Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters










Publication year range
1.
Cell Death Differ ; 22(1): 58-73, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25236395

ABSTRACT

Cells exposed to extreme physicochemical or mechanical stimuli die in an uncontrollable manner, as a result of their immediate structural breakdown. Such an unavoidable variant of cellular demise is generally referred to as 'accidental cell death' (ACD). In most settings, however, cell death is initiated by a genetically encoded apparatus, correlating with the fact that its course can be altered by pharmacologic or genetic interventions. 'Regulated cell death' (RCD) can occur as part of physiologic programs or can be activated once adaptive responses to perturbations of the extracellular or intracellular microenvironment fail. The biochemical phenomena that accompany RCD may be harnessed to classify it into a few subtypes, which often (but not always) exhibit stereotyped morphologic features. Nonetheless, efficiently inhibiting the processes that are commonly thought to cause RCD, such as the activation of executioner caspases in the course of apoptosis, does not exert true cytoprotective effects in the mammalian system, but simply alters the kinetics of cellular demise as it shifts its morphologic and biochemical correlates. Conversely, bona fide cytoprotection can be achieved by inhibiting the transduction of lethal signals in the early phases of the process, when adaptive responses are still operational. Thus, the mechanisms that truly execute RCD may be less understood, less inhibitable and perhaps more homogeneous than previously thought. Here, the Nomenclature Committee on Cell Death formulates a set of recommendations to help scientists and researchers to discriminate between essential and accessory aspects of cell death.


Subject(s)
Apoptosis , Signal Transduction , Animals , Humans , Terminology as Topic
2.
Cell Death Dis ; 5: e1552, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25476900

ABSTRACT

In Slavic folklore, Koschei the Immortal was bony, thin and lean. Was his condition caused by severe calorie restriction (CR)? CR deactivates the target of rapamycin pathway and slows down aging. But the life-extending effect of severe CR is limited by starvation. What if Koschei's anti-aging formula included rapamycin? And was rapamycin (or another rapalog) combined with commonly available drugs such as metformin, aspirin, propranolol, angiotensin II receptor blockers and angiotensin-converting enzyme inhibitors.


Subject(s)
Aging/drug effects , Caloric Restriction , Longevity/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Aging/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Aspirin/pharmacology , Exercise , Folklore , Gene Expression , Glucose/metabolism , Humans , Insulin Resistance , Longevity/physiology , Metformin/pharmacology , Propranolol/pharmacology , Russia , TOR Serine-Threonine Kinases/metabolism
3.
Cell Death Dis ; 5: e1214, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24810050

ABSTRACT

Mammalian target of rapamycin (mTOR) is involved in insulin resistance (IR) and diabetic retinopathy. In retinal pigment epithelial (RPE) cells, insulin activates the mTOR pathway, inducing hypoxia-inducible factor-1α (HIF-1α) and HIF-dependent transcription in serum-free minimum essential medium Eagle (MEM). Serendipitously, we found that insulin failed to induce the HIF-1α-dependent response, when RPE cells were cultured in Dulbecco's modification of Eagle's medium (DMEM). Whereas concentration of glucose in MEM corresponds to normal glucose levels in blood (5.5 mM), its concentration in DMEM corresponds to severe diabetic hyperglycemia (25 mM). Addition of glucose to MEM also caused IR. Glucose-mediated IR was characterized by basal activation of mTORC1 and its poor inducibility by insulin. Basal levels of phosphorylated S6 kinase (S6K), S6 and insulin receptor substrate 1 (IRS1) S635/639 were high, whereas their inducibilities were decreased. Insulin-induced Akt phosphorylation was decreased and restored by rapamycin and an inhibitor of S6K. IR was associated with de-phosphorylation of IRS1 at S1011, which was reversed by rapamycin. Both short (16-40 h) and chronic (2 weeks) treatment with rapamycin reversed IR. Furthermore, rapamycin did not impair Akt activation in RPE cells cultured in normoglycemic media. In contrast, Torin 1 blocked Akt activation by insulin. We conclude that by activating mTOR/S6K glucose causes feedback IR, preventable by rapamycin. Rapamycin does not cause IR in RPE cells regardless of the duration of treatment. We confirmed that rapamycin also did not impair phosphorylation of Akt at T308 and S473 in normal myoblast C2C12 cells. Our work provides insights in glucose-induced IR and suggests therapeutic approaches to treat patients with IR and severe hyperglycemia and to prevent diabetic complications such as retinopathy. Also our results prompt to reconsider physiological relevance of numerous data and paradigms on IR given that most cell lines are cultured with grossly super-physiological levels of glucose.


