151
|
Tan TT, White E. Therapeutic Targeting of Death Pathways in Cancer: Mechanisms for Activating Cell Death in Cancer Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 615:81-104. [DOI: 10.1007/978-1-4020-6554-5_5] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
152
|
Hidalgo M. Clinical Development of Epidermal Growth Factor Receptor (EGFR) Tyrosine Kinase Inhibitors: What Lessons Have We Learned? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2008; 610:128-43. [DOI: 10.1007/978-0-387-73898-7_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
153
|
Boruban C, Sencan O, Akmansu M, Atik ET, Ozbek S. Metastatic gastrointestinal stromal tumor with long-term response after treatment with concomitant radiotherapy and imatinib mesylate. Anticancer Drugs 2007; 18:969-72. [PMID: 17667604 DOI: 10.1097/cad.0b013e3280e94982] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Total surgical excision of the tumor is considered to be the only hope in treatment of malignant mesenchymal tumors. The roles of radiotherapy and/or chemotherapy have not yet been established. We report here a case of metastatic gastrointestinal stromal tumors with a dramatic long duration of response after treatment with concurrent radiotherapy and imatinib mesylate. The patient had a long-term complete response at the radiotherapy region with concomitant imatinib therapy although previous metastatic sides persisted with partial response.
Collapse
Affiliation(s)
- Cem Boruban
- Department of Medical Oncology, Selcuk University Faculty of Medicine, Konya, Turkey
| | | | | | | | | |
Collapse
|
154
|
Komarova NL, Wodarz D. Effect of cellular quiescence on the success of targeted CML therapy. PLoS One 2007; 2:e990. [PMID: 17912367 PMCID: PMC1991595 DOI: 10.1371/journal.pone.0000990] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2007] [Accepted: 09/04/2007] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Similar to tissue stem cells, primitive tumor cells in chronic myelogenous leukemia have been observed to undergo quiescence; that is, the cells can temporarily stop dividing. Using mathematical models, we investigate the effect of cellular quiescence on the outcome of therapy with targeted small molecule inhibitors. METHODS AND RESULTS According to the models, the initiation of treatment can result in different patterns of tumor cell decline: a biphasic decline, a one-phase decline, and a reverse biphasic decline. A biphasic decline involves a fast initial phase (which roughly corresponds to the eradication of cycling cells by the drug), followed by a second and slower phase of exponential decline (corresponding to awakening and death of quiescent cells), which helps explain clinical data. We define the time when the switch to the second phase occurs, and identify parameters that determine whether therapy can drive the tumor extinct in a reasonable period of time or not. We further ask how cellular quiescence affects the evolution of drug resistance. We find that it has no effect on the probability that resistant mutants exist before therapy if treatment occurs with a single drug, but that quiescence increases the probability of having resistant mutants if patients are treated with a combination of two or more drugs with different targets. Interestingly, while quiescence prolongs the time until therapy reduces the number of cells to low levels or extinction, the therapy phase is irrelevant for the evolution of drug resistant mutants. If treatment fails as a result of resistance, the mutants will have evolved during the tumor growth phase, before the start of therapy. Thus, prevention of resistance is not promoted by reducing the quiescent cell population during therapy (e.g., by a combination of cell activation and drug-mediated killing). CONCLUSIONS The mathematical models provide insights into the effect of quiescence on the basic kinetics of the response to targeted treatment of CML. They identify determinants of success in the absence of drug resistant mutants, and elucidate how quiescence influences the emergence of drug resistant mutants.
Collapse
Affiliation(s)
- Natalia L. Komarova
- Department of Mathematics, University of California Irvine, Irvine, California, United States of America
- Department of Ecology and Evolution, University of California Irvine, Irvine, California, United States of America
| | - Dominik Wodarz
- Department of Mathematics, University of California Irvine, Irvine, California, United States of America
- Department of Ecology and Evolution, University of California Irvine, Irvine, California, United States of America
| |
Collapse
|
155
|
Cui JJ. Inhibitors targeting hepatocyte growth factor receptor and their potential therapeutic applications. Expert Opin Ther Pat 2007. [DOI: 10.1517/13543776.17.9.1035] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
156
|
Bende G, Kollipara S, Kolachina V, Saha R. Development and Validation of an Stability Indicating RP-LC Method for Determination of Imatinib Mesylate. Chromatographia 2007. [DOI: 10.1365/s10337-007-0415-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
157
|
Komarova NL, Wodarz D. Stochastic modeling of cellular colonies with quiescence: an application to drug resistance in cancer. Theor Popul Biol 2007; 72:523-38. [PMID: 17915274 DOI: 10.1016/j.tpb.2007.08.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2007] [Revised: 05/19/2007] [Accepted: 08/08/2007] [Indexed: 10/22/2022]
Abstract
Several cancers are thought to be driven by cells with stem cell like properties. An important characteristic of stem cells, which also applies to primitive tumor cells, is the ability to undergo quiescence, where cells can temporarily stop the cell cycle. Cellular quiescence can affect the kinetics of tumor growth, and the susceptibility of the cells to therapy. To study how quiescence affects treatment, we formulate a stochastic birth-death process with quiescence, on a combinatorial cellular mutation network, and consider the pre-treatment (growth) and treatment (decay) regimes. We find that, in the absence of mutations, treatment (if sufficiently strong) will proceed as a biphasic decline with the first (faster) phase driven by the elimination of the cycling cells and the second (slower) phase limited by the process of cell awakening. Other regimes are possible for weaker treatments. We also describe how the process of mutant generation is influenced by quiescence. Interestingly, for single-drug treatments, the probability to have resistance at start of treatment is independent of quiescence. For two or more drugs, the probability to have generated resistant mutants before treatment grows with quiescence. Finally, we study the influence of quiescence on the treatment phase. Starting from a given composition of mutants, the chances of treatment success are not influenced by the presence of quiescence.
Collapse
Affiliation(s)
- Natalia L Komarova
- Department of Mathematics, University of California, Irvine, CA 92697, USA.
| | | |
Collapse
|
158
|
Rix U, Hantschel O, Dürnberger G, Remsing Rix LL, Planyavsky M, Fernbach NV, Kaupe I, Bennett KL, Valent P, Colinge J, Köcher T, Superti-Furga G. Chemical proteomic profiles of the BCR-ABL inhibitors imatinib, nilotinib, and dasatinib reveal novel kinase and nonkinase targets. Blood 2007; 110:4055-63. [PMID: 17720881 DOI: 10.1182/blood-2007-07-102061] [Citation(s) in RCA: 517] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The BCR-ABL tyrosine kinase inhibitor imatinib represents the current frontline therapy in chronic myeloid leukemia. Because many patients develop imatinib resistance, 2 second-generation drugs, nilotinib and dasatinib, displaying increased potency against BCR-ABL were developed. To predict potential side effects and novel medical uses, we generated comprehensive drug-protein interaction profiles by chemical proteomics for all 3 drugs. Our studies yielded 4 major findings: (1) The interaction profiles of the 3 drugs displayed strong differences and only a small overlap covering the ABL kinases. (2) Dasatinib bound in excess of 30 Tyr and Ser/Thr kinases, including major regulators of the immune system, suggesting that dasatinib might have a particular impact on immune function. (3) Despite the high specificity of nilotinib, the receptor tyrosine kinase DDR1 was identified and validated as an additional major target. (4) The oxidoreductase NQO2 was bound and inhibited by imatinib and nilotinib at physiologically relevant drug concentrations, representing the first nonkinase target of these drugs.
