HonCode

Go Back   HER2 Support Group Forums > Articles of Interest
Register Gallery FAQ Members List Calendar Search Today's Posts Mark Forums Read

Reply
 
Thread Tools Display Modes
Old 07-14-2009, 04:47 PM   #1
Rich66
Senior Member
 
Rich66's Avatar
 
Join Date: Feb 2008
Location: South East Wisconsin
Posts: 3,431
Antimitotic chemos promote adhesion in detached and circulating tumor cells

(Broad implications and possible relation to other chemo concerns, Taxol studied, filed patent shows reversal strategies: Vimentin, glu tubulin, kinesin inhibition, other theoretical remedies: green tea, Ashwaghanda etc)




1: Breast Cancer Res Treat. 2009 Jul 11. [Epub ahead of print]
Antimitotic chemotherapeutics promote adhesive responses in detached and circulating tumor cells.
(DOI: 10.1007/s10549-009-0457-3)

View Highlights from full text Here

Contact lead researcher (Martin) here

Balzer EM, Whipple RA, Cho EH, Matrone MA, Martin SS.
Program in Molecular Medicine, University of Maryland School of Medicine, Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, 21201, USA.
In the clinical treatment of breast cancer, antimitotic cytotoxic agents are one of the most commonly employed chemotherapies, owing largely to their antiproliferative effects on the growth and survival of adherent cells in studies that model primary tumor growth. Importantly, the manner in which these chemotherapeutics impact the metastatic process remains unclear. Furthermore, since dissemination of tumor cells through the systemic circulation and lymphatics necessitates periods of detached survival, it is equally important to consider how circulating tumor cells respond to such compounds. To address this question, we exposed both nontumorigenic and tumor-derived epithelial cell lines to two antitumor compounds, jasplakinolide and paclitaxel (Taxol), in a series of attached and detached states. We report here that jasplakinolide promoted the extension of microtubule-based projections and microtentacle protrusions in adherent and suspended cells, respectively. These protrusions were specifically enriched by upregulation of a stable post-translationally modified form of alpha-tubulin, and this occurred prior to, and independently of any reductions in cellular viability. Microtubule stabilization with Taxol significantly enhanced these effects. Additionally, Taxol promoted the attachment and spreading of suspended tumor cell populations on extracellular matrix. While the antiproliferative effects of these compounds are well recognized and clinically valuable, our findings that microfilament and microtubule binding chemotherapeutics rapidly increase the mechanisms that promote endothelial adhesion of circulating tumor cells warrant caution to avoid inadvertently enhancing metastatic potential, while targeting cell division.
PMID: 19593636 [PubMed - as supplied by publisher]
Some highlights:


Quote:
Because the cytoskeleton is a key regulator of cellular function and viability, some of the most clinically successful chemotherapies operate by directly targeting cytoskeletal filaments [36]. The most prominent examples of this approach are the taxanes, docetaxel, and paclitaxel, a family of microtubule (MT) stabilizing agents

Quote:
The vast majority of studies to date have examined these effects exclusively with regard to adherent cell populations. As a result, the effects of these chemotherapeutics on suspended cell populations have been largely overlooked. Since dissemination of tumor cells through the lymphatics or vasculature requires release from extracellular matrix (ECM) and indefinite periods of suspension, it is equally important to consider the cytoskeletal dynamics of circulating tumor cells (CTCs) and their contributions to survival and metastatic spread. Interestingly, taxane treatment has been shown to increase the concentration of CTCs in the bloodstream more than 1000-fold when applied in advance of surgery [16]. Additionally, taxanes can worsen progression-free survival compared to anthracycline treatment when used as a monotherapy [17], though a mechanism to explain such findings has not been delineated. Our recent work describing microtubule-based microtentacles (McTNs) in detached and circulating breast tumor cells [18, 19] provides a mechanism by which cytoskeletally directed chemotherapies could influence CTCs and highlights the need to investigate these effects.

Quote:
studies have focused principally on early steps of tumor progression that occur prior to release ECM release, transit through the circulation, extravasation, and colonization of secondary tissues [1], and therefore fail to address the manner in which these chemotherapies impact the behavior of circulating tumor cells, or those that are not actively circulating, but disseminated and resisting apoptosis.
taxane treatment has been shown to increase the concentration of CTCs in the bloodstream more than 1000-fold when applied in advance of surgery [16]. Additionally, taxanes can worsen progression-free survival compared to anthracycline treatment when used as a monotherapy [17], though a mechanism to explain such findings has not been delineated. Our recent work describing microtubule-based microtentacles (McTNs) in detached and circulating breast tumor cells [18, 19] provides a mechanism by which cytoskeletally directed chemotherapies could influence CTCs and highlights the need to investigate these effects.

Quote:
surgical resection of a primary tumor increases the concentration of epithelial cells in systemic circulation by 1000-fold, and tumor cells can still be detected in the bloodstream of patients many years after entering what is clinically recognized as remission [16, 30]
Quote:
periods of dormant survival require tumor cells to be resistant to apoptotic stimuli, and are thus ineffectively targeted by cytotoxic chemotherapies due to a combination of mitotic inactivity and apoptotic resistance [29, 33]. Circulating and dormant tumor cells may fail to die in response to MT and microfilament-binding chemotherapeutics, but the cytoskeletal behavior of these tumor cells is likely to be affected by continued exposure to these drugs.

Quote:
a two-step model for extravasation, wherein initial adhesion is tubulin-dependent and subsequent escape into surrounding tissue is driven by actin-mediated contractile forces. Our results support this model, and indicate that the initial phase of this adhesive transition depends upon microtentacles to facilitate endothelial or ECM contact. Therefore, exposure of CTCs to MT-stabilizing agents such as Taxol could enhance this initial MT-dependent adhesion and promote extravasation into secondary sites. In this study, Taxol served to enhance the adhesion and efficient cell spreading of MDA-436 tumor cells, relative to cells under vehicle control conditions.

Quote:
Though no apparent increase in McTN frequency was recorded in response to Taxol alone, there was a very significant increase in ECM adhesion and degree of cell spreading after 45 min. This suggests that while Taxol is insufficient to stimulate the formation of additional McTNs, it stabilizes existing structures and therefore promotes ECM contact and adhesion via a MT-dependent mechanism. It is likely because of the fact that the highly invasive MDA-436 cell line produces high levels of McTNs basally and that this enhancement was so pronounced in these cells[19].



Quote:
In light of our recent observations that microtentacles were present in a high percentage of circulating tumor cells isolated from the blood of breast cancer patients with advanced metastatic disease (Balzer, EM and Chumsri, S unpublished data), the work described here highlights the importance of determining whether Taxane treatment can promote extravasation of disseminated tumor cells. So, while stabilizing microtubules and disrupting actin filaments can each decrease primary tumor volume, it is important to consider the effects of these treatments on circulating tumor cells to avoid inadvertently enhancing the metastatic potential of disseminated tumor cells, while targeting cell division.


http://lifesciences.umaryland.edu/Pa...nt.aspx?SID=12
Research

Epithelial cells perform a critical barrier function in vivo, and the integrity of this barrier is maintained by rigid cell-cell and cell-ECM contacts. If such contacts are disrupted and the detached cells fail to re-adhere quickly, apoptosis is initiated. Our lab has shown that suspended epithelial cells extend microtubule-based plasma membrane protrusions (microtentacles) that promote reattachment to neighboring cells and extracellular substrates.


While this phenomenon appears to be a feature of healthy epithelial tissues, it is significantly enhanced in highly invasive tumor cells, and has been directly observed in circulating tumor cells isolated from blood samples of patients with advanced metastatic disease (Balzer, E. and Chumsri, S unpublished data). Our lab is seeking to test the hypothesis that microtentacles promote metastasis by facilitating the extravasation and/or survival of circulating tumor cells. Specifically, my research focuses on defining the molecular interplay between the actin and tubulin cytoskeletons that forms the mechanistic basis for the extension of these microtentacles, in an effort to understand how this process is naturally exploited by cancer cells.

Laboratory Techniques

We utilize a wide range of traditional cell and molecular genetic techniques. We also employ a variety of imaging applications, including live-cell microscopy, laser scanning confocal analysis of detached cells, epifluorescence microscopy of tumor-endothelial interactions (under flow conditions and in intact blood vessels), and whole-animal optical tracking of circulating tumor cells.



Publications

Balzer, E.M., Whipple, R.A., Cho, E.H., and Martin, S.S. (2009). Antimitotic chemotherapeutics promote adhesive responses in detached and circulating tumor cells. Breast Cancer Res Treat. epub July 11. (DOI: 10.1007/s10549-009-0457-3)

Whipple, R.A., Balzer, E.M., Cho, E.H., Matrone, M.A., Yoon, J.R., and Martin, S.S. (2007) Vimentin filaments support extension of tubulin-based microtentacles in detached breast tumor cell lines. Cancer Res. 2008. 68, 5678-5688

Cheng, Q., Balzer, E., Yoshida, M., Wong, J.C., and Miller, S.M. (2006) Effect of histone deacetylase inhibitors on tubulin acetylation in Volvox carteri (Volvocales). J. Phycol. 2006. 42, 417-422.

Balzer, E.M., et al.(2009). Microtentacles and Invadopodia: functionally distinct plasma membrane protrusions of metastatic breast tumor cells separated mechanistically by c-Src. (submitted to Cancer Research)

Whipple, R.A., Cho, E.H., Balzer, E.M., Matrone, M.A., Yang, J., and Martin, S.S. Metastasis-associated microtentacles are promoted by Epithelial-Mesenchymal Transition in human mammary epithelial cells and breast tumor cells. (submitted)



http://www.worldstemcellsummit.com/2009_abstracts.html
Epithelial-to-Mesenchymal Transition Promotes Breast Tumor Stem Cell Characteristics, Tubulin Detyrosination and Microtentacles

Stuart S. Martin, Rebecca Whipple Bettes, Monica Charpentier, Michael A. Matrone, Edward H. Cho, Eric M. Balzer, Kimberly C. Tuttle, Michele I. Vitolo, Jennifer R. Yoon, Jing Yang, Olga B. Ioffe
University of Maryland Baltimore, USA

Understanding the biology of breast tumor stem cells during passage through the bloodstream could provide new therapeutic opportunities to destroy circulating tumor cells. When detached from extracellular matrix, breast tumor cells with increased stem cell characteristics produce unique microtentacles composed of coordinated vimentin intermediate filaments and detyrosinated microtubules. These microtentacles promote the reattachment of circulating tumor cells and are associated with increased invasiveness in vitro and metastatic potential in vivo. Our previous work has shown that microtentacles are increased in Basal B breast tumor cell lines that display hallmarks of epithelial-to-mesenchymal transition (EMT) and stem cell markers (CD44+/CD24-). We report here that the direct induction of EMT by expression of the transcription factor Twist promotes increased stem cell characteristics (CD44+/CD24-) and microtentacle formation in detached human mammary epithelial cells (HMECs). Mechanistically, EMT results in an elevation of alpha-tubulin in which the c-terminal tyrosine residue has been removed (Glu-tubulin), and reorganization of such Glu-tubulin into microtentacle extensions. Conversely, siRNA-mediated downregulation of endogenous Twist in (CD44+/CD24-) breast carcinoma cells decreased Glu-tubulin levels, organization and the frequency of microtentacles. Twist siRNA decreased levels of Glu-tubulin before any changes in either vimentin or E-Cadherin expression, indicating that Twist can have a primary effect on tubulin detyrosination without requiring additional elements of the EMT program. This model is further supported by immunohistochemical staining of 66 patient tissue samples that show a high concordance between Glu-tubulin and Twist staining in invasive ductal carcinomas. In addition, tumor cells co-staining for Twist and detyrosinated tubulin are enriched at the invasive front where tumor cells encounter a changing microenvironment. Given the role of detyrosinated tubulin in cell motility and orientation, these EMT-induced microtubule alterations and stem cell induction may occur as part of an inherent program for cell movement during development or wound healing. However, this program may also prime tumor cells for metastatic success by increasing stem cell charactersitics, stabilizing microtubules, and promoting microtentacles. Functionally, expression of Twist led to an enhanced rate of tumor cell reattachment as monitored over time by electrical impedence or measured by fluorescence over an endothelial layer. Confocal microscopy captures microtentacles engaging and invading through an endothelial layer to facilitate reattachment. These data support a novel model in which the EMT that occurs during tumor invasion increases stem cell characteristics, alpha-tubulin detyrosination and promotes microtentacles which enhance the reattachment of circulating tumor cells during metastasis.


Vimentin Filaments Support Extension of Tubulin-Based Microtentacles in Detached Breast Tumor Cells

FULL TEXT

Rebecca A. Whipple1, Eric M. Balzer1,2, Edward H. Cho1,2, Michael A. Matrone1,2, Jennifer R. Yoon1,2 and Stuart S. Martin1,2 1 University of Maryland School of Medicine, Marlene and Stewart Greenebaum Cancer Center, Department of Physiology and 2 Graduate Program in Life Sciences Baltimore, Maryland
Requests for reprints: Stuart S. Martin, Bressler Building, Room 10-29, 655 W. Baltimore Street, Baltimore, MD 21201. Phone: 410-706-6601; Fax: 410-706-6600; E-mail: ssmartin@som.umaryland.edu.

Key Words: breast • microtentacle • vimentin

Solid tumor metastasis often involves detachment of epithelial carcinoma cells into the vasculature or lymphatics. However, most studies of cytoskeletal rearrangement in solid tumors focus on attached cells. In this study, we report for the first time that human breast tumor cells produce unique tubulin-based protrusions when detached from extracellular matrix. Tumor cell lines of high metastatic potential show significantly increased extension and frequency of microtubule protrusions, which we have termed tubulin microtentacles. Our previous studies in nontumorigenic mammary epithelial cells showed that such detachment-induced microtentacles are enriched in detyrosinated -tubulin. However, amounts of detyrosinated tubulin were similar in breast tumor cell lines despite varying microtentacle levels. Because detyrosinated -tubulin associates strongly with intermediate filament proteins, we examined the contribution of cytokeratin and vimentin filaments to tumor cell microtentacles. Increased microtentacle frequency and extension correlated strongly with loss of cytokeratin expression and up-regulation of vimentin, as is often observed during tumor progression. Moreover, vimentin filaments coaligned with microtentacles, whereas cytokeratin did not. Disruption of vimentin with PP1/PP2A-specific inhibitors significantly reduced microtentacles and inhibited cell reattachment to extracellular matrix. Furthermore, expression of a dominant-negative vimentin mutant disrupted endogenous vimentin filaments and significantly reduced microtentacles, providing specific genetic evidence that vimentin supports microtentacles. Our results define a novel model in which coordination of vimentin and detyrosinated microtubules provides structural support for the extensive microtentacles observed in detached tumor cells and a possible mechanism to promote successful metastatic spread. [Cancer Res 2008;68(14):5678–88]

/Live cell imaging and membrane microtentacle scoring. Cells were trypsinzed at f80% confluency after 24 h expression and resuspended in
serum/phenol red (PR)–free media over ultralow attachment plates in the presence or absence of 5 AM Latrunculin-A (LA; BioMol) for 1 h. For
microtentacle scoring involving phosphatase inhibitor treatment, MCF10A
and MDA-MB-436 were pretreated with either 1 AM okadiac acid
(OKA; Sigma) or 5 nM Calyculin-A (Cal-A
; Chemicon) in serum-containing
media for 1 h and resuspended in serum/PR-free media in the respective
phosphatase inhibitor with or without 5 AM L A./

<


Microtubule-destabilizing agents significantly reduced the frequency with which microtentacles were observed and impaired both cell cell
interaction and reattachment of cells to surfaces. Interestingly,
microtubule-destabilizing drugs prevent circulating colon carcinoma
cells from attaching to the microvascular endothelium in vivo,
although the precise mechanism is currently unclear (11). Strikingly,
actin depolymerization significantly increases binding of circulating
tumor cells to the vascular endothelium (11) and also strongly
promotes the extension of tubulin microtentacles
(10).

