Where To Start With Large Brain Tumor?

Blossom

Moderator
Forum Supporter
Joined
Nov 23, 2013
Messages
11,046
Location
Indiana USA
You are a very kind person, Lisa. Your friend is lucky to have you.
I agree.
Lisa if I had a friend or family member contemplating medical treatment for a serious condition I would see what information could be found in the Cochrane Library. I'm not insinuating at all that alternative/complementary measures shouldn't be explored as well but I think being as informed as possible is always a plus in making the best decisions. Here is a link to the Cochrane review website where you or your friend can look up what medical interventions for his particular diagnosis results in the most favorable outcomes.
Cochrane | Trusted evidence. Informed decisions. Better health.
 

Fletcher

Member
Joined
Aug 11, 2015
Messages
78
Hi Lisa, if it were me I would look to use multiple protocols that can help (rather than one fix).
Fasting / IF does seem to help, especially at the point where seizures occur.
DCA and cannabis, although both have side effects. More on DCA.
Ozone therapy def. has some benefits in some cancers, you might want to look into it (this is prob the #1 thing I would do / research if it were me).
Andrew Scarborough has a blog which may have some info that can help My brain cancer story .
Wishing your friend the best of luck.
 
B

Braveheart

Guest
Alirezaei, Mehrdad. "Short-term fasting induces profound neuronal autophagy." Autophagy (2010)

Autophagy is the modern term for autolysis: a process in which autophagosomes are induced inside each cell to recycle cellular components. This process occurs all the time to some degree, but it can be accelerated by fasting—even in the brain. The common belief had been that the brain was more‐or‐less spared from autophagy simply because is loses practically no weight during fasting, but not so:

'Published studies indicate that the brain is spared many of the effects of short-term food restriction, perhaps because it is a metabolically privileged site that, relative to other organs, is protected from the acute effects of nutrient deprivation, including autophagy.' ―Alirezaei

The brain does upregulate autophagosomes during fasting, yet is allowed to maintain its weight ostensibly by drawing components from the blood. Autophagy isn't apoptosis, nor is it necrosis, but merely a normal mechanism which degrades proteins to amino acids ⟶ to build other proteins. Cells are not destroyed in the process, yet many will shrink in size. The size reduction of each cell of course depends on its type and location, with adipocytes losing the most volume the fastest.

'Our findings also may have therapeutic implications, as outlined below. Autophagy is sometimes referred to as cellular “cleansing”, and our observations provide an attractive neuronal parallel to the organismal benefits that, historically, are perceived to derive from fasting.' ―Alirezaei

Fasting for 48 hours can induce autophagosomes in the brain, most likely through the mTOR pathway which can sense leucine. Leucine restriction appears more powerful for accelerating autophagy than glucose restriction, perhaps why Gerson had so much success. Tumors have been completely autolysed during 'water fasts,' yet this has also been done on 'juice fasts.'

View attachment 8447 View attachment 8448

By all accounts (juice) fasting appears to be the best cancer reversal strategy, but is unpopular because it's free: very few people can make money on it. Also adding to its unpopularity is (1) other people's discouragement (from simple ignorance), and (2) the food addictions that most people have.


'For example, in vitro models have shown that starvation in neuronal cell lines can remove toxic molecules and damaged mitochondria from neurons.' ―Alirezaei

Autophagy of the neurons could also be expected to decrease intracellular lipofuscin, the only known mechanism for reducing this besides simply avoiding iron, aluminum, and polyunsaturated fatty acids. I recently had fasted for 2¹⁄₂ days so I can guarantee that a fast this long is safe, and people surviving for weeks to months on water alone has been well documented.
nice...
 
OP
L

lollipop

Guest
I agree.
Lisa if I had a friend or family member contemplating medical treatment for a serious condition I would see what information could be found in the Cochrane Library. I'm not insinuating at all that alternative/complementary measures shouldn't be explored as well but I think being as informed as possible is always a plus in making the best decisions. Here is a link to the Cochrane review website where you or your friend can look up what medical interventions for his particular diagnosis results in the most favorable outcomes.
Cochrane | Trusted evidence. Informed decisions. Better health.
This is very helpful @Blossom!
 
OP
L

lollipop

Guest
Hi Lisa, if it were me I would look to use multiple protocols that can help (rather than one fix).
Fasting / IF does seem to help, especially at the point where seizures occur.
DCA and cannabis, although both have side effects. More on DCA.
Ozone therapy def. has some benefits in some cancers, you might want to look into it (this is prob the #1 thing I would do / research if it were me).
Andrew Scarborough has a blog which may have some info that can help My brain cancer story .
Wishing your friend the best of luck.
Thank you @Fletcher! Appreciate the interesting blog and ideas ❤️❤️
 

yerrag

Member
Joined
Mar 29, 2016
Messages
10,883
Location
Manila
Lisa, there is a lot of information here. Hope you get a lot of guidance and inspiration sifting through all of it. It is hard enough for most of us to process such information, what more it would be for the less initiated folks. And even more so if the cancer patient is to do the research, as well as the patient's immediate family. There are so many ways to slice and dice this, I am likely to just throw darts, or roll the dice. Godspeed to you.
 
OP
L

lollipop

Guest
Lisa, there is a lot of information here. Hope you get a lot of guidance and inspiration sifting through all of it. It is hard enough for most of us to process such information, what more it would be for the less initiated folks. And even more so if the cancer patient is to do the research, as well as the patient's immediate family. There are so many ways to slice and dice this, I am likely to just throw darts, or roll the dice. Godspeed to you.
Thank you @yerrag. Yes so am going to sift through and cherry pick a few directions that seem better for their life situation.
 

managing

Member
Joined
Jun 19, 2014
Messages
2,262
My father died of a glioblastoma. If I had it all to do over again (and yes, I was the one with legal authority to make decisions) there would be no surgery or radiation.

This review is interesting:

Metabolic management of brain cancer - ScienceDirect

To summarize the interesting points:

-surgery, chemo, radiation may prolong life, but will not ever cure
-tumors dependent on glycolysis, cannot utilize ketones
-brain tumors show distinct deficiency of mitochondrial lipids, especially cardiolipin, which are essential for oxidative respiration as well as ketone assimilation
-in the tumor cells, this mitochondrial defect is irreversible

They propose a ketogenic therapy in response to this. The idea is to feed the brain while starving the tumor. The problem is of course, from our perspective, that this stresses the organism systemically. Also, most conceptions of ketogenic diets are very high in PUFA.

What @Travis proposes above seems to address this energy issue (juice diet). But I wonder if a high SFA combined with small, frequent sugar (ie, juice diet or thereabouts) would provide similar benefits and even heightened protection.
 
OP
L

lollipop

Guest
My father died of a glioblastoma. If I had it all to do over again (and yes, I was the one with legal authority to make decisions) there would be no surgery or radiation.

This review is interesting:

Metabolic management of brain cancer - ScienceDirect

To summarize the interesting points:

-surgery, chemo, radiation may prolong life, but will not ever cure
-tumors dependent on glycolysis, cannot utilize ketones
-brain tumors show distinct deficiency of mitochondrial lipids, especially cardiolipin, which are essential for oxidative respiration as well as ketone assimilation
-in the tumor cells, this mitochondrial defect is irreversible

They propose a ketogenic therapy in response to this. The idea is to feed the brain while starving the tumor. The problem is of course, from our perspective, that this stresses the organism systemically. Also, most conceptions of ketogenic diets are very high in PUFA.

What @Travis proposes above seems to address this energy issue (juice diet). But I wonder if a high SFA combined with small, frequent sugar (ie, juice diet or thereabouts) would provide similar benefits and even heightened protection.
Very interesting @managing - thank you for sharing. Helpful perspective and highlights the importance of where a person is in awareness at the moment. Strong food for thought.
 

Travis

Member
Joined
Jul 14, 2016
Messages
3,189

tfcjesse

Member
Joined
Oct 18, 2016
Messages
122
Alirezaei, Mehrdad. "Short-term fasting induces profound neuronal autophagy." Autophagy (2010)

Autophagy is the modern term for autolysis: a process in which autophagosomes are induced inside each cell to recycle cellular components. This process occurs all the time to some degree, but it can be accelerated by fasting—even in the brain. The common belief had been that the brain was more‐or‐less spared from autophagy simply because is loses practically no weight during fasting, but not so:

'Published studies indicate that the brain is spared many of the effects of short-term food restriction, perhaps because it is a metabolically privileged site that, relative to other organs, is protected from the acute effects of nutrient deprivation, including autophagy.' ―Alirezaei

The brain does upregulate autophagosomes during fasting, yet is allowed to maintain its weight ostensibly by drawing components from the blood. Autophagy isn't apoptosis, nor is it necrosis, but merely a normal mechanism which degrades proteins to amino acids ⟶ to build other proteins. Cells are not destroyed in the process, yet many will shrink in size. The size reduction of each cell of course depends on its type and location, with adipocytes losing the most volume the fastest.

'Our findings also may have therapeutic implications, as outlined below. Autophagy is sometimes referred to as cellular “cleansing”, and our observations provide an attractive neuronal parallel to the organismal benefits that, historically, are perceived to derive from fasting.' ―Alirezaei

Fasting for 48 hours can induce autophagosomes in the brain, most likely through the mTOR pathway which can sense leucine. Leucine restriction appears more powerful for accelerating autophagy than glucose restriction, perhaps why Gerson had so much success. Tumors have been completely autolysed during 'water fasts,' yet this has also been done on 'juice fasts.'

View attachment 8447 View attachment 8448

By all accounts (juice) fasting appears to be the best cancer reversal strategy, but is unpopular because it's free: very few people can make money on it. Also adding to its unpopularity is (1) other people's discouragement (from simple ignorance), and (2) the food addictions that most people have.


'For example, in vitro models have shown that starvation in neuronal cell lines can remove toxic molecules and damaged mitochondria from neurons.' ―Alirezaei

Autophagy of the neurons could also be expected to decrease intracellular lipofuscin, the only known mechanism for reducing this besides simply avoiding iron, aluminum, and polyunsaturated fatty acids. I recently had fasted for 2¹⁄₂ days so I can guarantee that a fast this long is safe, and people surviving for weeks to months on water alone has been well documented.

Very helpful. Just wondering-which juice fast do you refer to? Breuss?
 

Travis

Member
Joined
Jul 14, 2016
Messages
3,189
Very helpful. Just wondering-which juice fast do you refer to? Breuss?
I haven't heard of him. I think this is theoretically sound—but not entirely for the common reasons given but because leucine inhibits autolysis. Of all amino acids tested—and they have tested them all—leucine was the most effective at inhibiting autolysis. Glutamine could do this too, but it had been shown to be through a different mechanism (probably by spontanous hydrolysis; releasing N₃ forming glutamate). Leucine is the most powerful signal for autolysis, which it likely mediates through the mTOR pathway (rapamycin also induces autolysis). Of course fruit is good for other reasons as well, but this is a big one never talked about. The two most powerful growth promoting amino acids are methionine and leucine, one increasing DNA replication by forming polyamines and the other inhibiting autolysis.
 