Collapse
MESH Headings
- Benzamides
- Dasatinib
- Discoidin Domain Receptor 1
- Dose-Response Relationship, Drug
- Drug Resistance, Neoplasm
- Drug Screening Assays, Antitumor
- Fusion Proteins, bcr-abl
- Humans
- Imatinib Mesylate
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/metabolism
- Piperazines/chemistry
- Piperazines/pharmacology
- Piperazines/therapeutic use
- Protein Kinase Inhibitors/chemistry
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/metabolism
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Protein-Tyrosine Kinases/metabolism
- Proteomics
- Pyrimidines/chemistry
- Pyrimidines/pharmacology
- Pyrimidines/therapeutic use
- Quinone Reductases/antagonists & inhibitors
- Quinone Reductases/metabolism
- Receptor Protein-Tyrosine Kinases/antagonists & inhibitors
- Receptor Protein-Tyrosine Kinases/metabolism
- Thiazoles/chemistry
- Thiazoles/pharmacology
- Thiazoles/therapeutic use
Collapse
Affiliation(s)
- Uwe Rix
- Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
159
|
Arvanitis C, Bendapudi PK, Bachireddy P, Felsher DW. Identifying critical signaling molecules for the treatment of cancer. Recent Results Cancer Res 2007; 172:5-24. [PMID: 17607933 DOI: 10.1007/978-3-540-31209-3_2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Constadina Arvanitis
- Department of Medicine, Stanford University School of Medicine, CA 94305-5151, USA
| | | | | | | |
Collapse
|
160
|
Sakamoto J, Annapragada A, Decuzzi P, Ferrari M. Antibiological barrier nanovector technology for cancer applications. Expert Opin Drug Deliv 2007; 4:359-69. [PMID: 17683250 DOI: 10.1517/17425247.4.4.359] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The advent of sophisticated drug delivery strategies for cancer applications has inundated the scientific and clinical community with new tactics and approaches such as molecular targeting, nanotechnology-based methods and personalized therapies. Unfortunately, the clinical impact has been moderate at best, falling significantly short from revolutionizing existing chemotherapeutic methodologies. To this day, a cancer patient has a higher probability of receiving traditional systemically administered drugs than a more sophisticated targeted or nanotechnology-based therapeutic. This is not a reflection upon the novelty or quality of the technologies, but an indication of opportunity for a new approach that offers the realisation of the full potential of these scientific advances. This approach acknowledges the significance of the numerous biological barriers presented in the human body and their sequential nature. It is then recommended that computational mathematical tools are used to predict which nanovectors, surface modifications, therapeutic agents and penetration enhancers to use for a multi-stage drug delivery strategy. An approach where several stages of micro-/nano-vectors are nested within each other and delivered to overcome specific biological barriers to ultimately release a concentrated dose of a therapeutic payload at the intended lesion site. This novel, multi-stage strategy enables efficient localised delivery of chemotoxic drugs that may lead to significant improvements in therapy efficacy, reduced systemic toxicity and decreased total amount of injected drugs.
Collapse
Affiliation(s)
- Jason Sakamoto
- Alliance for NanoHealth, 1825 Herman Pressler Street, Suite 537A, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
161
|
Orfao A, Garcia-Montero AC, Sanchez L, Escribano L. Recent advances in the understanding of mastocytosis: the role of KIT mutations. Br J Haematol 2007; 138:12-30. [PMID: 17555444 DOI: 10.1111/j.1365-2141.2007.06619.x] [Citation(s) in RCA: 154] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mastocytosis is a heterogeneous disorder characterised by the expansion and accumulation of mast cells in different organs and tissues. Mast cell physiology is closely dependent on activation of the stem cell factor/Kit signalling pathways and accumulating evidences confirm the physiopathological key role of activating KIT mutations (typically D816V) in mastocytosis and their relationship with the clinical manifestations of the disease. This paper reviews the most recent advances in the understanding of the molecular mechanisms associated with KIT mutations in mastocytosis, including recent data about the use of new therapies targeting the Kit molecule and its associated downstream signalling pathways.
Collapse
Affiliation(s)
- Alberto Orfao
- Centro de Investigación del Cáncer/IBMCC, Departamento de Medicina and Servicio General de Citometría, University of Salamanca, Salamanca, Spain.
| | | | | | | |
Collapse
|
162
|
Kurmasheva RT, Houghton PJ. Pediatric oncology. Curr Opin Chem Biol 2007; 11:424-32. [PMID: 17652007 PMCID: PMC2265418 DOI: 10.1016/j.cbpa.2007.05.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Revised: 05/03/2007] [Accepted: 05/04/2007] [Indexed: 11/24/2022]
Abstract
Intensive use of cytotoxic agents in multimodality therapeutic regimens has resulted in almost 80% five-year disease-free survival and cure in the majority of childhood cancer patients. However, such success has come at the expense of severe acute or delayed toxicities and an increased occurrence of secondary cancers. With an increasing understanding of the genetic changes that underlie transformation in childhood cancer, rational approaches using agents that target these transforming events are being developed. Current and future strategies in developing tumor-selective therapy using inhibitors of signaling pathways dysregulated in leukemias (FLT3, NOTCH1) and solid/brain tumors (ErbB1-4, IGF-IR, PTCH1), and the challenges in developing less toxic, but equally effective treatments in pediatric oncology are presented.
Collapse
Affiliation(s)
- Raushan T Kurmasheva
- Department of Molecular Pharmacology, St. Jude Children's Research Hospital, 332 North Lauderdale Street, Memphis, TN 38105-2794, United States
| | | |
Collapse
|
163
|
Shachaf CM, Perez OD, Youssef S, Fan AC, Elchuri S, Goldstein MJ, Shirer AE, Sharpe O, Chen J, Mitchell DJ, Chang M, Nolan GP, Steinman L, Felsher DW. Inhibition of HMGcoA reductase by atorvastatin prevents and reverses MYC-induced lymphomagenesis. Blood 2007; 110:2674-84. [PMID: 17622571 PMCID: PMC1988916 DOI: 10.1182/blood-2006-09-048033] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Statins are a class of drugs that inhibit 3-hydroxy-3-methylglutaryl-coenzyme A (HMGcoA) reductase, a critical enzyme in the mevalonate pathway. Several reports document that statins may prevent different human cancers. However, whether or not statins can prevent cancer is controversial due to discordant results. One possible explanation for these conflicting conclusions is that only some tumors or specific statins may be effective. Here, we demonstrate in an in vivo transgenic model in which atorvastatin reverses and prevents the onset of MYC-induced lymphomagenesis, but fails to reverse or prevent tumorigenesis in the presence of constitutively activated K-Ras (G12D). Using phosphoprotein fluorescence-activated cell sorter (FACS) analysis, atorvastatin treatment was found to result in the inactivation of the Ras and ERK1/2 signaling pathways associated with the dephosphorylation and inactivation of MYC. Correspondingly, tumors with a constitutively activated K-Ras (G12D) did not exhibit dephosphorylation of ERK1/2 and MYC. Atorvastatin's effects on MYC were specific to the inhibition of HMGcoA reductase, as treatment with mevalonate, the product of HMG-CoA reductase activity, abrogated these effects and inhibited the ability of atorvastatin to reverse or suppress tumorigenesis. Also, RNAi directed at HMGcoA reductase was sufficient to abrogate the neoplastic properties of MYC-induced tumors. Thus, atorvastatin, by inhibiting HMGcoA reductase, induces changes in phosphoprotein signaling that in turn prevent MYC-induced lymphomagenesis.
Collapse
Affiliation(s)
- Catherine M Shachaf
- Division of Medical Oncology, Department of Medicine, Stanford University, Stanford, CA 94305-5151, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
164
|
Chaturvedi A, Battmer K, Schaefer D, Ganser A, Eder M, Scherr M. Comparison between molecularly defined and conventional therapeutics in a conditional BCR-ABL cell culture model. Oligonucleotides 2007; 17:22-34. [PMID: 17461760 DOI: 10.1089/oli.2006.0054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Accumulating knowledge about the molecular mechanisms causing human diseases can support the development of targeted therapies such as imatinib, a BCR-ABL-specific tyrosine kinase inhibitor to treat chronic myeloid leukemia (CML). Here, we use lentivirus-mediated RNA interference (RNAi) targeting BCR-ABL and the downstream signaling molecules SHP2, STAT5, and Gab2 to compare the efficacy and specificity of molecularly defined therapeutics with that of conventional cytotoxic drugs (cytarabine, doxorubicin, etoposide) in a conditional BCR-ABL cell culture model. IC(50) values were determined for each drug in TonB cells cultured either with interleukin-3 (IL-3) or BCR-ABL, and molecularly defined therapies were studied using lentivirally expressed shRNAs. We demonstrate that conventional anti-leukemic drugs have small or no differential effects under different cell culture conditions, whereas both imatinib and specific RNAi significantly inhibit proliferation of TonB cells in the presence of BCR-ABL but not IL-3. To study molecularly defined combination therapy, we evaluated either imatinib in TonB cells with target-specific RNAi or we used lentiviral vectors to induce combinatorial RNAi through simultaneous expression of two shRNAs. These combination therapies result in increased efficacy without loss in specificity. Interestingly, combinatorial RNAi can specifically deplete TonB cell cultures in the presence of BCR-ABL, even without targeting the oncogene itself. This model provides a tool to evaluate potential therapeutic targets and to quantify efficacy and specificity preclinically of new combination therapies in BCR-ABL-positive cells.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Animals
- Antineoplastic Agents/therapeutic use
- Benzamides
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Genetic Therapy
- Imatinib Mesylate
- Inhibitory Concentration 50
- Interleukin-3/pharmacology
- Intracellular Signaling Peptides and Proteins/antagonists & inhibitors
- Intracellular Signaling Peptides and Proteins/genetics
- Lentivirus/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy
- Mice
- Models, Biological
- Phosphoproteins/antagonists & inhibitors
- Phosphoproteins/genetics
- Piperazines/therapeutic use
- Protein Tyrosine Phosphatase, Non-Receptor Type 11
- Protein Tyrosine Phosphatases/antagonists & inhibitors
- Protein Tyrosine Phosphatases/genetics
- Pyrimidines/therapeutic use
- RNA Interference
- RNA, Small Interfering/genetics
- STAT5 Transcription Factor/antagonists & inhibitors
- STAT5 Transcription Factor/genetics
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Anuhar Chaturvedi
- Hannover Medical School, Department of Hematology, Hemostasis, and Oncology, 30625 Hannover, Germany
| | | | | | | | | | | |
Collapse
|
165
|
Abstract
Biomarkers have been a buzz word in drug development for the last 5 years. But where do we stand now? This perspective article will demonstrate to which extent biomarkers have impacted drug development and the use of drugs. In particular, the different types of biomarkers, their identification, validation and use in different phases of drug development from drug discovery, to approval, to clinical application will be discussed as well as the state-of-the-art biomarker technologies and promising future methods. The high interest in biomarkers has generated the need for development of new technologies and refinement of existing ones. Besides discussing their perspectives of applications, the present article also illustrates the future of biomarker development in terms of qualification for regulatory use and co-development.
