23802] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 08/13/2014] [Indexed: 01/01/2023]
Abstract
Non-GIST soft tissue sarcomas are a heterogeneous grouping of mesenchymal tumors that comprise less than 1% of adult malignancies. Treatment continues to be based on cytotoxic chemotherapy regimens. However, characterization of the molecular pathway deregulations that drive these tumors has led to the emergence of more customized treatment options. In this review, we focus on the multitude of molecular inhibitors targeting angiogenesis and cell cycle pathways being tested in clinical trials.
Collapse
Affiliation(s)
- Douglas Sborov
- Hematology and Oncology Fellow, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - James L Chen
- Assistant Professor, Departments of Biomedical Informatics and Internal Medicine (Division of Medical Oncology), The Ohio State University, Columbus, OH, USA
| |
Collapse
|
12
|
Guan Z, Yu X, Wang H, Wang H, Zhang J, Li G, Cao J, Teng L. Advances in the targeted therapy of liposarcoma. Onco Targets Ther 2015; 8:125-36. [PMID: 25609980 PMCID: PMC4293924 DOI: 10.2147/ott.s72722] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Liposarcoma (LPS) is the most common type of soft-tissue sarcoma. Complete surgical resection is the only curative means for localized disease; however, both radiation and conventional cytotoxic chemotherapy remain controversial for metastatic or unresectable disease. An increasing number of trials with novel targeted therapy of LPS have provided encouraging data during recent years. This review will provide an overview of the advances in our understanding of LPS and summarize the results of recent trials with novel therapies targeting different genetic and molecular aberrations for different subtypes of LPS.
Collapse
Affiliation(s)
- Zhonghai Guan
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, People's Republic of China
| | - Xiongfei Yu
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, People's Republic of China
| | - Haohao Wang
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, People's Republic of China
| | - Haiyong Wang
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, People's Republic of China
| | - Jing Zhang
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, People's Republic of China
| | - Guangliang Li
- Department of Medicine Oncology, Zhejiang Cancer Hospital, Zhejiang, People's Republic of China
| | - Jiang Cao
- Clinical Research Center, The 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Lisong Teng
- Department of Surgical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, People's Republic of China
| |
Collapse
|
13
|
Honoki K, Tsujiuchi T. Senescence bypass in mesenchymal stem cells: a potential pathogenesis and implications of pro-senescence therapy in sarcomas. Expert Rev Anticancer Ther 2014; 13:983-96. [PMID: 23984899 DOI: 10.1586/14737140.2013.820010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Cellular senescence is a mechanism that limits the lifespan of somatic cells as the results of replicative proliferation and response to stresses, and that prevents undesired oncogenic changes constituting a barrier against immortalization and tumorigenesis. Mesenchymal stem cells (MSCs) reside in a variety of tissues, and participates in tissue maintenance with their multipotent differentiation ability. MSCs are also considered to be as cells of origin for certain type of sarcomas. We reviewed the mechanisms of cellular senescence in MSCs and hypothesized senescence bypass as the potential pathogenesis for sarcoma development, and proposed the possibility of senescence induction therapy for an alternative treatment strategy against sarcomas, especially cells with the resistance to conventional chemo and radiotherapy including sarcoma stem cells.
Collapse
Affiliation(s)
- Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan.
| | | |
Collapse
|
14
|
Schmitt T, Kasper B. New medical treatment options and strategies to assess clinical outcome in soft-tissue sarcoma. Expert Rev Anticancer Ther 2014; 9:1159-67. [PMID: 19671035 DOI: 10.1586/era.09.64] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Thomas Schmitt
- University of Heidelberg, Department of Internal Medicine V, Germany
| | | |
Collapse
|
15
|
Abstract
The past several decades have revealed certain challenges that are inherent to soft-tissue sarcomas with regards to devising, testing and setting treatment paradigms for such a rare and heterogeneous malignancy. Despite this, significant advances have been made through the efforts of scientists, clinicians and patients alike. We are now entering a molecular era of cancer and current biotechnology is beginning to unravel the pathogenic enigma of these often devastating tumors. As our understanding of these malignancies improves, so does our list of potential treatment options. The impetus now lies with the medical/scientific community to direct translational research and subsequently the development and clinical testing of novel compounds in a fashion that best serves this unique patient population. To do so, we must continue to integrate the lessons of the past with the resources and promise of the future. This review will outline current areas of therapeutic interest in soft-tissue sarcomas with regard to agents that have reached clinical testing.