LeeLemonoil

Member
Joined
Sep 24, 2016
Messages
4,265
So Leucine-restriction and not only Methionine-restriction is a way of mimicking fasting?

I have only very rudimentary understanding of the mTOR/AMPK pathway "dualism", a theme often variated in life-extender circles, but it seems to me that some aspects of this duality are vice-versa in the brain and the Rest of the body. What activates one pathway in the peripheria might activate the other in the CNS.
 

aquaman

Member
Joined
Aug 9, 2013
Messages
1,297
@lisaferraro , someone posted an excellent reply from Ray in the email advice thread [my bolding added]

Ray Peat said:
Tumors have multiple causes, so I think it’s best to use things that are known to be protective against known causes. I think cancers are continually produced and maintained by general metabolic conditions, and if those are ignored while trying to kill all of the abnormal cells, those cells release signals to recruit replacement cells, which, encountering the same or worse metabolic conditions, renew the tumor. Known causes involve inflammatory, excitatory processes, and when those are eliminated, tumors tend to disintegrate, undergoing “apoptosis,” a form of cell death that doesn’t create new inflammation. Here are some articles describing the effects of some of these antiinflammatory, antiexcitatory, substances—aspirin, caffeine, tetracylines, antihistamine/antiserotonin/anti-nitric oxide agents, progesterone, pregnenolone, antagonists of excitotoxicity, estrogen antagonists, vitamins D, E, and K, etc. Vitamin E (500 mg mixed tocopherols) and aspirin are things that the medical authorities might not object to.

Here are the VERY long references :)
1. Biomed Rep. 2016 Apr;4(4):444-448.
Cyclooxygenase inhibitor induces the upregulation of connexin-43 expression in C6
glioma cells.
Qin LJ(1), Jia YS(2), Zhang YB(1), Wang YH(1).
(1)Department of Physiology, School of Basic Medical Sciences, North China
University of Science and Technology, Tangshan, Hebei 063000, P.R. China.
(2)College of Traditional Chinese Medicine, North China University of Science and
Technology, Tangshan, Hebei 063000, P.R. China.
The present study was performed to determine whether aspirin, a cyclooxygenase
(COX) inhibitor, has an effect on the expression of connexin 43 (Cx43) in C6
glioma cells. Using an in vitro glioma invasion model, the expression of Cx43
protein in C6 cells was significantly increased following aspirin treatment at a
dose of 8 mmol/l for 30, 60 and 120 min via western blot analysis. The peak value
of the Cx43 expression was observed in C6 cells after 120 min of aspirin
treatment, which was significantly reduced by prostaglandin E2 (PGE2). In
addition, aspirin also significantly increased the gap junction intercellular
communication (GJIC) activity and reduced glioma invasion, which was induced by
PGE2. This led to the conclusion that the aspirin-induced glioma invasion
decrease may be associated with the increased expression of Cx43 protein and
formation of GJIC.
2. Eur J Cancer Prev. 2013 Nov;22(6):585-95.
Overexpression of S100A9 in human glioma and in-vitro inhibition by aspirin.
Huang N(1), Chen S, Deng J, Huang Q, Liao P, Wang F, Cheng Y.
(1)aDepartment of Neurosurgery, The Second Affiliated Hospital of Chongqing
Medical University bDepartment of Neurosurgery, The First Affiliated Hospital of
Chongqing Medical University cDepartment of Biomedical Engineering dInstitute of
Life Sciences, Chongqing Medical University, Chongqing, China.
Our previous work has shown that S100A9 promotes the growth of glioma cells. The
aim of this study was to investigate S100A9 expression in glioma cells and to
explore the potential of NSAIDs in the inhibition of S100A9. The levels of S100A9
were analyzed in five normal human brain tissues and 109 astrocytomas by
immunohistochemical analysis. In addition, S100A9 levels were detected in normal
human astrocytes, glioma cell lines, and six pairs of matched astrocytoma tissues
by reverse transcription-PCR or western blotting analysis. After treatment with
4, 8, and 16 mmol/l aspirin, cell viability, early apoptosis rate, and S100A9
levels were quantified. Cell viability and the changes in S100A9 levels were also
examined in glioma cells exposed to a cyclooxygenase-2 inhibitor, NS-398, alone
and in combination with prostaglandin E2. We found that S100A9 was upregulated in
astrocytomas and was significantly (P<0.05) correlated with histologic grades.
S100A9 protein levels were also elevated in six astrocytomas compared with
matched adjacent noncancerous tissues. Both S100A9 mRNA and protein levels were
higher in glioma cell lines than in normal human astrocytes (P<0.05). Aspirin
treatment inhibited cell proliferation and caused early apoptosis in glioma,
coupled with reduced S100A9 levels. Treatment with NS-398 decreased cell growth
and expression of S100A9 in glioma cells; these effects were partially reversed
by exogenous prostaglandin E2. These results suggest overexpression of S100A9 in
glioma cells. Aspirin may be a novel candidate for targeted prevention of S100A9
overexpression in glioma cells.
3. CNS Neurosci Ther. 2013 Feb;19(2):98-108.
Aspirin-/TMZ-coloaded microspheres exert synergistic antiglioma efficacy via
inhibition of β-catenin transactivation.
Shi ZD(1), Qian XM, Liu CY, Han L, Zhang KL, Chen LY, Zhang JX, Pu PY, Yuan XB,
Kang CS; Chinese Glioma Cooperative Group (CGCG).
(1)Laboratory of Neuro-Oncology, Department of Neurosurgery, Tianjin Neurological
Institute, Tianjin Medical University General Hospital, China.
BACKGROUND AND AIMS: Currently temozolomide (TMZ) as a potent agent is widely
used to treat the glioblastoma multiforme (GBM), whereas recurrence due to
intrinsic or acquired therapeutic resistance often occurs. Combination
chemotherapy with TMZ may be a promising therapeutic strategy to improve
treatment efficacy.
METHODS: Aspirin, TMZ, and aspirin-/TMZ-coloaded poly (L-lactide-co-glycolide)
(PLGA) microspheres were prepared by spray drying, and cytotoxicities of
glioblastoma cells were measured.
RESULTS: Aspirin microsphere treatment induced slight apoptosis and modestly
inhibited proliferation of LN229 and U87 cells in vitro and in vivo through
inhibition of β-catenin transactivation. However, aspirin-/TMZ-coloaded
microspheres presented synergistic antitumor efficacy compared with single
TMZ-loaded microspheres. Aspirin/TMZ microspheres induced more apoptosis and
repressed proliferation of LN229 and U87 cells. Corresponding to inhibition of
β-catenin signaling, β-catenin/TCF4 transcriptional activity and STAT3 luciferase
activity were strongly suppressed, and downstream targets expression was
decreased. Furthermore, aspirin/TMZ microsphere intratumoral injection
downregulated the expression of β-catenin, TCF4, pAKT, pSTAT3, and PCNA and
delayed tumor growth in nude mice harboring subcutaneous LN229 xenografts.
CONCLUSIONS: Aspirin sensitized TMZ chemotherapy efficacy through inhibition of
β-catenin transactivation; furthermore, the coloaded microspheres achieved a
sustained release action to reduce the TMZ dosage, offering the potential for
improved treatment of glioblastomas.
4. J Neurosurg. 2011 Oct;115(4):780-8.
Antitumor effect of aspirin in glioblastoma cells by modulation of
β-catenin/T-cell factor-mediated transcriptional activity.
Lan F(1), Yue X, Han L, Yuan X, Shi Z, Huang K, Yang Y, Zou J, Zhang J, Jiang T,
Pu P, Kang C.
(1)Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin Medical
University General Hospital, Taijin, People’s Republic of China.
OBJECT: The goal in this study was to investigate the antitumor effect of aspirin
in glioblastoma cells and the molecular mechanism involved in its antineoplastic
activities.
METHODS: The authors used the
3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide method, flow
cytometry, the annexin V method, and Transwell cell invasion test to detect the
proliferation and invasive activity of U87 and A172 glioma cells before and after
being treated with aspirin. To determine the effects of aspirin on
β-catenin/T-cell factor (TCF) transcription activity, reporter constructs
containing 3 repeats of the wild-type (TOPflash) or mutant (FOPflash) TCF-binding
sites were used. Reverse transcriptase polymerase chain reaction and Western blot
analyses were used to detect the expression of multiple β-catenin/TCF target
genes following aspirin treatment.
RESULTS: The transcriptional activity of the β-catenin/TCF complex was strongly
inhibited by aspirin. Increasing the concentration of aspirin resulted in
decreased expression of c-myc, cyclin D1, and fra-1 mRNA and protein in U87 and
A172 cells in a dose-dependent manner. Aspirin inhibited glioma cell
proliferation and invasive ability, and induced apoptotic cell death.
CONCLUSIONS: The results suggest that aspirin is a potent antitumor agent, and
that it exerts its antineoplastic action by inhibition of the β-catenin/TCF
signaling pathway in glioma cells.
5. Neurol Res. 2003 Jun;25(4):370-6.
Aspirin and indomethacin exhibit antiproliferative effects and induce apoptosis
in T98G human glioblastoma cells.
Amin R(1), Kamitani H, Sultana H, Taniura S, Islam A, Sho A, Ishibashi M, Eling
TE, Watanabe T.
(1)Department of Neurosurgery, Institute of Neurological Sciences, Faculty of
Medicine, Tottori University School of Medicine, 36-1 Nishi-cho, Yonago, Tottori
683-8504, Japan.
The in vitro antiproliferative and apoptosis inducing properties of the
nonsteroidal anti-inflammatory drugs (NSAIDs) like acetyl salicylic acid
(aspirin) and indomethacin were investigated in T98G human glioblastoma cells to
explore their potential role in the chemoprevention of human glioma. The
biological effects induced by aspirin and indomethacin on T98G cells, in which
the expression of cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2) were
confirmed by RT-PCR and immunostaining, were investigated by studying cell
proliferation and apoptosis assays. The antiproliferative effects occurred in a
dose- and time-dependent manner on T98G cells by the treatment with 0.1 -2 mM
aspirin and 25-100 microM indomethacin. Moreover, aspirin displayed the greatest
growth inhibition within 24 h. Approximately 90% growth inhibition occurred
following treatment either with 2 mM aspirin or 100 microM indomethacin by 72 h
and induction of apoptosis was confirmed by DNA laddering and TUNEL assay. Our in
vitro findings indicate that aspirin and indomethacin have an antiproliferative
effect on T98G human glioblastoma cells at toxic concentrations.
Mol Cells. 2011 Mar;31(3):275-9.
Caffeine inhibits cell proliferation and regulates PKA/GSK3β pathways in U87MG
human glioma cells.
Ku BM(1), Lee YK, Jeong JY, Ryu J, Choi J, Kim JS, Cho YW, Roh GS, Kim HJ, Cho
GJ, Choi WS, Kang SS.
(1)Department of Anatomy and Neurobiology, Institute of Health Science, School of
Medicine, Gyeongsang National University, Jinju, 660-751, Korea.
Caffeine is the most commonly ingested methylxanthine and has anti-cancer effects
in several types of cancer. In this study, we examined the anti-cancer effects of
caffeine on gliomas, both in vitro and in vivo. In vitro, caffeine treatment
reduced glioma cell proliferation through G(0)/G(1)-phase cell cycle arrest by
suppressing Rb phosphorylation. In addition, caffeine induced apoptosis through
caspase-3 activation and poly(ADP-ribose) polymerase (PARP) cleavage. Caffeine
also phosphorylated serine 9 of glycogen synthase kinase 3 beta (GSK3β).
Pretreatment with H89, a pharmacological inhibitor of protein kinase A (PKA), was
able to antagonize caffeine-induced GSK3β(ser9) phosphorylation, suggesting that
the mechanism might involve a cAMP-dependent PKA-dependent pathway. In vivo,
caffeine-treated tumors exhibited reduced proliferation and increased apoptosis
compared with vehicle-treated tumors. These results suggest that caffeine induces
cell cycle arrest and caspase-dependent cell death in glioma cells, supporting
its potential use in chemotherapeutic options for malignant gliomas.
Nutr Cancer. 2014;66(3):419-23.
Theobromine, the primary methylxanthine found in Theobroma cacao, prevents
malignant glioblastoma proliferation by negatively regulating
phosphodiesterase-4, extracellular signal-regulated kinase, Akt/mammalian target
of rapamycin kinase, and nuclear factor-kappa B.
Sugimoto N(1), Miwa S, Hitomi Y, Nakamura H, Tsuchiya H, Yachie A.
(1)a Department of Physiology, Graduate School of Medical Science , Kanazawa
University , Kanazawa , Japan.
Theobromine, a caffeine derivative, is the primary methylxanthine produced by
Theobroma cacao. We previously showed that methylxanthines, including caffeine
and theophylline, have antitumor and antiinflammatory effects, which are in part
mediated by their inhibition of phosphodiesterase (PDE). A member of the PDE
family, PDE4, is widely expressed in and promotes the growth of glioblastoma, the
most common type of brain tumor. The purpose of this study was to determine
whether theobromine could exert growth inhibitory effects on U87-MG, a cell line
derived from human malignant glioma. We show that theobromine treatment elevates
intracellular cAMP levels and increases the activity of p38 mitogen-activated
protein kinase and c-Jun N-terminal kinase, whereas it attenuates p44/42
extracellular signal-regulated kinase activity and the Akt/mammalian target of
rapamycin kinase and nuclear factor-kappa B signal pathways. It also inhibits
cell proliferation. These results suggest that foods and beverages containing
cocoa bean extracts, including theobromine, might be extremely effective in
preventing human glioblastoma.
Cancer Res. 2010 Feb 1;70(3):1173-83.
Caffeine-mediated inhibition of calcium release channel inositol
1,4,5-trisphosphate receptor subtype 3 blocks glioblastoma invasion and extends
survival.