Collapse
Affiliation(s)
- Estelle Marrer
- Novartis Pharma AG, Development, 4002 Basel, Switzerland.
| | | |
Collapse
|
166
|
Verkhivker GM. Exploring sequence-structure relationships in the tyrosine kinome space: functional classification of the binding specificity mechanisms for cancer therapeutics. Bioinformatics 2007; 23:1919-26. [PMID: 17537753 DOI: 10.1093/bioinformatics/btm277] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
MOTIVATION Evolutionary and structural conservation patterns shared by more than 500 of identified protein kinases have led to complex sequence-structure relationships of cross-reactivity for kinase inhibitors. Understanding the molecular basis of binding specificity for protein kinases family, which is the central problem in discovery of cancer therapeutics, remains challenging as the inhibitor selectivity is not readily interpreted from chemical proteomics studies, neither it is easily discernable directly from sequence or structure information. We present an integrated view of sequence-structure-binding relationships in the tyrosine kinome space in which evolutionary analysis of the kinases binding sites is combined with computational proteomics profiling of the inhibitor-protein interactions. This approach provides a functional classification of the binding specificity mechanisms for cancer agents targeting protein tyrosine kinases. RESULTS The proposed functional classification of the kinase binding specificities explores mechanisms in which structural plasticity of the tyrosine kinases and sequence variation of the binding-site residues are linked with conformational preferences of the inhibitors in achieving effective drug binding. The molecular basis of binding specificity for tyrosine kinases may be largely driven by conformational adaptability of the inhibitors to an ensemble of structurally different conformational states of the enzyme, rather than being determined by their phylogenetic proximity in the kinome space or differences in the interactions with the variable binding-site residues. This approach provides a fruitful functional linkage between structural bioinformatics analysis and disease by unraveling the molecular basis of kinase selectivity for the prominent kinase drugs (Imatinib, Dasatinib and Erlotinib) which is consistent with structural and proteomics experiments.
Collapse
Affiliation(s)
- Gennady M Verkhivker
- Department of Pharmaceutical Chemistry, School of Pharmacy, Center for Bioinformatics, The University of Kansas, Lawrence, KS 66047-1620, USA.
| |
Collapse
|
167
|
Komarova NL, Wu L, Baldi P. The fixed-size Luria-Delbruck model with a nonzero death rate. Math Biosci 2007; 210:253-90. [PMID: 17583754 DOI: 10.1016/j.mbs.2007.04.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2007] [Revised: 04/06/2007] [Accepted: 04/20/2007] [Indexed: 12/28/2022]
Abstract
What is the expected number of mutants in a stochastically growing colony once it reaches a given size, N? This is a variant of the famous Luria-Delbruck model which studies the distribution of mutants after a given time-lapse. Instead of fixing the time-lapse, we assume that the colony size is a measurable quantity, which is the case in many in-vivo oncological and other applications. We study the mean number of mutants for an arbitrary cell death rate, and give partial results for the variance. For a restricted set of parameters we provide analytical results; we also design a very efficient computational method to calculate the mean, which works for most of the parameter values, and any colony size, no matter how large. We find that a cellular population with a higher death rate will contain a larger number of mutants than a population of equal size with a smaller death rate. Also, a very large population will contain a larger percentage of mutants; that is, irreversible mutations act like a force of selection, even though here the mutants are assumed to have no selective advantage. Finally, we investigate the applicability of the traditional, 'fixed-time' approach and find that it approximates the 'fixed-size' problem whenever stochastic effects are negligible.
Collapse
Affiliation(s)
- Natalia L Komarova
- Department of Mathematics, University of California, Irvine, CA 92697, United States.
| | | | | |
Collapse
|
168
|
Kast RE, Altschuler EL. Consideration of use of phenothiazines in particular trifluorperazine for epidermal growth factor receptor associated cancers. Med Hypotheses 2007; 69:1074-5. [PMID: 17448610 DOI: 10.1016/j.mehy.2006.08.048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2006] [Accepted: 08/09/2006] [Indexed: 11/23/2022]
Abstract
Papers from a generation ago suggested that phenothiazines--in particular trifluorperazine (Stelazine) a medicinal approved by the FDA and still commonly used for schizophrenia--downregulate the epidermal growth factor receptor. As numerous cancers--e.g., colon cancer, breast cancer, pancreatic cancer and glioblastoma--are dependent on signaling via this receptor, we here suggest that phenothiazines such as trifluorperazine be considered for use in epidermal growth factor receptor associated cancers.
Collapse
Affiliation(s)
- Richard E Kast
- Department of Psychiatry, College of Medicine, University of Vermont, 2 Church Street, Burlington, VT 05401, United States
| | | |
Collapse
|
169
|
Abstract
Despite advancements in genetics, chemistry, and protein engineering, recent years have seen fewer approvals of new drugs, increases in development costs, and high-profile drug withdrawals. This article focuses on technologic methods for improving drug development efficiency. These technologies include high-content cell screening, expression profiling, mass spectroscopy, mouse models of disease, and a post-launch screening program that enables investigations of adverse drug effects. Implementation of these new technologies promises to improve performance in drug development and safety.
Collapse
Affiliation(s)
- C Thomas Caskey
- Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA.
| |
Collapse
|
170
|
Greene LM, Kelly L, Onnis V, Campiani G, Lawler M, Williams DC, Zisterer DM. STI-571 (imatinib mesylate) enhances the apoptotic efficacy of pyrrolo-1,5-benzoxazepine-6, a novel microtubule-targeting agent, in both STI-571-sensitive and -resistant Bcr-Abl-positive human chronic myeloid leukemia cells. J Pharmacol Exp Ther 2007; 321:288-97. [PMID: 17202400 DOI: 10.1124/jpet.106.116640] [Citation(s) in RCA: 18] [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
Interactions between the Bcr-Abl kinase inhibitor STI-571 (imatinib mesylate) and a novel microtubule-targeting agent (MTA), pyrrolo-1,5-benzoxazepine (PBOX)-6, were investigated in STI-571-sensitive and -resistant human chronic myeloid leukemia (CML) cells. Cotreatment of PBOX-6 with STI-571 induced significantly more apoptosis in Bcr-Abl-positive CML cell lines (K562 and LAMA-84) than either drug alone (P < 0.01). Cell cycle analysis of propidium iodide-stained cells showed that STI-571 significantly reduced PBOX-6-induced G2M arrest and polyploid formation with a concomitant increase in apoptosis. Similar results were obtained in K562 CML cells using lead MTAs (paclitaxel and nocodazole) in combination with STI-571. Potentiation of PBOX-6-induced apoptosis by STI-571 was specific to Bcr-Abl-positive leukemia cells with no cytoxic effects observed on normal peripheral blood cells. The combined treatment of STI-571 and PBOX-6 was associated with the down-regulation of Bcr-Abl and repression of proteins involved in Bcr-Abl transformation, namely the antiapoptotic proteins Bcl-x(L) and Mcl-1. Importantly, PBOX-6/STI-571 combinations were also effective in STI-571-resistant cells. Together, these findings highlight the potential clinical benefits in simultaneously targeting the microtubules and the Bcr-Abl oncoprotein in STI-571-sensitive and -resistant CML cells.