Subject(s)
Glucose/metabolism , Hyperglycemia/metabolism , Hypoglycemic Agents/pharmacology , Insulin Resistance , Myoblasts, Skeletal/drug effects , Protein Kinase Inhibitors/pharmacology , Retinal Pigment Epithelium/drug effects , Sirolimus/pharmacology , Animals , Cell Line , Enzyme Activation , Insulin/metabolism , Insulin Receptor Substrate Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes/metabolism , Myoblasts, Skeletal/metabolism , Oligonucleotides/genetics , Oligonucleotides/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Retinal Pigment Epithelium/metabolism , Ribosomal Protein S6 Kinases/antagonists & inhibitors , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Time Factors , Transfection
4.
Cell Death Dis ; 4: e964, 2013 Dec 12.
Article in English | MEDLINE | ID: mdl-24336084

ABSTRACT

This article is addressed to endocrinologists treating patients with diabetic complications as well as to basic scientists studying an elusive link between diseases and aging. It answers some challenging questions. What is the link between insulin resistance (IR), cellular aging and diseases? Why complications such as retinopathy may paradoxically precede the onset of type II diabetes. Why intensive insulin therapy may initially worsen retinopathy. How nutrient- and insulin-sensing mammalian target of rapamycin (mTOR) pathway can drive insulin resistance and diabetic complications. And how rapamycin, at rational doses and schedules, may prevent IR, retinopathy, nephropathy and beta-cell failure, without causing side effects.


Subject(s)
Diabetes Complications/metabolism , Insulin Resistance/physiology , Aging/genetics , Aging/metabolism , Animals , Diabetes Complications/genetics , Endocrinology/methods , Geriatrics/methods , Humans , Insulin Resistance/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
5.
Cell Death Differ ; 20(9): 1241-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23852369

ABSTRACT

When the cell cycle becomes arrested, MTOR (mechanistic Target of Rapamycin) converts reversible arrest into senescence (geroconversion). Hyperexpression of cyclin D1 is a universal marker of senescence along with hypertrophy, beta-Gal staining and loss of replicative/regenerative potential (RP), namely, the ability to restart proliferation when the cell cycle is released. Inhibition of MTOR decelerates geroconversion, although only partially decreases cyclin D1. Here we show that in p21- and p16-induced senescence, inhibitors of mitogen-activated/extracellular signal-regulated kinase (MEK) (U0126, PD184352 and siRNA) completely prevented cyclin D1 accumulation, making it undetectable. We also used MEL10 cells in which MEK inhibitors do not inhibit MTOR. In such cells, U0126 by itself induced senescence that was remarkably cyclin D1 negative. In contrast, inhibition of cyclin-dependent kinase (CDK) 4/6 by PD0332991 caused cyclin D1-positive senescence in MEL10 cells. Both types of senescence were suppressed by rapamycin, converting it into reversible arrest. We confirmed that the inhibitor of CDK4/6 caused cyclin D1 positive senescence in normal RPE cells, whereas U0126 prevented cyclin D1 expression. Elimination of cyclin D1 by siRNA did not prevent other markers of senescence that are consistent with the lack of its effect on MTOR. Our data confirmed that a mere inhibition of the cell cycle was sufficient to cause senescence, providing MTOR was active, and inhibition of MEK partially inhibited MTOR in a cell-type-dependent manner. Second, hallmarks of senescence may be dissociated, and hyperelevated cyclin D1, a marker of hyperactivation of senescent cells, did not necessarily determine other markers of senescence. Third, inhibition of MEK was sufficient to eliminate cyclin D1, regardless of MTOR.