<

We report for the first time that vimentin-expressing invasive breast carcinomas display a comparatively higher frequency of tubulin microtentacles following detachment than non–vimentin-expressing, noninvasive cell lines.
Microtentacle preservation following detachment indicates that
vimentin coaligns with Glu-tubulin in microtentacles, whereas
cytokeratin does not. Time-lapse video shows that membrane
microtentacles are rapidly disrupted by treatment with PP1/PP2A
inhibitors that disassemble vimentin, decreasing microtentacle
frequency and impairing reattachment of vimentin-expressing
tumor cell
s.

<

Detachment of invasive breast tumor cell lines Hs578t, HCC1395, MDA-MB-436, and MDA-MB-157 produced unusually long, semiflexible, and motile microtentacles compared with noninvasive MCF10A,
ZR-75-1, Bt-20, SkBr3, and MDA-MB-468 lines, which display relatively short, rigid, and semimotile microtentacles.
Our previous research has shown that inhibiting actin polymerization with LA (5 AM) enhances microtentacle frequency, motility, and length in both human and mouse mammary epithelial cells following detachment (10). LA similarly increases the length, flexibility, and motility of microtentacles in breast tumor cells (Fig. 1B and Supplementary Movies
S4–S6). However, the invasive breast tumor cell lines were clearly able to form dynamic and flexible microtentacles even in the absence of LA treatment.

Expression of vimentin is much higher in breast tumor cells which display long and numerous microtentacles, whereas cytokeratin expression predominated in those cell lines that displayed fewer and shorter microtentacles (Fig. 1D).
Vimentin extends into microtentacles. Given the correlation of vimentin expression in those cells that exhibit more extensive and frequent microtentacles, we examined intermediate filament localization with indirect immunofluorescence, finding that vimentin, and not cytokeratin, extends into microtentacles (Fig. 2).

<

Treatment with OKA resulted in a more dramatic change in vimentin morphology than Cal-A in MCF10A cells marked by cell rounding and vimentin fragmentation (Fig. 4A, d). MDA-MB-436 displayed similar morphologic changes with OKA as seen with Cal-A with a slight increase in retraction of the vimentin network to the perinuclear region (Fig. 4A, h)

<

While MCF10A attachment efficiency to either uncoated or laminin-coated surfaces was not significantly affected by Cal-A treatment, attachment was significantly sensitive to treatment with OKA (Fig. 6A). This result matches the less-pronounced vimentin disruption in MCF10A cells after Cal-A compared with OKA (Fig. 4A, b and d). Interestingly, MDA-MB-436 attachment was significantly impaired by the presence of either Cal-A or OKA (Fig. 5C). Figure 5.

<

Vimentin disassembly with PP1/PP2A inhibitors reduces microtentacles and attachment.

<

Our data showing colocalization of vimentin with Glu-tubulin in microtentacles, the extensive microtentacles formed in cells with high vimentin expression, and the inhibition of microtentacles with dominant negative vimentin or compounds targeting vimentin assembly, all support a model in which structural coordination of Glu-tubulin
and vimentin underlie the striking morphologic abnormalities we
observe in detached breast tumor cells
.
Our data also indicate that microtentacles in detached cells are distinct from other actin-based structures in attached cells, such as invadopodia or podosomes (30), which are inhibited by actin destablizing agents, such as LA or Cytochalasin-D. In contrast, microtentacles are enhanced by actin disruption and seem to become more flexible, motile, and longer (Fig. 1B).

<

Vimentin expression is a marker of the aggressive ‘‘basal cell’’ subtype of breast tumor (33, 34), and up-regulation of vimentin predicts metastatic progression and poor patient prognosis (35).
Beyond breast cancer, vimentin is an established independent predictor of poor patient prognosis in many tumors, including cervical (36), thyroid (37), hepatocellular (38), pancreatic (39), and renal carcinomas (40). Our results indicate that cell lines with the highest microtentacle counts display reduced expression of cytokeratins and increased vimentin, a change which is also observed during the epithelial-to-mesenchymal transition (EMT) that is associated with tumor progression and poor patient prognosis (41, 42). Interestingly, vimentin is also strongly upregulated when tumor cells are exposed to hypoxia (43). Our current evidence supports a direct role for vimentin in the
mechanism underlying microtentacles in detached breast tumor cells.


<

vimentin is rapidly hyperphosphorylated and disassembled after treatment with specific inhibitors of types 1 and 2A phosphatases,
providing evidence that vimentin is a major target of PP1 and PP2A
activity (23, 24).

<

we propose that, upon detachment, vimentin aligns with stable microtubules via associated proteins to provide a flexible scaffold to Glu-enriched microtubules, which allows microtentacles to protrude at great length from the cell body. The shorter, less frequent microtentacles observed in non–vimentin-expressing, less invasive cell lines likely rely on the population of stable microtubules but lack the increased resilience
and strength of vimentin filaments (48). Disruption of vimentin filaments with either PP1/PP2A inhibitors or dominant-negative mutant vimentin likely promotes the collapse of Glu-microtubules and ultimate deformation of the microtentacles.

<

While we continue to pursue the functional role of tubulin microtentacles in metastasis, the dramatic morphologic differences observed in detached breast tumor cell lines are compelling. Intriguing in vivo evidence exists that a yet-unidentified, tubulin-based (not actin-based) mechanism promotes the binding of circulating tumor cells to the vascular endothelium (11)

Available Vimentin Inhibitors:



The Intermediate Filament Protein Vimentin Is a New Target for Epigallocatechin Gallate

FULL TEXT

Hormel Institute, University of Minnesota, Austin, Minnesota 55912

To whom correspondence should be addressed: Hormel Institute, University of Minnesota, 801 16th Ave. NE, Austin, MN 55912. Tel.: 507-437-9600; Fax: 507-437-9606; E-mail: zgdong@hi.umn.edu.
Abstract

Epigallocatechin gallate (EGCG) is the major active polyphenol in green tea. Protein interaction with EGCG is a critical step in the effects of EGCG on the regulation of various key proteins involved in signal transduction. We have identified a novel molecular target of EGCG using affinity chromatography, two-dimensional electrophoresis, and mass spectrometry for protein identification. Spots of interest were identified as the intermediate filament, vimentin. The identification was confirmed by Western blot analysis using an anti-vimentin antibody. Experiments using a pull-down assay with [3H]EGCG demonstrate binding of EGCG to vimentin with a Kd of 3.3 nm. EGCG inhibited phosphorylation of vimentin at serines 50 and 55 and phosphorylation of vimentin by cyclin-dependent kinase 2 and cAMP-dependent protein kinase. EGCG specifically inhibits cell proliferation by binding to vimentin. Because vimentin is important for maintaining cellular functions and is essential in maintaining the structure and mechanical integration of the cellular space, the inhibitory effect of EGCG on vimentin may further explain its anti-tumor-promoting effect.




Chem Biol. 2007 Jun;14(6):623-34.
The tumor inhibitor and antiangiogenic agent withaferin A targets the intermediate filament protein vimentin.

FULL TEXT

Bargagna-Mohan P, Hamza A, Kim YE, Khuan Abby Ho Y, Mor-Vaknin N, Wendschlag N, Liu J, Evans RM, Markovitz DM, Zhan CG, Kim KB, Mohan R.
Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA.
Correspondence: royce.mohan@uky.edu

The natural product withaferin A (WFA) exhibits antitumor and antiangiogenesis activity in vivo, which results from this drug's potent growth inhibitory activities. Here, we show that WFA binds to the intermediate filament (IF) protein, vimentin, by covalently modifying its cysteine residue, which is present in the highly conserved alpha-helical coiled coil 2B domain. WFA induces vimentin filaments to aggregate in vitro, an activity manifested in vivo as punctate cytoplasmic aggregates that colocalize vimentin and F-actin. WFA's potent dominant-negative effect on F-actin requires vimentin expression and induces apoptosis. Finally, we show that WFA-induced inhibition of capillary growth in a mouse model of corneal neovascularization is compromised in vimentin-deficient mice. These findings identify WFA as a chemical genetic probe of IF functions, and illuminate a potential molecular target for withanolide-based therapeutics for treating angioproliferative and malignant diseases.

PMID: 17584610 [PubMed - indexed for MEDLINE]



Cancer Sci. 2006 Jul;97(7):658-64.
Chemotherapeutic efficacy of paclitaxel in combination with Withania somnifera on benzo(a)pyrene-induced experimental lung cancer.

Senthilnathan P, Padmavathi R, Magesh V, Sakthisekaran D.
Department of Medical Biochemistry, ALM Postgraduate Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai 600-113, India.
Lung cancer is one of the leading causes of cancer death in the world and is notoriously difficult to treat effectively. In the present study, male Swiss albino mice were divided into five groups of six animals each: group I animals received corn oil orally and served as a control; group II cancer-induced animals received benzo(a)pyrene (50 mg/kg bodyweight dissolved in corn oil, orally) twice weekly for four successive weeks; group III cancer-bearing animals (after 12 weeks of induction) were treated with paclitaxel (33 mg/kg bodyweight, i.p.) once weekly for 4 weeks; group IV cancer-bearing animals were treated with paclitaxel along with Withania somnifera (400 mg/kg bodyweight) orally once weekly for 4 weeks; and group V animals constituted the drug control treated with paclitaxel along with W. somnifera. The serum, lung and liver were investigated biochemically for aryl hydrocarbon hydroxylase, gamma-glutamyl transpeptidase, 5'-nucleotidase, lactate dehydrogenase and protein-bound carbohydrate components (hexose, hexosamine and sialic acid). These enzyme activities were increased significantly in cancer-bearing animals compared with control animals. The elevation of these in cancer-bearing animals was indicative of the persistent deteriorating effect of benzo(a)pyrene in cancer-bearing animals. Our data suggest that paclitaxel, administered with W. somnifera, may extend its chemotherapeutic effect through modulating protein-bound carbohydrate levels and marker enzymes, as they are indicators of cancer. The combination of paclitaxel with W. somnifera could effectively treat the benzo(a)pyrene-induced lung cancer in mice by offering protection from reactive oxygen species damage and also by suppressing cell proliferation.

PMID: 16827807 [PubMed - indexed for MEDLINE]







Chem Biol Interact. 2006 Feb 25;159(3):180-5. Epub 2005 Dec 22.
Enhancement of antitumor effect of paclitaxel in combination with immunomodulatory Withania somnifera on benzo(a)pyrene induced experimental lung cancer.

Senthilnathan P, Padmavathi R, Banu SM, Sakthisekaran D.
Department of Medical Biochemistry, Dr. ALM Postgraduate Institute of Basic Medical Sciences, University of Madras, Taramani, Chennai 600113, India. senthilnpm@rediffmail.com
The current experimental work deals with the immunomodulatory studies on the extract of Withania somnifera (L.) Dunal root powder against benzo(a)pyrene induced lung cancer in male Swiss albino mice. In our previous study, we reported the antioxidant and anticarcinogenic effect of W. somnifera (L.) Dunal along with paclitaxel. Immune dysfunction has been found to be associated with cancer and chemotherapy. Benzo(a)pyrene induced cancer animals were treated with 400mg/kg bodyweight of W. somnifera (L.) Dunal extract for 30 days significantly alters the levels of immunocompetent cells, immune complexes and immunoglobulins. Based on the data, the carcinogen as well as the paclitaxel affects the immune system, the toxic side effects on the immune system is more reversible and more controllable by W. somnifera (L.) Dunal. These results concluded the immunomodulatory activity of W. somnifera (L.) Dunal extract, which is a known immunomodulator in indigenous medicine.

PMID: 16375880 [PubMed - indexed for MEDLINE]


Altern Med Rev. 2006 Dec;11(4):269-77.
Ancient medicine, modern use: Withania somnifera and its potential role in integrative oncology.

Winters M.
Withania somnifera Dunal, commonly known as ashwagandha, has been used for centuries in Ayurvedic medicine to increase longevity and vitality. Western research supports its polypharmaceutical use, confirming antioxidant, anti-inflammatory, immune-modulating, and antistress properties in the whole plant extract and several separate constituents. This article reviews the literature pertaining to Withania somnifera and its botanical constituents as antitumor agents and in conjunction with radiation and chemotherapy treatment. Following a search of MEDLINE and EBSCO databases, it can be concluded that Withania somnifera reduces tumor cell proliferation while increasing overall animal survival time. Furthermore, it has been shown to enhance the effectiveness of radiation therapy while potentially mitigating undesirable side effects. Withania somnifera also reduces the side effects of chemotherapeutic agents cyclophosphamide and paclitaxel without interfering with the tumor-reducing actions of the drugs. These effects have been demonstrated in vitro on human cancer cell lines, and in vivo on animal subjects, but there have been no human trials to date. Given its broad spectrum of cytotoxic and tumor-sensitizing actions, Withania somnifera presents itself as a novel complementary therapy for integrative oncology care.

PMID: 17176166 [PubMed - indexed for MEDLINE]




Indian J Physiol Pharmacol. 2001 Apr;45(2):253-7.
Reversal of paclitaxel induced neutropenia by Withania somnifera in mice.

Gupta YK, Sharma SS, Rai K, Katiyar CK.
Department of Pharmacology, All India Institute of Medical Sciences, New Delhi-110 029.
The effect of aqueous extract of Withania somnifera (L. Solanaceae) was studied against paclitaxel induced neutropenia in mice. After paclitaxel 1 mg/kg, i.v. administration significant fall in total WBC and absolute neutrophil count was observed on day 3 and day 5. W. Somnifera (200 mg/kg, p.o.) per se produced significant increase in neutrophil counts. W. somnifera (200 mg/kg, p.o.) when administered for 4 days before paclitaxel treatment and continued for 12 days caused significant reversal of neutropenia of paclitaxel. The findings of the study suggest the potential of W. somnifera as an adjuvant during cancer chemotherapy for the prevention of bone marrow depression associated with anticancer drugs.

PMID: 11480235 [PubMed - indexed for MEDLINE]




Clin Cancer Res. 2007 Apr 1;13(7):2298-306.
Selective killing of cancer cells by leaf extract of Ashwagandha: identification of a tumor-inhibitory factor and the first molecular insights to its effect.