Collapse
Affiliation(s)
- Lisa M Greene
- School of Biochemistry and Immunology, Trinity College, Dublin 2, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
171
|
Kawasaki ES, Player A. Nanotechnology, nanomedicine, and the development of new, effective therapies for cancer. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2007; 1:101-9. [PMID: 17292064 DOI: 10.1016/j.nano.2005.03.002] [Citation(s) in RCA: 185] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2005] [Accepted: 03/25/2005] [Indexed: 12/20/2022]
Abstract
Cancer is the leading cause of death in the United States among people younger than 85 years, and for the first time has surpassed heart disease as the number one killer. This worrisome statistic has resulted not from an increase in the incidence of cancer, but because deaths from heart disease have dropped nearly in half while the number of cancer-related deaths has remained about the same. This fact accentuates the need for a new generation of more effective therapies for cancer. In this review, the development of new therapies will be discussed in the context of advances in nanotechnologies related to cancer detection, analysis, diagnosis, and therapeutic intervention. First, several nanoanalytical methods, such as the use of quantum dots in detection and imaging of cancer, will be described. These techniques will be essential to the process of precisely describing cancer at the level of the cell and whole organism. Second, examples of how nanotechnologies can be used in the development of new therapies will be given, including methods that might allow for more efficient and accurate drug delivery and rationally designed, targeted drugs. Finally, a new initiative--the National Cancer Institute Alliance for Nanotechnology in Cancer--will be described and discussed with respect to the scientific issues, policies, and funding.
Collapse
|
172
|
Bruna A, Darken RS, Rojo F, Ocaña A, Peñuelas S, Arias A, Paris R, Tortosa A, Mora J, Baselga J, Seoane J. High TGFbeta-Smad activity confers poor prognosis in glioma patients and promotes cell proliferation depending on the methylation of the PDGF-B gene. Cancer Cell 2007; 11:147-60. [PMID: 17292826 DOI: 10.1016/j.ccr.2006.11.023] [Citation(s) in RCA: 397] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2006] [Revised: 10/11/2006] [Accepted: 11/16/2006] [Indexed: 11/16/2022]
Abstract
TGFbeta acts as a tumor suppressor in normal epithelial cells and early-stage tumors and becomes an oncogenic factor in advanced tumors. The molecular mechanisms involved in the malignant function of TGFbeta are not fully elucidated. We demonstrate that high TGFbeta-Smad activity is present in aggressive, highly proliferative gliomas and confers poor prognosis in patients with glioma. We discern the mechanisms and molecular determinants of the TGFbeta oncogenic response with a transcriptomic approach and by analyzing primary cultured patient-derived gliomas and human glioma biopsies. The TGFbeta-Smad pathway promotes proliferation through the induction of PDGF-B in gliomas with an unmethylated PDGF-B gene. The epigenetic regulation of the PDGF-B gene dictates whether TGFbeta acts as an oncogenic factor inducing PDGF-B and proliferation in human glioma.
Collapse
Affiliation(s)
- Alejandra Bruna
- Medical Oncology Program, Vall d'Hebron University Hospital Research Institute, 08035 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
173
|
Mao JH, Wu D, Perez-Losada J, Jiang T, Li Q, Neve RM, Gray JW, Cai WW, Balmain A. Crosstalk between Aurora-A and p53: frequent deletion or downregulation of Aurora-A in tumors from p53 null mice. Cancer Cell 2007; 11:161-173. [PMID: 17292827 PMCID: PMC2730519 DOI: 10.1016/j.ccr.2006.11.025] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2006] [Revised: 09/22/2006] [Accepted: 11/28/2006] [Indexed: 10/23/2022]
Abstract
The Aurora-A kinase gene is amplified in a subset of human tumors and in radiation-induced lymphomas from p53 heterozygous mice. Normal tissues from p53-/- mice have increased Aurora-A protein levels, but lymphomas from these mice exhibit heterozygous deletions of Aurora-A and/or reduced protein expression. A similar correlation between low p53 levels and Aurora-A gene deletions and expression is found in human breast cancer cell lines. In vitro studies using mouse embryo fibroblasts demonstrate that inhibition of Aurora-A can have either positive or negative effects on cell growth as a function of p53 status. These data have implications for the design of approaches to targeted cancer therapy involving the crosstalk between Aurora-A kinase and p53 pathways.
Collapse
Affiliation(s)
- Jian-Hua Mao
- Cancer Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Di Wu
- Cancer Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jesus Perez-Losada
- Cancer Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tao Jiang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Qian Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Richard M. Neve
- Department of Laboratory Medicine and Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94270, USA
| | - Joe W. Gray
- Department of Laboratory Medicine and Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94143, USA
- Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94270, USA
| | - Wei-Wen Cai
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Allan Balmain
- Cancer Research Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
174
|
Coluccia AML, Gunby RH, Tartari CJ, Scapozza L, Gambacorti-Passerini C, Passoni L. Anaplastic lymphoma kinase and its signalling molecules as novel targets in lymphoma therapy. Expert Opin Ther Targets 2007; 9:515-32. [PMID: 15948671 DOI: 10.1517/14728222.9.3.515] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
A crucial issue in the development of molecularly-targeted anticancer therapies is the identification of appropriate molecules whose targeting would result in tumour regression with a minimal level of systemic toxicity. Anaplastic lymphoma kinase (ALK) is a transmembrane receptor tyrosine kinase, normally expressed at low levels in the nervous system. As a consequence of chromosomal translocations involving the alk gene (2p23), ALK is also aberrantly expressed and constitutively activated in approximately 60% of CD30+ anaplastic large cell lymphomas (ALCLs). Due to the selective overexpression of ALK in tumour cells, its direct involvement in the process of malignant transformation and its frequent expression in ALCL patients, the authors recognise ALK as a suitable candidate for the development of molecularly targeted strategies for the therapeutic treatment of ALK-positive lymphomas. Strategies targeting ALK directly or indirectly via the inhibition of the protein networks responsible for ALK oncogenic signalling are discussed.
Collapse
Affiliation(s)
- A M L Coluccia
- Oncogenic Fusion Genes and Proteins Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy
| | | | | | | | | | | |
Collapse
|
175
|
George S, Rini BI. Therapeutic considerations in patients with metastatic renal cell carcinoma previously treated with antiangiogenic therapies. Clin Genitourin Cancer 2007; 5 Suppl 1:S40-4. [PMID: 17239283 DOI: 10.3816/cgc.2006.s.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The recent application of antiangiogenic therapies has opened a new corridor of treatment in metastatic renal cell carcinoma. These therapies influence intracellular pathways that are important in angiogenesis and tumor progression. Although each of these agents produces objective responses as well as progression-free survival benefits, durable complete responses have not generally been observed, and thus, resistance to these chronic therapies is almost universal. The increasing use and eventual resistance to these agents has defined an increasing population of patients with targeted therapy-refractory disease. However, there are limited clinical and preclinical data to guide treatment decisions for these patients. This review will examine the existing data in this setting, describe ongoing clinical investigations, and discuss challenges unique to this patient population.
Collapse
Affiliation(s)
- Saby George
- Cleveland Clinic Taussig Cancer Center, Cleveland, OH 44195, USA
| | | |
Collapse
|
176
|
Imyanitov EN, Moiseyenko VM. Molecular-based choice of cancer therapy: realities and expectations. Clin Chim Acta 2007; 379:1-13. [PMID: 17306783 DOI: 10.1016/j.cca.2007.01.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2006] [Revised: 01/03/2007] [Accepted: 01/06/2007] [Indexed: 01/29/2023]
Abstract
Current choice of cancer therapy is usually empirical and relies mainly on the statistical prediction of the treatment success. Molecular research provides some opportunities to personalize antitumor treatment. For example, life-threatening toxic reactions can be avoided by the identification of subjects, who carry susceptible genotypes of drug-metabolizing genes (e.g. TPMT, UGT1A1, MTHFR, DPYD). Tumor sensitivity can be predicted by molecular portraying of targets and other molecules associated with drug response. Tailoring of antiestrogen and trastuzumab therapy based on hormone and HER2 receptor status has already become a classical example of customized medicine. Other predictive markers have been identified both for cytotoxic and for targeted therapies, and include, for example, expression of TS, TP, DPD, OPRT, ERCC1, MGMT, TOP2A, class III beta-tubulin molecules as well as genomic alterations of EGFR, KIT, ABL oncogenes.