Kang SS(1), Han KS, Ku BM, Lee YK, Hong J, Shin HY, Almonte AG, Woo DH, Brat DJ,
Hwang EM, Yoo SH, Chung CK, Park SH, Paek SH, Roh EJ, Lee SJ, Park JY, Traynelis
SF, Lee CJ.
(1)Center for Neural Science, Future Fusion Technology Laboratory, Seoul National
University, Seoul, Republic of Korea.
Calcium signaling is important in many signaling processes in cancer cell
proliferation and motility including in deadly glioblastomas of the brain that
aggressively invade neighboring tissue. We hypothesized that disturbing Ca(2+)
signaling pathways might decrease the invasive behavior of giloblastoma,
extending survival. Evaluating a panel of small-molecule modulators of Ca(2+)
signaling, we identified caffeine as an inhibitor of glioblastoma cell motility.
Caffeine, which is known to activate ryanodine receptors, paradoxically inhibits
Ca(2+) increase by inositol 1,4,5-trisphospate receptor subtype 3 (IP(3)R3), the
expression of which is increased in glioblastoma cells. Consequently, by
inhibiting IP(3)R3-mediated Ca(2+) release, caffeine inhibited migration of
glioblastoma cells in various in vitro assays. Consistent with these effects,
caffeine greatly increased mean survival in a mouse xenograft model of
glioblastoma. These findings suggest IP(3)R3 as a novel therapeutic target and
identify caffeine as a possible adjunct therapy to slow invasive growth of
glioblastoma.
Mol Cancer Ther. 2017 Jan;16(1):217-227.
Trifluoperazine, a Well-Known Antipsychotic, Inhibits Glioblastoma Invasion by
Binding to Calmodulin and Disinhibiting Calcium Release Channel IP3R.
Kang S(1), Hong J(2), Lee JM(2)(3), Moon HE(4), Jeon B(5)(6), Choi J(7), Yoon
NA(1), Paek SH(4), Roh EJ(5)(6), Lee CJ(8)(3)(9), Kang SS(10).
(1)Department of Anatomy and Convergence Medical Science, Institute of Health
Sciences, School of Medicine, Gyeongsang National University, Jinju, Republic of
Korea.
(2)Center for Neuroscience and Functional Connectomics, Brain Science Institute,
Korea Institute of Science and Technology, Seoul, Republic of Korea.
(3)KU-KIST Graduate School of Converging Science and Technology, Korea
University, Seoul, Republic of Korea.
(4)Department of Neurosurgery, Seoul National University College of Medicine,
Seoul, Republic of Korea.
(5)Chemical Kinomics Research Center, Korea Institute of Science and Technology,
Seoul, Republic of Korea.
(6)Department of Biological Chemistry, Korea University of Science and
Technology, Daejeon, Republic of Korea.
(7)Gyeongnam Department of Environmental Toxicology and Chemistry, Korea
Institute of Toxicology, Jinju, Republic of Korea.
(8)Center for Neuroscience and Functional Connectomics, Brain Science Institute,
Korea Institute of Science and Technology, Seoul, Republic of Korea.
[email protected] [email protected].
(9)Neuroscience Program, University of Science and Technology, Daejeon, Republic
of Korea.
(10)Department of Anatomy and Convergence Medical Science, Institute of Health
Sciences, School of Medicine, Gyeongsang National University, Jinju, Republic of
Korea. [email protected] [email protected].
Calcium (Ca(2+)) signaling is an important signaling process, implicated in
cancer cell proliferation and motility of the deadly glioblastomas that
aggressively invade neighboring brain tissue. We have previously demonstrated
that caffeine blocks glioblastoma invasion and extends survival by inhibiting
Ca(2+) release channel inositol 1,4,5-trisphosphate receptor (IP3R) subtype 3.

Trifluoperazine (TFP) is an FDA-approved antipsychotic drug for schizophrenia.
Interestingly, TFP has been recently reported to show a strong anticancer effect
on lung cancer, hepatocellular carcinoma, and T-cell lymphoma. However, the
possible anticancer effect of TFP on glioblastoma has not been tested. Here, we
report that TFP potently suppresses proliferation, motility, and invasion of
glioblastoma cells in vitro, and tumor growth in in vivo xenograft mouse model.
Unlike caffeine, TFP triggers massive and irreversible release of Ca(2+) from
intracellular stores by IP3R subtype 1 and 2 by directly interacting at the
TFP-binding site of a Ca(2+)-binding protein, calmodulin subtype 2 (CaM2). TFP
binding to CaM2 causes a dissociation of CaM2 from IP3R and subsequent opening of
IP3R. Compared with the control neural stem cells, various glioblastoma cell
lines showed enhanced expression of CaM2 and thus enhanced sensitivity to TFP. On
the basis of these findings, we propose TFP as a potential therapeutic drug for
glioblastoma by aberrantly and irreversibly increasing Ca(2+) in glioblastoma
cells. Mol Cancer Ther; 16(1); 217-27. ©2016 AACR.
1. Med Hypotheses. 2009 Jun;72(6):629-30.
Evo-Devo and the evolution of cancer: a hypothesis for metamorphic therapies for
the cancers of prolactin-influenced tumourigenesis: with special reference to
glioblastoma multiforme (GBM).
Pearson RD(1).
(1)Gerstein Science Information Centre, University of Toronto, 7 King's College
Circle, Toronto, Ontario, Canada. [email protected]
Recalling the remarkable developmental similarities between cancer cells and
embryonic tissues, this paper argues that, by the process of retrodifferentiation
and heterochronization, stem cells that have become neoplastic could be said to
have undergone "cellular heterochrony." It theorizes, therefore, that hormones
are the major factor in the non-random regulation of cellular heterochrony in
tumourigenesis. Two recent articles confirm that there is low thyroxine and high
prolactin in glioblastomas. Thyroxine metamorphoses vertebrates' tissues so as to
mature the tissues, e.g., in amphibian metamorphosis. In 1896, thyroxine (horse
thyroid extract) was the first successful hormonal product to be used against a
fulminating breast cancer.
Recent work confirms the important role of prolactin
in the induction and progression of mammary, prostate and colorectal tumours.
Although the pituitary is the main source of prolactin in vertebrates, there is
also placental production of prolactin, and paracrine production of prolactin by
tumours themselves. Since tumours produce their own prolactin, shutting down the
pituitary source has not proven wholly successful. Research to find prolactin
receptor antagonists is ongoing. Therefore, prolactin inhibitors (dopamine
agonists), prolactin receptor antagonists, plus thyroxine comprise a plausible
metamorphic therapy for shrinking solid tumour mass. By contrast with
"differentiation" therapies currently sought by stem cell oncologists, this paper
advocates "metamorphic" therapies, to introduce hormonal oncological knowledge of
how to modulate signalling pathways that are aberrant in the stem cells that give
rise to tumours. Despite subtle differences in these signalling translation
pathways and cascades, strategies exist that will allow these evolved
populations, going back to their stem precursors, to "metamorphose" or perhaps
apoptotically cease proliferation.
2. J Neurophysiol. 2004 Mar;91(3):1203-16.
Effects of prolactin on ionic membrane conductances in the human malignant
astrocytoma cell line U87-MG.
Ducret T(1), Vacher AM, Vacher P.
(1)INSERM EMI 0347 Signalisation et Mécanismes Moléculaires de l'Apoptose, and
Laboratoire de Physiologie et Physiopathologie de la Signalisation Cellulaire,
CNRS UMR 5543, Université de Bordeaux 2, 33076 Bordeaux Cedex, France.
Prolactin (PRL) is involved in numerous biological processes in peripheral
tissues and the brain. Although numerous studies have been conducted to elucidate
the signal transduction pathways associated with the PRL receptor, very few have
examined the role of ion conductances in PRL actions. We used the patch-clamp
technique in "whole cell" configuration and microspectrofluorimetry to
investigate the effects of PRL on membrane ion conductances in the U87-MG human
malignant astrocytoma cell line, which naturally expresses the PRL receptor. We
found that a physiological concentration (4 nM) of PRL exerted a biphasic action
on membrane conductances. First, PRL activated a Ca(2+)-dependent K(+) current
that was sensitive to CTX and TEA. This current depended on PRL-induced Ca(2+)
mobilization, through a JAK2-dependent pathway from a thapsigargin- and
2-APB-sensitive Ca(2+) pool. Second, PRL also activated an inwardly directed
current, mainly due to the stimulation of calcium influx via nickel- and
2-APB-sensitive calcium channels. Both phases resulted in membrane
hyperpolarizations, mainly through the activation of Ca(2+)-dependent K(+)
channels. As shown by combined experiments (electrophysiology and
microspectrofluorimetry), the PRL-induced Ca(2+) influx increased with cell
membrane hyperpolarization and conversely decreased with cell membrane
depolarization. Thus PRL-induced membrane hyperpolarizations facilitated Ca(2+)
influx through voltage-independent Ca(2+) channels. Finally, PRL also activated a
DIDS-sensitive Cl(-) current, which may participate in the PRL-induced
hyperpolarization. These PRL-induced conductance activations are probably related
to the PRL proliferative effect we have already described in U87-MG cells.
3. Glia. 2002 May;38(3):200-14.
Effects of prolactin on intracellular calcium concentration and cell
proliferation in human glioma cells.
Ducret T(1), Boudina S, Sorin B, Vacher AM, Gourdou I, Liguoro D, Guerin J,
Bresson-Bepoldin L, Vacher P.
(1)Laboratoire de Neurophysiologie, Centre National de la Recherche Scientifique
UMR 5543, Université de Bordeaux 2, France.
Prolactin (PRL) has several physiological effects on peripheral tissues and the
brain. This hormone acts via its membrane receptor (PRL-R) to induce cell
differentiation or proliferation. Using reverse transcription-polymerase chain
reaction (RT-PCR) combined with Southern blot analysis, we detected PRL-R
transcripts in a human glioma cell line (U87-MG) and in primary cultured human
glioblastoma cells. These transcripts were deleted or not in their extracellular
domains. We examined the effects of PRL on intracellular free Ca2+ concentration
([Ca2+](i)) in these cells in order to improve our understanding of the PRL
transduction mechanism, which is still poorly documented. [Ca2+](i) was measured
by microspectrofluorimetry using indo-1 as the Ca2+ fluorescent probe.
Spatiotemporal aspects of PRL-induced Ca2+ signals were investigated using
high-speed fluo-3 confocal imaging. We found that physiological concentrations
(0.4-4 nM) of PRL-stimulated Ca2+ entry and intracellular Ca2+ mobilization via a
tyrosine kinase-dependent mechanism. The two types of Ca2+ responses observed
were distinguishable by their kinetics: one showing a slow (type I) and the other
a fast (type II) increase in [Ca2+](i). The amplitude of PRL-induced Ca2+
increases may be sufficient to provoke several physiological responses, such as
stimulating proliferation. Furthermore, PRL induced a dose-dependent increase in
[3H]thymidine incorporation levels and in cellular growth and survival, detected
by the MTT method. These data indicate that PRL induced mitogenesis of human
glioma cells.