Widodo N, Kaur K, Shrestha BG, Takagi Y, Ishii T, Wadhwa R, Kaul SC.
Research Institute for Cell Engineering, GENE Therapeutics, Inc., National Institute of Advanced Industrial Science and Technology, Higashi, Tsukuba, Japan.
PURPOSE: Ashwagandha is regarded as a wonder shrub of India and is commonly used in Ayurvedic medicine and health tonics that claim its variety of health-promoting effects. Surprisingly, these claims are not well supported by adequate studies, and the molecular mechanisms of its action remain largely unexplored to date. We undertook a study to identify and characterize the antitumor activity of the leaf extract of ashwagandha. EXPERIMENTAL DESIGN: Selective tumor-inhibitory activity of the leaf extract (i-Extract) was identified by in vivo tumor formation assays in nude mice and by in vitro growth assays of normal and human transformed cells. To investigate the cellular targets of i-Extract, we adopted a gene silencing approach using a selected small hairpin RNA library and found that p53 is required for the killing activity of i-Extract. RESULTS: By molecular analysis of p53 function in normal and a variety of tumor cells, we found that it is selectively activated in tumor cells, causing either their growth arrest or apoptosis. By fractionation, purification, and structural analysis of the i-Extract constituents, we have identified its p53-activating tumor-inhibiting factor as with a none. CONCLUSION: We provide the first molecular evidence that the leaf extract of ashwagandha selectively kills tumor cells and, thus, is a natural source for safe anticancer medicine.

PMID: 17404115 [PubMed - indexed for MEDLINE]


Cancer Lett. 2008 Jan 9. [Epub ahead of print]
Selective killing of cancer cells by leaf extract of Ashwagandha: Components, activity and pathway analyses.

Widodo N, Takagi Y, Shrestha BG, Ishii T, Kaul SC, Wadhwa R.
National Institute of Advanced Industrial Science & Technology (AIST), Central 4, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8562, Japan; Department of Molecular and Cellular Physiology, University of Tsukuba, Ibaraki 305-8575, Japan.
Ashwagandha, also called as "Queen of Ayurveda" and "Indian ginseng", is a commonly used plant in Indian traditional medicine, Ayurveda. Its roots have been used as herb remedy to treat a variety of ailments and to promote general wellness. However, scientific evidence to its effects is limited to only a small number of studies. We had previously identified anti-cancer activity in the leaf extract (i-Extract) of Ashwagandha and demonstrated withanone as a cancer inhibitory factor (i-Factor). In the present study, we fractionated the i-Extract to its components by silica gel column chromatography and subjected them to cell based activity analyses. We found that the cancer inhibitory leaf extract (i-Extract) has, at least, seven components that could cause cancer cell killing; i-Factor showed the highest selectivity for cancer cells and i-Factor rich Ashwagandha leaf powder was non-toxic and anti-tumorigenic in mice assays. We undertook a gene silencing and pathway analysis approach and found that i-Extract and its components kill cancer cells by at least five different pathways, viz. p53 signaling, GM-CFS signaling, death receptor signaling, apoptosis signaling and G2-M DNA damage regulation pathway. p53 signaling was most common. Visual analysis of p53 and mortalin staining pattern further revealed that i-Extract, fraction F1, fraction F4 and i-Factor caused an abrogation of mortalin-p53 interactions and reactivation of p53 function while the fractions F2, F3, F5 work through other mechanisms.

PMID: 18191020 [PubMed - as supplied by publisher]


J Ethnopharmacol. 2006 May 24;105(3):336-41. Epub 2006 Jan 10.
Evaluation of the effect of Withania somnifera root extracts on cell cycle and angiogenesis.

Mathur R, Gupta SK, Singh N, Mathur S, Kochupillai V, Velpandian T.
Department of Pharmacology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India.
In the Indian System of Medicine, the medicinal plant, Withania somnifera Dunal (Solanaceae) finds application for numerous ailments including cancer. This study explores the mechanism(s) underlying this property. The hydroalcoholic extract of the roots (WS) was partitioned between chloroform (WS-chloroform) and water (WS-water). Further, WS-chloroform was fractionated (A1-A12) by reverse-phase column chromatography and their withanolide content was quantified by high-performance liquid chromatography (HPLC). Preliminarily, the anti-proliferative activity of all the extracts and fractions was screened against human laryngeal carcinoma (Hep2) cells by microculture tetrazolium assay (MTT). Two extracts (WS and WS-chloroform) and three fractions (A4, A5 and A6) negatively affected Hep2 viability at the concentration of 25 microg/ml and these were further investigated pharmacologically. Flow cytometry revealed cell cycle block and accumulation of hypoploid (sub G1) cells as the mode of anti-proliferative activity of all but A4. Their anti-angiogenic potential was investigated by a chickchorio-allantoic membrane (CAM) wherein a significant inhibition (p<0.0001) of vascular endothelium growth factor (VEGF), induced neovascularization was recorded. The effect was confirmed in vivo by mouse sponge implantation method. These findings suggest that the roots of Withania somnifera possess cell cycle disruption and anti-angiogenic activity, which may be a critical mediator for its anti-cancer action.

PMID: 16412596 [PubMed - indexed for MEDLINE]




Cancer Res. 2008 Sep 15;68(18):7661-9.


Nutr Cancer. 2008;60 Suppl 1:51-60.
Ayurvedic medicine constituent withaferin a causes G2 and M phase cell cycle arrest in human breast cancer cells.

Stan SD, Zeng Y, Singh SV.
University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Withaferin A (WA) is derived from the medicinal plant Withania somnifera that has been safely used for centuries in the Indian Ayurvedic medicine for treatment of various ailments. We now demonstrate that WA treatment causes G2 and mitotic arrest in human breast cancer cells. Treatment of MDA-MB-231 (estrogen-independent) and MCF-7 (estrogen-responsive) cell lines with WA resulted in a concentration- and time-dependent increase in G2-M fraction, which correlated with a decrease in levels of cyclin-dependent kinase 1 (Cdk1), cell division cycle 25C (Cdc25C) and/or Cdc25B proteins, leading to accumulation of Tyrosine15 phosphorylated (inactive) Cdk1. Ectopic expression of Cdc25C conferred partial yet significant protection against WA-mediated G2-M phase cell cycle arrest in MDA-MB-231 cells. The WA-treated MDA-MB-231 and MCF-7 cells were also arrested in mitosis as judged by fluorescence microscopy and analysis of Ser10 phosphorylated histone H3. Mitotic arrest resulting from exposure to WA was accompanied by an increase in the protein level of anaphase promoting complex/cyclosome substrate securin. In conclusion, the results of this study suggest that G2-M phase cell cycle arrest may be an important mechanism in antiproliferative effect of WA against human breast cancer cells.

PMID: 19003581 [PubMed - indexed for MEDLINE]



Phytother Res. 2009 Jul;23(7):987-92.
Withanolide sulfoxide from Aswagandha roots inhibits nuclear transcription factor-kappa-B, cyclooxygenase and tumor cell proliferation.

Mulabagal V, Subbaraju GV, Rao CV, Sivaramakrishna C, Dewitt DL, Holmes D, Sung B, Aggarwal BB, Tsay HS, Nair MG.
Bioactive Natural Products and Phytoceuticals, Department of Horticulture and National Food, Safety and Toxicology Center, Michigan State University, East Lansing, Michigan, USA.
Investigation of the methanol extract of Aswagandha (Withania somnifera) roots for bioactive constituents yielded a novel withanolide sulfoxide compound (1) along with a known withanolide dimer ashwagandhanolide (2) with an S-linkage. The structure of compound 1 was established by extensive NMR and MS experiments. Compound 1 was highly selective in inhibiting cyclooxygenase-2 (COX-2) enzyme by 60% at 100 microm with no activity against COX-1 enzyme. The IC(50) values of compound 1 against human gastric (AGS), breast (MCF-7), central nervous system (SF-268) and colon (HCT-116) cancer cell lines were in the range 0.74-3.63 microm. Both S-containing dimeric withanolides, 1 and 2, completely suppressed TNF-induced NF-kappaB activation when tested at 100 microm. The isolation of a withanolide sulfoxide from W. somnifera roots and its ability to inhibit COX-2 enzyme and to suppress human tumor cell proliferation are reported here for the first time. In addition, this is the first report on the abrogation of TNF-induced NF-kappaB activation for compounds 1 and 2. Copyright 2009 John Wiley & Sons, Ltd.

PMID: 19152372 [PubMed - indexed for MEDLINE]




Pharmazie. 2007 Apr;62(4):305-7.
Study of the anticancer potential of Yemeni plants used in folk medicine.

Mothana RA, Grünert R, Lindequist U, Bednarski PJ.
Department of Pharmacognosy, Faculty of Pharmacy, Sana'a-University, Yemen. r_mothana@yahoo.com
The present work evaluated the anticancer activity of methanol extracts from 24 plants used in Yemeni traditional medicine. To evaluate the in vitro cytotoxic potency of the investigated extracts, an established microtiter plate assay based on cellular staining with crystal violet was used with 5 human cancer cell lines: two lung cancer (A-427 and LCLC-103H), two urinary bladder carcinoma (5637 and RT-112) and one breast cancer (MCF-7) line. The methanolic extracts of Dendrosicyos socotrana, Withanina aduensis, Withania riebeckii, Dracena cinnabari and Buxus hildebrandtii exhibited the highest toxicity on all tumor cell lines with IC50 values ranging between 0.29 and 5.54 microg/ml. The extracts of Jatropha unicostata and Punica protopunica showed a moderate potency on the most tumor cell lines.

PMID: 17484289 [PubMed - indexed for MEDLINE]



Withaferin A causes FOXO3a- and Bim-dependent apoptosis and inhibits growth of human breast cancer cells in vivo.

Stan SD, Hahm ER, Warin R, Singh SV.
Department of Pharmacology and Chemical Biology, and University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
Withaferin A (WA) is derived from the medicinal plant Withania somnifera, which has been safely used for centuries in Indian Ayurvedic medicine for treatment of different ailments. We now show, for the first time, that WA exhibits significant activity against human breast cancer cells in culture and in vivo. The WA treatment decreased viability of MCF-7 (estrogen-responsive) and MDA-MB-231 (estrogen-independent) human breast cancer cells in a concentration-dependent manner. The WA-mediated suppression of breast cancer cell viability correlated with apoptosis induction characterized by DNA condensation, cytoplasmic histone-associated DNA fragmentation, and cleavage of poly-(ADP-ribose)-polymerase. On the other hand, a spontaneously immortalized normal mammary epithelial cell line (MCF-10A) was relatively more resistant to WA-induced apoptosis compared with breast cancer cells. The WA-mediated apoptosis was accompanied by induction of Bim-s and Bim-L in MCF-7 cells and induction of Bim-s and Bim-EL isoforms in MDA-MB-231 cells. The cytoplasmic histone-associated DNA fragmentation resulting from WA exposure was significantly attenuated by knockdown of protein levels of Bim and its transcriptional regulator FOXO3a in both cell lines. Moreover, FOXO3a knockdown conferred marked protection against WA-mediated induction of Bim-s expression. The growth of MDA-MB-231 cells implanted in female nude mice was significantly retarded by 5 weekly i.p. injections of 4 mg WA/kg body weight. The tumors from WA-treated mice exhibited reduced cell proliferation and increased apoptosis compared with tumors from control mice. These results point toward an important role of FOXO3a and Bim in regulation of WA-mediated apoptosis in human bre


Studies Suggest Potent Anti-Cancer Activity

http://www.lef.org/magazine/mag2006/...t_ashwa_01.htm


In addition to ashwagandha’s documented neuroprotective effects, exciting recent evidence suggests that it also has the potential to stop cancer cells in their tracks. For example, a recent analysis showed that ashwagandha extract inhibited the growth of human breast, lung, and colon cancer cell lines in the laboratory. This inhibition was comparable to that achieved with the common cancer chemotherapy drug doxorubicin (Caelyx®, Myocet®). In fact, researchers reported that withaferin A, a specific compound extracted from ashwagandha, was more effective than doxorubicin in inhibiting breast and colon cancer cell growth.11,14
Scientists in India recently conducted cell studies showing that ashwagandha extract disrupts cancer cells’ ability to reproduce—a key step in fighting cancer. Additionally, laboratory analysis indicates that ashwagandha extract possesses anti-angiogenic activity, also known as the ability to prevent cancer from forming new blood vessels to support its unbridled growth. These findings lend further support to ashwagandha’s potential role in fighting cancer.15 Based on these studies, research in this area continues.
In another study, orally administered ashwagandha extract significantly inhibited experimentally induced stomach cancer in laboratory animals. Tumor incidence was reduced by 60% and tumor multiplicity (number) by 92%. Similarly, in a rodent model of skin cancer, ashwagandha inhibited tumor incidence and multiplicity by 45% and 71%, respectively.16 Ashwagandha’s protective effect against skin cancer has been shown in other studies as well.17
A recent experiment demonstrated that ashwagandha extract produced a marked increase in life span and a decrease in tumor weight in animals with experimentally induced cancer of the lymphatic system.18 This is an exciting finding, suggesting that ashwagandha could enhance survival in individuals with cancer.
Ashwagandha's Pharmacological Activity

Scientists speculate that some of ashwagandha’s benefits stem from its antioxidant properties and ability to scavenge free radicals.10
Two main classes of compounds—steroidal alkaloids and steroidal lactones—may account for its broad range of beneficial effects. Steroidal lactones comprise a class of constituents called withanolides. To date, scientists have identified and studied at least 12 alkaloids and 35 withanolides. Much of ashwagandha’s pharmacological activity has been attributed to two primary withanolides, withaferin A and withanolide D.11
Other studies reveal that ashwagandha has antimicrobial properties, with antibacterial activity against potentially dangerous bacteria, including Salmonella, an organism associated with food poisoning. This activity was demonstrated in cell cultures as well as in infected laboratory animals.12
Additional studies show that ashwagandha root extract enhances the ability of macrophage immune cells to “eat” pathogens, as compared to macrophages from a control group that did not receive ashwagandha.13
Ashwagandha extract may also have applications as an adjunct to cancer chemotherapy treatment. One of the consequences of chemotherapy is neutropenia, a decrease in white blood cells called neutrophils that can leave patients dangerously vulnerable to infection. A study of animals demonstrated that orally administered ashwagandha extract protected against this decline in infection-fighting neutrophils. While further human studies are needed, these findings suggest that ashwagandha may be an excellent adjunctive therapy to chemotherapy.19
Another animal study investigated ashwagandha extract’s effects in normalizing the immune-suppressing effects of chemotherapy. When test animals received a common chemotherapy drug, levels of the desirable immune factors interferon-gamma and interleukin-2 decreased.
When the animals also received orally administered ashwagandha extract, however, their immune system parameters remained normal. These findings add support to the idea that ashwagandha may help protect immune function during chemotherapy treatment.20



Bad with cyclophosphamide??:


J Ethnopharmacol. 1998 Oct;62(3):209-14.
Suppressive effect of cyclophosphamide-induced toxicity by Withania somnifera extract in mice.

