1
|
Santaniello G, Nebbioso A, Altucci L, Conte M. Recent Advancement in Anticancer Compounds from Marine Organisms: Approval, Use and Bioinformatic Approaches to Predict New Targets. Mar Drugs 2022; 21:md21010024. [PMID: 36662197 PMCID: PMC9862894 DOI: 10.3390/md21010024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/23/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022] Open
Abstract
In recent years, the study of anticancer bioactive compounds from marine sources has received wide interest. Contextually, world regulatory authorities have approved several marine molecules, and new synthetic derivatives have also been synthesized and structurally improved for the treatment of numerous forms of cancer. However, the administration of drugs in cancer patients requires careful evaluation since their interaction with individual biological macromolecules, such as proteins or nucleic acids, determines variable downstream effects. This is reflected in a constant search for personalized therapies that lay the foundations of modern medicine. The new knowledge acquired on cancer mechanisms has certainly allowed advancements in tumor prevention, but unfortunately, due to the huge complexity and heterogeneity of cancer, we are still looking for a definitive therapy and clinical approaches. In this review, we discuss the significance of recently approved molecules originating from the marine environment, starting from their organism of origin to their structure and mechanism of action. Subsequently, these bio-compounds are used as models to illustrate possible bioinformatics approaches for the search of new targets that are useful for improving the knowledge on anticancer therapies.
Collapse
Affiliation(s)
- Giovanna Santaniello
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Vico L. De Crecchio 7, 80138 Naples, Italy
| | - Angela Nebbioso
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Vico L. De Crecchio 7, 80138 Naples, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Vico L. De Crecchio 7, 80138 Naples, Italy
- BIOGEM, Institute of Molecular Biology and Genetics, Via Camporeale, 83031 Ariano Irpino, Italy
- IEOS, Institute for Endocrinology and Experimental Oncology, CNR, Via Pansini 5, 80131 Napoli, Italy
- Correspondence: (L.A.); (M.C.); Tel.: +39-081-5667564 (M.C.)
| | - Mariarosaria Conte
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Vico L. De Crecchio 7, 80138 Naples, Italy
- Correspondence: (L.A.); (M.C.); Tel.: +39-081-5667564 (M.C.)
| |
Collapse
|
2
|
Brönstrup M, Sasse F. Natural products targeting the elongation phase of eukaryotic protein biosynthesis. Nat Prod Rep 2021; 37:752-762. [PMID: 32428051 DOI: 10.1039/d0np00011f] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Covering: 2000 to 2020 The translation of mRNA into proteins is a precisely regulated, complex process that can be divided into three main stages, i.e. initiation, elongation, termination, and recycling. This contribution is intended to highlight how natural products interfere with the elongation phase of eukaryotic protein biosynthesis. Cycloheximide, isolated from Streptomyces griseus, has long been the prototype inhibitor of eukaryotic translation elongation. In the last three decades, a variety of natural products from different origins were discovered to also address the elongation step in different manners, including interference with the elongation factors eEF1 and eEF2 as well as binding to A-, P- or E-sites of the ribosome itself. Recent advances in the crystallization of the ribosomal machinery together with natural product inhibitors allowed characterizing similarities as well as differences in their mode of action. Since aberrations in protein synthesis are commonly observed in tumors, and malfunction or overexpression of translation factors can cause cellular transformation, the protein synthesis machinery has been realized as an attractive target for anticancer drugs. The therapeutic use of the first natural products that reached market approval, plitidepsin (Aplidin®) and homoharringtonine (Synribo®), will be introduced. In addition, we will highlight two other potential indications for translation elongation inhibitors, i.e. viral infections and genetic disorders caused by premature termination of translation.
Collapse
Affiliation(s)
- Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany. and Center of Biomolecular Drug Research (BMWZ), Leibniz University, 30159 Hannover, Germany and German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Germany
| | - Florenz Sasse
- Department of Chemical Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany.
| |
Collapse
|
3
|
Randomized phase III study (ADMYRE) of plitidepsin in combination with dexamethasone vs. dexamethasone alone in patients with relapsed/refractory multiple myeloma. Ann Hematol 2019; 98:2139-2150. [PMID: 31240472 PMCID: PMC6700046 DOI: 10.1007/s00277-019-03739-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/12/2019] [Indexed: 12/28/2022]
Abstract
The randomized phase III ADMYRE trial evaluated plitidepsin plus dexamethasone (DXM) versus DXM alone in patients with relapsed/refractory multiple myeloma after at least three but not more than six prior regimens, including at least bortezomib and lenalidomide or thalidomide. Patients were randomly assigned (2:1) to receive plitidepsin 5 mg/m2 on D1 and D15 plus DXM 40 mg on D1, D8, D15, and D22 (arm A, n = 171) or DXM 40 mg on D1, D8, D15, and D22 (arm B, n = 84) q4wk. The primary endpoint was progression-free survival (PFS). Median PFS without disease progression (PD) confirmation (IRC assessment) was 2.6 months (arm A) and 1.7 months (arm B) (HR = 0.650; p = 0.0054). Median PFS with PD confirmation (investigator’s assessment) was 3.8 months (arm A) and 1.9 months (arm B) (HR = 0.611; p = 0.0040). Median overall survival (OS, intention-to-treat analysis) was 11.6 months (arm A) and 8.9 months (arm B) (HR = 0.797; p = 0.1261). OS improvement favoring arm A was found when discounting a crossover effect (37 patients crossed over from arm B to arm A) (two-stage method; HR = 0.622; p = 0.0015). The most common grade 3/4 treatment-related adverse events (% of patients arm A/arm B) were fatigue (10.8%/1.2%), myalgia (5.4%/0%), and nausea (3.6%/1.2%), being usually transient and reversible. The safety profile does not overlap with the toxicity observed with other agents used in multiple myeloma. In conclusion, efficacy data, the reassuring safety profile, and the novel mechanism of action of plitidepsin suggest that this combination can be an alternative option in patients with relapsed/refractory multiple myeloma after at least three prior therapy lines.
Collapse
|
4
|
Leisch M, Egle A, Greil R. Plitidepsin: a potential new treatment for relapsed/refractory multiple myeloma. Future Oncol 2019; 15:109-120. [DOI: 10.2217/fon-2018-0492] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Plitidepsin is a marine-derived anticancer compound isolated from the Mediterranean tunicate Applidium albicans. It exerts pleiotropic effects on cancer cells, most likely by binding to the eukaryotic translation eEF1A2. This ultimately leads to cell-cycle arrest, growth inhibition and induction of apoptosis via multiple pathway alterations. Recently, a Phase III randomized trial in patients with relapsed/refractory multiple myeloma reported outcomes for plitidepsin plus dexamethasone compared with dexamethasone. Median progression-free survival was 3.8 months in the plitidepsin arm and 1.9 months in the dexamethasone arm (HR: 0.611; p = 0.0048). Here, we review preclinical data regarding plitidepsins mechanism of action, give an overview of clinical trial results across different tumor types as well as the latest results in multiple myeloma.
Collapse
Affiliation(s)
- Michael Leisch
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology & Rheumatology, Oncologic Center, Salzburg Cancer Research Institute – Laboratory of Immunological & Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Salzburg, Austria, Cancer Cluster Salzburg, Austria
| | - Alexander Egle
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology & Rheumatology, Oncologic Center, Salzburg Cancer Research Institute – Laboratory of Immunological & Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Salzburg, Austria, Cancer Cluster Salzburg, Austria
| | - Richard Greil
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology & Rheumatology, Oncologic Center, Salzburg Cancer Research Institute – Laboratory of Immunological & Molecular Cancer Research (SCRI-LIMCR), Paracelsus Medical University, Salzburg, Austria, Cancer Cluster Salzburg, Austria
| |
Collapse
|
5
|
Kim SJ, Noh TH, Son S, Kim DH, Kim W, Lee Y, Choo J, Heo G, Kim MJ, Chung HY, Jung Y, Jung JH, Moon HR, Im E. Novel β-phenylacrylic acid derivatives exert anti-cancer activity by inducing Src-mediated apoptosis in wild-type KRAS colon cancer. Cell Death Dis 2018; 9:877. [PMID: 30158525 PMCID: PMC6115383 DOI: 10.1038/s41419-018-0942-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 06/19/2018] [Accepted: 07/30/2018] [Indexed: 11/09/2022]
Abstract
Many stress conditions including chemotherapy treatment is known to activate Src and under certain condition Src can induce the apoptotic signal via c-Jun N-terminal kinase (JNK) activation. Here we report that the newly synthesized β-phenylacrylic acid derivatives, MHY791 and MHY1036 (MHYs), bind to epidermal growth factor receptor (EGFR) tyrosine kinase domains and function as EGFR inhibitors, having anti-cancer activities selectively in wild-type KRAS colon cancer. Mechanistically, MHYs-induced Src/JNK activation which enhanced their pro-apoptotic effects and therefore inhibition of Src by the chemical inhibitor PP2 or Src siRNA abolished the response. In addition, MHYs generated reactive oxygen species and increased ER stress, and pretreatment with antioxidant-inhibited MHY-induced ER stress, Src activation, and apoptosis. Furthermore, the irreversible EGFR inhibitor PD168393 also activated Src while the reversible EGFR inhibitor gefitinib showed the opposite effect, indicating that MHYs are the irreversible EGFR inhibitor. Collectively, Src can play a key role in apoptosis induced by the novel EGFR inhibitor MHYs, suggesting that activation of Src might prove effective in treating EGFR/wild-type KRAS colon cancer.
Collapse
Affiliation(s)
- Su Jin Kim
- College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Tae Hwan Noh
- College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Sujin Son
- College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Do Hyun Kim
- College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Wooseong Kim
- College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Yunna Lee
- College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Jieun Choo
- College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Gwangbeom Heo
- College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Min Jae Kim
- College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Hae Young Chung
- College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Yunjin Jung
- College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Jee Hyung Jung
- College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Hyung Ryong Moon
- College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea
| | - Eunok Im
- College of Pharmacy, Pusan National University, Busan, 46241, Republic of Korea.
| |
Collapse
|
6
|
Xing H, Tong M, Jiang N, Zhang X, Hu H, Pan H, Li D. Antitumour bioactive peptides isolated from marine organisms. Clin Exp Pharmacol Physiol 2018; 44:1077-1082. [PMID: 28675498 DOI: 10.1111/1440-1681.12808] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 06/28/2017] [Accepted: 06/28/2017] [Indexed: 11/30/2022]
Abstract
Marine organisms are an important source of antitumour active substances. Thus, pharmaceutical research in recent years has focused on exploring new antitumour drugs derived from marine organisms, and, many peptide drugs with strong antitumour activities have been successfully extracted. Based on different mechanisms, this paper reviews the research on several typical antitumour bioactive peptides in marine drugs and the latest progress therein. Additionally, the development prospects for these antitumour bioactive peptide-based drugs are discussed so as to provide a reference for future research in this field.