Subject(s)
Cell Cycle Checkpoints/drug effects , Cellular Senescence/drug effects , Cyclin D1/metabolism , MAP Kinase Kinase 1/metabolism , TOR Serine-Threonine Kinases/metabolism , Antibiotics, Antineoplastic/pharmacology , Benzamides/pharmacology , Butadienes/pharmacology , Cell Cycle Checkpoints/genetics , Cell Division/drug effects , Cell Line, Tumor , Cellular Senescence/genetics , Cyclin D1/antagonists & inhibitors , Cyclin D1/biosynthesis , Cyclin D1/genetics , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p16 , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Enzyme Inhibitors/pharmacology , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , Neoplasm Proteins/metabolism , Nitriles/pharmacology , Piperazines/pharmacology , Pyridines/pharmacology , RNA Interference , RNA, Small Interfering , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/drug effects , TOR Serine-Threonine Kinases/genetics
6.
Cell Death Dis ; 4: e472, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23348586

ABSTRACT

High doses of rapamycin, an antiaging agent, can prevent obesity in mice on high fat diet (HFD). Obesity is usually associated with hyperinsulinemia. Here, we showed that rapamycin given orally, at doses that did not affect weight gain in male mice on HFD, tended to decrease fasting insulin levels. Addition of resveratrol, which alone did not affect insulin levels, potentiated the effect of rapamycin, so that the combination decreased obesity and prevented hyperinsulinemia. Neither rapamycin nor resveratrol, and their combination affected fasting levels of glucose (despite lowering insulin levels), implying that the combination might prevent insulin resistance. We and others previously reported that resveratrol at high doses inhibited the mTOR (Target of Rapamycin) pathway in cell culture. Yet, as we confirmed here, this effect was observed only at super-pharmacological concentrations. At pharmacological concentrations, resveratrol did not exert 'rapamycin-like effects' on cellular senescence and did not inhibit the mTOR pathway in vitro, indicating nonoverlapping therapeutic mechanisms of actions of rapamycin and resveratrol in vivo. Although, like rapamycin, resveratrol decreased insulin-induced HIF-1-dependent transcription in cell culture, resveratrol did not inhibit mTOR at the same concentrations. Given distinct mechanisms of action of rapamycin and resveratrol at clinically relevant doses, their combination warrants further investigation as a potential antiaging, antiobesity and antidiabetic modality.


Subject(s)
Diet, High-Fat , Hyperinsulinism/prevention & control , Obesity/prevention & control , Sirolimus/therapeutic use , Stilbenes/therapeutic use , Animals , Cell Line, Tumor , Cellular Senescence , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin/blood , Insulin Resistance , Male , Mice , Obesity/pathology , Resveratrol , Sirolimus/pharmacology , Stilbenes/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Transcription, Genetic , Weight Gain/drug effects
7.
Cell Death Differ ; 19(1): 107-20, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21760595

ABSTRACT

In 2009, the Nomenclature Committee on Cell Death (NCCD) proposed a set of recommendations for the definition of distinct cell death morphologies and for the appropriate use of cell death-related terminology, including 'apoptosis', 'necrosis' and 'mitotic catastrophe'. In view of the substantial progress in the biochemical and genetic exploration of cell death, time has come to switch from morphological to molecular definitions of cell death modalities. Here we propose a functional classification of cell death subroutines that applies to both in vitro and in vivo settings and includes extrinsic apoptosis, caspase-dependent or -independent intrinsic apoptosis, regulated necrosis, autophagic cell death and mitotic catastrophe. Moreover, we discuss the utility of expressions indicating additional cell death modalities. On the basis of the new, revised NCCD classification, cell death subroutines are defined by a series of precise, measurable biochemical features.


Subject(s)
Apoptosis , Autophagy , Cells/metabolism , Cells/pathology , Necrosis , Terminology as Topic , Animals , Caspases/metabolism , Humans , Mitosis
9.
Cell Death Differ ; 16(8): 1093-107, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19373242