Collapse
Affiliation(s)
- Evgeny N Imyanitov
- Laboratory of Molecular Oncology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia.
| | | |
Collapse
|
177
|
Buffa P, Manzella L, Consoli ML, Messina A, Vigneri P. Modelling of the ABL and ARG proteins predicts two functionally critical regions that are natively unfolded. Proteins 2007; 67:1-11. [PMID: 17211892 DOI: 10.1002/prot.21161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The ABL and ARG tyrosine kinases regulate many pivotal cellular processes and are implicated in the pathogenesis of several forms of leukemia. We have modelled the previously uncharacterized core domain (SH3-SH2-tyrosine kinase) and C-terminal actin-binding domain of ARG. We have also investigated the structural arrangement of the ABL and ARG Cap region and of the long multifunctional region located downstream of the tyrosine kinase domain. We report that the ARG core domain is homologous to the corresponding ABL region, therefore suggesting that ARG catalytic activity is likely regulated by the same SH3-SH2 clamp described for ABL. We also report that the Cap of both ABL and ARG is natively unfolded. Hence, biological events determining the folding of the Cap are critical to allow its interaction with the tyrosine kinase C-lobe. Furthermore, our results show that, with the exception of the C-terminal actin-binding domain, the entire region encoded by the ABL and ARG last exon is natively unfolded. Phosphorylation events or protein-protein interactions regulating the folding of this region will therefore modulate the activity of its numerous functional domains. Finally, our analyses show that the C-terminal actin-binding domain of ARG displays a four-helix bundle structure similar to the one reported for the corresponding ABL region. Our findings imply that many biological activities attributed to ABL, ARG, and their oncogenic counterparts are regulated by natively unfolded regions.
Collapse
Affiliation(s)
- Pietro Buffa
- Department of Biomedical Sciences, Section of General Pathology, University of Catania, Catania, Italy
| | | | | | | | | |
Collapse
|
178
|
Verkhivker GM. In silico profiling of tyrosine kinases binding specificity and drug resistance using Monte Carlo simulations with the ensembles of protein kinase crystal structures. Biopolymers 2007; 85:333-48. [PMID: 17167796 DOI: 10.1002/bip.20656] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The molecular basis of the tyrosine kinases binding specificity and drug resistance against cancer drugs Imatinib and Dasatinib is elucidated using Monte Carlo simulations of the inhibitor-receptor binding with the ensembles of protein kinase crystal structures. In silico proteomics analysis unravels mechanisms by which structural plasticity of the tyrosine kinases is linked with the conformational preferences of Imatinib and Dasatinib in achieving effective drug binding with a distinct spectrum of the tyrosine kinome. The differences in the inhibitor sensitivities to the ABL kinase mutants are rationalized based on variations in the binding free energy profiles with the conformational states of the ABL kinase. While Imatinib binding is highly sensitive to the activation state of the enzyme, the computed binding profile of Dasatinib is remarkably tolerant to the conformational state of ABL. A comparative analysis of the inhibitor binding profiles with the clinically important ABL kinase mutants has revealed an excellent agreement with the biochemical and proteomics data. We have found that conformational adaptability of the kinase inhibitors to structurally different conformational states of the tyrosine kinases may have pharmacological relevance in acquiring a specific array of potent activities and regulating a scope of the inhibitor resistance mutations. This study outlines a useful approach for understanding and predicting the molecular basis of the inhibitor sensitivity against potential kinase targets and drug resistance.
Collapse
Affiliation(s)
- Gennady M Verkhivker
- Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, LA Jolla, CA 92093-0392, USA.
| |
Collapse
|
179
|
Abstract
The advent of the Bcr-Abl selective tyrosine kinase inhibitor imatinib mesylate (Glivec, Gleevec, Novartis, East Hanover, NJ) has substantially changed the treatment landscape for chronic myelogenous leukemia (CML). However, some patients, primarily those with advanced disease, are either initially refractory to imatinib or eventually develop imatinib resistance. Imatinib resistance or intolerance frequently depends on the re-emergence of Bcr-Abl kinase activity, but can also indicate Bcr-Abl-independent disease progression. Results from phase II/III trials suggest rates of resistance and relapse correlate with stage of disease and with the monitoring parameters: hematologic, cytogenetic, and molecular responses. To date, more than 40 different point mutations that code for distinct single amino acid substitutions in the Bcr-Abl kinase domain have been isolated from imatinib-resistant patients. These mutations affect amino acids involved in imatinib binding or in regulatory regions of the Bcr-Abl kinase domain, resulting in decreased sensitivity to imatinib while retaining aberrant kinase activity. Early mutation detection may aid in risk stratification and molecular-based treatment decisions. To overcome imatinib-resistant disease, novel tyrosine kinase inhibitors with activity against imatinib-resistant mutations and/or with inhibition of alternative pathways, such as Src activation, have recently been developed. Additional strategies include imatinib dose escalation, combination therapy, and treatment interruption to stop clonal selection of resistant cells.
Collapse
MESH Headings
- Benzamides
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/physiology
- Fusion Proteins, bcr-abl/drug effects
- Fusion Proteins, bcr-abl/genetics
- Humans
- Imatinib Mesylate
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology
- Mutation/drug effects
- Piperazines/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Pyrimidines/pharmacology
- Randomized Controlled Trials as Topic
Collapse
Affiliation(s)
- Andreas Hochhaus
- III. Medizinische Klinik, Medizinische Fakultät Mannheim, University of Heidelberg, Mannheim, Germany.
| | | | | | | |
Collapse
|
180
|
Kneller JM, Ehlen T, Matisic JP, Miller D, Van Niekerk D, Lam WL, Marra M, Richards-Kortum R, Follen M, MacAulay C, Jones SJ. Using LongSAGE to Detect Biomarkers of Cervical Cancer Potentially Amenable to Optical Contrast Agent Labelling. Biomark Insights 2007. [DOI: 10.1177/117727190700200020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Sixteen longSAGE libraries from four different clinical stages of cervical intraepithelial neoplasia have enabled us to identify novel cell-surface biomarkers indicative of CIN stage. By comparing gene expression profiles of cervical tissue at early and advanced stages of CIN, several genes are identified to be novel genetic markers. We present fifty-six cell-surface gene products differentially expressed during progression of CIN. These cell surface proteins are being examined to establish their capacity for optical contrast agent binding. Contrast agent visualization will allow real-time assessment of the physiological state of the disease process bringing vast benefit to cancer care. The data discussed in this publication have been submitted to NCBIs Gene Expression Omnibus (GEO, http://www.ncbi.nlm.nih.gov/geo/ ) and are accessible through GEO Series accession number GSE6252.
Collapse
Affiliation(s)
- Julie M. Kneller
- Genome Sciences Centre, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Thomas Ehlen
- Department of Gynaecologic Oncology, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Jasenka P. Matisic
- Cancer Imaging, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Dianne Miller
- Department of Gynaecologic Oncology, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Dirk Van Niekerk
- Cervical Cancer Screening Program, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Wan L. Lam
- Cancer Genetics and Developmental Biology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Marco Marra
- Genome Sciences Centre, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | | | - Michelle Follen
- University of Texas M.D. Anderson Cancer Center, Department of Gynecologic Oncology and Biomedical Engineering Center, Houston, TX, U.S.A
| | - Calum MacAulay
- Cancer Imaging, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Steven J.M. Jones
- Genome Sciences Centre, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| |
Collapse
|
181
|
Verkhivker GM. Computational proteomics of biomolecular interactions in the sequence and structure space of the tyrosine kinome: Deciphering the molecular basis of the kinase inhibitors selectivity. Proteins 2006; 66:912-29. [PMID: 17173284 DOI: 10.1002/prot.21287] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Understanding and predicting the molecular basis of protein kinases specificity against existing therapeutic agents remains highly challenging and deciphering this complexity presents an important problem in discovery and development of effective cancer drugs. We explore a recently introduced computational approach for in silico profiling of the tyrosine kinases binding specificity with a class of the pyrido-[2,3-d]pyrimidine kinase inhibitors. Computational proteomics analysis of the ligand-protein interactions using parallel simulated tempering with an ensemble of the tyrosine kinases crystal structures reveals an important molecular determinant of the kinase specificity. The pyrido-[2,3-d]pyrimidine inhibitors are capable of dynamically interacting with both active and inactive forms of the tyrosine kinases, accommodating structurally different kinase conformations with a similar binding affinity. Conformational tolerance of the protein tyrosine kinases binding with the pyrido[2,3-d]pyrimidine inhibitors provides the molecular basis for the broad spectrum of potent activities and agrees with the experimental inhibition profiles. The analysis of the pyrido[2,3-d]pyrimidine sensitivities against a number of clinically relevant ABL kinase mutants suggests an important role of conformational adaptability of multitargeted kinase inhibitors in developing drug resistance mechanisms. The presented computational approach may be useful in complementing proteomics technologies to characterize activity signatures of small molecules against a large number of potential kinase targets.