Davis L, Kuttan G.
Professor Amala Cancer Research Centre, Amala Nagar P.O., Kerala, India.
Administration of Withania somnifera extract (Solanaceae) was found to significantly reduce leucopenia induced by cyclophosphamide (CTX) treatment. The total WBC count on the 12th day of the CTX-treated group was 3720 cells/mm3 and that of CTX along with Withania was 6120 cells/mm3. Treatment of Withania along with CTX was found to significantly (P < 0.001) increase the bone marrow cellularity (13.1 x 10(6) cells/femur) compared to CTX alone treated group (8 x 10(6) cells/femur). Administration of Withania extract increased the number of alpha-esterase positive cells (1130/4000 cells) in the bone marrow of CTX treated animals, compared to the CTX-alone treated group (687/4000 cells). The major activity of Withania somnifera may be the stimulation of stem cell proliferation. These studies indicate that Withania somnifera could reduce the cyclophosphamide induced toxicity and its usefulness in cancer therapy.

PMID: 9849630 [PubMed - indexed for MEDLINE]








Biochem Pharmacol. 2010 Feb 15;79(4):542-51.
Withaferin A targets heat shock protein 90 in pancreatic cancer cells.

Yu Y, Hamza A, Zhang T, Gu M, Zou P, Newman B, Li Y, Gunatilaka AA, Zhan CG, Sun D.
Department of Pharmaceutical Sciences, College of Pharmacy, The University of Michigan, 428 Church Street, Ann Arbor, MI 48109, USA.
The purpose of this study is to investigate the efficacy and the mechanism of Hsp90 inhibition of Withaferin A (WA), a steroidal lactone occurring in Withania somnifera, in pancreatic cancer in vitro and in vivo. Withaferin A exhibited potent antiproliferative activity against pancreatic cancer cells in vitro (with IC(50)s of 1.24, 2.93 and 2.78 microM) in pancreatic cancer cell lines Panc-1, MiaPaCa2 and BxPc3, respectively. Annexin V staining showed that WA induced significant apoptosis in Panc-1 cells in a dose-dependent manner. Western blotting demonstrated that WA inhibited Hsp90 chaperone activity to induce degradation of Hsp90 client proteins (Akt, Cdk4 and glucocorticoid receptor), which was reversed by the proteasomal inhibitor, MG132. WA-biotin pull down assay of Hsp90 using Panc-1 cancer cell lysates and purified Hsp90 showed that WA-biotin binds to C-terminus of Hsp90 which was competitively blocked by unlabeled WA. Co-immunoprecipitation exhibited that WA (10 microM) disrupted Hsp90-Cdc37 complexes from 1 to 24h post-treatment, while it neither blocked ATP binding to Hsp90, nor changed Hsp90-P23 association. WA (3, 6mg/kg) inhibited tumor growth in pancreatic Panc-1 xenografts by 30% and 58%, respectively. These data demonstrate that Withaferin A binds Hsp90, inhibits Hsp90 chaperone activity through an ATP-independent mechanism, results in Hsp90 client protein degradation, and exhibits in vivo anticancer activity against pancreatic cancer.

PMID: 19769945 [PubMed - in process]


Angiogenesis. 2004;7(2):115-22.
Withaferin A is a potent inhibitor of angiogenesis.

Mohan R, Hammers HJ, Bargagna-Mohan P, Zhan XH, Herbstritt CJ, Ruiz A, Zhang L, Hanson AD, Conner BP, Rougas J, Pribluda VS.
Discovery Research, EntreMed, Inc., Rockville, Maryland, USA. Royce.Mohan@uky.edu
The medicinal plant Withania somnifera is widely researched for its anti-inflammatory, cardioactive and central nervous system effects. In Ayurveda , the major Traditional Indian medicine system, extracts from W. somnifera are distinctively employed for the treatment of arthritis and menstrual disorders. Because these conditions involve angiogenic processes we hypothesized that the W. somnifera extracts might contain angiogenesis inhibitors. We employed an endothelial cell-sprouting assay to monitor the purification of substances from W. somnifera root extracts and isolated as the active principle the previously known natural product withaferin A. We show that withaferin A inhibits human umbilical vein endothelial cell (HUVEC) sprouting in three-dimensional collagen-I matrix at doses which are relevant to NF-kappa B-inhibitory activity. Withaferin A inhibits cell proliferation in HUVECs (IC50 =12 nM) at doses that are significantly lower than those required for tumor cell lines through a process associated with inhibition of cyclin D1 expression. We propose that the inhibition of NF-kappa B by withaferin A in HUVECs occurs by interference with the ubiquitin-mediated proteasome pathway as suggested by the increased levels of poly-ubiquitinated proteins. Finally, withaferin A is shown to exert potent anti-angiogenic activity in vivo at doses that are 500-fold lower than those previously reported to exert anti-tumor activity in vivo. In conclusion, our findings identify a novel mode of action of withaferin A, which highlights the potential use of this natural product for cancer treatment or prevention.

PMID: 15516832 [PubMed - indexed for MEDLINE]




http://www.raysahelian.com/ashwagandha.html

Ashwagandha, Withania somnifera, is an herb that is extensively used in Ayurveda, the traditional health care system in India. This herb is used as a general tonic and "adaptogen," helping the body adapt to stress. In addition, this herb has been shown to possess antioxidant activity as well as an ability to support a healthy immune system. Ashwagandha - also known as Indian Winter Cherry - is a shrub cultivated in India and North America whose roots have been used for thousands of years by Ayurvedic practitioners. The root contains flavonoids and many active ingredients of the withanolide class. Several studies over the past few years have looked into whether this herb has anti-inflammatory, anti-tumor, anti-stress, antioxidant, mind-boosting, immune-enhancing, and rejuvenating properties (see studies at bottom of page). Historically ashwagandha root has also been noted to have sex-enhancing properties. Many people notice a relaxing effect within hours of taking a 500 mg pill.

Anticarcinogenic activity of Withania somnifera Dunal against Dalton's Ascitic Lymphoma.
J Ethnopharmacol. 2004 Aug;93(2-3):359-61.
The effect of ethanolic extract of ashwagandha root against Dalton's Ascitic Lymphoma has been evaluated in Swiss albino mice. A significant increase in the life span and a decrease in the cancer cell number and tumour weight were noted in the tumour-induced mice after treatment with ashwagandha extract. These observations are suggestive of the protective effect of ashwagandha extract in Dalton's Ascitic Lymphoma.



http://www.ethnoleaflets.com/leaflets/withafer.htm
Conclusion: The studies so far indicate that W. somnifera could prove to be a good natural source of a potent and relatively safe radiosensitizer/chemotherapeutic agent.


http://www.drugs.com/npp/ashwaganda.html
Cancer
Withaferin A was first isolated as a cytotoxic agent, 3 and a considerable amount of investigation followed. The compound produced mitotic arrest in Ehrlich ascites carcinoma cells in vitro 31 while in vivo effects were mediated by macrophage activation. 26 , 32 Further investigations on peripheral blood lymphocytes determined that withaferin A destroyed spindle microtubules of cells in metaphase. 33

<
Despite a large volume of basic scientific studies on the plant, there is minimal evidence for a proper dose of this herb. A single study in which W. somnifera was the principal component of a polyherbal mixture administered 450 mg of root powder 4 times per day for arthritis. 49

The protective effect of a purified extract of Withania somnifera against doxorubicin-induced cardiac toxicity in rats 2007 LINK





www.raysahelian.com/silibinin.html
Silibinin is a flavonolignan found in certain herbs, particularly milk thistle

Acta Pharmacol Sin. 2009 Aug;30(8):1162-8. Epub 2009 Jul 6.
Silibinin inhibits prostate cancer invasion, motility and migration by suppressing vimentin and MMP-2 expression.

Wu KJ, Zeng J, Zhu GD, Zhang LL, Zhang D, Li L, Fan JH, Wang XY, He DL.
Department of Urology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an 710061, China.
AIM: Silibinin is known to exert growth inhibition and cell death together with cell cycle arrest and apoptosis in human prostate cancer cells. Whether silibinin could inhibit the invasion, motility and migration of prostate cancer cells remains largely unknown. This study was designed to evaluate this efficacy and possible mechanisms using a novel highly bone metastatic ARCaP(M) cell model. METHODS: Four prostate cancer cell lines, LNCaP, PC-3, DU145, and ARCaP(M), were used in this study. These cells were treated with increasing concentrations of silibinin (50, 100, and 200 micromol/L) for different periods of time. After treatment, cell viabilities of four prostate cancer cells were compared by MTT assay. Alterations of ARCaP(M) cell invasion, motility and migration were assessed by cell invasion, motility and wound healing assays. The changes of vimentin expression were observed by Western blotting and immunofluorescence staining, and the expression of MMP-2, MMP-9, and uPA was analyzed by reverse transcription-polymerase chain reaction (RT-PCR). RESULTS: ARCaP(M) cells showed less sensitivity to the growth inhibition of pharmacological doses of silibinin than LNCaP, PC-3, and DU145 cells. However, silibinin exerted significant dose- and time-dependent inhibitory effects on the invasion, motility and migration of ARCaP(M) cells. Furthermore, the expression of vimentin and MMP-2, but not MMP-9 or uPA, was down-regulated in a dose- and time-dependent manner after treatment of silibinin. CONCLUSION: This study shows that silibinin could inhibit the invasion, motility and migration of ARCaP(M) cells via down-regulation of vimentin and MMP-2 and therefore may be a promising agent against prostate cancer bone metastasis.

PMID: 19578386 [PubMed - indexed for MEDLINE]
> >
Clin Cancer Res. 2009 May 15;15(10):3557-67. Epub 2009 May 15.




Clin Cancer Res. 2008 Dec 1;14(23):7773-80.
Silibinin inhibits established prostate tumor growth, progression, invasion, and metastasis and suppresses tumor angiogenesis and epithelial-mesenchymal transition in transgenic adenocarcinoma of the mouse prostate model mice.

Singh RP, Raina K, Sharma G, Agarwal R.
Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado Denver, Denver, Colorado 80262, USA.
Comment in:
PURPOSE: The chronic nature of prostate cancer growth and progression leading to metastasis provides a large window for intervention. Herein, for the first time, we investigated the effect and associated mechanisms of silibinin phosphatidylcholine (silybin-phytosome) on established prostate tumors in transgenic adenocarcinoma of the mouse prostate (TRAMP) model. EXPERIMENTAL DESIGN: Twenty-week-old TRAMP male mice having palpable prostate tumor were fed with control or 0.5% and 1%, w/w, silybin-phytosome diets for 11 weeks and then sacrificed. RESULTS: Dietary silibinin inhibited the growth of prostate tumors (up to 60%, P < 0.001) and suppressed tumor progression from prostatic intraepithelial neoplasia to differentiated adenocarcinoma and poorly differentiated adenocarcinoma, with a complete absence of poorly differentiated adenocarcinoma at higher doses. It also inhibited the incidence of tumor invasion of seminal vesicle (up to 81%, P < 0.001) with complete absence of distant metastasis. Silibinin moderately inhibited tumor cell proliferation and induced apoptosis, but strongly suppressed tumor microvessel density (up to 60%, P < 0.001), vascular endothelial growth factor, and vascular endothelial growth factor receptor-2 expression. Antibody array analysis of plasma showed a decrease in the circulatory levels of vascular endothelial growth factor and basic fibroblast growth factor. Decreased levels of matrix metalloproteinases (MMP), snail-1, and vimentin, and an increased level of E-cadherin were also observed, indicating the anti-epithelial-mesenchymal transition effect of silibinin in tumors. CONCLUSIONS: Overall, silibinin treatment of TRAMP mice bearing prostate tumor inhibited tumor growth, progression, local invasion, and distant metastasis involving suppression of tumor angiogenesis and epithelial-mesenchymal transition. These findings would have greater relevance for the ongoing phase II clinical trial with silibinin-phytosome in prostate cancer patients.

PMID: 19047104 [PubMed - indexed for MEDLINE]




Nuclear factor-kappaB-mediated transforming growth factor-beta-induced expression of vimentin is an independent predictor of biochemical recurrence after radical prostatectomy.

Zhang Q, Helfand BT, Jang TL, Zhu LJ, Chen L, Yang XJ, Kozlowski J, Smith N, Kundu SD, Yang G, Raji AA, Javonovic B, Pins M, Lindholm P, Guo Y, Catalona WJ, Lee C.
Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA. q-zhang2@northwestern.edu .
PURPOSE: Transforming growth factor-beta (TGF-beta)-mediated epithelial-to-mesenchymal transition (EMT) has been shown to occur in some cancers; however, the pathway remains controversial and varies with different cancers. In addition, the mechanisms by which TGF-beta and the EMT contribute to prostate cancer recurrence are largely unknown. In this study, we elucidated TGF-beta-mediated EMT as a predictor of disease recurrence after therapy for prostate cancer, which has not been reported before. EXPERIMENTAL DESIGN: We analyzed TGF-beta-induced EMT using nuclear factor-kappaB (NF-kappaB) as an intermediate mediator in prostate cancer cell lines. A total of 287 radical prostatectomy specimens were evaluated using immunohistochemistry in a high-throughput tissue microarray analysis. Levels of TGF-beta signaling components and EMT-related factors were analyzed using specific antibodies. Results were expressed as the percentage of cancer cells that stained positive for a given antibody and were correlated with disease recurrence rates at a mean of 7 years following radical prostatectomy. RESULTS: In prostate cancer cell lines, TGF-beta-induced EMT was mediated by NF-kappaB signaling. Blockade of NF-kappaB or TGF-beta signaling resulted in abrogation of vimentin expression and inhibition of the invasive capability of these cells. There was high risk of biochemical recurrence associated with tumors that displayed high levels of expression of TGF-beta1, vimentin, and NF-kappaB and low level of cytokeratin 18. This was particularly true for vimentin, which is independent of patients' Gleason score. CONCLUSIONS: The detection of NF-kappaB-mediated TGF-beta-induced EMT in primary tumors predicts disease recurrence in prostate cancer patients following radical prostatectomy. The changes in TGF-beta signaling and EMT-related factors provide novel molecular markers that may predict prostate cancer outcomes following treatment.







Vimentin inhibitors:


http://www.faqs.org/patents/app/20090137473

Patent application title:
INHIBITION OF MICROTUBULE PROTRUSION IN CANCER CELLS
Inventors: Stuart Martin Rebecca Whipple-Bettes
Agents: FULBRIGHT & JAWORSKI, LLP
Assignees:
Origin: HOUSTON, TX US
IPC8 Class: AA61K3817FI
USPC Class: 514 12



Read more: http://www.faqs.org/patents/app/2009...#ixzz0c0JwIB7l

Abstract:


The present invention generally concerns microtubule protrusions in cancer cells, including detached cancer cells, and inhibition of the protrusions. In particular aspects, the inhibition of the protrusions interferes with attachment of the cell to a vessel wall, and in further aspects the cell is killed by forcing it to enter capillaries and be destroyed, for example by shearing. Inhibition by a variety of agents and methods is contemplated.