Collapse
Affiliation(s)
- William D Tap
- UCLA Sarcoma Program, Division of Hematology/Oncology, Los Angeles, CA 90095-7059, USA.
| | | | | |
Collapse
|
16
|
Kim S, Lee J, Jang BC, Kwon TK, Park JW. BAI, a novel cyclin-dependent kinase inhibitor induces apoptosis in A549 cells through activation of caspases and inactivation of Akt. J Cell Biochem 2013; 114:282-93. [PMID: 22887215 DOI: 10.1002/jcb.24314] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 07/30/2012] [Indexed: 11/06/2022]
Abstract
Previously, we have synthesized a novel cyclin-dependent kinase (CDK) inhibitor, 2-[1,1'biphenyl]-4-yl-N-[5-(1,1-dioxo-1λ(6) -isothiazolidin-2-yl)-1H-indazol-3-yl]acetamide (BAI) and reported its anti-cancer activity in head and neck cancer cells. In this study, we further evaluated the effect of BAI on growth of various human cancer cell lines, including A549 (nonsmall cell lung cancer), HCT116 (colon), and Caki (kidney). Profoundly, results of XTT and clonogenic assays demonstrated that BAI at nanomolar concentrations (20-60 nM) inhibited growth of A549, HCT116, and Caki cells, suggesting the anti-cancer potency. We show that BAI induced a dose-dependent apoptotic cell death in these human cancer cells, as measured by fluorescence-activated cell sorting (FACS). Interestingly, further biochemical analysis showed that treatment with BAI at 20 nM induced apoptosis in A549 cells in association with activation of caspases, cleavage of phospholipase C-γ1 (PLC-γ1), and inhibition of Akt in A549 cells. Importantly, pharmacological inhibition study revealed that pretreatment with z-VAD-fmk, a pan caspase inhibitor strongly blocked the BAI-induced apoptosis in A549 cells. Transfection analysis with Akt cDNA encoding constitutively active Akt further addressed the significance of Akt inhibition in the BAI-induced apoptosis in A549 cells. Notably, disruption of the PI3K/Akt pathway by LY294002, a PI3K/Akt inhibitor potentiated apoptosis in A549 cells by BAI at a subcytotoxic concentration. These findings collectively suggest that BAI potently inhibits growth of A549, HCT116, and Caki cells, and that the BAI-induced apoptosis in A549 cells is associated with activation of caspases, and inhibition of Akt.
Collapse
Affiliation(s)
- Shin Kim
- Department of Immunology, School of Medicine, Keimyung University, 1095 Dalgubeoldaero, Dalseo-Gu, Daegu 704-701, South Korea
| | | | | | | | | |
Collapse
|
17
|
Fabbro D, Cowan-Jacob SW, Möbitz H, Martiny-Baron G. Targeting cancer with small-molecular-weight kinase inhibitors. Methods Mol Biol 2012; 795:1-34. [PMID: 21960212 DOI: 10.1007/978-1-61779-337-0_1] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Protein and lipid kinases fulfill essential roles in many signaling pathways that regulate normal cell functions. Deregulation of these kinase activities lead to a variety of pathologies ranging from cancer to inflammatory diseases, diabetes, infectious diseases, cardiovascular disorders, cell growth and survival. 518 protein kinases and about 20 lipid-modifying kinases are encoded by the human genome, and a much larger proportion of additional kinases are present in parasite, bacterial, fungal, and viral genomes that are susceptible to exploitation as drug targets. Since many human diseases result from overactivation of protein and lipid kinases due to mutations and/or overexpression, this enzyme class represents an important target for the pharmaceutical industry. Approximately one third of all protein targets under investigation in the pharmaceutical industry are protein or lipid kinases.The kinase inhibitors that have been launched, thus far, are mainly in oncology indications and are directed against a handful of protein and lipid kinases. With one exception, all of these registered kinase inhibitors are directed toward the ATP-site and display different selectivities, potencies, and pharmacokinetic properties. At present, about 150 kinase-targeted drugs are in clinical development and many more in various stages of preclinical development. Kinase inhibitor drugs that are in clinical trials target all stages of signal transduction from the receptor protein tyrosine kinases that initiate intracellular signaling, through second-messenger-dependent lipid and protein kinases, and protein kinases that regulate the cell cycle.This review provides an insight into protein and lipid kinase drug discovery with respect to achievements, binding modes of inhibitors, and novel avenues for the generation of second-generation kinase inhibitors to treat cancers.