Collapse
Affiliation(s)
- Haibo Xing
- Department of ICU, Xiasha Campus, Sir Run Run Shaw Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| | - Mengting Tong
- Department of Medical Oncology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| | - Nanyu Jiang
- Department of Medical Oncology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| | - Xiaomin Zhang
- Department of Medical Oncology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| | - Hong Hu
- Department of Medical Oncology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| | - Da Li
- Department of Medical Oncology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University, School of Medicine, Hangzhou, China
| |
Collapse
|
7
|
Wang J, Tai G. Role of C-Jun N-terminal Kinase in Hepatocellular Carcinoma Development. Target Oncol 2017; 11:723-738. [PMID: 27392951 DOI: 10.1007/s11523-016-0446-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hepatocellular carcinoma (HCC) is among the most frequently occurring cancers and the leading causes of cancer mortality worldwide. Identification of the signaling pathways regulating liver carcinogenesis is critical for developing novel chemoprevention and targeted therapies. C-Jun N-terminal kinase (JNK) is a member of a larger group of serine/threonine (Ser/Thr) protein kinases known as the mitogen-activated protein kinase (MAPK) family. JNK is an important signaling component that converts external stimuli into a wide range of cellular responses, including cell proliferation, differentiation, survival, migration, invasion, and apoptosis, as well as the development of inflammation, fibrosis, cancer growth, and metabolic diseases. Because of the essential roles of JNK in these cellular functions, deregulated JNK is often found to contribute to the development of HCC. Recently, the functions and molecular mechanisms of JNK in HCC development have been addressed using mouse models and human HCC cell lines. Furthermore, recent studies demonstrate that the activation of JNK by oncogenes can promote the development of cancers by regulating the transforming growth factor (TGF)-β/Smad pathway, which makes the oncogenes/JNK/Smad signaling pathway an attractive target for cancer therapy. Additionally, JNK-targeted therapy has a broad potential for clinical applications. In summary, we are convinced that promising new avenues for the treatment of HCC by targeting JNK are on the horizon, which will undoubtedly lead to better, more effective, and faster therapies in the years to come.
Collapse
Affiliation(s)
- Juan Wang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Jilin, Changchun, 130021, China
| | - Guixiang Tai
- Department of Immunology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Street, Jilin, Changchun, 130021, China.
| |
Collapse
|
8
|
Palanisamy SK, Rajendran NM, Marino A. Natural Products Diversity of Marine Ascidians (Tunicates; Ascidiacea) and Successful Drugs in Clinical Development. NATURAL PRODUCTS AND BIOPROSPECTING 2017; 7:1-111. [PMID: 28097641 PMCID: PMC5315671 DOI: 10.1007/s13659-016-0115-5] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 12/14/2016] [Indexed: 06/06/2023]
Abstract
This present study reviewed the chemical diversity of marine ascidians and their pharmacological applications, challenges and recent developments in marine drug discovery reported during 1994-2014, highlighting the structural activity of compounds produced by these specimens. Till date only 5% of living ascidian species were studied from <3000 species, this study represented from family didemnidae (32%), polyclinidae (22%), styelidae and polycitoridae (11-12%) exhibiting the highest number of promising MNPs. Close to 580 compound structures are here discussed in terms of their occurrence, structural type and reported biological activity. Anti-cancer drugs are the main area of interest in the screening of MNPs from ascidians (64%), followed by anti-malarial (6%) and remaining others. FDA approved ascidian compounds mechanism of action along with other compounds status of clinical trials (phase 1 to phase 3) are discussed here in. This review highlights recent developments in the area of natural products chemistry and biotechnological approaches are emphasized.
Collapse
Affiliation(s)
- Satheesh Kumar Palanisamy
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 98166, Messina, Italy.
| | - N M Rajendran
- Key Laboratory of Engineering Plastics and Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Angela Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 98166, Messina, Italy
| |
Collapse
|
9
|
Losada A, Muñoz-Alonso MJ, García C, Sánchez-Murcia PA, Martínez-Leal JF, Domínguez JM, Lillo MP, Gago F, Galmarini CM. Translation Elongation Factor eEF1A2 is a Novel Anticancer Target for the Marine Natural Product Plitidepsin. Sci Rep 2016; 6:35100. [PMID: 27713531 PMCID: PMC5054363 DOI: 10.1038/srep35100] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 09/22/2016] [Indexed: 12/18/2022] Open
Abstract
eEF1A2 is one of the isoforms of the alpha subunit of the eukaryotic Elongation Factor 1. It is overexpressed in human tumors and is endowed with oncogenic properties, favoring tumor cell proliferation while inhibiting apoptosis. We demonstrate that plitidepsin, an antitumor agent of marine origin that has successfully completed a phase-III clinical trial for multiple myeloma, exerts its antitumor activity by targeting eEF1A2. The drug interacts with eEF1A2 with a KD of 80 nM and a target residence time of circa 9 min. This protein was also identified as capable of binding [14C]-plitidepsin in a cell lysate from K-562 tumor cells. A molecular modelling approach was used to identify a favorable binding site for plitidepsin at the interface between domains 1 and 2 of eEF1A2 in the GTP conformation. Three tumor cell lines selected for at least 100-fold more resistance to plitidepsin than their respective parental cells showed reduced levels of eEF1A2 protein. Ectopic expression of eEF1A2 in resistant cells restored the sensitivity to plitidepsin. FLIM-phasor FRET experiments demonstrated that plitidepsin localizes in tumor cells sufficiently close to eEF1A2 as to suggest the formation of drug-protein complexes in living cells. Altogether, our results strongly suggest that eEF1A2 is the primary target of plitidepsin.
Collapse
Affiliation(s)
- Alejandro Losada
- Departamento de Biología Celular y Farmacogenómica, Pharma Mar S.A., Colmenar Viejo, Madrid, Spain
| | - María José Muñoz-Alonso
- Departamento de Biología Celular y Farmacogenómica, Pharma Mar S.A., Colmenar Viejo, Madrid, Spain
| | - Carolina García
- Departamento de Química Física Biológica, Instituto de Química-Física "Rocasolano" (CSIC), Madrid, Spain
| | - Pedro A Sánchez-Murcia
- Departamento de Ciencias Biomédicas, Unidad Asociada al IQM-CSIC, Universidad de Alcalá, Madrid, Spain
| | | | - Juan Manuel Domínguez
- Departamento de Biología Celular y Farmacogenómica, Pharma Mar S.A., Colmenar Viejo, Madrid, Spain
| | - M Pilar Lillo
- Departamento de Química Física Biológica, Instituto de Química-Física "Rocasolano" (CSIC), Madrid, Spain
| | - Federico Gago
- Departamento de Ciencias Biomédicas, Unidad Asociada al IQM-CSIC, Universidad de Alcalá, Madrid, Spain
| | - Carlos M Galmarini
- Departamento de Biología Celular y Farmacogenómica, Pharma Mar S.A., Colmenar Viejo, Madrid, Spain
| |
Collapse
|
10
|
Abstract
Cyclic depsipeptides are polypeptides in which one or more amino acid is replaced by a hydroxy acid, resulting in the formation of at least one ester bond in the core ring structure. Many natural cyclic depsipeptides possessing intriguing structural and biological properties, including antitumor, antifungal, antiviral, antibacterial, anthelmintic, and anti-inflammatory activities, have been identified from fungi, plants, and marine organisms. In particular, the potent effects of cyclic depsipeptides on tumor cells have led to a number of clinical trials evaluating their potential as chemotherapeutic agents. Although many of the trials have not achieved the desired results, romidepsin (FK228), a bicyclic depsipeptide that inhibits histone deacetylase, has been shown to have clinical efficacy in patients with refractory cutaneous T-cell lymphoma and has received Food and Drug Administration approval for use in treatment. In this review, we discuss antitumor cyclic depsipeptides that have undergone clinical trials and focus on their structural features, mechanisms, potential applications in chemotherapy, and pharmacokinetic and toxicity data. The results of this study indicate that cyclic depsipeptides could be a rich source of new cancer therapeutics.
Collapse
|
11
|
Toulmonde M, Le Cesne A, Piperno-Neumann S, Penel N, Chevreau C, Duffaud F, Bellera C, Italiano A. Aplidin in patients with advanced dedifferentiated liposarcomas: a French Sarcoma Group Single-Arm Phase II study. Ann Oncol 2015; 26:1465-70. [PMID: 26041763 DOI: 10.1093/annonc/mdv195] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 04/17/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Preclinical data have suggested a therapeutic role of JUN pathway activation in dedifferentiated liposarcoma (DDLPS) tumorigenesis. Aplidin is a drug inducing apoptosis through a strong, sustained activation of c-Jun NH2-terminal kinase. METHODS This phase II trial included patients with progressive advanced DDLPS. They received Aplidin 5 mg/m(2) days 1-15, 28-day cycle until disease progression or unacceptable toxicity. The primary end point was the 3-month nonprogression rate (PFS3) defined as the proportion of patients with nonprogressive disease at 3 months. A PFS3 of 40% considered as a reasonable objective to claim drug efficacy. RESULTS Between August 2012 and May 2013, 24 patients were included. Sixteen had received prior chemotherapy. Twenty-two were assessable for efficacy. The PFS3 was 9.1% [95% confidence interval (CI) 1.1-29.2]. Median progression-free and overall survivals were 1.6 months (95% CI 1.4-2.6) and 9.2 months (95% CI 6.6-). The most frequent adverse events of any grade were nausea, fatigue, anorexia, vomiting and diarrhea. CONCLUSION Aplidin did not meet the primary end point of this trial and do not deserve further investigation in DDLPS. CLINICALTRIALSGOV IDENTIFIER NCT01876043.
Collapse
Affiliation(s)
- M Toulmonde
- Department of Medical Oncology, Institut Bergonié, Bordeaux
| | - A Le Cesne
- Department of Medicine, Institut Gustave Roussy, Villejuif
| | | | - N Penel
- Department of Medicine, Centre Oscar Lambret, Lille
| | - C Chevreau
- Department of Medicine, Institut Claudius Regaud, Toulouse
| | - F Duffaud
- Department of Medical Oncology, Hôpital La Timone, Marseille
| | - C Bellera
- Clinical and Epidemiological Research Unit, Institut Bergonié, Bordeaux Data Center for Cancer Clinical Trials, CTD-INCa, Bordeaux, France
| | - A Italiano
- Department of Medical Oncology, Institut Bergonié, Bordeaux
| |
Collapse
|
12
|
Xi HJ, Wu RP, Liu JJ, Zhang LJ, Li ZS. Role of acetylcholinesterase in lung cancer. Thorac Cancer 2015; 6:390-8. [PMID: 26273392 PMCID: PMC4511315 DOI: 10.1111/1759-7714.12249] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 02/07/2015] [Indexed: 12/14/2022] Open
Abstract
Acetylcholinesterase (AChE) plays a key role in catalytic hydrolysis of cholinergic neurotransmitters. Intensive research has proven the involvement of this protein in novel functions, such as cell adhesion, differentiation, and proliferation. In addition, several recent studies have indicated that acetylcholinesterase is potentially a marker and regulator of apoptosis. Importantly, AChE is also a promising tumor suppressor. In this review, we briefly summarize the involvement of AChE in apoptosis and cancer, focusing on the role of AChE in lung cancer, as well as the therapeutic consideration of AChE for cancer therapy.