In embodiments of the invention, one or more components of a microtubule protrusion are inhibited, directly or indirectly. In particular aspects of the invention, the components comprise Glu-tubulin, kinesin and/or vimentin. Therefore, in specific embodiments of methods of the invention, one targets Glu-tubulin, kinesin and/or vimentin to inhibit one or more microtubule protrusions, for example from a cancer cell. In certain embodiments, the protrusions may be targeted by inhibiting Glu-tubulin, such as by one or more of the following, for example: 1) increasing activity and/or expression of tubulin tyrosine ligase (TTL), which postranslationally converts Glu-tubulin back to Tyr-tubulin; 2) inhibition of a carboxypeptidase that produces Glu-tubulin from Tyr-tubulin, such as by delivery of okadaic acid, 3-nitrotyrosine, 1-nor-okadone, Cantharidin, Phoslactomycin B (Fostriecin), DL-benzylsuccinic acid, Sodium orthovanadate, or a combination thereof, and in specific embodiments the carboxypeptidase is hAGBL3 (Rodriguez de la Vega et al., 2007). In certain embodiments, the protrusions may be targeted by inhibiting vimentin, such as by one or more of the following, for example: a dominant negative mutant, such as R113C and/or .DELTA.C2B mutants; antisense or siRNA; and/or calyculin-A. In other embodiments, the protrusions may be targeted by inhibiting kinesin, such as by one or more of the following, for example: siRNA, including KIF5b siRNA, for example, lidocaine, and/or tetracaine.







Related?:

Cancer Cell. 2008 Sep 9;14(3):263-73.
Rapid chemotherapy-induced acute endothelial progenitor cell mobilization: implications for antiangiogenic drugs as chemosensitizing agents.

Shaked Y, Henke E, Roodhart JM, Mancuso P, Langenberg MH, Colleoni M, Daenen LG, Man S, Xu P, Emmenegger U, Tang T, Zhu Z, Witte L, Strieter RM, Bertolini F, Voest EE, Benezra R, Kerbel RS.
Molecular and Cellular Biology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada. yshaked@tx.technion.ac.il
Comment in:
Several hypotheses have been proposed to explain how antiangiogenic drugs enhance the treatment efficacy of cytotoxic chemotherapy, including impairing the ability of chemotherapy-responsive tumors to regrow after therapy. With respect to the latter, we show that certain chemotherapy drugs, e.g., paclitaxel, can rapidly induce proangiogenic bone marrow-derived circulating endothelial progenitor (CEP) mobilization and subsequent tumor homing, whereas others, e.g., gemcitabine, do not. Acute CEP mobilization was mediated, at least in part, by systemic induction of SDF-1alpha and could be prevented by various procedures such as treatment with anti-VEGFR2 blocking antibodies or paclitaxel treatment in CEP-deficient Id mutant mice, both of which resulted in enhanced antitumor effects mediated by paclitaxel, but not by gemcitabine.

PMID: 18772115


Especially interesting since Paclitaxel is an antimitotic, Gemcitabine is an antimetabolite.

But can metronomic approach reduce this?:

Maximum Tolerable Dose and Low-Dose Metronomic Chemotherapy Have Opposite Effects on the Mobilization and Viability of Circulating Endothelial Progenitor Cells1

http://cancerres.aacrjournals.org/cg...ull/63/15/4342

Francesco Bertolini2, Saki Paul, Patrizia Mancuso, Silvia Monestiroli, Alberto Gobbi, Yuval Shaked and Robert S. Kerbel Division of Hematology-Oncology, Department of Medicine [F. B., S. P., P. M.] and Department of Experimental Oncology, IFOM-Fondazione Italiana per la Ricerca sul Cancro Institute of Molecular Oncology [S. M., A. G.], European Institute of Oncology, 20141 Milan, Italy; Molecular and Cell Biology Research, Sunnybrook and Women’s College Health Sciences Centre, Department of Medical Biophysics, University of Toronto, Toronto, Ontario, M4N 3M5 Canada [Y. S., R. S. K.]
There is growing evidence that vasculogenesis (progenitor cell-derived generation of new blood vessels) is required for the growth of some neoplastic diseases. Here we show that the administration of cyclophosphamide (CTX) at the maximum tolerable dose with 21-day breaks or at more frequent low-dose (metronomic) schedules have opposite effects on the mobilization and viability of circulating endothelial progenitors (CEPs) in immunodeficient mice bearing human lymphoma cells. Animals treated with the maximum tolerable dose CTX experienced a robust CEP mobilization a few days after the end of a cycle of drug administration, and tumors rapidly became drug resistant. Conversely, the administration of metronomic CTX was associated with a consistent decrease in CEP numbers and viability and with more durable inhibition of tumor growth. Our findings suggest that metronomic low-dose chemotherapy regimens are particularly promising for avoiding CEP mobilization and, hence, to potentially reduce vasculogenesis-dependent mechanisms of tumor growth.


To Evade Chemotherapy, Some Cancer Cells Mimic Stem Cells

http://www.aacr.org/home/public--med...ses.aspx?d=822

Using the CellSearch System techique that quantifies circulating tumor cells, researchers had shown that chemotherapy with Taxol causes a massive release of cells into the circulation, while at the same time reducing the size of the tumor, explaining that complete pathologic responses do not correlate well with improvements in survival.

Circulating tumor cells (CTCs) are cancer cells that have detached from solid tumors and entered the blood stream. This can begin the process of metastasis, the most life-threatening aspect of cancer. To metastasize, or spread cancer to other sites in the body, CTCs travel through the blood and can take root in another tissue or organ.

In this stem cell research, anti-cancer treatments often effectively shrink the size of tumors, but some might have the opposite effect, actually expanding the small population of cancer stem cells that then are capable of metastasizing.

Even before the advent of the CellSearch technique, it had been observed in "cell death" cell culture assays, that there was an increase in the number of metabolic activity of mitochondria of the surviving cells from Taxol therapy, even in cases where the majority of the cells are being killed by Taxol. It may indeed give clinical response (tumor shrinkage), however, these are mostly short-lived and relapses after a response are often dramatic.



To evade chemotherapy, some cancer cells mimic stem cells

September 2007 ATLANTA - Anti-cancer treatments often effectively shrink the size of tumors, but some might have an opposite effect, actually expanding the small population of cancer stem cells believed to drive the disease, according to findings presented today in Atlanta, Georgia at the American Association for Cancer Research's second International Conference on Molecular Diagnostics in Cancer Therapeutic Development.

"Our experiments suggest that some treatments could be producing more cancer stem cells that then are capable of metastasizing, because these cells are trying to find a way to survive the therapy," said one of the study's investigators, Vasyl Vasko, M.D. Ph.D., a pathologist at the Uniformed Services University of the Health Sciences in Bethesda, Md.

"This may help explain why the expression of stem cell markers has been associated with resistance to chemotherapy and radiation treatments and poor outcome for patients with cancers including prostate, breast and lung cancers," Dr. Vasko said. "That tells us that understanding how to target these markers and these cells could prove useful in treating these cancers."

The cancer stem cell markers include Nanog and BMI1, both of which contribute to stem cells' defining ability to renew themselves and differentiate into different cell types, Dr. Vasko said. These same molecules are found in embryonic stem cells.

Researchers have recently debated the notion that some therapies are not capable of eradicating cancer because they do not target the cancer stem cells responsible for tumor development. To test this hypothesis, Dr. Vasko, along with scientists from the CRTRC Institute for Drug Development in San Antonio and from the Johns Hopkins University, set out to measure both stem cells markers and tumor volume before and after treatment in a mouse model.

They selected a rare form of cancer, mesenchymal chondrosarcoma (MCS), which has not been well described and for which there is no effective treatment. The researchers first determined that Nanog and BMI1 stem cell markers were more highly expressed in metastatic tumors compared to primary tumors. "This suggests that expression of the marker plays some role in development of metastasis," Dr. Vasko said.

They then applied various therapies - from VEGF inhibitors such as Avastin to the proteasome inhibitor Velcade - in mice implanted with human MSC, and analyzed the effects on tumors. Some of the treatments seemed to work, because they led to a dramatic decrease in the size of the tumors, Dr. Vasko said. But analysis of stem cell expression before and after treatment revealed that even as some anti-cancer treatments shrank tumors, they increased expression of Nanog and BMI1. "These treatments were not enough to completely inhibit tumor growth, and the cancer stem cell markers were still present," Dr. Vasko said.

Use of the agents Velcade and Docetaxel led to the most significant increase in stem cell markers within the treated tumor, while ifosfamide and Avastin inhibited expression of the markers in this cancer subtype.

"We hypothesize that the tumor escapes from chemotherapy by induction of stem cell marker expression," he said. "The small number of cells that survive the treatment could then generate another tumor that metastasizes."

Dr. Vasko doesn't know how this happens, but theorizes that "dying cells could secrete a lot of factors that induce expression of stem cell markers in other cancer cells. I think they are trying to survive and they use a mechanism from their experience of embryonic life."

American Association for Cancer Research. September 2007.
Rich66 is offline   Reply With Quote
Old 07-14-2009, 04:51 PM   #2
Laurel
Senior Member
 
Laurel's Avatar
 
Join Date: May 2008
Location: Hershey, PA. Live The Sweet Life!
Posts: 2,005
Okay, not liken that.....
__________________

Smile On!
Laurel


Dx'd w/multifocal DCIS/IDS 3/08
7mm invasive component
Partial mast. 5/08
Stage 1b, ER 80%, PR 90%, HER-2 6.9 on FISH
0/5 nodes
4 AC, 4 TH finished 9/08
Herceptin every 3 weeks. Finished 7/09
Tamoxifen 10/08. Switched to Femara 8/09
Bilat SPM w/reconstruction 10/08
Clinical Trial w/Clondronate 12/08
Stopped Clondronate--too hard on my gizzard!
Switched back to Tamoxifen due to tendon pain from Femara

15 Years NED
I think I just might hang around awhile....

Laurel is offline   Reply With Quote
Old 07-14-2009, 05:09 PM   #3
Rich66
Senior Member
 
Rich66's Avatar
 
Join Date: Feb 2008
Location: South East Wisconsin
Posts: 3,431
GDPawel's related points on Taxol:

Is Taxol the Vioxx of Cancer Drugs?In the last decade, the incidence of central nervous system (CNS) metastasis has increased. The very first reference I found of this was a NCI observational study in 1995 that reported experience in their clinic where recurrent systemic disease occurred in all patients for which they received dose-intense Paclitaxel (Taxol) therapy. Brain metastasis was the only site of disease recurrence. The cerebellum was involved in two out of three patients, presenting with headache, dizziness, unsteady gait, nausea and vomiting (all the things that happened to my wife in 1998, after her adjunct Taxol treatment in 1997).

This is what led me to research this further and found out about the rarity of ovarian cancer cells metastasizing to the brain. Ovarian cancer uncommonly involves the nervous system. Brain metastasis was a "rare" complication of ovarian cancer with only 67 well-documented cases in medical literature, until 1994. A multi-ins***utional study of 4027 ovarian cancer patients over 30 years identified only 32 cases while an autopsy study of ovarian cancer reported an incidence of 0.9%. Even more "rare" is the occurrance of Carcinomatous Meningitis. Until 1994, there have been only 14 cases reported. This presentation is similar to metastases from other solid tumors (breast, lung). (1)

In 2002, I came across a study by Christos Kosmas, M.D., consultant medical oncologist, Department of Medicine and Medical Oncology Unit at Helena-Venizelou Hospital, Athens, Greece en***led, "Carcinomatous Meningitis: Taxane-Induced," which found what is called "dissemination after taxane-based (Taxol) chemotherapy." The study conclusions stated that Carcinomatous Meningitis (a CNS metastasis) after a major response to front-line taxane-based regimens represents a grave disease manifestation and its incidence appears increased when compared retrospectively to non-taxane-treated patients. (2)

A commentary by Dr. Lawrence N. Shulman, Vice Chair for Clinical Services and Adult Oncology, Dana-Farber Cancer Ins***ute, Harvard Medical School, Boston, in the September, 2002 issue of The American Journal of Oncology Review, describes the complete lack of progress in the chemothe****utic treatment of metastatic breast cancer since 1970. Dr. Shulman noted that a retrospective com*****ion of a well-characterized "standard-dose" database with a less well-characterized "high-dose" database suggested that there was increased early mortality for "high-dose" therapy. (3)

An editorial by Drs. V. Valero and G.N. Hortobagyi in the March 15, 2003 issue of the Journal of Clinical Oncology, reviewed all of the large, prospective, randomized trials published comparing taxane-based chemotherapy regimens. They conclude that none of these regimens have increased either complete response rates or overall survival, with median survivals remaining at two years or less, or precisely the same results which were being obtained over thirty years ago. (4)

In 2004, as reported at the 27th Annual San Antonio Breast Cancer Symposium, using a technique that quantifies circulating tumor cells, German investigators from Friedrich-Schiller University in Jena, have shown that neoadjuvant chemotherapy with paclitaxel (taxol) causes a massive release of cells into the circulation, while at the same time reducing the size of the tumor. The finding could help explain the fact that complete pathologic responses do not correlate well with improvements in survival.

In the study, according to Katharina Pachmann, M.D., professor of experimental oncology and hematology, breast cancer patients undergoing neoadjuvant chemotherapy gave blood samples in which epithelial antigen-positive cells were isolated. Such cells are detected in most breast cancer patients but are rarely found in normal subjects. The investigators measured the levels of cirulating tumor cells before and during primary chemotherapy with several different cytotoxic agents.

Paclitaxel (taxol) produces the greatest degree of tumor shrinkage but also the greatest release of circulating tumor cells. In three different paclitaxel-containing regimens, circulating cell numbers massively increased, whereas tumor size decreased. These cells remained in the circulation for at least five months after surgery.

The tumor shrinks, but more cells are found in the circulation. This corresponds with a high pathologic complete response during paclitaxel treatment, but in the end, this is not reflected in improved survival. These cells are alive in the circulation. What this study has shown, so far, that in three different paclitaxel (taxol) containing regimens, as the tumor collapses (a clinical response, not cure), it produces the greatest release of circulating tumor cells. The study has not looked at any other combination regimens. (5)

The results of these kinds of study are coming out slowly and quietly (now that Taxol is off-patent) and indicate that taxol containing regimens didn't prolong survival over other more conventional and less expensive cytotoxic drugs. Even before the advent of the CellSearch technique, it had been observed in various "cell death" assays, that there was an increase in the number of metabolic activity of mitochondria of the surviving cells from taxane therapy, even in cases where the majority of the cells are being killed by taxanes. It may indeed give clincial response (tumor shrinkage), sometimes impressive, however, these are mostly short-lived and relapses after a response to taxanes (Taxol) are often dramatic. (6)

With these cells being alive in the circulation, it may mean that a patient with invasive breast cancer without lymph node involvement (where systemic treatment "may" benefit), or a patient with invasive breast cancer that involves lymph nodes (where systemic treatment is "usually" recommended), would need additional (anti-estrogen) treatment, such as Tamoxifen (it may be given alone or in addition to chemotherapy, if given).

It has been shown that Tamoxifen treatment will reduce circulating tumor cells in some patients, but not all. So they develop a drug called Herceptin. Why? It has been shown that Herceptin treatment will reduce circulating tumor cells in patients with HER2-negative tumors, but less pronounced in HER2-positive tumors.

Does Herceptin really work on these circulating tumor cells? A study from the Dana Farber Cancer Ins***ute identified central nervous system metastases in women who receive trastuzumab-based (Herceptin) therapy for metastatic breast carcinoma. Central nervous system disease is defined as one or more brain metastases or leptomeningeal carcinomatosis (carcinomatous meningitis).