5. J Neurochem. 1999 Dec;73(6):2272-7.
Prolactin induction of nitric oxide synthase in rat C6 glioma cells.
Raso GM(1), Meli R, Gualillo O, Pacilio M, Di Carlo R.
(1)Department of Experimental Pharmacology, University of Naples Frederico II,
Italy.
We have examined the neuroimmunoregulatory function of prolactin (PRL) on
astrocytic inducible nitric oxide synthase (iNOS) expression in the C6 glioma
cell line. After 24 h of PRL (5-100 nM) stimulation, a concentration-dependent
increase of NO release, evaluated as nitrite, was observed in C6 culture medium.
Moreover, both NO release and iNOS expression induced by interferon-gamma (250
U/ml) were enhanced by PRL (18-100 nM). PRL-induced NO release was inhibited by
dexamethasone, an inhibitor of de novo iNOS synthesis. We used erbstatin (5
microg/ml), a potent inhibitor of protein tyrosine kinases, to test whether these
proteins were required for signaling events evoked by PRL in these cells. This
inhibitor was able to inhibit completely the PRL-induced NO production and iNOS
expression. In conclusion, we provide evidence that NO production in glial cells
can be regulated not only by cytokines but also by neuroimmunoregulatory hormones
such as PRL, which is present in normal brain but may be elevated in several
pathological states.
4. J Neurosurg Sci. 2001 Jun;45(2):70-4.
Hyperprolactinaemia and prolactin binding in benign intracranial tumours.
Ciccarelli E(1), Razzore P, Gaia D, Todaro C, Longo A, Forni M, Ghè C, Camanni F,
Muccioli G, Faccani G, Lanotte MM.
(1)Department of Internal Medicine, Division of Endocrinology, University of
Turin, Turin, Italy.
BACKGROUND: Meningiomas have been found to have receptors for several hormones,
such as oestrogen, progesterone, somatostatin, dopamine and recently also for
prolactin.
METHODS: To investigate any possible role of prolactin in the growth of those
tumours we detected the presence of prolactin-receptors (PRL-R) in 22 meningiomas
and we correlated these data with PRL serum levels in patients before surgery. We
also studied 13 patients with schwannomas and 7 with other cerebral tumours (4
glioblastomas, 2 ependymomas and 1 astrocytoma).
RESULTS: Increased prolactin binding was present in 10 (45.4 percent;)
meningiomas, 9 (69.2 percent;) schwannomas and in the patient with astrocytoma.
The presence of high PRL levels was present in 6 (27.2 percent;) patients with
meningiomas, 8 (61.5 percent;) with schwannomas and in 3 (42.8 percent;) with
other tumours. No direct correlation was present between serum PRL levels and PRL
binding in all groups.
CONCLUSIONS: In conclusion we confirmed the presence of PRL receptors in patients
with meningiomas and we have also shown the presence of PRL receptors also in
schwannomas. Moreover increased serum PRL were shown in some patients with
different tumours of nervous tissue before surgery. Our data could suggest that
PRL might have a role in the growth of meningiomas and schwannomas.
1. Int J Physiol Pathophysiol Pharmacol. 2017 Apr 15;9(2):8-15. eCollection 2017.
Lidocaine suppresses glioma cell proliferation by inhibiting TRPM7 channels.
Leng T(1), Lin S(2), Xiong Z(1), Lin J(3).
(1)Neurobiology, Neuroscience Institute, Morehouse School of MedicineAtlanta GA
30329, USA.
(2)Guangzhou Cellproteck Pharmaceutical CO., Ltd3 Lanyue Road, Science City,
Guangzhou 510663, PR China.
(3)Department of Anesthesiology, Stony Brook University Health Sciences Center,
Stony BrookNY 11794-8480, USA.
BACKGROUND: Malignant glioma is the most common brain cancer with devastating
prognosis. Recurrence of malignant glioma following surgery is very common with
few preventive and therapeutic options. Novel targets and therapeutic agents are
constantly sought for better outcome. Our previous study established that
inhibition of transient receptor potential melastatin 7 (TRPM7) channels resulted
in significant decrease of human glioma cell growth and proliferation. As local
anesthetic lidocaine has been shown to inhibit TRPM7 currents, we hypothesize
that lidocaine may suppress glioma cell proliferation through TRPM7 channel
inhibition.
METHODS: TRPM7 currents were recorded in rat C6 glioma cells using the whole cell
patch clamp technique. Cell growth and proliferation were assessed under
microscopic examination and biochemical assays.
RESULTS: Lidocaine inhibits TRPM7-like currents in a dose-dependent and
reversible manner. At 1 and 3 mM, it inhibits ~30% and ~50% of TRPM7 currents. At
these concentrations, it is effective in inhibiting the proliferation of C6
cells. As expected, the TRPM7 inhibitors gadolinium and 2-Aminoethoxydiphenyl
borate have similar effects on TRPM7 currents and proliferation of C6 cells.
Similar to its effect on C6 cells, lidocaine inhibits the proliferation of A172
cells, a human glioblastoma cell line.
CONCLUSIONS: Lidocaine significantly inhibits the proliferation of glioma cells.
The effect of lidocaine is mediated, at least in part, by inhibiting TRPM7
channels.
Anesth Analg. 2016 Apr;122(4):1038-47.
Clinical Concentrations of Local Anesthetics Bupivacaine and Lidocaine Differentially Inhibit Human Kir2.x Inward Rectifier K+ Channels.
Nakahira K, Oshita K, Itoh M, Takano M, Sakaguchi Y, Ishihara K.
BACKGROUND:
Inward rectifier K channels of the Kir2.x subfamily are widely expressed in neuronal tissues, controlling neuronal excitability. Previous studies reported that local anesthetics (LAs) do not affect Kir2 channels. However, the effects have not been studied at large concentrations used in regional anesthesia.
METHODS:
This study used the patch-clamp technique to examine the effects of bupivacaine and lidocaine on Kir2.1, Kir2.2, and Kir2.3 channels expressed in human embryonic kidney 293 cells.
RESULTS:
When applied extracellularly in whole-cell recordings, both LAs inhibited Kir2.x currents in a voltage-independent manner. Inhibition with bupivacaine was slow and irreversible, whereas that with lidocaine was fast and reversible. Kir2.3 displayed a greater sensitivity to bupivacaine than Kir2.1 and Kir2.2 (50% inhibitory concentrations at approximately 5 minutes, 0.6 vs 8-10 mM), whereas their sensitivities to lidocaine were similar (50% inhibitory concentrations, 1.5-2.7 mM). Increases in the charged/neutral ratio of the LAs at an acidic extracellular pH attenuated their inhibitory effects, and a permanently charged lidocaine derivative QX-314 exhibited no effects when applied extracellularly. Inside-out experiments demonstrated that inhibition of Kir2.1 with cytoplasmic lidocaine and QX-314 was rapid and reversible, whereas that induced by bupivacaine was slow and irreversible. Furthermore, dose-inhibition relations for the charged form of bupivacaine and lidocaine obtained at different cytoplasmic pHs could be approximated by a single relation for each LA.
CONCLUSIONS:
The results indicate that both LAs at clinical concentrations equilibrated rapidly with the intracellular milieu, differentially inhibiting Kir2.x channel function from the cytoplasmic side.
Anesthesiology. 2001 Jun;94(6):1089-95.
Local anesthetic inhibition of voltage-activated potassium currents in rat dorsal root ganglion neurons.
Komai H, McDowell TS.
BACKGROUND:
Local anesthetic actions on the K+ channels of dorsal root ganglion (DRG) and dorsal horn neurons may modulate sensory blockade during neuraxial anesthesia. In dorsal horn neurons, local anesthetics are known to inhibit transient but not sustained K+ currents. The authors characterized the effects of local anesthetics on K+ currents of isolated DRG neurons.
METHODS:
The effects of lidocaine, bupivacaine, and tetracaine on K+ currents in isolated rat DRG neurons were measured with use of a whole cell patch clamp method. The currents measured were fast-inactivating transient current (I(Af)), slow-inactivating transient current (I(As)), and noninactivating sustained current (I(Kn)).
RESULTS:
One group of cells (type 1) expressed I(Af) and I(Kn). The other group (type 2) expressed I(As) and I(Kn). The diameter of type 2 cells was smaller than that of type 1 cells. Lidocaine and bupivacaine inhibited all three K+ currents. Tetracaine inhibited I(As) and I(Kn) but not I(Af) For bupivacaine, the concentration for half-maximal inhibition (IC50) of I(Kn) in type 2 cells was lower than that for I(Kn) in type 1 cells (57 vs. 121 microM). Similar results were obtained for tetracaine (0.6 vs. 1.9 mM) and for lidocaine (2.2 vs. 5.1 mM).
CONCLUSIONS:
Local anesthetics inhibited both transient and sustained K+ currents in DRG neurons. Because K+ current inhibition is known to potentiate local anesthetic-induced impulse inhibition, the lower IC50 for I(Kn) of small type 2 cells may reflect preferential inhibition of impulses in nociceptive neurons. The overall modulatory actions of local anesthetics probably are determined by their differential effects on presynaptic (DRG) and postsynaptic (dorsal horn neurons) K+ currents.