Collapse
Affiliation(s)
- Hui-Jun Xi
- Digestive Endoscopy Center, Changhai Hospital, Second Military Medical University Shanghai, China
| | - Ren-Pei Wu
- Digestive Endoscopy Center, Changhai Hospital, Second Military Medical University Shanghai, China
| | - Jing-Jing Liu
- School of Nursing, Second Military Medical University Shanghai, China
| | - Ling-Juan Zhang
- Department of Nursing, Changhai Hospital, Second Military Medical University Shanghai, China
| | - Zhao-Shen Li
- Digestive Endoscopy Center, Changhai Hospital, Second Military Medical University Shanghai, China ; Department of Gastroenterology, Changhai Hospital, Second Military Medical University Shanghai, China
| |
Collapse
|
13
|
Beesoo R, Neergheen-Bhujun V, Bhagooli R, Bahorun T. Apoptosis inducing lead compounds isolated from marine organisms of potential relevance in cancer treatment. Mutat Res 2014; 768:84-97. [PMID: 24685981 DOI: 10.1016/j.mrfmmm.2014.03.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 03/17/2014] [Accepted: 03/20/2014] [Indexed: 06/03/2023]
Abstract
Apoptosis is a critical defense mechanism against the formation and progression of cancer and exhibits distinct morphological and biochemical traits. Targeting apoptotic pathways becomes an intriguing strategy for the development of chemotherapeutic agents particularly if the process is selective to cancer cells. Marine natural products have become important sources in the discovery of antitumour drugs, especially when recent technological and methodological advances have increased the scope of investigations of marine organisms. A high number of individual compounds from diverse organisms have induced apoptosis in several tumour cell lines via a number of mechanisms. Here, we review the effects of selected marine natural products and their synthetic derivatives on apoptosis signalling pathways in association with their pharmacological properties. Providing an outlook into the future, we also examine the factors that contribute to new discoveries and the difficulties associated with translating marine-derived compounds into clinical trials.
Collapse
Affiliation(s)
- Rima Beesoo
- ANDI Centre of Excellence for Biomedical and Biomaterials Research, University of Mauritius, Reduit, Mauritius; Department of Health Sciences, Faculty of Science, University of Mauritius, Reduit, Mauritius; Department of Biosciences, Faculty of Science, University of Mauritius, Reduit, Mauritius
| | - Vidushi Neergheen-Bhujun
- ANDI Centre of Excellence for Biomedical and Biomaterials Research, University of Mauritius, Reduit, Mauritius; Department of Health Sciences, Faculty of Science, University of Mauritius, Reduit, Mauritius
| | - Ranjeet Bhagooli
- Department of Biosciences, Faculty of Science, University of Mauritius, Reduit, Mauritius
| | - Theeshan Bahorun
- ANDI Centre of Excellence for Biomedical and Biomaterials Research, University of Mauritius, Reduit, Mauritius.
| |
Collapse
|
14
|
Nano-encapsulation of plitidepsin: in vivo pharmacokinetics, biodistribution, and efficacy in a renal xenograft tumor model. Pharm Res 2013; 31:983-91. [PMID: 24287622 DOI: 10.1007/s11095-013-1220-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 09/22/2013] [Indexed: 12/26/2022]
Abstract
PURPOSE Plitidepsin is an antineoplasic currently in clinical evaluation in a phase III trial in multiple myeloma (ADMYRE). Presently, the hydrophobic drug plitidepsin is formulated using Cremophor®, an adjuvant associated with unwanted hypersensitivity reactions. In search of alternatives, we developed and tested two nanoparticle-based formulations of plitidepsin, aiming to modify/improve drug biodistribution and efficacy. METHODS Using nanoprecipitation, plitidepsin was loaded in polymer nanoparticles made of amphiphilic block copolymers (i.e. PEG-b-PBLG or PTMC-b-PGA). The pharmacokinetics, biodistribution and therapeutic efficacy was assessed using a xenograft renal cancer mouse model (MRI-H-121 xenograft) upon administration of the different plitidepsin formulations at maximum tolerated multiple doses (0.20 and 0.25 mg/kg for Cremophor® and copolymer formulations, respectively). RESULTS High plitidepsin loading efficiencies were obtained for both copolymer formulations. Considering pharmacokinetics, PEG-b-PBLG formulation showed lower plasma clearance, associated with higher AUC and Cmax than Cremophor® or PTMC-b-PGA formulations. Additionally, the PEG-b-PBLG formulation presented lower liver and kidney accumulation compared with the other two formulations, associated with an equivalent tumor distribution. Regarding the anticancer activity, all formulations elicited similar efficacy profiles, as compared to the Cremophor® formulation, successfully reducing tumor growth rate. CONCLUSIONS Although the nanoparticle formulations present equivalent anticancer activity, compared to the Cremophor® formulation, they show improved biodistribution profiles, presenting novel tools for future plitidepsin-based therapies.
Collapse
|
15
|
c-Jun N-terminal kinase phosphorylation is a biomarker of plitidepsin activity. Mar Drugs 2013; 11:1677-92. [PMID: 23697951 PMCID: PMC3707168 DOI: 10.3390/md11051677] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/01/2013] [Accepted: 04/18/2013] [Indexed: 01/30/2023] Open
Abstract
Plitidepsin is an antitumor drug of marine origin currently in Phase III clinical trials in multiple myeloma. In cultured cells, plitidepsin induces cell cycle arrest or an acute apoptotic process in which sustained activation of c-Jun N-terminal kinase (JNK) plays a crucial role. With a view to optimizing clinical use of plitidepsin, we have therefore evaluated the possibility of using JNK activation as an in vivo biomarker of response. In this study, we show that administration of a single plitidepsin dose to mice xenografted with human cancer cells does indeed lead to increased phosphorylation of JNK in tumors at 4 to 12 h. By contrast, no changes were found in other in vitro plitidepsin targets such as the levels of phosphorylated-ERK, -p38MAPK or the protein p27KIP1. Interestingly, plitidepsin also increased JNK phosphorylation in spleens from xenografted mice showing similar kinetics to those seen in tumors, thereby suggesting that normal tissues might be useful for predicting drug activity. Furthermore, plitidepsin administration to rats at plasma concentrations comparable to those achievable in patients also increased JNK phosphorylation in peripheral mononuclear blood cells. These findings suggest that changes in JNK activity provide a reliable biomarker for plitidepsin activity and this could be useful for designing clinical trials and maximizing the efficacy of plitidepsin.
Collapse
|
16
|
Bharate SB, Sawant SD, Singh PP, Vishwakarma RA. Kinase inhibitors of marine origin. Chem Rev 2013; 113:6761-815. [PMID: 23679846 DOI: 10.1021/cr300410v] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Sandip B Bharate
- Medicinal Chemistry Division, Indian Institute of Integrative Medicine (Council of Scientific and Industrial Research), Canal Road, Jammu-180001, India
| | | | | | | |
Collapse
|
17
|
Barboza NM, Medina DJ, Budak-Alpdogan T, Aracil M, Jimeno JM, Bertino JR, Banerjee D. Plitidepsin (Aplidin) is a potent inhibitor of diffuse large cell and Burkitt lymphoma and is synergistic with rituximab. Cancer Biol Ther 2012; 13:114-22. [PMID: 22336911 DOI: 10.4161/cbt.13.2.18876] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Plitidepsin (Aplidin), an antitumor agent of marine origin, presently is undergoing phase II/III clinical trials, and has shown promise for the treatment of lymphoma. Here, we describe the antitumor effects of plitidepsin alone and in combination with rituximab and investigated the effects of each drug and the combination on the cell cycle and mechanism of cell death. Several Diffuse Large Cell Lymphoma (DLCL) lines and Burkitt cell lines were tested for sensitivity to plitidepsin and rituximab. All DLCL and Burkitt lymphoma cell lines were inhibited by plitidepsin in nanomolar concentrations, while rituximab sensitivity varied among different cell lines. Ramos and the RL cell lines proved sensitive to rituximab and were used to test the effects of each of the two drugs. The two agents exhibited synergism at all tested concentrations. For in vivo studies, irradiated athymic nude mice were engrafted with the Ramos lymphoma. Treatment was initiated when the tumors were ~0.5 cm in diameter, and toxic and therapeutic effects were monitored. In the in vivo study, additive effects of the combined two drugs, was demonstrated without an increase in host toxicity. The in vitro synergy and the in vivo additive antitumor effects without an increase in host toxicity with two relatively non-marrow suppressive agents encourages further development of this combination for treatment of aggressive B-cell lymphomas.
Collapse
Affiliation(s)
- Nora M Barboza
- Department of Medicine, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Lee J, Currano JN, Carroll PJ, Joullié MM. Didemnins, tamandarins and related natural products. Nat Prod Rep 2012; 29:404-24. [PMID: 22270031 DOI: 10.1039/c2np00065b] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Since the discovery and isolation of the didemnin family of marine depsipeptides in 1981, the synthesis and biological activity of its congeners have been of great interest to the scientific community. The didemnins have demonstrated antitumor, antiviral, and immunosuppressive activity at low nano- and femtomolar levels. Of the congeners, didemnin B was the first marine natural product to reach phase II clinical trials in the United States, stimulating many analogue syntheses to date. About two decades later, tamandarins A and B were isolated, and were found to possess very similar structure and biological activity to that of the didemnin B. These compounds have shown impressive biological activity and some progress has been made in establishing structure-activity relationships. However, their molecular mechanism of action still remains unclear. This review highlights the long-standing study of didemnins and its critical application towards the understanding of the molecular mechanism of action of tamandarins and their potential use as therapeutic agents.
Collapse
Affiliation(s)
- Jisun Lee
- University of Pennsylvania, Department of Chemistry, Philadelphia, Pennsylvania, United States
| | | | | | | |
Collapse
|
19
|
Soto-Matos A, Szyldergemajn S, Extremera S, Miguel-Lillo B, Alfaro V, Coronado C, Lardelli P, Roy E, Corrado CS, Kahatt C. Plitidepsin has a safe cardiac profile: a comprehensive analysis. Mar Drugs 2011; 9:1007-1023. [PMID: 21747745 PMCID: PMC3131558 DOI: 10.3390/md9061007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2011] [Revised: 05/25/2011] [Accepted: 05/31/2011] [Indexed: 12/11/2022] Open
Abstract
Plitidepsin is a cyclic depsipeptide of marine origin in clinical development in cancer patients. Previously, some depsipeptides have been linked to increased cardiac toxicity. Clinical databases were searched for cardiac adverse events (CAEs) that occurred in clinical trials with the single-agent plitidepsin. Demographic, clinical and pharmacological variables were explored by univariate and multivariate logistic regression analysis. Forty-six of 578 treated patients (8.0%) had at least one CAE (11 patients (1.9%) with plitidepsin-related CAEs), none with fatal outcome as a direct consequence. The more frequent CAEs were rhythm abnormalities (n = 31; 5.4%), mostly atrial fibrillation/flutter (n = 15; 2.6%). Of note, life-threatening ventricular arrhythmias did not occur. Myocardial injury events (n = 17; 3.0%) included possible ischemic-related and non-ischemic events. Other events (miscellaneous, n = 6; 1.0%) were not related to plitidepsin. Significant associations were found with prostate or pancreas cancer primary diagnosis (p = 0.0017), known baseline cardiac risk factors (p = 0.0072), myalgia present at baseline (p = 0.0140), hemoglobin levels lower than 10 g/dL (p = 0.0208) and grade ≥2 hypokalemia (p = 0.0095). Treatment-related variables (plitidepsin dose, number of cycles, schedule and/or total cumulative dose) were not associated. Electrocardiograms performed before and after plitidepsin administration (n = 136) detected no relevant effect on QTc interval. None of the pharmacokinetic parameters analyzed had a significant impact on the probability of developing a CAE. In conclusion, the most frequent CAE type was atrial fibrillation/atrial flutter, although its frequency was not different to that reported in the age-matched healthy population, while other CAEs types were rare. No dose-cumulative pattern was observed, and no treatment-related variables were associated with CAEs. Relevant risk factors identified were related to the patient's condition and/or to disease-related characteristics rather than to drug exposure. Therefore, the current analysis supports a safe cardiac risk profile for single-agent plitidepsin in cancer patients.