Collapse
Affiliation(s)
- Doriano Fabbro
- Novartis Institutes for Biomedical Research, Expertise Platform Kinases, Basel, Switzerland.
| | | | | | | |
Collapse
|
18
|
Skropeta D, Pastro N, Zivanovic A. Kinase inhibitors from marine sponges. Mar Drugs 2011; 9:2131-2154. [PMID: 22073013 PMCID: PMC3210622 DOI: 10.3390/md9102131] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 10/01/2011] [Accepted: 10/14/2011] [Indexed: 01/08/2023] Open
Abstract
Protein kinases play a critical role in cell regulation and their deregulation is a contributing factor in an increasing list of diseases including cancer. Marine sponges have yielded over 70 novel compounds to date that exhibit significant inhibitory activity towards a range of protein kinases. These compounds, which belong to diverse structural classes, are reviewed herein, and ordered based upon the kinase that they inhibit. Relevant synthetic studies on the marine natural product kinase inhibitors have also been included.
Collapse
Affiliation(s)
- Danielle Skropeta
- School of Chemistry, University of Wollongong, Wollongong, NSW 2522, Australia; E-Mails: (N.P.); (A.Z.)
- Centre for Medicinal Chemistry, University of Wollongong, Wollongong, NSW 2522, Australia
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +61-2-42214360; Fax: +61-2-42214287
| | - Natalie Pastro
- School of Chemistry, University of Wollongong, Wollongong, NSW 2522, Australia; E-Mails: (N.P.); (A.Z.)
| | - Ana Zivanovic
- School of Chemistry, University of Wollongong, Wollongong, NSW 2522, Australia; E-Mails: (N.P.); (A.Z.)
| |
Collapse
|
19
|
Cicenas J, Valius M. The CDK inhibitors in cancer research and therapy. J Cancer Res Clin Oncol 2011; 137:1409-18. [PMID: 21877198 DOI: 10.1007/s00432-011-1039-4] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Accepted: 08/12/2011] [Indexed: 12/21/2022]
Abstract
Chemical compounds that interfere with an enzymatic function of kinases are useful for gaining insight into the complicated biochemical processes in mammalian cells. Cyclin-dependent kinases (CDK) play an essential role in the control of the cell cycle and/or proliferation. These kinases as well as their regulators are frequently deregulated in different human tumors. Aberrations in CDK activity have also been observed in viral infections, Alzheimer's, Parkinson's diseases, ischemia and some proliferative disorders. This led to an intensive search for small-molecule CDK inhibitors not only for research purposes, but also for therapeutic applications. Here, we discuss seventeen CDK inhibitors and their use in cancer research or therapy. This review should help researchers to decide which inhibitor is best suited for the specific purpose of their research. For this purpose, the targets, commercial availability and IC(50) values are provided for each inhibitor. The review will also provide an overview of the clinical studies performed with some of these inhibitors.
Collapse
Affiliation(s)
- Jonas Cicenas
- Department of Medicine, Institute of Anatomy, University of Fribourg, Rte. Albert- Gockel 1, 1700, Fribourg, Switzerland.
| | | |
Collapse
|
20
|
Haque A, Koide N, Iftakhar-E-Khuda I, Noman ASM, Odkhuu E, Badamtseren B, Naiki Y, Komatsu T, Yoshida T, Yokochi T. Flavopiridol inhibits lipopolysaccharide-induced TNF-α production through inactivation of nuclear factor-κB and mitogen-activated protein kinases in the MyD88-dependent pathway. Microbiol Immunol 2011; 55:160-7. [PMID: 21204955 DOI: 10.1111/j.1348-0421.2010.00304.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Flavopiridol is a cyclin-dependent kinase inhibitor and inhibits the growth of various cancer cells. The effect of flavopiridol on lipopolysaccharide (LPS)-induced proinflammatory mediator production was examined in RAW 264.7 macrophage-like cells. Flavopiridol significantly reduced the production of tumor necrosis factor-α and, to a lesser extent, nitric oxide in LPS-stimulated cells. Flavopiridol inhibited the activation of nuclear factor-κB and IκB kinase in response to LPS. Flavopiridol also inhibited the activation of a series of mitogen-activated protein kinases, such as p38, stress-activated protein kinase/c-Jun N-terminal kinase and extracellular signal-regulated kinase 1/2 in response to LPS. However, flavopiridol did not alter the expression of tumor necrosis factor receptor-associated factor 6, myeloid differentiation factor 88 (MyD88) or CD14/toll-like receptor (TLR) 4. Flavopiridol inhibited nitric oxide production induced by a MyD88-dependent TLR2 ligand, but not a MyD88-independent TLR3 ligand. Further, flavopiridol did not alter the phosphorylation of interferon regulatory factor 3 in the MyD88-independent pathway. Therefore, it was suggested that flavopiridol exclusively inhibited the activation of nuclear factor-κB and mitogen-activated protein kinases in the MyD88-dependent pathway. Flavopiridol might be useful for the prevention of LPS-induced inflammatory response.