Arch Biochem Biophys. 1985 Mar;237(2):415-22.
Induction of intracellular acyl-CoA:cholesterol acyltransferase activity in
glioblastoma cells by lidocaine.
Jeng I, Klemm N, Bressie S, Samson L.
The perturbation of cellular cholesteryl ester biosynthesis in glioblastoma C-6
cells by lidocaine was investigated. Lidocaine specifically inhibited the
incorporation of radioactive oleic acid into cellular cholesteryl ester but had
no significant effect on the incorporation of oleic acid into
phosphatidylcholine. Oxygenated cholesterol-enhanced cholesteryl ester formation
was less sensitive to lidocaine inhibition. Several other local anesthetics were
compared. Lidocaine altered cholesteryl ester formation in time- and
dose-dependent manners. Lidocaine was a powerful inhibitor initially and its
potency declined with time. Lidocaine was capable of directly inhibiting
acyl-CoA:cholesterol acyltransferase (ACAT) activity in broken cell homogenates.
The lidocaine-mediated inhibition of cellular cholesteryl ester formation
triggered an enhanced intracellular ACAT activity that was not fully expressed in
the presence of lidocaine. The activation of ACAT activity by lidocaine might
represent a compensatory mechanism by which the inhibitory effect of lidocaine
was partially overcome with time.
Int J Neurosci. 2007 Oct;117(10):1465-80.
Medroxyprogesterone acetate induces c6 glioma chemosensitization via
antidepressant-like lysosomal phospholipidosis/myelinosis in vitro.
Altinoz MA(1), Gedikoglu G, Sav A, Ozcan E, Ozdilli K, Bilir A, Del Maestro RF.
(1)Institute of Health, Halic University, Istanbul, Turkey. [email protected]
The authors have previously shown that medroxyprogesterone acetate (MPA) inhibits
growth and increases drug sensitivity in C6 glioma with myeloid bodies. Myeloid
bodies can occur in cells either due to robust toxicity with mitochondrial
membrane disruption or due to milder events such as seen in
lysosomal-phospholipidosis. Exact patterns of myelinosis accompanying to MPA
chemo-sensitization is important, because uncoupling of nuclear versus
mitochondrial toxicity of anti-neoplastics by MPA would lead to safer employment
of glioma chemotherapy with reduced neurotoxicity. By monitoring and comparing
cell kinetics with fine structural features of cell death, the authors estimated
subcellular effects accompanying growth-inhibitory drug actions in C6 glioma. The
analysis revealed that MPA induced mainly lysosomal phospholipidosis, while
inhibiting clonogenicity alone and augmenting procarbazine efficacy. It induced
apoptosis in combination with cisplatin. It reduced mitochondrial-damage-based
early cytotoxicity of methotrexate, yet it did not hinder its anti-clonogenic
efficacy. Progesterone analogues - similar to antidepressants - inhibit
cholesterol esterification, and this efficacy relates with their P-glycoprotein
inhibition. Reducing esterification and plasma-membrane localization of
cholesterol may lead MPA induction of lysosomal phospholipidosis, growth
indolency, and drug sensitization in glioma.
Int J Dev Neurosci. 2001 Oct;19(6):541-7.
Medroxyprogesterone acetate alone or synergistic with chemotherapy suppresses
colony formation and DNA synthesis in C6 glioma in vitro.
Altinoz MA(1), Bilir A, Ozar E, Onar FD, Sav A.
(1)Histology and Embryology Department, Istanbul Medical School, 34390, Istanbul,
Turkey. [email protected]
We have studied the effects of medroxyprogesterone acetate (MPA) on C6 glioma
growth in vitro in order to prove the hypothesis that it could arrest growth and
induce drug sensitisation in a glial tumour as it does in breast cancer cells.
Plating, thymidine-labelling index, ultra-structure, and soft agar colony growth
were determined after incubation with MPA, and/or cisplatin, procarbazine and
methotrexate (MTX). MPA (microg/ml) reduced the thymidine-labelling index by 41
and 73% at 48 and 96 h, respectively, and decreased colony growth by 61%. Soft
agar colony inhibition by MPA was almost as potent as MTX (0.3 microg/ml), but
the latter drug showed very high cytotoxicity. Electron microscopy revealed that
in medroxyprogesterone treated cells myeloid bodies developed, but MTX treatment
caused mainly necrosis. Medroxyprogesterone increased procarbazine and
cisplatin-induced colony growth and S-phase inhibition, but reduced MTX-induced
thymidine-labelling inhibition. In conclusion, progesterone may inhibit growth
and sensitize to drugs.
Brain Res. 1988 Mar 8;443(1-2):338-44.
Memantine (1-amino-3,5-dimethyladamantane) blocks the serotonin-induced
depolarization response in a neuronal cell line.
Reiser G(1), Binmöller FJ, Koch R.
(1)Physiologisch-chemisches Institut der Universität Tübingen, F.R.G.
The influence of memantine on several properties of a neuronal cell line was
tested. The aim was to get some insight into possible mechanisms of action of
this drug which is therapeutically applicable in treatment of spasticity,
Parkinson's disease, and cerebral coma. In neuroblastoma X glioma hybrid cells,
memantine, at micromolar concentrations, blocked the depolarization induced by
iontophoretically applied serotonin (5-hydroxytryptamine, 5-HT). In the hybrid
cells, receptors of the 5-HT3 type mediated the depolarization, which was
frequently accompanied by a series of action potentials. The inhibition by
memantine of the serotonin response occurred fast and was completely reversible,
irrespective of whether the cell showed a stable membrane potential or
spontaneous action potentials. However, memantine did not alter spontaneous or
electrically evoked action potential activity in the hybrid cells, and apparently
did not block the underlying ionic conductances. Furthermore memantine did not
affect either the cation permeability activated by substance P in the hybrid
cells or the K+ channel triggered by bradykinin in a glioma cell line. Thus,
memantine appears specifically to suppress the ion channel opened by serotonin in
the hybrid cells. The interaction of memantine with serotonin receptors and the
associated ion channels reported here, might give an important clue, as to a site
of action of memantine in the nervous system.
J Neurosurg. 2014 Apr;120(4):854-63.
Efficacy of local polymer-based and systemic delivery of the anti-glutamatergic
agents riluzole and memantine in rat glioma models.
Yohay K(1), Tyler B, Weaver KD, Pardo AC, Gincel D, Blakeley J, Brem H, Rothstein
JD.
(1)Department of Pediatrics, Weill Cornell Medical College;
OBJECT: The poor outcome of malignant gliomas is largely due to local
invasiveness. Previous studies suggest that gliomas secrete excess glutamate and
destroy surrounding normal peritumoral brain by means of excitotoxic mechanisms.
In this study the authors assessed the effect on survival of 2 glutamate
modulators (riluzole and memantine) in rodent glioma models.
METHODS: In an in vitro growth inhibition assay, F98 and 9L cells were exposed to
riluzole and memantine. Mouse cerebellar organotypic cultures were implanted with
F98 glioma cells and treated with radiation, radiation + riluzole, or vehicle and
assessed for tumor growth. Safety and tolerability of intracranially implanted
riluzole and memantine CPP:SA polymers were tested in F344 rats. The efficacy of
these drugs was tested against the 9L model and riluzole was further tested with
and without radiation therapy (RT).
RESULTS: In vitro assays showed effective growth inhibition of both drugs on F98
and 9L cell lines. F98 organotypic cultures showed reduced growth of tumors
treated with radiation and riluzole in comparison with untreated cultures or
cultures treated with radiation or riluzole alone. Three separate efficacy
experiments all showed that localized delivery of riluzole or memantine is
efficacious against the 9L gliosarcoma tumor in vivo. Systemic riluzole
monotherapy was ineffective; however, riluzole given with RT resulted in improved
survival.
CONCLUSIONS: Riluzole and memantine can be safely and effectively delivered
intracranially via polymer in rat glioma models. Both drugs demonstrate efficacy
against the 9L gliosarcoma and F98 glioma in vitro and in vivo. Although systemic
riluzole proved ineffective in increasing survival, riluzole acted
synergistically with radiation and increased survival compared with RT or
riluzole alone.
Bull Cancer. 2013 Sep;100(9):829-35.
[Glutamate and malignant gliomas, from epilepsia to biological aggressiveness:
therapeutic implications].
[Article in French]
Blecic S(1), Rynkowski M, De Witte O, Lefranc F.
(1)Service de neurologie, Epicura, 7800 Ath, Belgique.
In this review article, we describe the unrecognized roles of glutamate and
glutamate receptors in malignant glioma biology. The neurotransmitter glutamate
released from malignant glioma cells in the extracellular matrix is responsible
for seizure induction and at higher concentration neuronal cell death. This
neuronal cell death will create vacated place for tumor growth. Glutamate also
stimulates the growth and the migration of glial tumor cells by means of the
activation of glutamate receptors on glioma cells in a paracrine and autocrine
manner. The multitude of effects of glutamate in glioma biology supports the
rationale for pharmacological targeting of glutamate receptors and transporters
in the adjuvant treatment of malignant gliomas in neurology and neuro-oncology.
Using the website www.clinicaltrials.gov/ as a reference - a service developed by
the National Library of Medicine for the National Health Institute in USA - we
have evoked the few clinical trials completed and currently ongoing with
therapies targeting the glutamate receptors.
Oncol Lett. 2014 Aug; 8(2): 645–650.
Pregnenolone, a cholesterol metabolite, induces glioma cell apoptosis via activating extrinsic and intrinsic apoptotic pathways
XIAO XIAO,1,* LIJUN CHEN,1,* YING OUYANG,2,* WENBO ZHU,1 PENGXIN QIU,1 XINWEN SU,1 YUNLING DOU,3 LIPENG TANG,1 MIN YAN,1 HAIPENG ZHANG,1 XIAOXIAO YANG,1 DONG XU,1 and GUANGMEI YAN1
Gliomas are one of the most common types of malignant tumors worldwide, however, an effective therapeutic strategy not yet been fully determined. The present study aimed to investigate the anti-glioma activity and underlying mechanisms of pregnenolone, which originates from cholesterol and is metabolized into important steroid hormones in the body. The results demonstrated that 100 μM pregnenolone induced a significant loss of cell viability in various malignant glioma cell lines. In the U-87 MG, LN-18 and C6 cell lines, the loss of cell viability resulted from cell apoptosis, which was evidenced by apoptotic nuclear morphology changes and caspase 3 activation. Moreover, the increased activities of caspase 8 and 9 strongly indicated that pregnenolone activated the extrinsic and intrinsic pathways of apoptosis. Additionally, glioma cell apoptosis was prevented by the general caspase inhibitor, Z-VAD-FMK. In the C6 cells, upregulation of Fas and Fas ligand triggered the activation of the extrinsic pathway, whereas knockdown of Fas significantly abrogated the cell apoptosis that was induced by pregnenolone. Furthermore, downregulation of the anti-apoptotic protein, B-cell lymphoma 2 and upregulation of pro-apoptotic proteins, such as Bax and Bak, activated the intrinsic pathway. In conclusion, pregnenolone induced glioma cell apoptosis in a caspase-dependent manner, which was mediated by activation of the extrinsic and intrinsic apoptotic pathways.
Introduction
Gliomas originate from glial cells and are the most common type of primary brain tumors accounting for 80% of all malignant primary brain tumors (1). According to the pathological and clinical criteria established by the World Health Organization, gliomas are classified as grades I–IV (2,3). Grade IV tumors, such as glioblastomas (GBMs), are the most devastating and aggressive form comprising >50% of gliomas, and have a poor prognosis (4). The current treatment standard of GBMs is surgical resection to a feasible extent, followed by radiotherapy and chemotherapy. Among the currently available chemotherapy agents, temozolomide is the most popular; doctors and patients favor it as it is administrated orally and efficiently crosses the blood-brain barrier (BBB) (5). However, 67.2–76% of patients are resistant to this agent and therefore do not benefit from it (6). Regardless of systemic therapeutic strategies, including surgery, temozolomide and radiotherapy, patient median survival is only 14.6 months and the five-year survival rate is ~9.8% (7,8). The poor prognosis of glioma fuels the requirement for identifying therapeutic agents with the merits of temozolomide, such as high lipophilicity and strong anti-glioma activity.