Collapse
Affiliation(s)
- Gennady M Verkhivker
- Department of Pharmacology, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093-0392, USA.
| |
Collapse
|
182
|
Olney HJ, Le Beau MM. Evaluation of recurring cytogenetic abnormalities in the treatment of myelodysplastic syndromes. Leuk Res 2006; 31:427-34. [PMID: 17161457 DOI: 10.1016/j.leukres.2006.10.023] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2006] [Revised: 10/16/2006] [Accepted: 10/18/2006] [Indexed: 11/28/2022]
Abstract
Myelodysplastic syndromes (MDS) are clinically heterogeneous, but the presence of specific cytogenetic abnormalities can predict disease manifestations, provide a basis for prognosis, and direct treatment. Conventional cytogenetic analysis is instrumental in identifying chromosomal abnormalities in MDS and novel genetic methods may provide supplementary information. Treatment with lenalidomide was recently shown to be effective in MDS, particularly in those cases with del(5q), resulting in durable cytogenetic remission and hematological responses. In this paradigm, diagnosis of the del(5q) abnormality would be essential to predicting response to therapy.
Collapse
Affiliation(s)
- Harold J Olney
- Université de Montréal, CHUM Hospital Notre-Dame, 1560 Sherbrooke St E, Montréal, Québec, H2L 4M1, Canada.
| | | |
Collapse
|
183
|
Fiskus W, Pranpat M, Balasis M, Bali P, Estrella V, Kumaraswamy S, Rao R, Rocha K, Herger B, Lee F, Richon V, Bhalla K. Cotreatment with vorinostat (suberoylanilide hydroxamic acid) enhances activity of dasatinib (BMS-354825) against imatinib mesylate-sensitive or imatinib mesylate-resistant chronic myelogenous leukemia cells. Clin Cancer Res 2006; 12:5869-78. [PMID: 17020995 DOI: 10.1158/1078-0432.ccr-06-0980] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We determined the effects of vorinostat [suberoylanilide hydroxamic acid (SAHA)] and/or dasatinib, a dual Abl/Src kinase (tyrosine kinase) inhibitor, on the cultured human (K562 and LAMA-84) or primary chronic myelogenous leukemia (CML) cells, as well as on the murine pro-B BaF3 cells with ectopic expression of the unmutated and kinase domain-mutant forms of Bcr-Abl. EXPERIMENTAL DESIGN Following exposure to dasatinib and/or vorinostat, apoptosis, loss of clonogenic survival, as well as the activity and levels of Bcr-Abl and its downstream signaling proteins were determined. RESULTS Treatment with dasatinib attenuated the levels of autophosphorylated Bcr-Abl, p-CrkL, phospho-signal transducer and activator of transcription 5 (p-STAT5), p-c-Src, and p-Lyn; inhibited the activity of Lyn and c-Src; and induced apoptosis of the cultured CML cells. Combined treatment of cultured human CML and BaF3 cells with vorinostat and dasatinib induced more apoptosis than either agent alone, as well as synergistically induced loss of clonogenic survival, which was associated with greater depletion of Bcr-Abl, p-CrkL, and p-STAT5 levels. Cotreatment with dasatinib and vorinostat also attenuated the levels of Bcr-AblE255K and Bcr-AblT315I and induced apoptosis of BaF3 cells with ectopic expression of the mutant forms of Bcr-Abl. Finally, cotreatment of the primary CML cells with vorinostat and dasatinib induced more loss of cell viability and depleted Bcr-Abl or Bcr-AblT315I, p-STAT5, and p-CrkL levels than either agent alone. CONCLUSIONS As shown here, the preclinical in vitro activity of vorinostat and dasatinib against cultured and primary CML cells supports the in vivo testing of the combination in imatinib mesylate-sensitive and imatinib mesylate-resistant CML cells.
Collapse
MESH Headings
- Animals
- Antineoplastic Combined Chemotherapy Protocols
- Apoptosis/drug effects
- Benzamides
- Dasatinib
- Drug Resistance, Neoplasm
- Drug Synergism
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/metabolism
- Histone Deacetylase Inhibitors
- Humans
- Hydroxamic Acids/pharmacology
- Imatinib Mesylate
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Mice
- Piperazines/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Pyrimidines/pharmacology
- STAT5 Transcription Factor/metabolism
- Thiazoles/pharmacology
- Tumor Cells, Cultured
- Vorinostat
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- Warren Fiskus
- Department of Interdisciplinary Oncology, H. Lee Moffitt Cancer Center, University of South Florida, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Hidalgo M, Amador ML, Jimeno A, Mezzadra H, Patel P, Chan A, Nielsen ME, Maitra A, Altiok S. Assessment of gefitinib- and CI-1040-mediated changes in epidermal growth factor receptor signaling in HuCCT-1 human cholangiocarcinoma by serial fine needle aspiration. Mol Cancer Ther 2006; 5:1895-903. [PMID: 16891476 DOI: 10.1158/1535-7163.mct-05-0525] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
One specific limitation to the clinical development of targeted cancer therapeutics is the lack of well-validated pharmacodynamic markers. Such tools might conceivably provide a framework within which to better evaluate the selection of specific molecules as therapeutic targets. Nevertheless, the practical application of this hypothesis in clinical development remains elusive. In this study, we present a minimally invasive pharmacodynamic assay for monitoring therapy-mediated changes in the activity of target signaling pathways by using fine needle aspiration (FNA) samples and quantitative ELISA methods. To this end, we used the HuCCT-1 cholangiocarcinoma cell line treated with gefitinib (ZD1839, Iressa), a selective blocker of the epidermal growth factor receptor (EGFR), and CI-1040, a selective inhibitor of the mitogen extracellular regulated kinase [mitogen-activated protein/extracellular signal-regulated kinase (ERK) kinase 1/2]. HuCCT-1 cells were resistant to gefitinib and CI-1040 alone but susceptible to the combination of these drugs in vitro and in vivo. This effect was associated with a greater inhibition of ERK1/2 activation, a downstream mediator in the EGFR-mitogen-activated protein/ERK kinase pathway. Using this model, we sought to assess whether FNA-obtained tumor biopsies could be used to measure signaling pathway activation. Cellular extracts prepared from FNA samples yielded adequately cellular, high-quality samples to assess therapy-mediated changes in EGFR and ERK1/2 phosphorylation by Western blotting and quantitative ELISA assays. Treatment with gefitinib alone effectively inhibited EGFR activation but failed to block ERK1/2 phosphorylation and tumor growth. Blocking was achieved by the addition of CI-1040 to the treatment regimen. These results show that the combination of serial FNA sampling with highly sensitive quantitative ELISA assays permits assessment of therapy-mediated changes in signaling pathways, which correlate well with antitumor effects. This assay is simple to implement and broadly applicable to diverse tumor types in clinical studies with cancer patients and may be useful in the development of targeted anticancer agents.
Collapse
Affiliation(s)
- Manuel Hidalgo
- Department of Pathology. Johns Hopkins School of Medicine, 600 North Wolfe Street, Baltimore, MD 21287, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Patyna S, Laird AD, Mendel DB, O'farrell AM, Liang C, Guan H, Vojkovsky T, Vasile S, Wang X, Chen J, Grazzini M, Yang CY, Haznedar JO, Sukbuntherng J, Zhong WZ, Cherrington JM, Hu-Lowe D. SU14813: a novel multiple receptor tyrosine kinase inhibitor with potent antiangiogenic and antitumor activity. Mol Cancer Ther 2006; 5:1774-82. [PMID: 16891463 DOI: 10.1158/1535-7163.mct-05-0333] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Receptor tyrosine kinases (RTK), such as vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR), stem cell factor receptor (KIT), and fms-like tyrosine kinase 3 (FLT3), are expressed in malignant tissues and act in concert, playing diverse and major roles in angiogenesis, tumor growth, and metastasis. With the exception of a few malignancies, seemingly driven by a single genetic mutation in a signaling protein, most tumors are the product of multiple mutations in multiple aberrant signaling pathways. Consequently, simultaneous targeted inhibition of multiple signaling pathways could be more effective than inhibiting a single pathway in cancer therapies. Such a multitargeted strategy has recently been validated in a number of preclinical and clinical studies using RTK inhibitors with broad target selectivity. SU14813, a small molecule identified from the same chemical library used to isolate sunitinib, has broad-spectrum RTK inhibitory activity through binding to and inhibition of VEGFR, PDGFR, KIT, and FLT3. In cellular assays, SU14813 inhibited ligand-dependent and ligand-independent proliferation, migration, and survival of endothelial cells and/or tumor cells expressing these targets. SU14813 inhibited VEGFR-2, PDGFR-beta, and FLT3 phosphorylation in xenograft tumors in a dose- and time-dependent fashion. The plasma concentration required for in vivo target inhibition was estimated to be 100 to 200 ng/mL. Used as monotherapy, SU14813 exhibited broad and potent antitumor activity resulting in regression, growth arrest, or substantially reduced growth of various established xenografts derived from human or rat tumor cell lines. Treatment in combination with docetaxel significantly enhanced both the inhibition of primary tumor growth and the survival of the tumor-bearing mice compared with administration of either agent alone. In summary, SU14813 inhibited target RTK activity in vivo in association with reduction in angiogenesis, target RTK-mediated proliferation, and survival of tumor cells, leading to broad and potent antitumor efficacy. These data support the ongoing phase I clinical evaluation of SU14813 in advanced malignancies.