Collapse
Affiliation(s)
| | | | | | | | - Vicente Alfaro
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +34-93-4037094; Fax: +34-93-4491079
| | | | | | | | | | | |
Collapse
|
20
|
Zhou P, Gross S, Liu JH, Yu BY, Feng LL, Nolta J, Sharma V, Piwnica-Worms D, Qiu SX. Flavokawain B, the hepatotoxic constituent from kava root, induces GSH‐sensitive oxidative stress through modulation of IKK/NF‐κB and MAPK signaling pathways. FASEB J 2010. [DOI: 10.1096/fj.10.163311] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Ping Zhou
- Division of OncologyMallinckrodt Institute of Radiology, Washington University, School of Medicine St. Louis MO USA
| | - Shimon Gross
- Division of OncologyMallinckrodt Institute of Radiology, Washington University, School of Medicine St. Louis MO USA
- Department of Internal Medicine, Stem Cell ProgramUniversity of California Davis Sacramento CA USA
| | - Ji-Hua Liu
- Department of Developmental BiologyMallinckrodt Institute of Radiology, Washington University, School of Medicine St. Louis MO USA
- Molecular Imaging CenterMallinckrodt Institute of Radiology, Washington University, School of Medicine St. Louis MO USA
| | - Bo-Yang Yu
- School of Traditional Chinese PharmacyChina Pharmaceutical University Nanjing China
| | - Ling-Ling Feng
- Key Laboratory of Plant Resources Conservation and Sustainable UtilizationSouth China Botanical GardenChinese Academy of Sciences Guangzhou China
| | - Jan Nolta
- Division of OncologyMallinckrodt Institute of Radiology, Washington University, School of Medicine St. Louis MO USA
- Department of Internal Medicine, Stem Cell ProgramUniversity of California Davis Sacramento CA USA
| | - Vijay Sharma
- Molecular Imaging CenterMallinckrodt Institute of Radiology, Washington University, School of Medicine St. Louis MO USA
| | - David Piwnica-Worms
- Department of Developmental BiologyMallinckrodt Institute of Radiology, Washington University, School of Medicine St. Louis MO USA
- Molecular Imaging CenterMallinckrodt Institute of Radiology, Washington University, School of Medicine St. Louis MO USA
| | - Samuel X. Qiu
- Key Laboratory of Plant Resources Conservation and Sustainable UtilizationSouth China Botanical GardenChinese Academy of Sciences Guangzhou China
| |
Collapse
|
21
|
Zhou P, Gross S, Liu JH, Yu BY, Feng LL, Nolta J, Sharma V, Piwnica-Worms D, Qiu SX. Flavokawain B, the hepatotoxic constituent from kava root, induces GSH-sensitive oxidative stress through modulation of IKK/NF-kappaB and MAPK signaling pathways. FASEB J 2010; 24:4722-32. [PMID: 20696856 DOI: 10.1096/fj.10-163311] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Kava (Piper methysticum Foster, Piperaceae) organic solvent-extract has been used to treat mild to moderate anxiety, insomnia, and muscle fatigue in Western countries, leading to its emergence as one of the 10 best-selling herbal preparations. However, several reports of severe hepatotoxicity in kava consumers led the U.S. Food and Drug Administration and authorities in Europe to restrict sales of kava-containing products. Herein we demonstrate that flavokawain B (FKB), a chalcone from kava root, is a potent hepatocellular toxin, inducing cell death in HepG2 (LD(50)=15.3 ± 0.2 μM) and L-02 (LD(50)=32 μM) cells. Hepatocellular toxicity of FKB is mediated by induction of oxidative stress, depletion of reduced glutathione (GSH), inhibition of IKK activity leading to NF-κB transcriptional blockade, and constitutive TNF-α-independent activation of mitogen-activated protein kinase (MAPK) signaling pathways, namely, ERK, p38, and JNK. We further demonstrate by noninvasive bioluminescence imaging that oral consumption of FKB leads to inhibition of hepatic NF-κB transcriptional activity in vivo and severe liver damage. Surprisingly, replenishment with exogenous GSH normalizes both TNF-α-dependent NF-κB as well as MAPK signaling and rescues hepatocytes from FKB-induced death. Our data identify FKB as a potent GSH-sensitive hepatotoxin, levels of which should be specifically monitored and controlled in kava-containing herb products.
Collapse
Affiliation(s)
- Ping Zhou
- Division of Oncology, Washington University, School of Medicine, St. Louis, MO, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Salazar R, Plummer R, Oaknin A, Robinson A, Pardo B, Soto-Matos A, Yovine A, Szyldergemajn S, Calvert AH. Phase I study of weekly plitidepsin as 1-hour infusion combined with carboplatin in patients with advanced solid tumors or lymphomas. Invest New Drugs 2010; 29:1406-13. [PMID: 20623160 DOI: 10.1007/s10637-010-9488-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 06/24/2010] [Indexed: 11/28/2022]
Abstract
This dose-escalating phase I clinical trial was designed to determine the recommended dose (RD) and to assess the safety and feasibility of weekly plitidepsin (1-hour i.v. infusion, Days 1, 8 and 15) combined with carboplatin (1-hour i.v. infusion, Day 1, after plitidepsin) in 4-week (q4wk) cycles given to patients with advanced solid tumors or lymphomas. Twenty patients were enrolled and evaluable for both safety and efficacy. The starting dose was plitidepsin 1.8 mg/m(2) and carboplatin area under the curve (AUC) = 5 min*mg/ml; dose escalation proceeded based on worst toxicity in the previous cohort. The maximum tolerated dose (MTD) was plitidepsin 3.0 mg/m(2) and carboplatin AUC = 5 min*mg/ml, with grade 3 transaminase increases as the most common dose-limiting toxicities (DLTs). The RD for phase II studies was plitidepsin 2.4 mg/m(2) and carboplatin AUC = 5 min*mg/ml, with fatigue, myalgia and nausea as the most common drug-related adverse events (AEs). No unexpected toxicity was seen. Twelve patients (60%), ten of whom were heavily pretreated (≥2 previous chemotherapy lines) showed stable disease (SD), with a median time to progression (TTP) of 4.4 months. In conclusion, plitidepsin 2.4 mg/m(2) and carboplatin AUC = 5 min*mg/ml is a safe dose for future phase II studies evaluating the use of this combination in cancer patients potentially sensitive to platinum-based therapy.
Collapse
Affiliation(s)
- Ramón Salazar
- Instituto Catalán de Oncología, Ctra. Gran Vía, s/n, L'Hospitalet de Llobregat, 08907, Barcelona, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Mateos MV, Cibeira MT, Richardson PG, Prosper F, Oriol A, de la Rubia J, Lahuerta JJ, García-Sanz R, Extremera S, Szyldergemajn S, Corrado C, Singer H, Mitsiades CS, Anderson KC, Bladé J, San Miguel J. Phase II Clinical and Pharmacokinetic Study of Plitidepsin 3-Hour Infusion Every Two Weeks Alone or with Dexamethasone in Relapsed and Refractory Multiple Myeloma. Clin Cancer Res 2010; 16:3260-9. [DOI: 10.1158/1078-0432.ccr-10-0469] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
24
|
Le Tourneau C, Faivre S, Ciruelos E, Domínguez MJ, López-Martín JA, Izquierdo MA, Jimeno J, Raymond E. Reports of clinical benefit of plitidepsin (Aplidine), a new marine-derived anticancer agent, in patients with advanced medullary thyroid carcinoma. Am J Clin Oncol 2010; 33:132-6. [PMID: 19687728 DOI: 10.1097/coc.0b013e318199fb6e] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES To assess clinical benefit of plitidepsin (Aplidine) in patients with advanced medullary thyroid carcinoma (MTC). MATERIALS AND METHODS We retrospectively reported the outcome of 10 patients with advanced MTC among 215 patients who have entered the phase I program with plitidepsin. RESULTS Median number of cycles was 5. Using World Health Organization criteria, 1 among 5 patients with measurable disease displayed a confirmed partial response, whereas 8 patients experienced a stable disease, and 1 patient had a progressive disease, corresponding to a disease control rate of 90%. Two patients treated at the maximum tolerated dose experienced muscular dose-limiting toxicity possibly related to palmitoyl transferase inhibition. One of these 2 patients was able to continue therapy with no dose reduction with the prophylactic addition of l-carnitine, which is used in the treatment of the carnitine palmitoyl transferase deficiency type 2. DISCUSSION Plitidepsin seems to be able to induce clinical benefit in patients with pretreated MTC, and its toxicity has been manageable at the recommended dose.
Collapse
|
25
|
Dumez H, Gallardo E, Culine S, Galceran JC, Schöffski P, Droz JP, Extremera S, Szyldergemajn S, Fléchon A. Phase II study of biweekly plitidepsin as second-line therapy for advanced or metastatic transitional cell carcinoma of the urothelium. Mar Drugs 2009; 7:451-63. [PMID: 19841725 PMCID: PMC2763111 DOI: 10.3390/md7030451] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 08/28/2009] [Accepted: 09/14/2009] [Indexed: 01/24/2023] Open
Abstract
The objective of this exploratory, open-label, single-arm, phase II clinical trial was to evaluate plitidepsin (5 mg/m(2)) administered as a 3-hour continuous intravenous infusion every two weeks to patients with locally advanced/metastatic transitional cell carcinoma of the urothelium who relapsed/progressed after first-line chemotherapy. Treatment cycles were repeated for up to 12 cycles or until disease progression, unacceptable toxicity, patient refusal or treatment delay for >2 weeks. The primary efficacy endpoint was objective response rate according to RECIST. Secondary endpoints were the rate of SD lasting > or = 6 months and time-to-event variables. Toxicity was assessed using NCI-CTC v. 3.0. Twenty-one patients received 57 treatment cycles. No objective tumor responses occurred. SD lasting <6 months was observed in two of 18 evaluable patients. With a median follow-up of 4.6 months, the median PFR and the median OS were 1.4 months and 2.3 months, respectively. The most common AEs were mild to moderate nausea, fatigue, myalgia and anorexia. Anemia, lymphopenia, and increases in transaminases, alkaline phosphatase and creatinine were the most frequent laboratory abnormalities. No severe neutropenia occurred. Treatment was feasible and generally well tolerated in this patient population; however the lack of antitumor activity precludes further studies of plitidepsin in this setting.