Steroid hormones are generally divided into the following five groups: Estrogens, androgens, progestogens, glucocorticoids and mineralocorticoids (9,10). The natural steroid hormones are predominantly synthesized from cholesterol in the gonads and adrenal glands, and readily diffuse through the cell membrane due to their lipophilic properties (11,12). Accumulating evidence has indicated that certain steroid hormones possess antitumor activities, such as the 17β-estradiol metabolite, 2-methoxyestradiol (2ME), which exerts the strongest activity. 2ME inhibits proliferation and induces apoptosis of various types of cancer, including gliomas, and breast and gastric cancer, independently of estrogen receptors α and β (13–15). However, findings from a clinical trial identified low oral bioavailability of 2ME, which prevents the transfer of this promising agent from bench to bed side (16). The example of 2ME implies the application potential of steroid hormones and provides the basis for the subsequent investigation of other endogenous steroid hormones, such as pregnenolone (Fig. 1).
The present study aimed to investigate the pharmacological effects of the endogenous steroid, pregnenolone, on GBM cells, and the mechanisms underlying its pro-apoptotic activity via the extrinsic and intrinsic apoptotic pathways. Pregnenolone may be a leading compound in the treatment of gliomas and may be modified and developed for clinical application.
J Neurosurg. 1995 Apr;82(4):635-40.
The role of minocycline in the treatment of intracranial 9L glioma.
Weingart JD(1), Sipos EP, Brem H.
(1)Department of Neurological Surgery, Johns Hopkins University School of
Medicine, Baltimore, Maryland.
This study was designed to explore the question of whether minocycline, a
semisynthetic tetracycline shown to inhibit tumor-induced angiogenesis, could
control the growth of the rat intracranial 9L gliosarcoma. Minocycline was tested
alone and in combination with 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) in
vivo. Treatment was started at the time of intracranial implantation of 9L
gliosarcoma into male Fischer 344 rats, 5 days later, or after tumor resection.
Minocycline was delivered locally with a controlled-release polymer or
systemically by intraperitoneal injection. Systemic minocycline did not extend
survival time. Local treatment with minocycline by a controlled-release polymer
implanted at the time of tumor implantation extended median survival time by 530%
(p < 0.001) compared to treatment with empty polymer. When treatment was begun 5
days after tumor implantation, minocycline delivered locally or systemically had
no effect on survival. However, after tumor resection, treatment with locally
delivered minocycline resulted in a 43% increase in median survival time (p <
0.002) compared to treatment with empty polymer. Treatment with a combination of
minocycline delivered locally in a controlled-release polymer and systemic BCNU 5
days after tumor implantation resulted in a 93% extension of median survival time
compared to BCNU alone (p < 0.002). In contrast, treatment with a combination of
systemic minocycline and BCNU did not increase survival time compared to systemic
BCNU alone. These results demonstrate that minocycline affects tumor growth when
delivered locally and suggest that minocycline may be a clinically effective
modulator of intracranial tumor growth when used in combination with a
chemotherapeutic agent and surgical resection.
J Neurooncol. 2003 Sep;64(3):203-9.
Local delivery of minocycline and systemic BCNU have synergistic activity in the
treatment of intracranial glioma.
Frazier JL(1), Wang PP, Case D, Tyler BM, Pradilla G, Weingart JD, Brem H.
(1)Department of Neurological Surgery, Johns Hopkins University School of
Medicine, Baltimore, MD 21205, USA.
Minocycline, a tetracycline derivative, has been shown to inhibit tumor
angiogenesis through inhibitory effects on matrix metalloproteinases. Previous
studies have shown this agent to be effective against a rodent brain tumor model
when delivered intracranially and to potentiate the efficacy of standard
chemotherapeutic agents. In the present study, the in vivo efficacy of
intracranial minocycline delivered by a biodegradable controlled-release polymer
against rat intracranial 9L gliosarcoma was investigated to determine whether it
potentiates the effects of systemic 1,3-bis (2-chloroethyl)-1-nitrosourea (BCNU).
Minocycline was incorporated into the biodegradable polymer polyanhydride
poly[bis(p-carboxyphenoxy)propane-sebacic acid] (pCPP:SA) at a ratio of 50:50 by
weight. The release kinetics of minocycline from the polymer were assessed. For
the efficacy studies, female Fischer 344 rats were implanted with 9L glioma.
Treatment with minocycline delivered by the pCPP:SA polymer at the time of tumor
implantation resulted in 100% survival in contrast to untreated control animals
that died within 21 days. Treatment with the minocycline-polymer 5 days after
tumor implantation provided only modest increases in survival. The combination of
intracranial minocycline and systemic BCNU extended median survival by 82%
compared to BCNU alone (p < 0.0001) and 200% compared to no treatment (p <
0.004). We conclude that local intracranial delivery of minocycline from
biodegradable controlled-release polymers inhibits tumor growth and may have
clinical utility when combined with a chemotherapeutic agent.
Int J Cancer. 2016 Jun 1;138(11):2678-87.
Investigating the role of tumour cell derived iNOS on tumour growth and
vasculature in vivo using a tetracycline regulated expression system.
Papaevangelou E(1), Whitley GS(1), Johnstone AP(1), Robinson SP(2), Howe FA(1).
(1)Cardiovascular and Cell Sciences Research Institute, St. George's, University
of London, Cranmer Terrace, London, SW17 0RE, United Kingdom. (2)Division of
Radiotherapy and Imaging, The Institute of Cancer Research, Belmont, Sutton,
Surrey, SM2 5NG, United Kingdom.
Nitric oxide (NO) is a free radical signalling molecule involved in various
physiological and pathological processes, including cancer. Both tumouricidal and
tumour promoting effects have been attributed to NO, making its role in cancer
biology controversial and unclear. To investigate the specific role of
tumour-derived NO in vascular development, C6 glioma cells were genetically
modified to include a doxycycline regulated gene expression system that controls
the expression of an antisense RNA to inducible nitric oxide synthase (iNOS) to
manipulate endogenous iNOS expression. Xenografts of these cells were propagated
in the presence or absence of doxycycline. Susceptibility magnetic resonance
imaging (MRI), initially with a carbogen (95% O2 /5% CO2 ) breathing challenge
and subsequently an intravascular blood pool contrast agent, was used to assess
haemodynamic vasculature (ΔR2 *) and fractional blood volume (fBV), and
correlated with histopathological assessment of tumour vascular density,
maturation and function. Inhibition of NO production in C6 gliomas led to
significant growth delay and inhibition of vessel maturation. Parametric fBV maps
were used to identify vascularised regions from which the carbogen-induced ΔR2 *
was measured and found to be positively correlated with vessel maturation,
quantified ex vivo using fluorescence microscopy for endothelial and perivascular
cell staining. These data suggest that tumour-derived iNOS is an important
mediator of tumour growth and vessel maturation, hence a promising target for
anti-vascular cancer therapies. The combination of ΔR2 * response to carbogen and
fBV MRI can provide a marker of tumour vessel maturation that could be applied to
non-invasively monitor treatment response to iNOS inhibitors.
Cancer Lett. 1995 Jan 27;88(2):141-5.
Methylene blue induces cytotoxicity in human brain tumor cells.
Lee YS(1), Wurster RD.
(1)Department of Neurological Surgery, Loyola University Medical Center, Maywood,
IL 60153.
Methylene blue (MB), a known inhibitor of guanylyl cyclase, induced cytotoxicity
in SK-N-MC human neuroblastoma and U-373 MG human astrocytoma cells in a
dose-dependent manner. MB did not significantly alter cellular levels of cGMP in
both cells. 8-Br cGMP, a membrane-permeable analogue of cGMP, did not decrease
MB-induced cytotoxicity, indicating that cGMP may not be a major target of the
cytotoxic action of MB. However, hydroxyl radical scavengers or intracellular
Ca2+ modulators effectively blocked the MB-induced cytotoxicity. These results
suggest that hydroxyl radical and intracellular Ca2+ may have an important
involvement in the cytotoxic action of MB. These results further suggest that the
treatment with MB may be useful for the therapeutic applications of human brain
tumors.
Prog Neurobiol. 2015 Nov 18. pii: S0301-0082(15)30060-5.
Alternative mitochondrial electron transfer for the treatment of
neurodegenerative diseases and cancers: Methylene blue connects the dots.
Yang SH(1), Li W(2), Sumien N(2), Forster M(2), Simpkins JW(3), Liu R(2).
(1)Center for Neuroscience Discovery, University of North Texas Health Science
Center, Fort Worth, TX 76107, USA. Electronic address: [email protected].
(2)Center for Neuroscience Discovery, University of North Texas Health Science
Center, Fort Worth, TX 76107, USA. (3)Department of Physiology and Pharmacology,
Center for Neuroscience, Health Science Center, West Virginia University, Medical
Center Drive, Morgantown, WV 26506, USA.
Brain has exceptional high requirement for energy metabolism with glucose as the
exclusive energy source. Decrease of brain energy metabolism and glucose uptake
has been found in patients of Alzheimer's, Parkinson's and other
neurodegenerative diseases, providing a clear link between neurodegenerative
disorders and energy metabolism. On the other hand, cancers, including
glioblastoma, have increased glucose uptake and rely on aerobic glycolysis for
energy metabolism. The switch of high efficient oxidative phosphorylation to low
efficient aerobic glycolysis pathway (Warburg effect) provides macromolecule for
biosynthesis and proliferation. Current research indicates that methylene blue, a
century old drug, can receive electron from NADH in the presence of complex I and
donates it to cytochrome c, providing an alternative electron transfer pathway.
Methylene blue increases oxygen consumption, decrease glycolysis, and increases
glucose uptake in vitro. Methylene blue enhances glucose uptake and regional
cerebral blood flow in rats upon acute treatment. In addition, methylene blue
provides protective effect in neuron and astrocyte against various insults in
vitro and in rodent models of Alzheimer's, Parkinson's, and Huntington's disease.
In glioblastoma cells, methylene blue reverses Warburg effect by enhancing
mitochondrial oxidative phosphorylation, arrests glioma cell cycle at s-phase,
and inhibits glioma cell proliferation. Accordingly, methylene blue activates
AMP-activated protein kinase, inhibits downstream acetyl-coA carboxylase and
cyclin-dependent kinases. In summary, there is accumulating evidence providing a
proof of concept that enhancement of mitochondrial oxidative phosphorylation via
alternative mitochondrial electron transfer may offer protective action against
neurodegenerative diseases and inhibit cancers proliferation.
J Clin Endocrinol Metab. 2002 Nov;87(11):5325-31.
Progesterone production and actions in the human central nervous system and
neurogenic tumors.
Inoue T, Akahira J, Suzuki T, Darnel AD, Kaneko C, Takahashi K, Hatori M,
Shirane R, Kumabe T, Kurokawa Y, Satomi S, Sasano H.
Department of Pathology, Second Department of Surgery, Tohoku University School
of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan.
[email protected]
Progesterone has been suggested to be involved in the functions of the nervous
system, but it has yet to be examined in humans. Progesterone has also been
postulated to be involved in the biological behavior of various human neurogenic
tumors via progesterone receptors A and B (PR-A and PR-B). In this study we
examined the expression of PR and the enzymes responsible for progesterone