Collapse
Affiliation(s)
- Shem Patyna
- Pfizer Global Research and Development, 10777 Science Center Drive, San Diego, CA 92024, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
186
|
|
187
|
Abstract
The accumulation of multiple mutations and alterations in the cancer genome underlies the complexity of cancer phenotypes. A consequence of these alterations is the deregulation of various cell-signalling pathways that control cell function. Molecular-profiling studies, particularly DNA microarray analyses, have the potential to describe this complexity. These studies also provide an opportunity to link pathway deregulation with potential therapeutic strategies. This approach, when coupled with other methods for identifying pathway activation, provides an opportunity to both match individual patients with the most appropriate therapeutic strategy and identify potential options for combination therapy.
Collapse
Affiliation(s)
- Andrea H Bild
- Duke Institute for Genome Sciences and Policy, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | |
Collapse
|
188
|
Abstract
The 'magic bullet' concept of specifically targeting cancer cells at the same time as sparing normal tissues is now proven, as several monoclonal antibodies and targeted small-molecule compounds have been approved for cancer treatment. Both antibodies and small-molecule compounds are therefore promising tools for target-protein-based cancer therapy. We discuss and compare the distinctive properties of these two therapeutic strategies so as to provide a better view for the development of new drugs and the future direction of cancer therapy.
Collapse
Affiliation(s)
- Kohzoh Imai
- Sapporo Medical University, South 1, West 17, Chuo-ku, Sapporo, 060-8556, Japan.
| | | |
Collapse
|
189
|
Hochhaus A. Chronic myelogenous leukemia (CML): resistance to tyrosine kinase inhibitors. Ann Oncol 2006; 17 Suppl 10:x274-9. [PMID: 17018738 DOI: 10.1093/annonc/mdl273] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- A Hochhaus
- III Medizinische Klinik, Medizinische Fakultät Mannheim der Universität Heidelberg, Mannheim, Germany
| |
Collapse
|
190
|
Céspedes MV, Casanova I, Parreño M, Mangues R. Mouse models in oncogenesis and cancer therapy. Clin Transl Oncol 2006; 8:318-29. [PMID: 16760006 DOI: 10.1007/s12094-006-0177-7] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Animal models have been critical in the study of the molecular mechanisms of cancer and in the development of new antitumor agents; nevertheless, there is still much room for improvement. The relevance of each particular model depends on how close it replicates the histology, physiological effects, biochemical pathways and metastatic pattern observed in the same human tumor type. Metastases are especially important because they are the main determinants of the clinical course of the disease and patient survival, and are the target of systemic therapy. The generation of clinically relevant models using the mouse requires their humanization, since differences exist in transformation and oncogenesis between human and mouse. Although genetically modified (GM) mice have been instrumental in understanding the molecular mechanisms involved in tumor initiation, they have been less successful in replicating advanced cancer. Moreover, a particular genetic alteration frequently leads to different tumor types in human and mouse and to lower metastastatic rates in GM mice than in humans. These findings question the capacity of current GM mouse carcinoma models to predict clinical response to therapy. On the other hand, orthotopic (ORT) xenografts of human tumors, or tumor cell lines, in nude mice reproduce the histology and metastatic pattern of most human tumors at advanced stage. Using ex vivo genetic manipulation of human tumor cells, ORT models can be used to molecularly dissect the metastatic process and to evaluate in vivo tumor response to therapy, using non-invasive procedures. Nevertheless, this approach is not useful in the study of the initial stages of tumorigenesis or the contribution of the immune system in this process. Despite ORT models are more promising than the most commonly used subcutaneous xenografts in preclinical drug development, their capacity to predict clinical response to antitumor agents remains to be studied. Humanizing mouse models of cancer will most likely require the combined use of currently available methodologies.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/therapeutic use
- Cell Line, Tumor/drug effects
- Cell Line, Tumor/transplantation
- Cell Transformation, Neoplastic
- Humans
- Immunocompromised Host
- Mice
- Mice, Mutant Strains
- Mice, Nude
- Mice, SCID
- Models, Animal
- Neoplasm Metastasis
- Neoplasm Transplantation
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/pathology
- Species Specificity
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- M V Céspedes
- Grup d'Oncogenesi i Antitumorals, Institut de Recerca, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | | | | |
Collapse
|
191
|
Abstract
It is an exciting time in the treatment of systemic mastocytosis. Major advances in the past 2 decades have helped to define the molecular abnormalities associated with this disease and to delineate pathways involved in its pathogenesis. This has directly translated into the development of novel targeted therapies. These therapies hold great promise to patients and health care providers that a "cure" for systemic mastocytosis may someday be obtainable.
Collapse
Affiliation(s)
- Todd M Wilson
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 10, Room 11C205, 10 Center Drive, MSC 1881, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
192
|
Talano JAM, Margolis DA. Recent molecular and cellular advances in pediatric bone marrow transplantation. Pediatr Clin North Am 2006; 53:685-98. [PMID: 16872999 DOI: 10.1016/j.pcl.2006.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The field of allogeneic transplantation has made vast improvements since its inception in 1968. Improvements in supportive care have greatly improved survival. Delayed immune reconstitution, graft versus host disease, and relapse of disease still pose great obstacles. This article has highlighted novel strategies for using cellular therapy in conjunction with hematopoietic cell transplantation (HCT) that potentially may lead to improved clinical outcomes for patients undergoing HCT in the future.
Collapse
Affiliation(s)
- Julie-An M Talano
- Division of Pediatric Hematology and Oncology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| | | |
Collapse
|
193
|
Maehara Y, Egashira A, Kakeji Y. Current topics and the clinical effects of target-based antineoplastic agents. Int J Clin Oncol 2006; 11:164-6. [PMID: 16850121 DOI: 10.1007/s10147-006-0581-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2006] [Indexed: 10/24/2022]
Affiliation(s)
- Yoshihiko Maehara
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| | | | | |
Collapse
|
194
|
Green J, Ikram M, Vyas J, Patel N, Proby CM, Ghali L, Leigh IM, O'toole EA, Storey A. Overexpression of the Axl tyrosine kinase receptor in cutaneous SCC-derived cell lines and tumours. Br J Cancer 2006; 94:1446-51. [PMID: 16641895 PMCID: PMC2361292 DOI: 10.1038/sj.bjc.6603135] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The molecular mechanisms that underlie the development of squamous cell skin cancers (SSC) are poorly understood. We have used oligonucleotide microarrays to compare the differences in cellular gene expression between a series of keratinocyte cell that mimic disease progression with the aim of identifying genes that may potentially contribute towards squamous cell carcinoma (SCC) progression in vivo, and in particular to identify markers that may serve as potential therapeutic targets for SCC treatment. Gene expression differences were corroborated by polymerase chain reaction and Western blotting. We identified Axl, a receptor tyrosine kinase with transforming potential that has also been shown to have a role in cell survival, adhesion and chemotaxis, was upregulated in vitro in SCC-derived cells compared to premalignant cells. Extending the investigation to tumour biopsies showed that the Axl protein was overexpressed in vivo in a series of SCCs.