Collapse
Affiliation(s)
- Herlinde Dumez
- University Hospitals Gasthuisberg, Catholic University Leuven, Herestraat 49, 3000 Leuven, Belgium; E-Mail: (P.S.)
| | - Enrique Gallardo
- Corporació Parc Taulí, C/Parc Taulí, s/n, 08208 Sabadell (Barcelona), Spain; E-Mail: (E.G.)
| | - Stephane Culine
- Parc Euromedicine, 323 rue des Apothicaires, 34298 Montpellier, France; E-Mail: (S.C.)
| | - Joan Carles Galceran
- Hospital Universitari del Mar, Passeig Marítim, 25-29, 08003 Barcelona, Spain; E-Mail: (J.C.G.)
| | - Patrick Schöffski
- University Hospitals Gasthuisberg, Catholic University Leuven, Herestraat 49, 3000 Leuven, Belgium; E-Mail: (P.S.)
| | - Jean P. Droz
- Centre Léon Bérard, 28 Rue Laennec, F-69008, Lyon, France; E-Mail: (J.P.D.); (A.F.)
| | - Sonia Extremera
- Pharma Mar S.A., Av. de los Reyes, 1, PI La Mina-Norte, 28770 Colmenar Viejo, Madrid, Spain; E-Mails: (S.E.); (S.S.)
| | - Sergio Szyldergemajn
- Pharma Mar S.A., Av. de los Reyes, 1, PI La Mina-Norte, 28770 Colmenar Viejo, Madrid, Spain; E-Mails: (S.E.); (S.S.)
| | - Aude Fléchon
- Centre Léon Bérard, 28 Rue Laennec, F-69008, Lyon, France; E-Mail: (J.P.D.); (A.F.)
| |
Collapse
|
26
|
Morton CL, Houghton PJ, Gorlick R, Kolb EA, Lock R, Carol H, Keir ST, Reynolds CP, Kang MH, Maris JM, Billups C, Smith MA. Initial testing of aplidin by the pediatric pre-clinical testing program. Pediatr Blood Cancer 2009; 53:509-12. [PMID: 19418543 PMCID: PMC2777698 DOI: 10.1002/pbc.21976] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Aplidin was tested in vitro at concentrations ranging from from 0.1 nM to 1.0 microM and in vivo at a dose of 0.6 mg/kg administered intraperitoneally on an every 4 days x 3-schedule that was repeated at day 21. In vitro, Aplidin was most active against acute lymphoblastic leukemia (ALL) cell lines. In vivo, Aplidin induced significant differences in EFS distribution in 12 of 28 (43%) solid tumor models and 2 of 6 evaluable ALL models. Aplidin showed potent in vitro activity and induced significant in vivo tumor growth inhibition in some xenografts, but did not induce tumor regressions.
Collapse
Affiliation(s)
| | | | | | | | - Richard Lock
- Children’s Cancer Institute Australia for Medical Research, Randwick, NSW, Australia
| | - Hernan Carol
- Children’s Cancer Institute Australia for Medical Research, Randwick, NSW, Australia
| | | | | | - Min H. Kang
- Texas Tech University Health Sciences Center, Lubbock, TX
| | - John M. Maris
- Children’s Hospital of Philadelphia, University of Pennsylvania School of Medicine and Abramson Family Cancer Research Institute, Philadelphia, PA
| | | | | |
Collapse
|
27
|
Well-differentiated and dedifferentiated liposarcomas. Virchows Arch 2009; 456:167-79. [PMID: 19688222 DOI: 10.1007/s00428-009-0815-x] [Citation(s) in RCA: 169] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 07/04/2009] [Accepted: 07/18/2009] [Indexed: 01/10/2023]
Abstract
Atypical lipomatous tumor or well-differentiated liposarcoma (ALT-WDLPS) and dedifferentiated liposarcoma (DDLPS) share the same basic genetic abnormality characterized by a simple genomic profile with a 12q14-15 amplification involving MDM2 gene. These tumors are the most frequent LPS. This paper reviews the molecular pathology, general clinical and imaging features, histopathology, new diagnostic tools, and prognosis of ALT-WDLPS and DDLPS.
Collapse
|
28
|
Phase II study of biweekly plitidepsin as second-line therapy in patients with advanced malignant melanoma. Melanoma Res 2009; 19:185-92. [PMID: 19436178 DOI: 10.1097/cmr.0b013e32832bbde6] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The objective of this study was to evaluate the antitumor activity and safety profile of 5 mg/m2 plitidepsin administered as a 3-h continuous intravenous infusion every 2 weeks to patients with advanced malignant melanoma who relapsed or progressed after one line of systemic therapy. Objective response rate (primary efficacy endpoint) was evaluated according to Response Evaluation Criteria In Solid Tumors and toxicity was assessed using National Cancer Institute -Common Toxicity Criteria Version 2.0. Of 39 enrolled patients (median age: 53 years), 37 patients were treated who received a total of 167 treatment cycles (median: 3 cycles per patient; range: 1-32). All patients had received prior systemic therapy with a median of one line per patient (range: 1-6 lines). Of the 35 evaluable patients, two dacarbazine-resistant patients (5.7%) with metastatic cutaneous melanoma achieved partial responses. Five other patients (14.3%) reported stable disease (median stable disease duration: 3.5 months; range: 2.2-15.8 months). Therefore, the rate of tumor growth control was 20.0%. With a median follow-up of 11.0 months, the median progression-free survival was 1.3 months and the median overall survival was 3.5 months. Six patients (16.2%) had the following treatment-related grade 3/4 adverse events: myalgia (n = 3), injection-site reaction (n = 2), hypersensitivity, hypotension, and fatigue (n = 1 each). One patient was withdrawn from the trial because of grade 4 hypersensitivity reaction and hypotension. No severe neutropenia was reported. Plitidepsin showed a minor degree of antitumor activity in patients with refractory advanced malignant melanoma. Further evaluation of plitidepsin in combination schedules may be warranted.
Collapse
|
29
|
Abstract
Understanding the molecular mechanisms underlying synergistic, potentiative and antagonistic effects of drug combinations could facilitate the discovery of novel efficacious combinations and multi-targeted agents. In this article, we describe an extensive investigation of the published literature on drug combinations for which the combination effect has been evaluated by rigorous analysis methods and for which relevant molecular interaction profiles of the drugs involved are available. Analysis of the 117 drug combinations identified reveals general and specific modes of action, and highlights the potential value of molecular interaction profiles in the discovery of novel multicomponent therapies.
Collapse
|
30
|
Abstract
Drug discovery from marine natural products has enjoyed a renaissance in the past few years. Ziconotide (Prialt; Elan Pharmaceuticals), a peptide originally discovered in a tropical cone snail, was the first marine-derived compound to be approved in the United States in December 2004 for the treatment of pain. Then, in October 2007, trabectedin (Yondelis; PharmaMar) became the first marine anticancer drug to be approved in the European Union. Here, we review the history of drug discovery from marine natural products, and by describing selected examples, we examine the factors that contribute to new discoveries and the difficulties associated with translating marine-derived compounds into clinical trials. Providing an outlook into the future, we also examine the advances that may further expand the promise of drugs from the sea.
Collapse
|
31
|
Izquierdo MA, Bowman A, García M, Jodrell D, Martinez M, Pardo B, Gómez J, López-Martin JA, Jimeno J, Germá JR, Smyth JF. Phase I clinical and pharmacokinetic study of plitidepsin as a 1-hour weekly intravenous infusion in patients with advanced solid tumors. Clin Cancer Res 2008; 14:3105-12. [PMID: 18483378 DOI: 10.1158/1078-0432.ccr-07-1652] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Plitidepsin, given as a 1-hour weekly i.v. infusion for 3 consecutive weeks during a 4-week treatment cycle, was investigated in patients with solid tumors to determine the maximum tolerated dose and the recommended dose (RD) using this administration schedule. EXPERIMENTAL DESIGN Consecutive cohorts of patients with metastatic solid tumors or non-Hodgkin's lymphomas were to be treated at escalating doses of plitidepsin in a conventional phase I study including pharmacokinetic analyses of plitidepsin in plasma, whole blood, and blood cell pellets. RESULTS Forty-nine patients with solid tumors were enrolled, and 48 were treated with plitidepsin (doses from 0.133 to 3.6 mg/m2/week). Dose-limiting toxicities (defining 3.6 mg/m2/week as the maximum tolerated dose) included myalgia, increased creatine phosphokinase levels, and sustained grade 3/4 increases of hepatic enzyme levels. The RD was established at 3.2 mg/m2/week. The most common toxicities were fatigue, vomiting/nausea, anorexia, injection site reaction, and pain, mostly of mild or moderate severity. Muscular toxicity manifested by mild-moderate myalgia, weakness, and/or creatine phosphokinase elevations occurred in approximately 25% of patients and seemed to be dose related. Transient transaminase elevations were frequent but achieved grade 3 or 4 in only approximately 10% of patients. Plitidepsin lacked significant hematologic toxicity. No complete or partial tumor responses were observed; however, five patients had disease stabilization (including one patient with medullary thyroid carcinoma with an unconfirmed partial response and one patient with renal carcinoma with major tumor shrinkage in lung metastases). Pharmacokinetic results for the RD indicated a long plasma half-life give value (16.8 +/- 7.7 hour) and a high volume of distribution value (525.2 +/- 219.3 L). CONCLUSIONS The recommended dose for plitidepsin given as a weekly 1-hour schedule was 3.2 mg/m2/week. Muscular and liver toxicity were dose limiting at 3.6 mg/m2/week. Additional evaluation of this dose dense schedule is warranted.
Collapse
Affiliation(s)
- Miguel A Izquierdo
- Department of Clinical Oncology, Catalan Institute of Oncology, Hospitalet de Llobregat, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Phase II study of weekly plitidepsin as second-line therapy for small cell lung cancer. Lung Cancer 2008; 64:60-5. [PMID: 18692272 DOI: 10.1016/j.lungcan.2008.06.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2008] [Revised: 06/20/2008] [Accepted: 06/27/2008] [Indexed: 11/21/2022]
Abstract
OBJECTIVE To evaluate the antitumor activity and safety profile of plitidepsin administered as a 1h weekly intravenous (i.v.) infusion of 3.2mg/m(2) to patients with small cell lung cancer (SCLC) who relapsed or progressed after one line of chemotherapy. PATIENTS AND METHODS This was a multicenter, open-label, single-arm, exploratory, phase II clinical trial. Treatment lasted until disease progression, unacceptable toxicity, patient refusal or treatment delay for >2 weeks. Objective response rate (primary efficacy endpoint) was evaluated according to response evaluation criteria in solid tumors (RECIST). The rate of stable disease (SD) lasting for at least 6 months and time-to-event variables were secondary endpoints of efficacy. Toxicity was assessed using National Cancer Institute Common Toxicity Criteria (NCI-CTC) version 2.0. RESULTS Twenty pretreated SCLC patients (median age, 60 years) with extensive (n = 13) or limited-stage disease (n = 7) received a total of 24 treatment cycles (median, one cycle per patient; range, 1-2). Objective tumor responses were not observed and only one of the 17 evaluable patients had SD. With a median follow-up of 11.8 months, the progression-free survival and the median overall survival were 1.3 months and 4.8 months, respectively. The most troubling or common toxicities were fatigue, muscle weakness, lymphopenia, anemia (no patients showed neutropenia), and asymptomatic, non-cumulative increase of transaminases levels and alkaline phosphatase. CONCLUSION This clinical trial shows that a cycle of 1h weekly i.v. infusion of plitidepsin (3.2mg/m(2)) was generally well tolerated other than fatigue and muscle weakness in patients with pretreated SCLC. One patient died due to multi-organ failure. The absence of antitumor activity found here precludes further studies of this plitidepsin schedule as second-line single-agent treatment of SCLC.