biosynthesis (P450scc, 3betahydroxysteroid dehydrogenase, and steroidogenic
acute regulatory protein) in human brain. We also examined the distribution of
PR isoforms in neurogenic tumors using immunohistochemistry and RT-PCR analysis.
The presence of PR and mRNA for P450scc, 3beta-hydroxysteroid dehydrogenase, and
steroidogenic acute regulatory protein was detected in human brain. PR isoforms
were detected in neurogenic tumors. PR-A and PR-B were equally expressed in
meningiomas, but PR-B was the predominant isoform compared with PR-A in
astrocytic tumors and Schwannomas. There was a statistically significant inverse
correlation between PR-A and the proliferation index in meningiomas and
astrocytic tumors. These findings suggest that progesterone is locally
synthesized and exerts its actions through PR in the human central nervous
system, and that progesterone may be involved in regulation of the growth and
development of neurogenic tumors via PR, especially in the inhibition of tumor
cell proliferation via PR-A.

Eur J Pharm Biopharm. 2016 Dec;109:81-92.
Inhibition of carbonic anhydrase IX in glioblastoma multiforme.
Amiri A(1), Le PU(2), Moquin A(1), Machkalyan G(1), Petrecca K(2), Gillard JW(3),
Yoganathan N(4), Maysinger D(5).
(1)Department of Pharmacology and Therapeutics, McGill University, Montreal,
Quebec H3G 1Y6, Canada.
(2)Department of Neurology and Neurosurgery, Montreal Neurological Institute and
Hospital, McGill University, Montreal H3A 2B4, Quebec, Canada.
(3)Department of Pharmacology and Therapeutics, McGill University, Montreal,
Quebec H3G 1Y6, Canada; Kalgene Pharmaceuticals, Innovation Park at Queens
University, Kingston K7L 3N6, Ontario, Canada.
(4)Kalgene Pharmaceuticals, Innovation Park at Queens University, Kingston K7L
3N6, Ontario, Canada.
(5)Department of Pharmacology and Therapeutics, McGill University, Montreal,
Quebec H3G 1Y6, Canada. Electronic address: [email protected].
Carbonic anhydrase IX (CAIX) is a transmembrane enzyme upregulated in several
types of tumors including glioblastoma multiforme (GBM). GBM is among the most
aggressive tumors among gliomas. Temozolomide (TMZ) therapy combined with
surgical or radiation approaches is the standard treatment but not effective in
long term. In this study we tested the treatment with acetazolamide (ATZ), an
inhibitor of CAIX, alone or combined with TMZ. The experiments were performed in
2D and 3D cultures (spheroids) using glioblastoma U251N and human brain tumor
stem cells (BTSCs). Several proteins implicated in tumor cell death were also
investigated. The key results from these studies suggest the following: (1) Cell
death of human glioblastoma spheroids and BTSC is significantly increased with
combined treatment after 7 days, and (2) the effectiveness of ATZ is
significantly enhanced against BTSC and U251N when incorporated into
nano-carriers. Collectively, these results point toward the usefulness of
nano-delivery of CAIX inhibitors and their combination with chemotherapeutics for
glioblastoma treatment.
Bioorg Med Chem. 2013 Jul 1;21(13):3949-57.
Hypoxia induced CA9 inhibitory targeting by two different sulfonamide derivatives
including acetazolamide in human glioblastoma.

Said HM(1), Hagemann C, Carta F, Katzer A, Polat B, Staab A, Scozzafava A,
Anacker J, Vince GH, Flentje M, Supuran CT.
(1)Dept. of Radiation Oncology, University of Würzburg, Germany.
[email protected]
HIF-1α regulated genes are mainly responsible for tumour resistance to radiation-
and chemo-therapy. Among these genes, carbonic anhydrase isoform IX (CA9) is
highly over expressed in many types of cancer especially in high grade brain
cancer like Glioblastoma (GBM). Inhibition of the enzymatic activity by
application of specific chemical CA9 inhibitor sulphonamides (CAI) like
Acetazolamide (Aza.), the new sulfonamide derivative carbonic anhydrase inhibitor
(SU.D2) or indirect inhibitors like the HIF-1α inhibitor Chetomin or molecular
inhibitors like CA9-siRNA are leading to an inhibition of the functional role of
CA9 during tumorigenesis. Human GBM cells were treated with in vitro hypoxia (1,
6, or 24 h at 0.1%, O2). Aza. application was at a range between 250 and 8000 nM
and the HIF-1α inhibitor Chetomin at a concentration range of 150-500 nM. Cell
culture plates were incubated for 24 h under hypoxia (0.1% O2). Further,
CA9-siRNA constructs were transiently transfected into GBM cells exposed to
extreme hypoxic aeration conditions. CA9 protein expression level was detectable
in a cell-type specific manner under normoxic conditions. Whereas U87-MG
exhibited a strong aerobic expression, U251 and U373 displayed moderate and GaMG
very weak normoxic CA9 protein bands. Aza. as well as SU.D2 displayed inhibitory
characteristics to hypoxia induced CA9 expression in the four GBM cell lines for
24 h of hypoxia (0.1% O2) at concentrations between 3500 and 8000 nM, on both the
protein and mRNA level. Parallel experiments using CA9-siRNA confirmed these
results. Application of 150-500 nM of the glycolysis inhibitor Chetomin under
similar oxygenation conditions led to a sharply reduced expression of both CA IX
protein and CA9 mRNA levels, indicating a clear glucose availability involvement
for the hypoxic HIF-1α and CA9 expression in GBM cells.
Hypoxia significantly
influences the behaviour of human tumour cells by activation of genes involved in
the adaptation to hypoxic stress. The main objective in malignant GBM therapy is
either to eradicate the tumour or to convert it into a controlled, quiescent
chronic disease.
Aza., SU.D2, Chetomin or CA9-siRNA possesses functional CA9
inhibitory characteristics when applied against human cancers with hypoxic
regions like GBM. They may be used as alternative or in conjunction with other
direct inhibitors possessing similar functionality, thereby rendering them as
potential optimal tools for the development of an optimized therapy in human
brain cancer treatment.
Neurosci Lett. 2004 Sep 30;368(3):279-84.
Aromatase, the enzyme responsible for estrogen biosynthesis, is expressed by
human and rat glioblastomas.
Yague JG(1), Lavaque E, Carretero J, Azcoitia I, Garcia-Segura LM.
(1)Instituto Cajal, C.S.I.C., Avenida Doctor Arce 37, E-28002 Madrid, Spain.
The biosynthesis of estradiol and related estrogens is catalyzed by the enzyme
aromatase. Among other tissues, aromatase is expressed in the brain, where it is
involved in the regulation of neuroendocrine events and reproduction. Under
physiological conditions, the expression of aromatase in the mammalian brain is
restricted to neurons. However, recent studies have shown that reactive
astrocytes express aromatase after brain injury. This opens the possibility for
the expression of the enzyme in other altered forms of glial cell, such as
gliomas. In the present study, the expression of aromatase has been assessed, by
RT-PCR and immunocytochemistry, in the rat glioblastoma C6 and in two human
glioblastoma cell lines T98G and U373MG. The three cell lines expressed aromatase
mRNA and showed a cytoplasmic pattern of aromatase immunoreactivity. In addition,
the three cell lines express estrogen receptor alpha, suggesting that estradiol
formed by aromatase may act as an autocrine
or paracrine factor for glioblastoma
cells. By analogy to the implication of aromatase into the growth of other forms
of estrogen-sensitive tumors, such as some breast cancers, it is conceivable that
the expression of aromatase may play a role in the growth of glioblastomas.
Oncotarget. 2017 Mar 7;8(10):16605-16620.
Anti-GD2-ch14.18/CHO coated nanoparticles mediate glioblastoma (GBM)-specific
delivery of the aromatase inhibitor, Letrozole, reducing proliferation, migration
and chemoresistance in patient-derived GBM tumor cells.