ABSTRACT

Cell death is essential for a plethora of physiological processes, and its deregulation characterizes numerous human diseases. Thus, the in-depth investigation of cell death and its mechanisms constitutes a formidable challenge for fundamental and applied biomedical research, and has tremendous implications for the development of novel therapeutic strategies. It is, therefore, of utmost importance to standardize the experimental procedures that identify dying and dead cells in cell cultures and/or in tissues, from model organisms and/or humans, in healthy and/or pathological scenarios. Thus far, dozens of methods have been proposed to quantify cell death-related parameters. However, no guidelines exist regarding their use and interpretation, and nobody has thoroughly annotated the experimental settings for which each of these techniques is most appropriate. Here, we provide a nonexhaustive comparison of methods to detect cell death with apoptotic or nonapoptotic morphologies, their advantages and pitfalls. These guidelines are intended for investigators who study cell death, as well as for reviewers who need to constructively critique scientific reports that deal with cellular demise. Given the difficulties in determining the exact number of cells that have passed the point-of-no-return of the signaling cascades leading to cell death, we emphasize the importance of performing multiple, methodologically unrelated assays to quantify dying and dead cells.


Subject(s)
Cell Death , Apoptosis , Eukaryotic Cells/cytology , Flow Cytometry , Guidelines as Topic , Humans , Immunoblotting , Microscopy, Electron, Scanning , Microscopy, Fluorescence , Spectrometry, Fluorescence
10.
Cell Death Differ ; 16(1): 3-11, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18846107

ABSTRACT

Different types of cell death are often defined by morphological criteria, without a clear reference to precise biochemical mechanisms. The Nomenclature Committee on Cell Death (NCCD) proposes unified criteria for the definition of cell death and of its different morphologies, while formulating several caveats against the misuse of words and concepts that slow down progress in the area of cell death research. Authors, reviewers and editors of scientific periodicals are invited to abandon expressions like 'percentage apoptosis' and to replace them with more accurate descriptions of the biochemical and cellular parameters that are actually measured. Moreover, at the present stage, it should be accepted that caspase-independent mechanisms can cooperate with (or substitute for) caspases in the execution of lethal signaling pathways and that 'autophagic cell death' is a type of cell death occurring together with (but not necessarily by) autophagic vacuolization. This study details the 2009 recommendations of the NCCD on the use of cell death-related terminology including 'entosis', 'mitotic catastrophe', 'necrosis', 'necroptosis' and 'pyroptosis'.


Subject(s)
Cell Death , Terminology as Topic , Animals , Humans
11.
Oncogene ; 27(32): 4402-10, 2008 Jul 24.
Article in English | MEDLINE | ID: mdl-18469851

ABSTRACT

Paclitaxel (PTX) and other microtubule inhibitors cause mitotic arrest. However, low concentrations of PTX (low PTX) paradoxically cause G1 arrest (without mitotic arrest). Here, we demonstrated that unexpectedly, low PTX did not cause G1 arrest in the first cell cycle and did not prevent cells from passing through S phase and entering mitosis. Mitosis was prolonged but cells still divided, producing either two or three cells (tripolar mitosis), thus explaining a sub G1 peak caused by low PTX. Importantly, sub G1 cells were viable and non-apoptotic. Some cells fused back and then progressed to mitosis, frequently producing three cells again before becoming arrested in the next cell-cycle interphase. Thus, low PTX caused postmitotic arrest in second and even the third cell cycles. By increasing concentration of PTX, tripolar mitosis was transformed to mitotic slippage, thus eliminating a sub G1 peak. Time-lapse microscopy revealed that prolonged mitosis ensured a p53-dependent postmitotic arrest. We conclude that PTX directly affects cells only in mitosis and the duration of mitosis determines cell fate, including p53-dependent G1-like arrest.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , DNA/analysis , G1 Phase/drug effects , Mitosis/drug effects , Paclitaxel/pharmacology , Tumor Suppressor Protein p53/physiology , Apoptosis/drug effects , Cell Line, Tumor , Doxorubicin/pharmacology , Humans , S Phase/drug effects , Time Factors
12.
Oncogene ; 26(48): 6954-8, 2007 Oct 18.
Article in English | MEDLINE | ID: mdl-17486059