Collapse
|
33
|
Caers J, Menu E, De Raeve H, Lepage D, Van Valckenborgh E, Van Camp B, Alvarez E, Vanderkerken K. Antitumour and antiangiogenic effects of Aplidin in the 5TMM syngeneic models of multiple myeloma. Br J Cancer 2008; 98:1966-74. [PMID: 18521088 PMCID: PMC2441967 DOI: 10.1038/sj.bjc.6604388] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Aplidin® is an antitumour drug, currently undergoing phase II evaluation in different haematological and solid tumours. In this study, we analysed the antimyeloma effects of Aplidin in the syngeneic 5T33MM model, which is representable for the human disease. In vitro, Aplidin inhibited 5T33MMvv DNA synthesis with an IC50 of 3.87 nM. On cell-cycle progression, the drug induced an arrest in transition from G0/G1 to S phase, while Western blot showed a decreased cyclin D1 and CDK4 expression. Furthermore, Aplidin induced apoptosis by lowering the mitochondrial membrane potential, by inducing cytochrome c release and by activating caspase-9 and caspase-3. For the in vivo experiment, 5T33MM-injected C57Bl/KaLwRij mice were intraperitoneally treated with vehicle or Aplidin (90 μg kg−1 daily). Chronic treatment with Aplidin was well tolerated and reduced serum paraprotein concentration by 42% (P<0.001), while BM invasion with myeloma cells was decreased by 35% (P<0.001). Aplidin also reduced the myeloma-associated angiogenesis to basal values. This antiangiogenic effect was confirmed in vitro and explained by inhibition of endothelial cell proliferation and vessel formation. These data indicate that Aplidin is well tolerated in vivo and its antitumour and antiangiogenic effects support the use of the drug in multiple myeloma.
Collapse
Affiliation(s)
- J Caers
- Laboratory of Hematology and Immunology, Vrije Universiteit Brussel, Laarbeeklaan 103, Jette, Brussels 1090, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Peschel C, Hartmann JT, Schmittel A, Bokemeyer C, Schneller F, Keilholz U, Buchheidt D, Millan S, Izquierdo MÁ, Hofheinz RD. Phase II study of plitidepsin in pretreated patients with locally advanced or metastatic non-small cell lung cancer. Lung Cancer 2008; 60:374-80. [DOI: 10.1016/j.lungcan.2007.10.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Revised: 10/10/2007] [Accepted: 10/22/2007] [Indexed: 10/22/2022]
|
35
|
Muñoz-Alonso MJ, González-Santiago L, Zarich N, Martínez T, Alvarez E, Rojas JM, Muñoz A. Plitidepsin has a dual effect inhibiting cell cycle and inducing apoptosis via Rac1/c-Jun NH2-terminal kinase activation in human melanoma cells. J Pharmacol Exp Ther 2008; 324:1093-101. [PMID: 18089842 DOI: 10.1124/jpet.107.132662] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Melanoma is the most aggressive skin cancer and a serious health problem worldwide because of its increasing incidence and the lack of satisfactory chemotherapy for late stages of the disease. The marine depsipeptide Aplidin (plitidepsin) is an antitumoral agent under phase II clinical development against several neoplasias, including melanoma. We report that plitidepsin has a dual effect on the human SK-MEL-28 and UACC-257 melanoma cell lines; at low concentrations (=45 nM), it inhibits the cell cycle by inducing G(1) and G(2)/M arrest, whereas at higher concentrations it induces apoptosis as assessed by poly-(ADP-ribose) polymerase cleavage and the appearance of a hypodiploid peak in flow cytometry analyses. Plitidepsin activates Rac1 GTPase and c-Jun NH(2)-terminal kinase (JNK). In addition, it induces AKT and p38 mitogen-activated protein kinase (MAPK) phosphorylation. By using inhibitors, we found that JNK and p38 MAPK activation depends on Rac1 but not on phosphatidylinositol 3-kinase (PI3K), whereas AKT activation is independent of Rac1 but requires PI3K activity. Plitidepsin cytotoxicity diminishes by Rac1 inhibition or by the blockage of JNK and p38 MAPK using 4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)1H-imidazole (SB203580), but not by PI3K inhibition using wortmannin or 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (LY294002). It is remarkable that plitidepsin and dacarbazine, the alkylating agent most active for treating metastatic melanoma, show a synergistic antiproliferative effect that was paralleled at the level of JNK activation. These results indicate that Rac1/JNK activation is critical for cell cycle arrest and apoptosis induction by plitidepsin in melanoma cells. They also support the combined use of plitidepsin and dacarbazine in in vivo studies.
Collapse
Affiliation(s)
- María J Muñoz-Alonso
- Instituto de Investigaciones Biomédicas "Alberto Sols," Arturo Duperier, 4, E-28029 Madrid, Spain.
| | | | | | | | | | | | | |
Collapse
|
36
|
Bowling JJ, Kochanowska AJ, Kasanah N, Hamann MT. Nature's bounty - drug discovery from the sea. Expert Opin Drug Discov 2007; 2:1505-22. [PMID: 23484601 PMCID: PMC4928193 DOI: 10.1517/17460441.2.11.1505] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
With ∼ 40 years of research completed after the development of self-contained underwater breathing apparatus, drug discovery opportunities in the sea are still too numerous to count. Since the FDA approval of the direct-from-the-sea calcium channel blocker ziconotide, marine natural products have been validated as a source for new medicines. However, the demand for natural products is extremely high due to the development of high-throughput assays and this bottleneck has created the need for an intense focus on increasing the rate of isolating and elucidating the structures of new bioactive secondary metabolites. In addition to highlighting the drug discovery potential of the marine environment, this review discusses several of the pressing needs to increase the rate of drug discovery in marine natural products, and describes some of the work and new technologies that are contributing in this regard.
Collapse
Affiliation(s)
- John J Bowling
- The University of Mississippi, Department of Pharmacognosy, School of Pharmacy, University, MS 38677, USA +1 662 915 5730 ; +1 662 915 6975 ;
| | | | | | | |
Collapse
|
37
|
Humeniuk R, Menon LG, Mishra PJ, Saydam G, Longo-Sorbello GSA, Elisseyeff Y, Lewis LD, Aracil M, Jimeno J, Bertino JR, Banerjee D. Aplidin synergizes with cytosine arabinoside: functional relevance of mitochondria in Aplidin-induced cytotoxicity. Leukemia 2007; 21:2399-405. [PMID: 17713546 DOI: 10.1038/sj.leu.2404911] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Aplidin (plitidepsin) is a novel marine-derived antitumor agent presently undergoing phase II clinical trials in hematological malignancies and solid tumors. Lack of bone marrow toxicity has encouraged further development of this drug for treatment of leukemia and lymphoma. Multiple signaling pathways have been shown to be involved in Aplidin-induced apoptosis and cell cycle arrest in G1 and G2 phase. However, the exact mechanism(s) of Aplidin action remains to be elucidated. Here we demonstrate that mitochondria-associated or -localized processes are the potential cellular targets of Aplidin. Whole genome gene-expression profiling (GEP) revealed that fatty acid metabolism, sterol biosynthesis and energy metabolism, including the tricarboxylic acid cycle and ATP synthesis are affected by Aplidin treatment. Moreover, mutant MOLT-4, human leukemia cells lacking functional mitochondria, were found to be resistant to Aplidin. Cytosine arabinoside (araC), which also generates oxidative stress but does not affect the ATP pool, showed synergism with Aplidin in our leukemia and lymphoma models in vitro and in vivo. These studies provide new insights into the mechanism of action of Aplidin. The efficacy of the combination of Aplidin and araC is currently being evaluated in clinical phase I/II program for the treatment of patients with relapsed leukemia and high-grade lymphoma.
Collapse
MESH Headings
- Adenosine Triphosphate/biosynthesis
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Cell Cycle/drug effects
- Cell Line, Tumor/drug effects
- Cell Line, Tumor/transplantation
- Cytarabine/administration & dosage
- Cytarabine/pharmacology
- Depsipeptides/administration & dosage
- Depsipeptides/pharmacology
- Doxorubicin/pharmacology
- Drug Screening Assays, Antitumor
- Drug Synergism
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic/drug effects
- Humans
- K562 Cells/drug effects
- Leukemia-Lymphoma, Adult T-Cell/drug therapy
- Leukemia-Lymphoma, Adult T-Cell/pathology
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Methylprednisolone/pharmacology
- Mice
- Mice, SCID
- Mitochondria/drug effects
- Mitochondria/physiology
- Mitoxantrone/pharmacology
- Oxidative Stress/drug effects
- Peptides, Cyclic
- Specific Pathogen-Free Organisms
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- R Humeniuk
- Department of Medicine and Pharmacology, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, New Brunswick, NJ 08903, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
González-Santiago L, Alfonso P, Suárez Y, Núñez A, García-Fernández LF, Alvarez E, Muñoz A, Casal JI. Proteomic Analysis of the Resistance to Aplidin in Human Cancer Cells. J Proteome Res 2007; 6:1286-94. [PMID: 17338558 DOI: 10.1021/pr060430+] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Aplidin (plitidepsin) is an antitumoral agent that induces apoptosis via Rac1-JNK activation. A proteomic approach using 2D-DIGE technology found 52 cytosolic and 39 membrane proteins differentially expressed in wild-type and Aplidin-resistant HeLa cells, of which 39 and 27 were identified by MALDI-TOF mass spectrometry and database interrogation. A number of proteins involved in apoptosis pathways were found to be deregulated. Alterations in Rab geranylgeranyltransferase, protein disulfide isomerase (PDI), cystathionine gamma-lyase, ezrin, and cyclophilin A (CypA) were confirmed by immunoblotting. Moreover, the role of PDI and CypA in Aplidin resistance was functionally confirmed by using the inhibitor bacitracin and overexpression, respectively. These deregulated proteins are candidates to mediate, at least partially, Aplidin action and might provide a route to the cells to escape the induction of apoptosis by this drug.
Collapse
Affiliation(s)
- Laura González-Santiago
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Arturo Duperier, 4, E-28029 Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Kowalik AS, Johnson CL, Chadi SA, Weston JY, Fazio EN, Pin CL. Mice lacking the transcription factor Mist1 exhibit an altered stress response and increased sensitivity to caerulein-induced pancreatitis. Am J Physiol Gastrointest Liver Physiol 2007; 292:G1123-32. [PMID: 17170023 DOI: 10.1152/ajpgi.00512.2006] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Several animal models have been developed to investigate the pathobiology of pancreatitis, but few studies have examined the effects that altered pancreatic gene expression have in these models. In this study, the sensitivity to secretagogue-induced pancreatitis was examined in a mouse line that has an altered acinar cell environment due to the targeted deletion of Mist1. Mist1 is an exocrine specific transcription factor important for the complete differentiation and function of pancreatic acinar cells. Mice lacking the Mist1 gene [Mist1 knockout (KO) mice] exhibit cellular disorganization and functional defects in the exocrine pancreas but no gross morphological defects. Following the induction of pancreatitis with caerulein, a CCK analog, we observed elevated serum amylase levels, necrosis, and tissue damage in Mist1 KO mice, indicating increased pancreatic damage. There was also a delay in the regeneration of acinar tissue in Mist1 KO animals. Molecular profiling revealed an altered activation of stress response genes in Mist1 KO pancreatic tissue compared with wild-type (WT) tissue following the induction of pancreatitis. In particular, Western blot analysis for activating transcription factor 3 and phosphorylated eukaryotic initiation factor 2alpha (eIF2alpha), mediators of endoplasmic reticulum (ER) stress, indicated limited activation of this pathway in Mist1 KO animals compared with WT controls. Conversely, Mist1 KO pancreatic tissue exhibits increased expression of growth arrest and DNA damage inducible 34 protein, an inhibitor of eIF2alpha phosphorylation, before and after the induction of pancreatitis. These finding suggest that activation of the ER stress pathway is a protective event in the progression of pancreatitis and highlight the Mist1 KO mouse line as an important new model for studying the molecular events that contribute to the sensitivity to pancreatic injury.