Collapse
Affiliation(s)
- Abedul Haque
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Penel N, Van Glabbeke M, Marreaud S, Ouali M, Blay J, Hohenberger P. Testing new regimens in patients with advanced soft tissue sarcoma: analysis of publications from the last 10 years. Ann Oncol 2011; 22:1266-1272. [DOI: 10.1093/annonc/mdq608] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
22
|
Nitta N, Sonoda A, Seko A, Ohta S, Nagatani Y, Tsuchiya K, Otani H, Tanaka T, Kanasaki S, Takahashi M, Murata K. A combination of cisplatin-eluting gelatin microspheres and flavopiridol enhances anti-tumour effects in a rabbit VX2 liver tumour model. Br J Radiol 2009; 83:428-32. [PMID: 20019172 DOI: 10.1259/bjr/17506834] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The aim of this study was to investigate whether the combination of cisplatin-eluting gelatin microspheres (GMSs) and flavopiridol enhances anti-tumour effects in a rabbit VX2 liver tumour model. Tumour-bearing rabbits (n = 21) were divided into five groups and infused from the proper hepatic artery. Group 1 (n = 5) received cisplatin-eluting GMSs (1 mg kg(-1)) and flavopiridol (3 mg kg(-1)), group 2 (n = 5) cisplatin-eluting GMSs alone (1 mg kg(-1)), Group 3 (n = 5) flavopiridol (3 mg kg(-1)), Group 4 (n = 3) GMSs alone (1 mg kg(-1)), and Group 5 (n = 3) was the control group receiving physiological saline (1 ml kg(-1)). On days 0 and 7 after procedures the liver tumour volume was measured using a horizontal open MRI system and the relative tumour volume growth rates for 7 days after treatment were calculated. On T(1) weighted images, the tumours were visualised as circular, low-intensity areas just below the liver surface. After treatment, the signals remained similar. The relative tumour volume growth rate for 7 days after treatment was 54.2+/-22.4% in Group 1, 134.1+/-40.1% in Group 2,166.7+/-48.1% in Group 3, 341.8+/-8.6% in Group 4 and 583.1+/-46.9% in Group 5; the growth rate was significantly lower in Group 1 than the other groups (p<0.05). We concluded that in our rabbit model of liver tumours the combination of cisplatin-eluting GMSs and flavopiridol was effective.
Collapse
Affiliation(s)
- N Nitta
- Department of Radiology, Shiga University of Medical Science, Tsukinowa-cho, Seta, Otsu, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Jin Y, Chen Q, Shi X, Lu Z, Cheng C, Lai Y, Zheng Q, Pan J. Activity of triptolide against human mast cells harboring the kinase domain mutant KIT. Cancer Sci 2009; 100:1335-43. [PMID: 19383029 PMCID: PMC11159779 DOI: 10.1111/j.1349-7006.2009.01159.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Revised: 02/17/2009] [Accepted: 03/04/2009] [Indexed: 01/18/2023] Open
Abstract
Gain-of-function mutations of the receptor tyrosine kinase KIT can cause systemic mastocytosis (SM) and gastrointestinal stromal tumors. Most of the constitutively active KIT can be inhibited by imatinib; D816V KIT cannot. In this study, we investigated the activity of triptolide, a diterpenoid isolated from the Chinese herb Tripterygium wilfordii Hook. f., in cells expressing mutant KIT, including D816V KIT. Imatinib-sensitive HMC-1.1 cells harboring the mutation V560G in the juxtamembrane domain of KIT, imatinib-resistant HMC-1.2 cells harboring both V560G and D816V mutations, and murine P815 cells, were treated with triptolide, and analyzed in terms of growth, apoptosis, and signal transduction. The in vivo antitumor activity was evaluated by using the nude mouse xenograft model. Our results demonstrated that triptolide potently inhibits the growth of both human and murine mast cells harboring not only imatinib-sensitive KIT mutation but also imatinib-resistant D816V KIT. Triptolide markedly inhibited KIT mRNA levels and strikingly reduced the levels of phosphorylated and total Stat3, Akt, and Erk1/2, downstream targets of KIT. Triptolide triggered apoptosis by inducing depolarization of mitochondrial potential and release of cytochrome c, downregulation of Mcl-1 and XIAP. Furthermore, triptolide significantly abrogated the growth of imatinib-resistant HMC-1.2 cell xenografts in nude mice and decreased KIT expression in xenografts. Our data demonstrate that triptolide inhibits imatinib-resistant mast cells harboring D816V KIT. Further investigation of triptolide for treatment of human neoplasms driven by gain-of-function KIT mutations is warranted.