Central nervous system metastases was identified in 34% of patients at a median of 16 months after diagnosis of metastatic breast cancer and 6 months from the beginning of Herceptin treatment. Patients receiving Herceptin as first-line therapy for metastatic disease frequently developd brain metastases while responding to or stable on Herceptin. (7)

In 2006, another report that CNS relapses are common among breast cancer patients treated with a taxane-based chemotherapy regimen. Central Nervous System Relapse in Patients With Breast Cancer Is Associated With Advanced Stages, With CK-19 mRNA-positive Circulating Occult Tumor Cells and With Her2/neu-positive tumors.

During the past years it has been frequently observed that patients with breast cancer treated with a taxane-containing chemotherapy regimen, either in the adjuvant setting or in the metastatic setting, presenting central nervous system (CNS) involvement as the only evidence of disease progression. More studies were therefore interested to evaluate the incidence of CNS metastases in patients with early and advanced breast cancer treated with a taxane-containing chemotherapy regimen and to identify predictive factors for CNS relapse.

Recent studies reported that breast cancer patients who received a taxane-containing chemotherapy regimen had a significantly higher incidence of CNS metastases compared with that of patients treated with a nontaxane-containing regimen. There are also data indicating an increased risk for brain metastases in breast cancer patients receiving trastuzumab (Herceptin).

In the present study it was also possible to confirm the initial clinical observation that breast cancer patients who receive a taxane-containing chemotherapy regimen have a significantly higher incidence of CNS metastases compared with that of patients treated with a nontaxane-containing regimen.

The reasons for the association between treatment of breast cancer with a taxane-containing chemotherapy regimen and an increased incidence of CNS involvement could be that taxanes are very lipophilic, their concentration in the CNS is very low after their intravenous administration. Taxanes are unable to penetrate the intact blood-brain barrier, the concentration of radiolabeled paclitaxel in the cerebrospinal fluid is found to be significantly lower than in other organs, and thus undetectable in the brain, in the spinal cord or in any other site of the CNS.

Also, paclitaxel is exported from the p-glycoprotein and other ATP-binding cassette transporters placed at the luminal membrane of brain capillaries, as an explanation for the low concentrations of taxanes in the CNS.

Furthermore, the detection of cytokeratin 19 (CK-19) and of mRNA-positive circulating tumor cells (CTCs) in the peripheral blood and the bone marrow of patients with breast cancer is correlated with increased incidence of relapse.

The aforementioned data suggest that taxanes may not penetrate well into the CNS, and therefore the CNS may represent tumor 'sanctuary' sites for taxane-containing chemotherapy regimens. A difference in the incidence of CNS relapses between patients with breast cancer and other solid tumors treated with taxanes was observed. (8)

The percentage of patients that must respond to a drug before it is approved by the FDA varies from as high as 80% to as low as 20%. Thereafter, it is used routinely for all patients with the same form of cancer, though unfortunately a drug that helps one person does not necessarily mean it will help all patients with the same diagnosis. The response rate for Taxol for FDA approval was 30%.

Taxol (Paclitaxel) is known as a taxane type of chemotherapy drug. Taxol is given into a vein, but in order for the body to absorb the drug, it must first be dissolved in a solution. The compound wouldn't dissolve very much in any solution. It was discovered that something Taxol would dissove in the might work in a reasonably safe intravenous solution in humans. It was an elixir made of castor oil and marketed as Cremophor EL. It was the only answer. However, this castor-oil carrier is suspected as the culprit behind the misery which includes nausea, vomiting, joint pain, appe***e loss, brittle hair and tingling sensations in hands and feet (neuropathy). (9)

The American Cancer Society has mentioned that the solution can cause dangerous allergic reactions in many people, so patients "must" first take other drugs like steroids and antihistamines in "hopes" to prevent a bad reaction. The solution can also leach chemicals from regular plastic tubes used to deliver medication, so Taxol must be given through special tubing.

A new drug for breast cancer (Abraxane) is a new form of Taxol (Paclitaxel). Abraxane does not need to be dissolved in the castor oil solution and does not require special equipment to be given to patients. However, more of the women on Abraxane had numbness and tingling in their hands and feet. And more suffered nausea and vomiting, diarrhea, muscle and joint pain and anemia.

Taxol (Paclitaxel) is an extremely potent chemotherapy drug, often producing a number of side effects in patients. Side effects of Taxol (Paclitaxel) include severe allergic reactions, cardiovascular problems (such as changes in blood pressure), infections developing from white blood cell deficiencies, complete hair loss (apolecia), joint and muscle pain, irritation at the Taxol and other chemotherapy drugs injection site, low red blood cell count, mouth or lip sore, numbness or burning in the hands and feet, and stomach upset/diarrhea. (10)

There is a molecular basis for the peripheral pain caused by Taxol. It appears to be caused when the drug binds to a protein and initiates improper calcium signaling, researchers at Yale School of Medicine reported in a study published in the Proceedings of the National Academy of Sciences. This response leads to side effects such as acute hypersensitivity, slower heart rhythms, tingling, numbness, and other symptoms. These serious side effects limit the drug's effectiveness. Peripheral pain becomes worse with continued use and increased dosages lead to persistent and irreversible pain.

The binding protein is called neuronal calcium sensor (NCS-1). When paclitaxel (taxol) binds to NCS-1, it makes the cell more sensitive to normal signals and increases the magnitude and frequency of changes in calcium. Over time, increased calcium levels activate an enzyme (calpain) that degrades proteins, especially NCS-1. Calcium signals are needed for nerves to be stimulated and to respond and the loss of NCS-1 makes it more difficult to generate any calcium signals. While the loss of NCS-1 stops the protein interaction that is causing the inappropriate calcium signals, it also decreases the ability to have normal responses. (11)

Sources:

(1) National Cancer Ins***ute
(2) American Journal Clinical Oncology 2002;63:6-15
(3) Am J Oncology Rev 1(3):169-170, '02
(4) J Clin Oncol 21(6): 959-962, '03
(5) Oncol News Int'l, Vol 14, #5, May '05
(6) Cell Function ****ysis
(7) Cancer 2003 Jun 15;97(12):2972-7
(8) Breast Cancer Res. 2006;8(4)2006 BioMed Central, Ltd.
(9) Office of Research, Florida State University
(10) Bionumerik
(11) PNAS 104: 11103-11108 June 20, 2007


The original article about Pachmann's findings(mentioned above), a follow up by Fehm et al which makes the distinction regarding what type of cells are released and relevance, and further commentary on the distinctions by Pachmann:



CH321 • 03/29/05 • Scheduled for MAY 2005
Neoadjuvant Chemotherapy for Breast Cancer Can Cause Release of Tumor Cells

SAN ANTONIO—Using a technique that quantifies circulating tumor cells, German investigators have shown that neoadjuvant chemotherapy with paclitaxel causes a massive release of cells into the circulation, while at the same time reducing the size of the tumor.
Reporting her findings at the 27th Annual San Antonio Breast Cancer Symposium (abstract 6014), Katharina Pachmann, MD, professor of experimental oncology and hematology, Friedrich-Schiller University, Jena, Germany, said that the impact of these released cells on relapse is not clear. The finding could help explain the fact that complete pathologic responses do not correlate well with improvements in survival.
“During the applied combination therapy, three different phases could be observed,” she said. “An initial decline in the number of circulating cells during the epirubicin (Ellence)-containing part of the regimen, followed by a steep increase during paclitaxel treatment, and a subsequent re-decrease if a third segment with CMF cyclophosphamide/methotrexate/fluorouracil) was administered before surgery.”

Ironically, she said, paclitaxel produces the greatest degree of tumor shrinkage but also the greatest release of circulating tumor cells. In three different paclitaxel containing regimens, with five to eight patients in each group, circulating cell numbers massively increased whereas tumor size decreased. These cells remained in the circulation for at least 5 months after surgery, Dr. Pachmann reported.
“The tumor collapses, but we find more cells in the circulation. This corresponds with a high pathologic complete response during paclitaxel treatment, but in the end, this is not reflected in improved survival,” she said. “These cells are alive in the circulation, and they would be accessible to an additional treatment, such as tamoxifen. We have shown that tamoxifen treatment will reduce circulating tumor cells in some patients but not all. In this study, the patients received no further treatment after surgery.”

She noted that the initial decrease in circulating tumor cells correctly predicted final tumor reduction in patients with HER2-negative tumors, but this correlation was less pronounced in HER2-positive patients who additionally received trastuzumab (Herceptin).



Presence of apoptotic and nonapoptotic disseminated tumor cells reflects the response to neoadjuvant systemic therapy in breast cancer

Tanja Fehm1 , Sven Becker1, Graziella Becker-Pergola1, Karl Sotlar2, Gerhard Gebauer3, Silke Dürr-Störzer1, Hans Neubauer1, Diethelm Wallwiener1 and Erich-Franz Solomayer1
1Department of Obstetrics and Gynecology, University of Tuebingen, Calwerstrasse 7, D-72076 Tuebingen, Germany
2Department of Pathology, University of Tuebingen, Calwerstrasse 7, D-72076 Tuebingen, Germany
3Department of Gynecology and Obstetrics, University of Heidelberg, Vossstrasse 9, 69115 Heidelberg, Germany


Breast Cancer Research 2006, 8:R60doi:10.1186/bcr1611
The electronic version of this article is the complete one and can be found online at: http://breast-cancer-research.com/content/8/5/R60

............

Patients with complete remission of the primary tumor have a better clinical outcome compared with those with partial remission or compared with nonresponders [5,6]. Local therapy response is normally assessed by palpation and imaging techniques including ultrasound and mammography. The efficacy of neoadjuvant treatment on disseminated tumor cells (DTC) is assumed to be correlated to local therapy response. Complete remission is considered a surrogate marker for complete eradication of micrometastatic disease. However, 13–25% of patients with pathological complete response develop metastatic disease over 5 years [1,5-8].
Eradication of a minimal residual disease may be monitored by serial bone marrow analysis during NST, offering new insights into the effects of systemic therapy on minimal residual disease. DTC can be detected in 30–40% of untreated primary breast cancer patients prior to surgery [9-12]. Tumor cell persistence after removal of the primary tumor and adjuvant chemotherapy has been suggested to indicate chemotherapy resistance and poor clinical outcome [13-15]. The efficacy of neoadjuvant chemotherapy may therefore be indicated by bone marrow negativity or by the presence of disseminated apoptotic cells that were susceptible to cytotoxic agents.


...........


Response of the primary tumor is reflected by bone marrow status

DTC were detected in 53% of patients after completion of neoadjuvant therapy. The highest positivity rate was observed in patients with progressive disease (80%), followed by patients with stable disease (68%) and those with partial remission (48%). Only 10 of 24 (42%) patients with pathological complete remission were DTC positive. The response of the primary tumor was therefore reflected by the presence of DTC. Interestingly, the overall incidence was higher compared with the preoperative positivity rates reported in primary breast cancer patients prior to surgery and not treated by chemotherapy, ranging from 30% to 50% [9-12]. Most of the patients, however, had initially advanced stages with nodal involvement. In general, these patients tend to have higher positivity rates in the bone marrow. In addition, most of the patients still have tumor left despite neoadjuvant chemotherapy. It has been shown that regressing tumors still shed tumor cells into the blood circulation to the same extent as progressing tumors [21]. Therefore, even if patients respond to chemotherapy, cells may be still shed into the bone marrow until removal of the primary tumor. Most of these cells may have no relevance and undergo apoptosis induced by cytotoxic agents.

Presence of apoptotic cells in bone marrow after systemic treatment


Apoptosis is programmed cell death, which represents a signaling pathway that leads to cellular suicide in an organized manner. Apoptosis is fundamentally different from the necrotic mode of cell death, in which the cells suffer a major insult, resulting in a loss of membrane integrity, swelling and disrupture of the cell. The gold standard for detection of apoptosis has been morphological assessment either with electronic microscopy or with light microscopy [22-28]. Cells are characterized by specific morphologic signs of celllular shrinkage, membrane blebbing (cleavage of cytoskeletal proteins leading to blebbing), nuclear condensation and fragmentation [26]. The alteration of the cytoskeleton takes place first [42] and the intact intermediate filament network is then replaced by cytokeratin inclusions (Figure 1c). The final stages are apoptotic bodies (Figure 1c,d) [28].
Given that apoptosis is the principle mechanism of chemotherapy-induced regression [29], we analyzed the bone marrow aspirates for the presence of apoptotic DTC by immunocytochemical staining with M30 antibody. This antibody reacts with a caspase cleaved epitope of cytokeratin 18 expressed during early apoptosis [18].
No apoptotic DTC could be detected in the bone marrow of patients with progressive disease, whereas the positivity rate of only apoptotic DTC was 4% in patients with stable disease versus 14% in patients with partial remission or complete remission. The presence of only apoptotic DTC was therefore reflected by the therapy response. A subset of patients with stable disease, partial remission or complete remission had both apoptotic cells and nonapoptotic cells, reflecting heterogeneity in terms of therapy responsiveness also observed in the primary tumor [30].

Clinical relevance of disseminated tumor cells in patients with complete remission


The most interesting group of patients to elucidate the biology of chemoresistance and metastatic disease are those with complete remission of the primary tumor and persisting tumor cells in the bone marrow. The aim of (neo)adjuvant chemotherapy is to eradicate micrometastatic disease to improve disease-free survival and overall survival. Theoretically, if complete remission indicates complete eradication of metastatic disease, no patients with complete remission should have disseminated DTC or should develop metastatic disease during the course of disease. Several studies have shown, in fact, that patients without DTC have the best outcome. However, 13–25% of these patients suffer from systemic relapse. Since no primary tumor is left, one of the sources of metastatic cells is likely to be in the bone marrow. In our study, nonapoptotic DTC could be detected in 10 out of 24 (42%) patients with complete remission. Similar positive rates of tumor cell persistence have also been observed in breast cancer patients after surgery and completion of adjuvant therapy [13-15]. These DTC have the capability to survive in the blood circulation despite cell-matrix loss and are apparently resistant to systemic therapy. Two important steps of the metastatic cascade are therefore fulfilled [21]. Since it has already been demonstrated that patients with persistent DTC have a poor prognosis [13-15], determination of the bone marrow status after neoadjuvant chemotherapy might identify those patients at a high risk for metastatic disease and identify patients who may need secondary therapy.
Clearly not all disseminated cells may necessarily be cells with metastatic potential, however, since the positivity rate of 42% is higher than the expected relapse rate for patients with complete remission, ranging from 13 to 25%. Meng and colleagues [31] have shown that circulating tumor cells can be detected in the blood of breast cancer patients more than 20 years after primary diagnosis without evidence for recurrence, supporting the theory of (lifelong) tumor cell dormancy. An important task will therefore be the identification of those persistent and temporarily dormant cells that will cause a metastatic disease. As a very provocative theory it has been suggested that this subpopulation of persistent DTC may also be seen as cancer stem cells since they are dormant but have the ability to regrow following treatment [32,33]. Identification and further characterization of this subset will offer the chance to understand the mechanism of tumor cell growth and metastatic disease. Furthermore, development of new drugs based on these results will optimize treatment strategies since these disseminated cells are directly linked to metastatic spread.