Collapse
MESH Headings
- Activating Transcription Factor 3/genetics
- Activating Transcription Factor 3/metabolism
- Acute Disease
- Amylases/blood
- Animals
- Antigens, Differentiation/genetics
- Antigens, Differentiation/metabolism
- Apoptosis/drug effects
- Basic Helix-Loop-Helix Transcription Factors/deficiency
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cells, Cultured
- Ceruletide
- Cholecystokinin/metabolism
- Cholecystokinin/pharmacology
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Endoplasmic Reticulum/metabolism
- Eukaryotic Initiation Factor-2/genetics
- Eukaryotic Initiation Factor-2/metabolism
- Gene Expression
- Immediate-Early Proteins/genetics
- Immediate-Early Proteins/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Pancreas, Exocrine/metabolism
- Pancreas, Exocrine/pathology
- Pancreatitis/chemically induced
- Pancreatitis/genetics
- Pancreatitis/metabolism
- Pancreatitis/pathology
- Pancreatitis/physiopathology
- Protein Phosphatase 1
- RNA, Messenger/metabolism
- Regeneration
- Severity of Illness Index
- Stress, Physiological/chemically induced
- Stress, Physiological/genetics
- Stress, Physiological/metabolism
- Stress, Physiological/pathology
- Stress, Physiological/physiopathology
- Time Factors
Collapse
Affiliation(s)
- Agnes S Kowalik
- Department of Physiology, The University of Western Ontario, London, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
Recent leaps in elucidating the biology of myeloma, particularly the intracellular pathways and the complex interaction with the bone marrow microenvironment, have resulted in an unprecedented surge of novel, targeted therapies and therapeutic regimens. There are currently over 30 new agents being tested in the treatment of multiple myeloma (MM). Many of these are novel, targeted agents that have demonstrated significant efficacy and prolonged survival. In this review, we summarize the current understanding of the mechanisms of action of novel therapies being tested in the preclinical and clinical settings in MM. These include agents that act directly on the intracellular signaling pathways, cell maintenance processes, and cell surface receptors. Finally, we present the clinical responses to some of these agents when used alone or in combination in clinical trials of patients with MM. Indeed, MM has become a model disease for the development of novel, therapeutic agents.
Collapse
Affiliation(s)
| | - Irene M. Ghobrial
- Harvard Medical School,
Boston, MA,
USA
- Jerome Lipper Multiple Myeloma Center,
Department of Medical Oncology,
Dana Farber Cancer Institute,
Boston, MA,
USA
| | - Kenneth C. Anderson
- Harvard Medical School,
Boston, MA,
USA
- Jerome Lipper Multiple Myeloma Center,
Department of Medical Oncology,
Dana Farber Cancer Institute,
Boston, MA,
USA
- *Kenneth C. Anderson:
| |
Collapse
|
41
|
Bogoyevitch MA, Kobe B. Uses for JNK: the many and varied substrates of the c-Jun N-terminal kinases. Microbiol Mol Biol Rev 2006; 70:1061-95. [PMID: 17158707 PMCID: PMC1698509 DOI: 10.1128/mmbr.00025-06] [Citation(s) in RCA: 455] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The c-Jun N-terminal kinases (JNKs) are members of a larger group of serine/threonine (Ser/Thr) protein kinases from the mitogen-activated protein kinase family. JNKs were originally identified as stress-activated protein kinases in the livers of cycloheximide-challenged rats. Their subsequent purification, cloning, and naming as JNKs have emphasized their ability to phosphorylate and activate the transcription factor c-Jun. Studies of c-Jun and related transcription factor substrates have provided clues about both the preferred substrate phosphorylation sequences and additional docking domains recognized by JNK. There are now more than 50 proteins shown to be substrates for JNK. These include a range of nuclear substrates, including transcription factors and nuclear hormone receptors, heterogeneous nuclear ribonucleoprotein K, and the Pol I-specific transcription factor TIF-IA, which regulates ribosome synthesis. Many nonnuclear substrates have also been characterized, and these are involved in protein degradation (e.g., the E3 ligase Itch), signal transduction (e.g., adaptor and scaffold proteins and protein kinases), apoptotic cell death (e.g., mitochondrial Bcl2 family members), and cell movement (e.g., paxillin, DCX, microtubule-associated proteins, the stathmin family member SCG10, and the intermediate filament protein keratin 8). The range of JNK actions in the cell is therefore likely to be complex. Further characterization of the substrates of JNK should provide clearer explanations of the intracellular actions of the JNKs and may allow new avenues for targeting the JNK pathways with therapeutic agents downstream of JNK itself.
Collapse
Affiliation(s)
- Marie A Bogoyevitch
- Cell Signalling Laboratory, Biochemistry and Molecular Biology (M310), School of Biomedical, Biomolecular and Chemical Sciences, University of Western Australia, 35 Stirling Highway, Crawley, Western Australia 6009, Australia.
| | | |
Collapse
|
42
|
Suárez Y, González-Santiago L, Zarich N, Dávalos A, Aranda JF, Alonso MA, Lasunción MA, Rojas JM, Muñoz A. Plitidepsin cellular binding and Rac1/JNK pathway activation depend on membrane cholesterol content. Mol Pharmacol 2006; 70:1654-63. [PMID: 16928956 DOI: 10.1124/mol.106.025569] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Plitidepsin (aplidin) is a marine cyclic depsipeptide in phase II clinical development against several neoplasias. Plitidepsin is a potent inducer of apoptosis through the sustained activation of Jun N-terminal kinase (JNK). We have reported that this activation depends on the early induction of oxidative stress, activation of Rac1 small GTPase, and the later down-regulation of MKP-1 phosphatase. Using Scatchard and saturation binding analyses, we have found that (14)C-labeled plitidepsin binds to a moderately high-affinity receptor (K(d) of 44.8 +/- 3.1 and 35.5 +/- 4.8 nM, respectively) in MDA-MB-231 breast cancer cells. Two minutes after addition to cells, half of the drug was membrane-bound and was subsequently found in the cytosolic fraction. At 4 degrees C, plitidepsin cellular binding was around 10-fold lower than at 37 degrees C but sufficed to induce cell death, suggesting that this process is triggered from the membrane. Depletion of plasma membrane cholesterol by short treatment with methyl-beta-cyclodextrin diminished plitidepsin binding and Rac1 and JNK activation. Rac1 is targeted to the plasma membrane by plitidepsin as shown by subcellular fractioning and immunofluorescence analysis followed by confocal microscopy. Methyl-beta-cyclodextrin blocked this effect. A subline of HeLa cells (HeLa-R), partially resistant to plitidepsin, showed similar affinity (K(d) of 79.5 +/- 2.5 versus 37.7 +/- 8.2 nM) but 7.5-fold lower binding capacity than wild-type HeLa cells. Moreover, HeLa-R cells had lower total (71%) and membrane (67%) cholesterol content and membrane-bound Rac1, and showed no Rac1 activation upon plitidepsin treatment. In conclusion, cellular plitidepsin uptake and induction of apoptosis via activation of the Rac1-JNK pathway is membrane-cholesterol dependent.
Collapse
Affiliation(s)
- Yajaira Suárez
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Cientificas, Universidad Autónoma de Madrid, Arturo Duperier, 4, E-28029 Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Zarich N, Oliva JL, Martínez N, Jorge R, Ballester A, Gutiérrez-Eisman S, García-Vargas S, Rojas JM. Grb2 is a negative modulator of the intrinsic Ras-GEF activity of hSos1. Mol Biol Cell 2006; 17:3591-7. [PMID: 16760435 PMCID: PMC1525251 DOI: 10.1091/mbc.e05-12-1104] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
hSos1 is a Ras guanine-nucleotide exchange factor. It was suggested that the carboxyl-terminal region of hSos1 down-regulates hSos1 functionality and that the intrinsic guanine-nucleotide exchange activity of this protein may be different before and after stimulation of tyrosine kinase receptors. Using different myristoylated hSos1 full-length and carboxyl-terminal truncated mutants, we show that Grb2 function accounts not only for recruitment of hSos1 to the plasma membrane but also for modulation of hSos1 activity. Our results demonstrate that the first two canonical Grb2 binding sites, inside the carboxyl-terminal region of hSos1, are responsible for this regulation. Following different approaches, such as displacement of Grb2 from the hSos1-Grb2 complex or depletion of Grb2 levels by small interfering RNA, we found that the full-length Grb2 proteins mediate negative regulation of the intrinsic Ras guanine-nucleotide exchange activity of hSos1.
Collapse
Affiliation(s)
- Natasha Zarich
- Unidad de Biología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| | - José Luis Oliva
- Unidad de Biología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| | - Natalia Martínez
- Unidad de Biología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| | - Rocío Jorge
- Unidad de Biología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| | - Alicia Ballester
- Unidad de Biología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| | - Silvia Gutiérrez-Eisman
- Unidad de Biología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| | - Susana García-Vargas
- Unidad de Biología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| | - José M. Rojas
- Unidad de Biología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain
| |
Collapse
|
44
|
Deng R, Li W, Guan Z, Zhou JM, Wang Y, Mei YP, Li MT, Feng GK, Huang W, Liu ZC, Han Y, Zeng YX, Zhu XF. Acetylcholinesterase expression mediated by c-Jun-NH2-terminal kinase pathway during anticancer drug-induced apoptosis. Oncogene 2006; 25:7070-7. [PMID: 16715131 DOI: 10.1038/sj.onc.1209686] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
It has been shown that acetylcholinesterase (AChE) expression was induced during apoptosis and the anti-sense oligonucleotides and siRNA of AChE may prevent apoptosis in various cell types. However, the mechanisms underlying AChE upregulation remain elusive. We demonstrated here that c-Jun NH2-terminal kinase (JNK) could mediate AChE expression. In this study, both etoposide and excisanin A, two anticancer agents, induced apoptosis in colon cancer cell line SW620 as determined by Annexin V staining, the cleavage of caspase-3 and the proteolytic degradation of poly (ADP-ribose) polymerase (PARP). The results showed that both the agents upregulated AChE in SW620 cells. In the meantime, JNK was also activated and the expression and phosphorylation of c-Jun increased in SW620 cells exposed to the two agents. The induced AChE mRNA and protein expression could be blocked by SP600125, a specific inhibitor of SAPK/JNK, and small interfering RNA directed against JNK1/2. Transfection with adenovirus-mediated dominant negative c-Jun also blocked the upregulation of AChE expression. Together, these results suggest that AChE expression may be mediated by the activation of JNK pathway during apoptosis through a c-Jun-dependent mechanism.