Collapse
Affiliation(s)
- J Green
- Cancer Research UK, Skin Tumour Laboratory, London E1 2AT, UK
- Centre for Cutaneous Research, Institute for Cell and Molecular Science, 4 Newark Street, London E1 2AT, UK
| | - M Ikram
- Cancer Research UK, Skin Tumour Laboratory, London E1 2AT, UK
- Centre for Cutaneous Research, Institute for Cell and Molecular Science, 4 Newark Street, London E1 2AT, UK
| | - J Vyas
- Centre for Cutaneous Research, Institute for Cell and Molecular Science, 4 Newark Street, London E1 2AT, UK
| | - N Patel
- Centre for Cutaneous Research, Institute for Cell and Molecular Science, 4 Newark Street, London E1 2AT, UK
| | - C M Proby
- Cancer Research UK, Skin Tumour Laboratory, London E1 2AT, UK
- Centre for Cutaneous Research, Institute for Cell and Molecular Science, 4 Newark Street, London E1 2AT, UK
| | - L Ghali
- Cancer Research UK, Skin Tumour Laboratory, London E1 2AT, UK
- Centre for Cutaneous Research, Institute for Cell and Molecular Science, 4 Newark Street, London E1 2AT, UK
| | - I M Leigh
- Cancer Research UK, Skin Tumour Laboratory, London E1 2AT, UK
- Centre for Cutaneous Research, Institute for Cell and Molecular Science, 4 Newark Street, London E1 2AT, UK
| | - E A O'toole
- Centre for Cutaneous Research, Institute for Cell and Molecular Science, 4 Newark Street, London E1 2AT, UK
| | - A Storey
- Cancer Research UK, Skin Tumour Laboratory, London E1 2AT, UK
- Centre for Cutaneous Research, Institute for Cell and Molecular Science, 4 Newark Street, London E1 2AT, UK
- Cancer Research UK, Skin Tumour Laboratory, London E1 2AT, UK. E-mail:
| |
Collapse
|
195
|
Lacal JC. Changing the course of oncogenesis: The development of tyrosine kinase inhibitors. EUROPEAN JOURNAL OF CANCER SUPPLEMENTS 2006. [DOI: 10.1016/j.ejcsup.2006.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
196
|
|
197
|
Klumpp M, Boettcher A, Becker D, Meder G, Blank J, Leder L, Forstner M, Ottl J, Mayr LM. Readout Technologies for Highly Miniaturized Kinase Assays Applicable to High-Throughput Screening in a 1536-Well Format. ACTA ACUST UNITED AC 2006; 11:617-33. [PMID: 16760365 DOI: 10.1177/1087057106288444] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This article discusses the development of homogeneous, miniaturized assays for the identification of novel kinase inhibitors from very large compound collections. In particular, the suitability of time-resolved fluorescence resonance energy transfer (TR-RET) based on phospho-specific antibodies, an antibody-independent fluorescence polarization (FP) approach using metal-coated beads (IMAP™ technology), and the determination of adenosine triphosphate consumption through chemiluminescence is evaluated. These readouts are compared with regard to assay sensitivity, compound interference, reagent consumption, and performance in a 1536-well format, and practical considerations for their application in primary screening or in the identification of kinase substrates are discussed. All of the tested technologies were found to be suitable for miniaturized high-throughput screening (HTS) in principle, but each of them has distinct limitations and advantages. Therefore, the target-specific selection of the most appropriate readout technology is recommended to ensure maximal relevance of HTS campaigns.
Collapse
Affiliation(s)
- Martin Klumpp
- Novartis Institute for Biomedical Research, Basel, Switzerland.
| | | | | | | | | | | | | | | | | |
Collapse
|
198
|
Tashiro H, Shirasaki R, Noguchi M, Gotoh M, Kawasugi K, Shirafuji N. Molecular Analysis of Chronic Eosinophilic Leukemia with t(4;10) Showing Good Response to Imatinib Mesylate. Int J Hematol 2006; 83:433-8. [PMID: 16787876 DOI: 10.1532/ijh97.05180] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
A 38-year-old Japanese man was referred to our hospital in June 2003 for treatment of acute respiratory failure with severe eosinophilia. Idiopathic hypereosinophilic syndrome had been diagnosed in 1994. However, karyotypic examination of bone marrow cells revealed that chromosomal translocation with t(4;10)(q12;p11) had occurred in 2000, and chronic eosinophilic leukemia was diagnosed. At admission, the patient's respiratory condition was extremely serious, and mechanical support was necessary. Despite treatment with steroid pulse therapy and cytarabine, the blood eosinophil count did not decrease, and the patient's respiratory condition worsened. After obtaining informed consent, we administered imatinib mesylate at a dose of 200 mg/day for 2 days and 100 mg/day for 3 days. The blood eosinophil count decreased dramatically over 5 days, and the patient's condition rapidly improved, such that the patient could be discharged. In this case, we performed molecular analysis using peripheral blood. The FIP1-like 1 (FIP1L1)-platelet-derived growth factor receptor alpha (PDGFRalpha) fusion transcript was found with the reverse transcriptase polymerase chain reaction analysis. In this case, eosinophilia was possibly caused by constitutive activation of tyrosine kinase produced by the FIP1L1-PDGFRalpha fusion transcript.
Collapse
MESH Headings
- Adult
- Asian People
- Benzamides
- Chromosomes, Human, Pair 10/genetics
- Chromosomes, Human, Pair 4/genetics
- Chronic Disease
- Enzyme Activation/drug effects
- Eosinophilia/drug therapy
- Eosinophilia/enzymology
- Eosinophilia/genetics
- Follow-Up Studies
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Leukemic/drug effects
- Humans
- Hypereosinophilic Syndrome/drug therapy
- Hypereosinophilic Syndrome/enzymology
- Hypereosinophilic Syndrome/genetics
- Imatinib Mesylate
- Leukocyte Count
- Male
- Oncogene Proteins, Fusion/antagonists & inhibitors
- Oncogene Proteins, Fusion/biosynthesis
- Piperazines/administration & dosage
- Protein Kinase Inhibitors/administration & dosage
- Pyrimidines/administration & dosage
- RNA, Messenger/biosynthesis
- RNA, Messenger/genetics
- RNA, Neoplasm/biosynthesis
- RNA, Neoplasm/genetics
- Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors
- Receptor, Platelet-Derived Growth Factor alpha/biosynthesis
- Reverse Transcriptase Polymerase Chain Reaction/methods
- Time Factors
- Translocation, Genetic/genetics
- mRNA Cleavage and Polyadenylation Factors/antagonists & inhibitors
- mRNA Cleavage and Polyadenylation Factors/biosynthesis
Collapse
Affiliation(s)
- Haruko Tashiro
- Department of Hematology/Oncology, Teikyo University School of Medicine, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
199
|
Verkhivker GM. Imprint of evolutionary conservation and protein structure variation on the binding function of protein tyrosine kinases. Bioinformatics 2006; 22:1846-54. [PMID: 16720585 DOI: 10.1093/bioinformatics/btl199] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
MOTIVATION According to the models of divergent molecular evolution, the evolvability of new protein function may depend on the induction of new phenotypic traits by a small number of mutations of the binding site residues. Evolutionary relationships between protein kinases are often employed to infer inhibitor binding profiles from sequence analysis. However, protein kinases binding profiles may display inhibitor selectivity within a given kinase subfamily, while exhibiting cross-activity between kinases that are phylogenetically remote from the prime target. The emerging insights into kinase function and evolution combined with a rapidly growing number of publically available crystal structures of protein kinases complexes have motivated structural bioinformatics analysis of sequence-structure relationships in determining the binding function of protein tyrosine kinases. RESULTS In silico profiling of Imatinib mesylate and PD-173955 kinase inhibitors with protein tyrosine kinases is conducted on kinome scale by using evolutionary analysis and fingerprinting inhibitor-protein interactions with the panel of all publically available protein tyrosine kinases crystal structures. We have found that sequence plasticity of the binding site residues alone may not be sufficient to enable protein tyrosine kinases to readily evolve novel binding activities with inhibitors. While evolutionary signal derived solely from the tyrosine kinase sequence conservation can not be readily translated into the ligand binding phenotype, the proposed structural bioinformatics analysis can discriminate a functionally relevant kinase binding signal from a simple phylogenetic relationship. The results of this work reveal that protein conformational diversity is intimately linked with sequence plasticity of the binding site residues in achieving functional adaptability of protein kinases towards specific drug binding. This study offers a plausible molecular rationale to the experimental binding profiles of the studied kinase inhibitors and provides a theoretical basis for constructing functionally relevant kinase binding trees.
Collapse
Affiliation(s)
- Gennady M Verkhivker
- Department of Pharmacology, University of California San Diego 9500 Gilman Drive, La Jolla, CA 92093-0392, USA.
| |
Collapse
|
200
|
Abstract
Prostate cancer is the leading cause of nonskin malignancy and the second leading cause of cancer death in men. Androgen deprivation therapy is the first-line of systemic therapy against advanced prostate cancer. All advanced prostate cancers eventually grow despite castrate levels of testosterone. We review the evidence that androgen independent prostate cancer continues to require androgen receptor activity for growth, the mechanisms of androgen receptor activation in the castrate setting, and possible points of intervention for novel therapies targeting the androgen receptor and prostate cancer.
Collapse
Affiliation(s)
- Nima Sharifi
- Cytokine Molecular Mechanisms Section, Laboratory of Molecular Immunoregulation, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA.
| | | |
Collapse
|