Conclusion


The presence of apoptotic tumor cells in bone marrow is reflected by therapy response of the primary tumor to systemic therapy. Patients with complete response, however, still may have nonapoptotic DTC, indicating that the response of the primary tumor does not necessarily reflect the therapeutic effect on DTC. Even patients with complete remission but positive bone marrow status after primary systemic therapy may therefore need secondary adjuvant therapy, which may be based on bisphosphonates or antibody-based strategies, to completely eradicate minimal residual disease.




Comments(1)

Presence of apoptotic and nonapoptotic disseminated tumor cells reflects the response to neoadjuvant systemic therapy in breast cancer

Tanja Fehm, Sven Becker, Graziella Becker-Pergola, Karl Sotlar, Gerhard Gebauer, Silke Dürr-Störzer, Hans Neubauer, Diethelm Wallwiener and Erich-Franz Solomayer
Breast Cancer Research 2006, 8:R60doi:10.1186/bcr1611


Release of tumor cells during neoadjuvant therapy

Katharina Pachmann (01 November 2006) Friedrich Schiller University of Jena, Germany
The present report by Dr. Fehm confirms our previous results about dissemination of tumor cells during the course of neoadjuvant therapy presented already at the 2004 San Antonio Breast cancer symposium (Pachmann K., Camara O., Pachmann UA. Influence of primary tumor chemotherapy in breast cancer on circulating tumor cells. Indications for massive cell release into circulations concurrent with tumor size reduction. (2004) Breast Cancer Research and Treatment 88: S224), which was commented in several publications: "An article in the May 2005 issue of Oncology News International (Vol. 14, No. 5) reports that neoadjuvant chemotherapy* may cause the release of cancer cells into the blood. Katharina Pachmann, MD, of Friedrich-Schiller University in Germany said that "ironically, paclitaxel produces the greatest degree of tumor shrinkage but also the greatest release of circulating tumor cells." The cells remained in the circulation for at least 5 months after subsequent surgery. Dr. Pachmann said that this observation corresponds with results found in patients, that is, tumor response does not mean increased survival". In accordance with our results Dr. Fehm shows that after neoadjuvant therapy disseminated cells are found at a higher frequency than before. We would like to emphasize in addition, that most of these cells are not apoptotic but even of these non-apoptotic cells not all are necessarily metastatic cells. Rather, a minimal fraction of them (perhaps less than 0.01% as shown by the group of Dr. Chambers) is expected to form metastases, but release of such cells is the first step and should be avoided or the released cells be destroyed by additional chemotherapy after surgery, as suggested by us.
With best regards
Katharina Pachmann




Central Nervous System Relapse in Patients With Breast Cancer Is Associated With Advanced Stages, With CK-19 mRNA-positive Circulating Occult Tumor Cells and With HER2/neu-positive tumor
Full Text PDF: http://www.biomedcentral.com/content/pdf/bcr1516.pdf

John Souglakos; Lambros Vamvakas; Stella Apostolaki; Maria Perraki; Zacharenia Saridaki; Irine Kazakou; Athanasios Pallis; Charalambos Kouroussis; Nikos Androulakis; Kostas Kalbakis; Georgia Millaki; Dimitris Mavroudis; Vassilis Georgoulias

Abstract

Introduction: To evaluate the incidence of central nervous system (CNS) involvement in patients with breast cancer treated with a taxane-based chemotherapy regimen and to determine predictive factors for CNS relapse.

Methods: The medical files of patients with early breast cancer (n = 253) or advanced stage breast cancer (n = 239) as well of those with other solid tumors (n = 336) treated with or without a taxane-based chemotherapy regimen during a 42-month period were reviewed. HER2/neu overexpression was identified by immunohistochemistry, whereas cytokeratin 19 (CK-19) mRNA-positive circulating tumor cells (CTCs) in the peripheral blood were identified by real-time PCR.

Results: The incidence of CNS relapse was similar in patients suffering from breast cancer or other solid tumors (10.4% and 11.4%, respectively; P = 0.517). The incidence of CNS relapse was significantly higher in breast cancer patients with advanced disease (P = 0.041), visceral disease and bone disease (P = 0.036), in those who were treated with a taxane-containing regimen (P = 0.024), in those with HER2/neu-overexpressing tumors (P = 0.022) and, finally, in those with detectable CK-19 mRNA-positive CTCs (P = 0.008). Multivariate analysis revealed that the stage of disease (odds ratio, 0.23; 95% confidence interval, 0.007-0.23; P = 0.0001), the HER2/neu status (odds ratio, 29.4; 95% confidence interval, 7.51-101.21; P = 0.0001) and the presence of CK-19 mRNA-positive CTCs (odds ratio, 8.31; 95% confidence interval, 3.97-12.84; P = 0.001) were independent predictive factors for CNS relapse.

Conclusion: CNS relapses are common among breast cancer patients treated with a taxane-based chemotherapy regimen, patients with HER2/neu-positive tumor and patients with CK-19 mRNA-positive CTCs.

Breast Cancer Res. 2006;8(4) ©2006 BioMed Central, Ltd.

Quote:
The reasons for the association between treatment of breast cancer with a taxane-containing chemotherapy regimen and an increased incidence of CNS involvement are not clear. Although taxanes are very lipophilic, their concentration in the CNS is very low after their intravenous administration [12,13]. Moreover, preclinical data suggest that taxanes are unable to penetrate the intact blood–brain barrier; in these models, the concentration of radiolabeled paclitaxel in the cerebrospinal fluid was found to be significantly lower than in other organs, and thus was undetectable in the brain, in the spinal cord or in any other site of the CNS [14]. It has more recently been proposed that paclitaxel is exported from the p-glycoprotein and other ATP-binding cassette transporters placed at the luminal membrane of brain capillaries, as an explanation for the low concentrations of taxanes in the CNS [15].
Furthermore, the detection of cytokeratin 19 (CK-19) and of mRNA-positive circulating tumor cells (CTCs) in the peripheral blood and the bone marrow of patients with breast cancer was correlated with increased incidence of relapse in several studies [16,17]. The possible correlation of this finding with an increased incidence of CNS relapse was not reported in any of these studies.
.......
Quote:
In the present study it was also possible to confirm the initial clinical observation that breast cancer patients who receive a taxane-containing chemotherapy regimen have a significantly higher incidence of CNS metastases compared with that of patients treated with a nontaxane-containing regimen. Nevertheless, this parameter did not emerge as an independent predictive factor in the multivariate analysis. It is important to note, however, that a similar difference was not observed in patients with other types of solid tumor treated or not with a taxanecontaining chemotherapy regimen.
Taxanes are highly effective in breast cancer and they prolong the survival of patients with advanced disease. It is also well known that CNS involvement is more frequent in the late stages of the disease. A possible explanation could therefore be that the increase in survival and the prolonged period of advanced disease is correlated with the increased frequency of CNS relapse. It is important to note that such a correlation
could represent a confounding factor, in a retrospective analysis. For example, someone can hypothesize that physicians may have chosen more aggressive treatment for patients with aggressive tumors. These patients also showed increased incidence for CNS relapse. Although this observation could be explained with the 'sanctuary site' hypothesis, experimental studies in mice have demonstrated that the blood–brain barrier is intact inside and around brain metastases smaller than 0.2 mm2 but not in larger lesions [29]. This finding implies that the barrier should not be a major obstacle to using chemotherapy in the treatment of brain metastases, at least in the advanced stages of the disease. In addition, more recent studies have shown that paclitaxel is exported by the p-glycoprotein and other ATP-binding cassette transporters placed at the luminal membrane of brain capillaries [15]
Quote:
We have previously reported that the use of Transtuzumab could decrease or eradicate CK-19 mRNA-positive CTCs [30]. Based on these results we have initiated a randomized clinical trial to evaluate the effect of Transtuzumab administration on the disease-free survival of patients with early breast cancer and CK-19 and HER-2 mRNA-positive CTCs. The incidence of CNS relapse in correlation with the detection of CK-19 mRNA-positive CTCs will be prospectively evaluated in this ongoing study.
PDF of full text attached

Advanced Breast Cancer: Treatment
with Docetaxel/Epirubicin
Quote:
An interesting observation regarding a higher incidence of central nervous system
(CNS) involvement in patients treated with docetaxel/epirubicin was reported by an
Italian group, based on a pooled analysis of their phase I and II studies (Pagani et
al., 1999, 2000). A total of 92 patients were included in these two studies and
the authors reported that 28 (30%) of the 92 patients treated with this combination
developed CNS metastases; 25 patients developed cerebral metastasis, two leptomeningeal, and one both (Crivellari et al., 2001). Median time for the development
of CNS metastases from the start of chemotherapy was 15 months (range 5–42), when the six patients presenting CNS progression within 3 months from start of treatment were excluded. It is noteworthy that 11 patients (39%) had disease progression
only in the CNS. Although, this observation could be easily explained by the sanctuary site ‘hypothesis’, as a consequence of an intact blood-brain barrier, this is not proven and the exact explanation remains to be elucidated. The authors conclude that as anthracycline- and taxanecontaining regimens are increasingly used both in the metastatic and in the adjuvant setting, a careful monitoring of any neurological
symptoms should be advisable.
Rich66 is offline   Reply With Quote
Old 11-12-2009, 06:05 AM   #4
Rich66
Senior Member
 
Rich66's Avatar
 
Join Date: Feb 2008
Location: South East Wisconsin
Posts: 3,431
Re: Antimitotic chemos promote adhesion in detached and circulating tumor cells

Ann Oncol. 2008 Nov;19(11):1837-41. Epub 2008 Jun 18.
Is risk of central nervous system (CNS) relapse related to adjuvant taxane treatment in node-positive breast cancer? Results of the CNS substudy in the intergroup Phase III BIG 02-98 Trial.

Pestalozzi BC, Francis P, Quinaux E, Dolci S, Azambuja E, Gelber RD, Viale G, Balil A, Andersson M, Nordenskjöld B, Gnant M, Gutierrez J, Láng I, Crown JP, Piccart-Gebhart M; BIG 02-98 Collaborative Group.
Department of Oncology, University Hospital, Zurich, Switzerland. bernhard.pestalozzi@usz.ch
BACKGROUND: Breast cancer central nervous system (CNS) metastases are an increasingly important problem because of high CNS relapse rates in patients treated with trastuzumab and/or taxanes. PATIENTS AND METHODS: We evaluated data from 2887 node-positive breast cancer patients randomised in the BIG 02-98 trial comparing anthracycline-based adjuvant chemotherapy (control arms) to anthracycline-docetaxel-based sequential or concurrent chemotherapy (experimental arms). After a median follow-up of 5 years, 403 patients had died and detailed information on CNS relapse was collected for these patients. RESULTS: CNS relapse occurred in 4.0% of control patients and 3.7% of docetaxel-treated patients. CNS relapse occurred in 27% of deceased patients in both treatment groups. CNS relapse was usually accompanied by neurologic symptoms (90%), and 25% of patients with CNS relapse died without evidence of extra-CNS relapse. Only 20% of patients survived 1 year from the diagnosis of CNS relapse. Prognosis of CNS relapse was worse for patients with meningeal carcinomatosis when compared with brain metastases. Unexpected findings included a higher rate of positive cerebrospinal fluid cytology (8% versus 3%) and more frequent use of magnetic resonance imaging for diagnosis (47% versus 30%) in the docetaxel-treated patients. CONCLUSION: There is no evidence that adjuvant docetaxel treatment is associated with an increased frequency of CNS relapse.

PMID: 18562328 [PubMed - indexed for MEDLINE]
__________________

Mom's treatment history (link)
Rich66 is offline   Reply With Quote
Old 11-12-2009, 12:57 PM   #5
Ellie F
Senior Member
 
Join Date: Feb 2009
Posts: 1,526
Re: Antimitotic chemos promote adhesion in detached and circulating tumor cells

Phew that's better news Rich especially as it compares a large number of patients
Ellie
Ellie F is offline   Reply With Quote
Old 11-15-2009, 11:20 PM   #6
Rich66
Senior Member
 
Rich66's Avatar
 
Join Date: Feb 2008
Location: South East Wisconsin
Posts: 3,431
Re: Antimitotic chemos promote adhesion in detached and circulating tumor cells

Maybe it's still is an issue in metastatic BC:

From the above study:

Quote:
The objective of the present analysis was to evaluate the frequency of CNS relapse in patients treated with or without adjuvant taxanes. This question was studied because taxane treatment in advanced breast cancer was associated with high
CNS relapse rates in some studies (up to 30%) [5, 6].
Quote:
In recent years, high rates of CNS relapse have been observed in smaller studies of patients treated with taxane-based (14% and 30%) [5,6] or with trastuzumab-based (34% and 39%) [10,11,12] therapy for metastatic breast cancer. The reason for these higher rates are unknown, although efficacy in the systemic ‘compartment’ in conjunction with restricted efficacy of these treatments in the CNS ‘compartment’ [11], heterogeneity in tumour cell biology [13] and/or drug interactions with the blood brain barrier can be considered.
Advanced Breast Cancer: Treatment
with Docetaxel/Epirubicin
:

Quote:
An interesting observation regarding a higher incidence of central nervous system (CNS) involvement in patients treated with docetaxel/epirubicin was reported by an Italian group, based on a pooled analysis of their phase I and II studies (Pagani et al., 1999, 2000). A total of 92 patients were included in these two studies and the authors reported that 28 (30%) of the 92 patients treated with this combination
developed CNS metastases; 25 patients developed cerebral metastasis, two leptomeningeal, and one both (Crivellari et al., 2001). Median time for the development of CNS metastases from the start of chemotherapy was 15 months (range 5–42), when the six patients presenting CNS progression within 3 months from start of treatment were excluded. It is noteworthy that 11 patients (39%) had disease progression only in the CNS. Although, this observation could be easily explained by the sanctuary site ‘hypothesis’, as a consequence of an intact blood-brain barrier, this is not proven and the exact explanation remains to be elucidated. The authors conclude that as anthracycline and taxane containing regimens are increasingly used both in the metastatic and in the adjuvant setting, a careful monitoring of any neurological symptoms should be advisable.
5. Freilich RJ, Seidman AD, DeAngelis LM. Central nervous system progression of metastatic breast cancer in patients treated with paclitaxel. Cancer 1995; 76:232–236.

6. Crivellari D, Pagani O, Veronesi A et al. High incidence of central nervous system involvement in patients with metastatic or locally advanced breast cancer treated with epirubicin and docetaxel. Ann Oncol. 2001; 12: 353–356.