ABSTRACT

Damage-induced G1 checkpoint in mammalian cells involves upregulation of p53, which activates transcription of p21(Waf1) (CDKN1A). Inhibition of cyclin-dependent kinase (CDK)2 and CDK4/6 by p21 leads to dephosphorylation and activation of Rb. We now show that ectopic p21 expression in human HT1080 fibrosarcoma cells causes not only dephosphorylation but also depletion of Rb; this effect was p53-independent and susceptible to a proteasome inhibitor. CDK inhibitor p27 (CDKN1B) also caused Rb dephosphorylation and depletion, but another CDK inhibitor p16 (CDKN2A) induced only dephosphorylation but not depletion of Rb. Rb depletion was observed in both HT1080 and HCT116 colon carcinoma cells, where p21 was induced by DNA-damaging agents. Rb depletion after DNA damage did not occur in the absence of p21, and it was reduced when p21 induction was inhibited by p21-targeting short hairpin RNA or by a transdominant inhibitor of p53. These results indicate that p21 both activates Rb through dephosphorylation and inactivates it through degradation, suggesting negative feedback regulation of damage-induced cell-cycle checkpoint arrest.


Subject(s)
Colonic Neoplasms/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Fibrosarcoma/metabolism , Retinoblastoma Protein/metabolism , Antibiotics, Antineoplastic/pharmacology , Colonic Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Damage/drug effects , Doxorubicin/pharmacology , Fibrosarcoma/pathology , Humans , Immunoblotting , Phosphorylation/drug effects , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Tumor Suppressor Protein p53/metabolism
13.
Cell Death Differ ; 13(9): 1434-41, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16311509

ABSTRACT

Selective modulation of cell death is important for rational chemotherapy. By depleting Hsp90-client oncoproteins, geldanamycin (GA) and 17-allylamino-17-demethoxy-GA (17-AAG) (heat-shock protein-90-active drugs) render certain oncoprotein-addictive cancer cells sensitive to chemotherapy. Here we investigated effects of GA and 17-AAG in apoptosis-prone cells such as HL60 and U937. In these cells, doxorubicin (DOX) caused rapid apoptosis, whereas GA-induced heat-shock protein-70 (Hsp70) (a potent inhibitor of apoptosis) and G1 arrest without significant apoptosis. GA blocked caspase activation and apoptosis and delayed cell death caused by DOX. Inhibitors of translation and transcription and siRNA Hsp70 abrogated cytoprotective effects of GA. Also GA failed to protect HL60 cells from cytotoxicity of actinomycin D and flavopiridol (FL), inhibitors of transcription. We next compared cytoprotection by GA-induced Hsp70, caspase inhibitors (Z-VAD-fmk) and cell-cycle arrest. Whereas cell-cycle arrest protected HL60 cells from paclitaxel (PTX) but not from FL and DOX, Z-VAD-fmk prevented FL-induced apoptosis but was less effective against DOX and PTX. Thus, by inducing Hsp70, GA protected apoptosis-prone cells in unique and cell-type selective manner. Since GA does not protect apoptosis-reluctant cancer cells, we envision a therapeutic strategy to decrease side effects of chemotherapy without affecting its therapeutic efficacy.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Benzoquinones/pharmacology , Caspase Inhibitors , Doxorubicin/pharmacology , HSP70 Heat-Shock Proteins/biosynthesis , Lactams, Macrocyclic/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cytoprotection , Dactinomycin/pharmacology , Enzyme Activation , Flavonoids/pharmacology , Humans , Paclitaxel/pharmacology , Piperidines/pharmacology , Protein Biosynthesis/drug effects , RNA, Small Interfering/genetics , Transcriptional Activation/drug effects
14.
Leukemia ; 20(3): 385-91, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16357832

ABSTRACT

Tumor stem cells are quiescent and, therefore, resistant to therapy, yet harbor the capacity to replenish a tumor after therapy. Therefore, it is tempting to explain all therapeutic failures by the persistence of tumor stem cells. Yet, this explanation is relevant only to initial stages of stem-cell-dependent tumors (such as chronic myeloid leukemia) that, actually, are well controlled by therapy. In advanced cancers that poorly respond to therapy, quiescent tumor stem cells play a negligible role. Instead, proliferating cells determine disease progression, prognosis, therapeutic failures, and resistance to therapy. And therapy fails not because it eliminates only proliferating tumor cells, but because it does not eliminate them. With noticeable exceptions, it is the proliferating cell that should be targeted, whereas resting cancer cells including stem and dormant cells need to be targeted only when they 'wake up'. Finally, I discuss a strategy of selectively killing dominant proliferating clones, including proliferating stem-like and drug-resistant cancer cells, while sparing normal cells.