Collapse
Affiliation(s)
- R Deng
- State Key Laboratory of Oncology in Southern China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Bravo SB, García-Rendueles MER, Seoane R, Dosil V, Cameselle-Teijeiro J, López-Lázaro L, Zalvide J, Barreiro F, Pombo CM, Alvarez CV. Plitidepsin has a cytostatic effect in human undifferentiated (anaplastic) thyroid carcinoma. Clin Cancer Res 2006; 11:7664-73. [PMID: 16278386 DOI: 10.1158/1078-0432.ccr-05-0455] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Undifferentiated (anaplastic) thyroid carcinoma is a highly aggressive human cancer with very poor prognosis. Although there have been a few studies of candidate treatments, the fact that it is an infrequent tumor makes it very difficult to design clinical trials. A strong association has been observed between undifferentiated thyroid carcinoma and TP53 mutations in numerous molecular genetic and expression studies. Plitidepsin (Aplidin, PharmaMar, Madrid, Spain) is a novel anticancer compound obtained from a sea tunicate. This compound has been reported to induce apoptosis independently of TP53 status. We investigated the actions of plitidepsin in human thyroid cancer cells. In initial experiments using primary cultured cells from a differentiated (papillary) carcinoma, we found that 100 nmol/L plitidepsin induced apoptosis, whereas lower doses were cytostatic. Because our aim was to study the effects of plitidepsin at clinically relevant concentrations, subsequent experiments were done with a dosage regimen reflecting plasma concentrations observed in previously reported clinical trials: 100 nmol/L for 4 hours, followed by 10 nmol/L for 20 hours (4(100)/20(10) plitidepsin). This plitidepsin dosage regimen blocked the proliferation of a primary undifferentiated/anaplastic thyroid carcinoma culture obtained in our laboratory and of a commercial cell line (8305C) obtained from an undifferentiated thyroid carcinoma; however, it did not induce apoptosis. The proportion of cells in the G(1) phase of the cell cycle was greatly increased and the proportion in the S/G(2)-M phases greatly reduced, suggesting that plitidepsin blocks G(1)-to-S transition. Levels of the cyclin D1/cyclin-dependent kinase 4/p21 complex proteins were decreased and, in line with this, the levels of unphosphorylated Rb1 increased. The decrease in cell cycle proteins correlated with hypoacetylation of histone H3. Finally, we did experiments to assess how rapidly tumor cells return to their initial pretreatment proliferative behavior after 4(100)/20(10) plitidepsin treatment. Cells from undifferentiated tumors needed more than 3 days to recover logarithmic growth, and after 7 days, cell number was still significantly lower than in control cultures. 4(100)/20(10) plitidepsin inhibited the growth in soft agar. Together, our data show that plitidepsin is able to block in vitro cell cycle progression at concentrations similar to serum concentrations observed in vivo, and that this effect is persistent for several days after plitidepsin removal. Whether plitidepsin will prove to be clinically useful in the treatment of undifferentiated thyroid cancers remains to be established. However, our results raise the possibility that plitidepsin might be effective alone or in combination with radiotherapy and/or other drug treatments.
Collapse
Affiliation(s)
- Susana B Bravo
- Department of Physiology, School of Medicine, University of Santiago de Compostela, Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
González-Santiago L, Suárez Y, Zarich N, Muñoz-Alonso MJ, Cuadrado A, Martínez T, Goya L, Iradi A, Sáez-Tormo G, Maier JV, Moorthy A, Cato ACB, Rojas JM, Muñoz A. Aplidin® induces JNK-dependent apoptosis in human breast cancer cells via alteration of glutathione homeostasis, Rac1 GTPase activation, and MKP-1 phosphatase downregulation. Cell Death Differ 2006; 13:1968-81. [PMID: 16543941 DOI: 10.1038/sj.cdd.4401898] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aplidin is an antitumor agent in phase II clinical trials that induces apoptosis through the sustained activation of Jun N-terminal kinase (JNK). We report that Aplidin alters glutathione homeostasis increasing the ratio of oxidized to reduced forms (GSSG/GSH). Aplidin generates reactive oxygen species and disrupts the mitochondrial membrane potential. Exogenous GSH inhibits these effects and also JNK activation and cell death. We found two mechanisms by which Aplidin activates JNK: rapid activation of Rac1 small GTPase and downregulation of MKP-1 phosphatase. Rac1 activation was diminished by GSH and enhanced by L-buthionine (SR)-sulfoximine, which inhibits GSH synthesis. Downregulation of Rac1 by transfection of small interfering RNA (siRNA) duplexes or the use of a specific Rac1 inhibitor decreased Aplidin-induced JNK activation and cytotoxicity. Our results show that Aplidin induces apoptosis by increasing the GSSG/GSH ratio, a necessary step for induction of oxidative stress and sustained JNK activation through Rac1 activation and MKP-1 downregulation.
Collapse
Affiliation(s)
- L González-Santiago
- Instituto de Investigaciones Biomédicas 'Alberto Sols', Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Arturo Duperier, 4, Madrid E-28029, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lin HJ, Hsieh FC, Song H, Lin J. Elevated phosphorylation and activation of PDK-1/AKT pathway in human breast cancer. Br J Cancer 2006; 93:1372-81. [PMID: 16288304 PMCID: PMC2361529 DOI: 10.1038/sj.bjc.6602862] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Activation of kinases signalling pathways contributes to various malignant phenotypes in human cancers, including breast tumour. To examine the possible activation of these signalling molecules, we examined the phosphorylation status in 12 protein kinases and transcription factors in normal primary human mammary epithelial cells, telomerase-immortalised human breast epithelial cell line, and two breast cancer lines, MDA-MB-468 and MCF-7, using Kinexus phosphorylated protein screening assays. The phosphorylation of FAK, mTOR, p70S6K, and PDK-1 were elevated in both breast cancer cell lines, whereas the phosphorylation of AKT, EGFR, ErbB2/Her2, PDGFR, Shc, and Stat3 were elevated in only one breast cancer line compared to normal primary mammary epithelial cells and telomerase-immortalised breast epithelial cells. The same findings were confirmed by Western blotting and by kinase assays. We further substantiated the phosphorylation status of these molecules in tissue microarray slides containing 89 invasive breast cancer tissues as well as six normal mammary tissues with immunohistochemistry staining using phospho-specific antibodies. Consistent findings were obtained as greater than 70% of invasive breast carcinomas expressed moderate to high levels of phosphorylated PDK-1, AKT, p70S6K, and EGFR. In sharp contrast, phosphorylation of the same proteins was nearly undetectable or was at low levels in normal mammary tissues under the same assay. Elevated phosphorylation of PDK-1, AKT, mTOR, p70S6K, S6, EGFR, and Stat3 were highly associated with invasive breast tumours (P<0.05). Taken together, our results suggest that activation of these kinase pathways by phosphorylation may in part account for molecular pathogenesis of human breast carcinoma. Particularly, moderate to high level of PDK-1 phosphorylation was found in 86% of high-grade metastasised breast tumours. This is the first report demonstrating phosphorylation of PDK-1 is frequently elevated in breast cancer with concomitantly increased phosphorylation of downstream kinases, including AKT, mTOR, p70S6K, S6, and Stat3. This finding thus suggested PDK-1 may promote oncogenesis in part through the activation of AKT and p70S6K and rationalised that PDK-1 as well as downstream components of PDK-1 signalling pathway may be promising therapeutic targets to treat breast cancer.
Collapse
Affiliation(s)
- H-J Lin
- Division of Medical Technology, School of Allied Medical Professions, College of Medicine and Public Health, The Ohio State University, Suite 535A, Atwell Hall, 453 West 10th Street, Columbus, OH 43210, USA.
| | | | | | | |
Collapse
|
48
|
Xu C, Shen G, Yuan X, Kim JH, Gopalkrishnan A, Keum YS, Nair S, Kong ANT. ERK and JNK signaling pathways are involved in the regulation of activator protein 1 and cell death elicited by three isothiocyanates in human prostate cancer PC-3 cells. Carcinogenesis 2005; 27:437-45. [PMID: 16272172 DOI: 10.1093/carcin/bgi251] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Many isothiocyanates (ITCs) such as sulforaphane (SFN), phenethyl isothiocyanate (PEITC) and allyl isothiocyanate (AITC) are highly effectively in chemoprevention or reduction of the risk of cancer and possess antitumor activities in vitro and in vivo. The activator protein 1 (AP-1) and MAPK signaling pathways are believed to play an important role in cancer chemoprevention and chemotherapy due to their involvement in tumor cell growth, proliferation, apoptosis and survival. In the present study, we determined the effects of SFN, PEITC and AITC on AP-1 activation, and investigated the roles of extracellular signal-regulated protein kinase (ERK) and c-Jun N-terminal kinase (JNK) signaling pathways in the regulation of AP-1 activation and cell death elicited by these ITCs in human prostate cancer PC-3 cells. SFN, PEITC and AITC each induced AP-1 activity potently and caused a significant elevation in the phosphorylation of ERK1/2, JNK1/2, Elk-1 and c-Jun. Transfection with ERK2 and upstream kinase DNEE-MEK1 activated AP-1 activity, and transfection with dominant-negative mutant ERK2 (dnERK2) potently decreased AP-1 activation induced by SFN, PEITC and AITC. Transfection with JNK1 and upstream kinase MKK7 activated AP-1 activity, and transfection with dominant-negative mutant JNK1-APF significantly attenuated AP-1 activation induced by SFN, PEITC and AITC. Pretreatment with MEK1-ERK inhibitor U0126 and JNK inhibitor SP600125 substantially attenuated the decrease in cell viability induced by SFN, PEITC and AITC. Transfection with dnERK2 and JNK1-APF significantly reversed the decrease of Bcl-2 expression elicited by these ITCs. Furthermore, transfection with dnERK2 and JNK1-APF blocked the apoptosis induced by these ITCs in PC-3 cells. Taken together, our results indicate that the activation of the ERK and JNK signaling pathways is important for transcriptional activity of AP-1 and is involved in the regulation of cell death elicited by ITCs in PC-3 cells.
Collapse
Affiliation(s)
- Changjiang Xu
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Road, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Faivre S, Chièze S, Delbaldo C, Ady-Vago N, Guzman C, Lopez-Lazaro L, Lozahic S, Jimeno J, Pico F, Armand JP, Martin JAL, Raymond E. Phase I and Pharmacokinetic Study of Aplidine, a New Marine Cyclodepsipeptide in Patients With Advanced Malignancies. J Clin Oncol 2005; 23:7871-80. [PMID: 16172454 DOI: 10.1200/jco.2005.09.357] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PurposeTo establish the safety, pharmacokinetic parameters, maximum-tolerated dose, and recommended dose of aplidine, a novel marine cyclodepsipeptide, in patients with advanced cancer.Patients and MethodsUsing a modified Fibonacci method, we performed a phase I and pharmacokinetic study of aplidine administered as a 24-hour intravenous infusion every 2 weeks.ResultsSixty-seven patients received aplidine at a dose ranging from 0.2 to 8 mg/m2. Dose-limiting myotoxicity corresponding to grade 2 to 3 creatine phosphokinase elevation and grade 1 to 2 myalgia and muscle weakness occurred in two of six patients at 6 mg/m2. No cardiac toxicity was observed. Electron microscopy analysis showed the disappearance of thick filaments of myosin. Grade 3 muscle toxicity occurred in three of 14 patients at the recommended dose of 5 mg/m2and seemed to be more readily reversible with oral carnitine (1 g/10 kg). Therefore, dose escalation was resumed using carnitine prophylactically, allowing an increase in the recommended dose to 7 mg/m2. Other toxicities were nausea and vomiting, diarrhea, asthenia, and transaminase elevation with mild hematologic toxicity. Aplidine displayed a long half-life (21 to 44 hours), low clearance (45 to 49 L/h), and a high volume of distribution (1,036 to 1,124 L) with high interpatient variability in plasma, whereas in whole blood, clearance ranged from 3.0 to 6.2 L/h. Minor responses and prolonged tumor stabilizations were observed in patients with medullary thyroid carcinoma.ConclusionMuscle toxicity was dose limiting in this study. Recommended doses of aplidine were 5 and 7 mg/m2without and with carnitine, respectively. The role of carnitine will be further explored in phase II studies.
Collapse
Affiliation(s)
- Sandrine Faivre
- Department of Medicine, Institute Gustave-Roussy, Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
|