7. Pestalozzi BC, Zahrieh D, Price KN et al. for the International Breast Cancer Study Group (IBCSG). Identifying breast cancer patients at risk for central nervous system (CNS) metastases in trials of the International Breast Cancer Study Group (IBCSG). Ann Oncol. 2006; 17: 935–944.
Quote:
In advanced breast cancer, a high incidence of CNS metastases was found in women treated with taxanes, 14% and 30%, respectively [21, 22]
Quote:
21. Freilich RJ, Seidman AD, DeAngelis LM. Central nervous system progression
of metastatic breast cancer in patients treated with paclitaxel. Cancer 1995; 76:
232–236.
22. Crivellari D, Pagani O, Veronesi A et al. High incidence of central nervous system
involvment in patients with metastatic or locally advanced breast cancer treated
with epirubicin and docetaxel. Ann Oncol 2001; 12: 353–356.
. Similarly, a high incidence (34%) of CNS metastases was found among patients treated with trastuzumab for stage IV breast cancer [23, 24]. Because trastuzumab does not cross the blood-brain barrier [25], this higher incidence may reflect prolongation of disease control in the systemic compartment but not in the CNS. In addition, HER2-positive tumors are known to behave more aggressively, and it may be their biological behavior that results in an increased rate of CNS metastasis...Prophylactic cranial irradiation is not justified for any cohort of breast cancer patients at this time.
8. Gonzales-Angulo AM, Cristofanilli M, Strom EA et al. Central nervous system metastases in patients with high-risk breast carcinoma after multimodality treatment. Cancer 2004; 101: 1760–1766.
Central nervous system metastases in women after multimodality therapy for high risk breast cancer.

Carey LA, Ewend MG, Metzger R, Sawyer L, Dees EC, Sartor CI, Moore DT, Graham ML.
Division of Hematology/Oncology, Department of Medicine, University of North Carolina, 3009 Old Clinic Building, Chapel Hill, NC, 27599-7305, USA. lisa_carey@med.unc.edu
Comment in:
Quote:
BACKGROUND: Central nervous system (CNS) relapse is increasing in breast cancer. This increase may reflect altered failure patterns from adjuvant therapy, more effective systemic therapy with improved control in non-CNS sites, or a resistant breast cancer subtype. METHODS: To determine the factors associated with clinical CNS relapse, we examined response to neoadjuvant chemotherapy (chemosensitivity), time to relapse and sites of relapse in a cohort of 140 patients without evidence of metastasis at presentation. RESULTS: At 5 years (interquartile range 3-6 years), 44 (31%) patients developed distant metastases, including 13 with CNS metastases. CNS relapse was early (median 24 months after diagnosis) and associated with relapse in bone and liver, suggesting hematogenous dissemination. Those with CNS relapse were younger at diagnosis (40 versus 49 years) and more likely to have lymphovascular invasion in the primary tumor compared with non-CNS metastases. Response to neoadjuvant chemotherapy was not different (69% versus 73% response rate) between the two groups. Extent of residual disease after chemotherapy was strongly associated with relapse outside the CNS but not CNS relapses. The CNS was an isolated or dominant site of metastasis in 8 of 13. Despite treatment, most patients with CNS involvement died of neurologic causes a median of 6 months later. CONCLUSION: Breast cancers that develop CNS metastases differ from those that develop metastases elsewhere. Both tumor behavior and reduced chemotherapy accessibility to the CNS may contribute to increased CNS involvement in breast cancer patients treated with multimodality therapy.

PMID: 15609130 [PubMed - indexed for MEDLINE]
9. Carey LA, Ewend MG, Metzger R et al. Central nervous system metastases in women after multimodality therapy for high risk breast cancer. Breast Cancer Res Treat. 2004; 88: 273–280.

10. Bendell JC, Domchek S, Burstein HJ et al. Central nervous system metastases in
women who receive trastuzumab-based therapy for metastatic breast carcinoma. Cancer 2003; 97: 2972–2977.

11. Burstein HJ, Lieberman G, Slamon DJ et al. Isolated central nervous system metastases in patients with HER2-overexpressing advanced breast cancer treated with first-line trastuzumab-based therapy. Ann Oncol. 2005; 16(11):1772–1777.

12. Dawson SJ, Ranieri NF, Snyder RD et al. Central nervous system metastases in
women with HER-2 positive metastatic breast cancer after treatment with trastuzumab. Asia Pac J Clin Oncol. 2006; 2: 50–56.

13. Minn AJ, Kang Y, Serganova I et al. Distinct organ-specific metastatic potential of
individual breast cancer cells and primary tumors. J Clin Invest. 2005; 115:44–55.

14. Pestalozzi BC, Brignoli S. Trastuzumab in CSF. J Clin Oncol. 2000; 18: 2350–2351.

Research May Provide Potential Target For New Therapies To Limit Metastasis Of Primary Breast Cancers


http://www.medicalnewstoday.com/articles/182239.php


Quote:
15 Mar 2010

Researchers at the University of Maryland Marlene and Stewart Greenebaum Cancer Center have discovered that "microtentacles," or extensions of the plasma membrane of breast cancer cells, appear to play a key role in how cancers spread to distant locations in the body. Targeting these microtentacles might prove to be a new way to prevent or slow the growth of these secondary cancers, the scientists say.

They report in an article to be published online March 15, 2010, in the journal Oncogene that a protein called "tau" promotes the formation of these microtentacles on breast tumor cells which break away from primary cancers and circulate in the bloodstream. While twisted remnants of tau protein have been seen in the brain tissue of patients with Alzheimer's disease, this is the first report that tau could play a role in tumor metastasis by changing the shape of cancer cells. These tau-induced microtentacles can help the cells reattach to the walls of small blood vessels to create new pockets of cancer.
Quote:
researchers analyzed breast tumor cells from 102 patients and found that 52 percent had tau in their metastatic tumors and 26 percent (27 patients) showed a significant increase in tau as their cancer progressed. Twenty-two of these patients even had tau in metastatic tumors despite having none in their primary tumors.

Oncogene. 2010 Jun 3;29(22):3217-27. Epub 2010 Mar 15.
Metastatic breast tumors express increased tau, which promotes microtentacle formation and the reattachment of detached breast tumor cells.

Matrone MA, Whipple RA, Thompson K, Cho EH, Vitolo MI, Balzer EM, Yoon JR, Ioffe OB, Tuttle KC, Tan M, Martin SS.
Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.


TEXT

Abstract

The cytoskeletal organization of detached and circulating tumor cells (CTCs) is currently not well defined and may provide potential targets for new therapies to limit metastatic tumor spread. In vivo, CTCs reattach in distant tissues by a mechanism that is tubulin-dependent and suppressed by polymerized actin. The cytoskeletal mechanisms that promote reattachment of CTCs match exactly with the mechanisms supporting tubulin microtentacles (McTN), which we have recently identified in detached breast tumor cells. In this study, we aimed to investigate how McTN formation is affected by the microtubule-associated protein, tau, which is expressed in a subset of chemotherapy-resistant breast cancers. We demonstrate that endogenous tau protein localizes to McTNs and is both necessary and sufficient to promote McTN extension in detached breast tumor cells. Tau-induced McTNs increase reattachment of suspended cells and retention of CTCs in lung capillaries. Analysis of patient-matched primary and metastatic tumors reveals that 52% possess tau expression in metastases and 26% display significantly increased tau expression over disease progression. Tau enrichment in metastatic tumors and the ability of tau to promote tumor cell reattachment through McTN formation support a model in which tau-induced microtubule stabilization provides a selective advantage during tumor metastasis.

PMID: 20228842 [PubMed - in process]




Quote:
In summary, MAPs have powerful implications for chemotherapy efficacy and metastatic progression. Tau provides taxane-resistance through binding-competition on microtubules (Wagner et al., 2005). Furthermore, taxanes have been determined to increase CTCs by 10 000-fold among breast cancer patients (Camara et al.,2007). As CTC persistence is indicative of an increased risk of disease recurrence (Janni et al., 2005), and tau increases the trapping and reattachment of detached breast tumor cells, pathological evaluation of tau may benefit patients by minimizing metastasis through CTC mobilization. Elucidating the role of tau and other MAPs in cancer may lead to new therapeutics that target MAP expression or the regulation of MAPtubulin interactions. It has been suggested that the expression of neuronal MAPs in non-neuronally derived cancers speak to their origin from stem cell-like precursors (Bhat and Setaluri, 2007). This raises the potential for collaboration between the neurodegenerative and cancer drug fields. Aberrant regulation of tau leads to neurodegenerative tauopathies. Consequently, numerous tau-targeted compounds have been characterized, some of which are current therapeutics (Bulic et al.,2009). Promising are two drugs, aziridinylbenzoquinone and albendazole, which can reduce tau expression by 38 and 52%, respectively, without detectable cytotoxicity (Dickey et al., 2006). As cancer therapeutics, these and other anti-tau drugs may hold potential to inhibit tumor metastasis by reducing tau-induced McTNs.




Breast Cancer Res. 2010 Jun 28;12(3):R43. [Epub ahead of print]
The estrogen receptor influences microtubule-associated protein tau (MAPT) expression and the selective estrogen receptor inhibitor fulvestrant downregulates MAPT and increases the sensitivity to taxane in breast cancer cells.

Ikeda H, Taira N, Hara F, Fujita T, Yamamoto H, Soh J, Toyooka S, Nogami T, Shien T, Doihara H, Miyoshi S.
Corresponding author: Naruto Taira ntaira@md.okayama-u.ac.jp

FREE TEXT


ABSTRACT:

INTRODUCTION: Microtubule-associated protein tau (MAPT) inhibits the function of taxanes and high expression of MAPT decreased the sensitivity to taxanes. The relationship between estrogen receptor (ER) and MAPT in breast cancer is unclear. In this study, we examined the correlation of MAPT expression with the sensitivity of human breast cancer cells to taxanes, and the relationship between ER and MAPT. METHODS: The correlation between MAPT expression and sensitivity to taxanes was investigated in 12 human breast cancer cell lines. Alterations in cellular sensitivity to taxanes were evaluated after knockdown of MAPT expression. ER expression was knocked down or stimulated in MAPT- and ER-positive cell lines to examine the relationship between ER and MAPT. The cells were also treated with hormone drugs (fulvestrant and tamoxifen) and taxanes. RESULTS: mRNA expression of MAPT did not correlate with protein expression or the sensitivity to taxanes. However, expression of MAPT protein isoforms of less than 70 kDa was correlated with a low sensitivity to taxanes. Downregulation of MAPT increased cellular sensitivity to taxanes. MAPT protein expression was decreased by downregulation of ER and by fulvestrant, an ER inhibitor; but increased by stimulation with 17-beta-estradiol or tamoxifen. The combination of fulvestrant with taxanes had a synergistic effect, whereas tamoxifen and taxanes had an antagonistic effect. CONCLUSIONS: Expression of MAPT protein isoforms of less than 70 kDa is correlated with a low sensitivity to taxanes in breast cancer cells. ER influences MAPT expression and fulvestrant increased the sensitivity to taxanes in MAPT- and ER-positive breast cancer cells.

PMID: 20579400 [PubMed - as supplied by publisher]


Quote:
Several mechanisms of taxane resistance have been described, including overexpression of the drug efflux pump MDR-1/P-gp, HER-2 overexpression, tubulin mutation, and variable expression of tubulin isotypes and stathmin [4, 7-12]. Microtubule-associated protein-tau (MAPT), which is implicated in the pathogenesis of Alzheimer’s disease, is associated with another mechanism of taxane resistance. MAPT binds to both the outer and inner surfaces of microtubules, leading to tubulin assembly and microtubule stabilization. Since
taxanes also bind to the inner surface of microtubules, MAPT obstructs the function of the drug [5, 6, 13, 14].
Rouzier et al. found that low MAPT expression was associated with higher rates of a pathologic complete response to preoperative paclitaxel and 5-fluorouracil, doxorubicin, cyclophosphamide (paclitaxel/FAC) chemotherapy [5]. This group also showed that MAPT overexpression was correlated with resistance to paclitaxel and that knockdown of MAPT with small interfering RNA (siRNA) reversed the resistance to taxanes in vitro [5].
<

Quote:
Hormonal drugs play an important role in breast cancer therapy. The selective ER inhibitor, fulvestrant, inhibits estrogen signaling through the ER in two ways: by competing with estradiol binding to the ER, and by increasing the turnover of ER to decrease the ER protein level in breast cancer cells. In contrast, tamoxifen, a selective ER modulator, is an ER antagonist but often displays estrogen-like agonist activity [22-24].
Previous in vitro studies show that tamoxifen has an antagonistic effect on anti-cancer drugs [25, 26]. Several clinical studies that used tamoxifen for hormone therapy have found that it has an antagonistic effect on chemotherapy drugs when it is used concurrently with them, and that the results of the combined use of tamoxifen with chemotherapy drugs is inferior, compared with using the drugs sequentially [27-30]. The effect of combination treatment using other modern hormone therapies, such as aromatase inhibitors or
fulvestrant, has not been examined thoroughly.
Quote:
Our results suggested that the effect of tamoxifen on ER signaling differs depending on the dose. MAPT protein expression was increased at low concentrations of tamoxifen of 500 nM - 1 μM, but decreased at higher concentrations. Several factors associated with resistance to chemotherapy via regulation by ER signaling have been identified [4, 22, 38-40]. Tamoxifen is thought to exert an antagonistic effect in concomitant use with chemotherapeutic drugs by increasing the expression of these factors via an agonistic effect on the ER. Active metabolites of tamoxifen also have different functions compared with the parent drug [41-43], and more detailed studies are needed to
determine how tamoxifen and its metabolites influence chemotherapy in vivo and in vitro.
Fulvestrant decreased ER and MAPT expression at all concentrations. An MTS assay, flow cytometry, and immunofluorescence all showed that the combination of fulvestrant and taxanes had a synergistic effect, consistent with the finding of Sui et al. that fulvestrant combined with paclitaxel was effective in breast cancer cells in vitro [24]. Fulvestrant assists taxane function by downregulating the ER and ER-regulated factors associated with taxane resistance, and the combination of fulvestrant with taxanes increases the sensitivity of MAPT- and ER-positive breast cancer cells to taxanes.
ER-positive breast cancers clinically show a lower sensitivity to chemotherapy than do ER-negative breast cancers. This may be caused by the ER itself or by ER modulation of factors that result in resistance to chemotherapy. Our study indicates that the combination of modern hormone therapy with modern chemotherapy may become an effective therapy to ER-positive breast cancers.
__________________

Mom's treatment history (link)
Rich66 is offline   Reply With Quote
Old 09-10-2010, 05:53 PM   #7
Rich66
Senior Member
 
Rich66's Avatar
 
Join Date: Feb 2008
Location: South East Wisconsin
Posts: 3,431
Re: Antimitotic chemos promote adhesion in detached and circulating tumor cells

.................................................. ..........
__________________

Mom's treatment history (link)
Rich66 is offline   Reply With Quote
Old 09-10-2010, 05:53 PM   #8
Rich66
Senior Member
 
Rich66's Avatar
 
Join Date: Feb 2008
Location: South East Wisconsin
Posts: 3,431
Re: Antimitotic chemos promote adhesion in detached and circulating tumor cells

.................................................. ....
__________________

Mom's treatment history (link)
Rich66 is offline   Reply With Quote
Reply

Thread Tools
Display Modes

Posting Rules
You may not post new threads
You may not post replies
You may not post attachments
You may not edit your posts

BB code is On
Smilies are On
[IMG] code is On
HTML code is On

Forum Jump


All times are GMT -7. The time now is 02:57 PM.


Powered by vBulletin® Version 3.8.7
Copyright ©2000 - 2024, vBulletin Solutions, Inc.
Copyright HER2 Support Group 2007 - 2021
free webpage hit counter