Subject(s)
Cell Division , Neoplasms/therapy , Stem Cell Transplantation , Cell Line , Drug Resistance, Neoplasm , Humans , Neoplasms/pathology , Recurrence
16.
Cell Death Differ ; 12(6): 592-602, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15818400

ABSTRACT

Carcinogenesis and cancer therapy are two sides of the same coin, such that the same cytotoxic agent can cause cancer and be used to treat cancer. This review links carcinogenesis, chemoprevention and cancer therapy in one process driven by cytotoxic agents (carcinoagents) that select either for or against cells with oncogenic alterations. By unifying therapy and cancer promotion and by distinguishing nononcogenic and oncogenic mechanisms of resistance, I discuss anticancer- and chemopreventive agent-induced carcinogenesis and tumor progression and, vice versa, carcinogens as anticancer drugs, anticancer drugs as chemopreventive agents and exploiting oncogene-addiction and drug resistance for chemoprevention and cancer therapy.


Subject(s)
Carcinogens/adverse effects , Chemoprevention , Neoplasms/drug therapy , Animals , Carcinogens/pharmacology , Carcinogens/therapeutic use , Chemoprevention/trends , Disease Progression , Drug Resistance, Neoplasm , Humans , Neoplasms/pathology
18.
Br J Cancer ; 89(7): 1147-51, 2003 Oct 06.
Article in English | MEDLINE | ID: mdl-14520435

ABSTRACT

Instead of exploiting the differences between normal and cancer cells, seemingly unrelated anticancer modalities (from immunotherapy to hormones) exploit (a). the differences between various normal tissues and (b). tissue-specific similarities of normal and cancer cells. Although these therapies are successfully used for years to treat leukaemia and cancer, their unifying principles have never been explicitly formulated: namely, they are aimed at differentiated cells and normal tissues and target both normal and cancer cells in a tissue-specific manner. Whereas tiny differences between cancer and normal cells have yet to be successfully exploited for selective anticancer therapy, numerous tissue-specific differences (e.g. differences between melanocytes, prostate, thyroid and breast cells) provide a means to attack selectively that exact tissue that produced cancer. Despite inherent limitations, such as fostering resistance and dedifferentiation, tissue-selective therapy have enormous potentials to control cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/therapy , Organ Specificity , Animals , Humans
19.
Leukemia ; 16(4): 455-62, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11960322

ABSTRACT

Geldanamycin (GA), herbimycin A and radicicol bind heat-shock protein-90 (Hsp90) and destabilize its client proteins including v-Src, Bcr-Abl, Raf-1, ErbB2, some growth factor receptors and steroid receptors. Thus, Hsp90-active agents induce ubiquitination and proteasomal degradation of numerous oncoproteins. Depending on the cellular context, HSP90-active agents cause growth arrest, differentiation and apoptosis, or can prevent apoptosis. HSP-active agents are undergoing clinical trials. Like targets of most chemotherapeutics, Hsp90 is not a cancer-specific protein. By attacking a nonspecific target, HSP-90-active compounds still may preferentially kill certain tumor cells. How can this be achieved? How can therapeutic potentials be exploited? This article starts the discussion.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Neoplasms/metabolism , Oncogene Proteins/metabolism , Quinones/pharmacology , Receptors, Growth Factor/metabolism , Animals , Benzoquinones , Clinical Trials as Topic , Growth Inhibitors/pharmacology , Humans , Lactams, Macrocyclic , Neoplasms/drug therapy , Neoplasms/pathology , Protein Kinases/metabolism , Signal Transduction , Transcription Factors/metabolism
20.
Leukemia ; 16(4): 570-2, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11960334

ABSTRACT

Seemingly disappointing, the Bcr-Abl kinase inhibitor STI-571 shares an 'unfortunate' characteristic with conventional cancer drugs: the development of drug resistance. I argue that the resistance must develop even faster to STI-571 than to conventional drugs, because STI-571 is so effective. This is predictable, but is it inevitable? And how do mechanisms of resistance in relapse depend on a degree of remission. In addition to mutation rate and number of tumor cells, one additional factor determines relapse vs. 'extinction' of the leukemia cell population.


Subject(s)
Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Piperazines/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Benzamides , Fusion Proteins, bcr-abl , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...