Collapse
Affiliation(s)
- Yanli Jin
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Gurumurthy M, Tan CH, Ng R, Zeiger L, Lau J, Lee J, Dey A, Philp R, Li Q, Lim TM, Price DH, Lane DP, Chao SH. Nucleophosmin interacts with HEXIM1 and regulates RNA polymerase II transcription. J Mol Biol 2008; 378:302-17. [PMID: 18371977 DOI: 10.1016/j.jmb.2008.02.055] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2007] [Revised: 02/18/2008] [Accepted: 02/25/2008] [Indexed: 11/17/2022]
Abstract
Hexamethylene bis-acetamide-inducible protein 1 (HEXIM1) was identified earlier as an inhibitor of positive transcription elongation factor b (P-TEFb), which is a key transcriptional regulator of RNA polymerase II (Pol II). Studies show that more than half of P-TEFb in cells is associated with HEXIM1, which results in the inactivation of P-TEFb. Here, we identify a nucleolar protein, nucleophosmin (NPM), as a HEXIM1-binding protein. NPM binds to HEXIM1 in vitro and in vivo, and functions as a negative regulator of HEXIM1. Over-expression of NPM leads to proteasome-mediated degradation of HEXIM1, resulting in activation of P-TEFb-dependent transcription. In contrast, an increase in HEXIM1 protein levels and a decrease in transcription are detected when NPM is knocked down. We show that a cytoplasmic mutant of NPM, NPMc+, associates with and sequesters HEXIM1 in the cytoplasm resulting in higher RNA Pol II transcription. Correspondingly, cytoplasmic localization of endogenous HEXIM1 is detected in an acute myeloid leukemia (AML) cell line containing the NPMc+ mutation, suggesting the physiological importance of HEXIM1-NPMc+ interaction. Over-expression of NPM has been detected in tumors of various histological origins and our results may provide a possible molecular mechanism for the proto-oncogenic function of NPM. Furthermore, considering that 35% of AML patients are diagnosed with NPMc+ mutation, our findings suggest that in some cases of AML, RNA Pol II transcription may be disregulated by the malfunction of NPM and the mislocation of HEXIM1.
Collapse
Affiliation(s)
- Meera Gurumurthy
- Expression Engineering Group, Bioprocessing Technology Institute, 20 Biopolis Way, #06-01 Centros, Singapore 138668, Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Rosa DD, Ismael G, Lago LD, Awada A. Molecular-targeted therapies: lessons from years of clinical development. Cancer Treat Rev 2007; 34:61-80. [PMID: 17826917 DOI: 10.1016/j.ctrv.2007.07.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2007] [Revised: 07/18/2007] [Accepted: 07/21/2007] [Indexed: 12/26/2022]
Abstract
Over the past decade, molecular-targeted therapies have been added to cytotoxic and anti-endocrine drugs in the treatment of cancer, with the aim to target the molecular pathways that underlie the carcinogenic process and maintain the cancer phenotype. Success with some of these agents has suggested that identification and validation of the drug target is the starting point for the route of development of active, safe and effective drugs. Main molecular targets used to the development of anticancer drugs are cell surface receptors, signal transduction pathways, gene transcription targets, ubiquitin-proteasome/heat shock proteins and tumour microenvironment components (especially antiangiogenic agents). Here, we review the development of the main molecular targeted non-cytotoxic agents studied in cancer, highlighting lessons derived from the development of these novel drugs and proposing new horizons for the clinical development of molecular-targeted therapies.
Collapse
Affiliation(s)
- Daniela D Rosa
- Medical Oncology Clinic, Jules Bordet Institute, and L Universite Libre de Bruxelles (ULB), Brussels, Belgium.
| | | | | | | |
Collapse
|