Tivnan A(1), Heilinger T(1)(2), Ramsey JM(3), O'Connor G(3), Pokorny JL(4)(5),
Sarkaria JN(4), Stringer BW(6), Day BW(6), Boyd AW(6), Kim EL(7), Lode HN(8),
Cryan SA(3), Prehn JH(1).
(1)Centre for Systems Medicine, Department of Physiology and Medical Physics,
Royal College of Surgeons in Ireland, York House, Dublin 2, Ireland.
(2)IMC Fachhochschule Krems, University of Applied Sciences, Krems, Austria.
(3)School of Pharmacy, Royal College of Surgeons in Ireland, York House, Dublin
2, Ireland & Tissue Engineering Research Group, Department of Anatomy, RCSI and
Centre for Research in Medical Devices (CURAM), NUIG, Ireland.
(4)Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States of
America.
(5)Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA.
(6)Brain Cancer Research Unit, QIMR Berghofer Medical Research Institute,
Brisbane, Australia.
(7)Laboratory of Neurooncology, Department of Neurosurgery, Johannes Gutenberg
University Medical Center, Mainz, Germany.
(8)Department of Paediatrics and Paediatric Haematology/Oncology, University of
Greifswald, Greifswald, Germany.
Aromatase is a critical enzyme in the irreversible conversion of androgens to
oestrogens, with inhibition used clinically in hormone-dependent malignancies. We
tested the hypothesis that targeted aromatase inhibition in an aggressive brain
cancer called glioblastoma (GBM) may represent a new treatment strategy. In this
study, aromatase inhibition was achieved using third generation inhibitor,
Letrozole, encapsulated within the core of biodegradable poly lactic-co-glycolic
acid (PLGA) nanoparticles (NPs). PLGA-NPs were conjugated to human/mouse chimeric
anti-GD2 antibody ch14.18/CHO, enabling specific targeting of GD2-positive GBM
cells. Treatment of primary and recurrent patient-derived GBM cells with
free-Letrozole (0.1 μM) led to significant decrease in cell proliferation and
migration; in addition to reduced spheroid formation. Anti-GD2-ch14.18/CHO-NPs
displayed specific targeting of GBM cells in colorectal-glioblastoma co-culture,
with subsequent reduction in GBM cell numbers when treated with
anti-GD2-ch14.18-PLGA-Let-NPs in combination with temozolomide. As miR-191 is an
estrogen responsive microRNA, its expression, fluctuation and role in Letrozole
treated GBM cells was evaluated, where treatment with premiR-191 was capable of
rescuing the reduced proliferative phenotype induced by aromatase inhibitor. The
repurposing and targeted delivery of Letrozole for the treatment of GBM, with the
potential role of miR-191 identified, provides novel avenues for target
assessment in this aggressive brain cancer.
J Neurooncol. 2014 Sep;119(2):275-84.
Aromatase and estrogen receptor alpha mRNA expression as prognostic biomarkers in
patients with astrocytomas.
Dueñas Jiménez JM(1), Candanedo Arellano A, Santerre A, Orozco Suárez S, Sandoval
Sánchez H, Feria Romero I, López-Elizalde R, Alonso Venegas M, Netel B, de la
Torre Valdovinos B, Dueñas Jiménez SH.
(1)Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara,
Guadalajara, Jalisco, Mexico, [email protected].
Estrogens are oncogenic hormones at a high level in breast, prostate, endometrial
and lung cancer. Estrogens are synthesized by aromatase which has been used as a
biomarker both in breast and lung cancer. Estrogen biological activities are
executed by their classic receptors (ERα and ERβ). ERα has been described as a
cancer promoter and ERβ, as a possible tumor suppressor. Both receptors are
present at low levels in primary multiforme glioblastoma (GBM). The GBM frequency
is 50 % higher in men than in women. The GBM patient survival period ranges from
7 to 18 months. The purpose of this pilot study was to evaluate aromatase and
estrogen receptor expression, as well as 17ß-estradiol concentration in
astrocytoma patients biopsies to obtain a prognosis biomarker for these patients.
We analyzed 36 biopsies of astrocytoma patients with a different grade (I-IV) of
malignity. Aromatase and estrogen receptor mRNA expression were analyzed by
semiquantitative RT-PCR, and the E2 levels, by ELISA. E2 concentration was higher
in GBM, compared to grade II or III astrocytomas.
The number of cells
immunoreactive to aromatase and estrogen receptors decreased as the grade of
tumor malignity increased. Aromatase mRNA expression was present in all biopsies,
regardless of malignity grade or patient age or gender.
The highest expression of
aromatase mRNA in GBM patients was associated to the worst survival prognostic
(6.28 months). In contrast lowest expression of ERα mRNA in astrocytoma patients
had a worst prognosis. In conclusion, aromatase and ERα expression could be used
as prognosis biomarkers for astrocytoma patients.
Bioorg Med Chem. 2013 Jul 1;21(13):3949-57.
Hypoxia induced CA9 inhibitory targeting by two different sulfonamide derivatives
including acetazolamide in human glioblastoma.

Said HM(1), Hagemann C, Carta F, Katzer A, Polat B, Staab A, Scozzafava A,
Anacker J, Vince GH, Flentje M, Supuran CT.
(1)Dept. of Radiation Oncology, University of Würzburg, Germany.
[email protected]
HIF-1α regulated genes are mainly responsible for tumour resistance to radiation-
and chemo-therapy. Among these genes, carbonic anhydrase isoform IX (CA9) is
highly over expressed in many types of cancer especially in high grade brain
cancer like Glioblastoma (GBM). Inhibition of the enzymatic activity by
application of specific chemical CA9 inhibitor sulphonamides (CAI) like
Acetazolamide (Aza.)
, the new sulfonamide derivative carbonic anhydrase inhibitor
(SU.D2) or indirect inhibitors like the HIF-1α inhibitor Chetomin or molecular
inhibitors like CA9-siRNA are leading to an inhibition of the functional role of
CA9 during tumorigenesis. Human GBM cells were treated with in vitro hypoxia (1,
6, or 24 h at 0.1%, O2). Aza. application was at a range between 250 and 8000 nM
and the HIF-1α inhibitor Chetomin at a concentration range of 150-500 nM. Cell
culture plates were incubated for 24 h under hypoxia (0.1% O2). Further,
CA9-siRNA constructs were transiently transfected into GBM cells exposed to
extreme hypoxic aeration conditions. CA9 protein expression level was detectable
in a cell-type specific manner under normoxic conditions. Whereas U87-MG
exhibited a strong aerobic expression, U251 and U373 displayed moderate and GaMG
very weak normoxic CA9 protein bands. Aza. as well as SU.D2 displayed inhibitory
characteristics to hypoxia induced CA9 expression in the four GBM cell lines for
24 h of hypoxia (0.1% O2) at concentrations between 3500 and 8000 nM, on both the
protein and mRNA level. Parallel experiments using CA9-siRNA confirmed these
results. Application of 150-500 nM of the glycolysis inhibitor Chetomin under
similar oxygenation conditions led to a sharply reduced expression of both CA IX
protein and CA9 mRNA levels, indicating a clear glucose availability involvement
for the hypoxic HIF-1α and CA9 expression in GBM cells. Hypoxia significantly
influences the behaviour of human tumour cells by activation of genes involved in
the adaptation to hypoxic stress. The main objective in malignant GBM therapy is
either to eradicate the tumour or to convert it into a controlled, quiescent
chronic disease. Aza., SU.D2, Chetomin or CA9-siRNA possesses functional CA9
inhibitory characteristics when applied against human cancers with hypoxic
regions like GBM.
They may be used as alternative or in conjunction with other
direct inhibitors possessing similar functionality, thereby rendering them as
potential optimal tools for the development of an optimized therapy in human
brain cancer treatment.

1. Tumour Biol. 2015 May;36(5):3407-15.
Fisetin suppresses ADAM9 expression and inhibits invasion of glioma cancer cells
through increased phosphorylation of ERK1/2.
Chen CM(1), Hsieh YH, Hwang JM, Jan HJ, Hsieh SC, Lin SH, Lai CY.
(1)Division of Neurosurgery, Department of Surgery, Changhua Christian Hospital,
Changhua, Taiwan.
Fisetin (3,3',4',7-tetrahydroxyflavone) is a naturally occurring flavonoid which
is widely distributed in plants. It has been reported to possess some anticancer
and anti-invasive capabilities. We set out to explore the effects of fisetin on
antimetastatic and its mechanism of action in GBM8401 cells. The results
indicated that fisetin exhibited effective inhibition of cell migration and
inhibited the invasion of GBM8401 cells under non-cytotoxic concentrations. To
identify the potential targets of fisetin, human proteinase antibody array
analysis was performed, and the results indicated that the fisetin treatment
inhibited the expression of ADAM9 protein and mRNA, which are known to contribute
to the progression of glioma cancer. Our results showed that fisetin
phosphorylated ERK1/2 in a sustained way that contributed to the inhibited ADAM9
protein and mRNA expression determined by Western blot and RT-PCR. Moreover,
inhibition of ERK1/2 by U0126 or transfection with the siERK plasmid
significantly abolished the fisetin-inhibited migration and invasion through
activation of the ERK1/2 pathway. In summary, our results suggest that fisetin
might be a potential therapeutic agent against human glioma cells based on its
capacity to activate ERK1/2 and to inhibit ADAM9 expression.
2. Nutr Cancer. 2016 Nov-Dec;68(8):1357-1368.
Inhibitory Kinetics and Mechanism of Flavonoids Extracted from Cotinus coggygria
Scop. Against Glioblastoma Cancer.
Wang G(1), Wang JJ(1)(2), Du L(1), Fei L(1), To ST(3). (1)a Department of Pharmaceutics , Shanghai Eighth People's Hospital, Jiangsu
University , Shanghai , China.
(2)b Hubei University of Medicine , Shiyan , Hubei , China.
(3)c Department of Health Technology and Informatics , The Hong Kong Polytechnic
University , Hong Kong.
This proposal seeks to study the potential therapeutic modality of
chemoprevention and anticancer effects and mechanisms of the flavonoids from
Cotinus coggygria Scop. on glioblastoma cancer. In the current study, the total
flavonoids (TFs) isolated from Cotinus coggygria Scop. var. cinerea Engl.
(Cotinus coggygria Scop.) and the major flavonoids of Cotinus coggygria Scop.
(CCFs) were identified, and the inhibitory kinetics of TF and CCF on glioblastoma
cell lines were calculated. We also investigated whether TF or CCF regulated the
apoptotic mechanism in cellular models of glio-blastoma cells. Finally, we
evaluated whether treatment with TF or CCF suppressed tumor growth and inhibited
migration in orthotopic mouse models of glioblastoma in vivo. In this study, the
CCFs were identified as rutin, myricetin, and fisetin. TF and CCF remarkably
inhibited cell proliferation and downregulated the PI3K/Akt and ERK signaling
pathway in glioblastoma cell lines. Furthermore, the mitochondrial
caspase-dependent cascade was regulated by TF and myricetin. In addition, TF and
myricetin exhibited significant antitumor effects on glioblastoma in vivo. Taken
together, these results suggest that phytochemical and biological data provide
evidence for the active components in Cotinus coggygria, and that the TFs are
responsible for the anticancer effects on glioblastoma cell growth via induction
of apoptosis. In addition, the representative compound myricetin could provide a
clinically relevant therapeutic opportunity. Therefore, our data strongly suggest
that myricetin-deprived CCF can serve as a potent chemopreventive herbal
medicine.
Biochem Biophys Res Commun. 2006 Apr 21;342(4):1329-33.
The effect of gamma-tocopherol on proliferation, integrin expression, adhesion, and migration of human glioma cells.
Samandari E, Visarius T, Zingg JM, Azzi A.
The effect of vitamin E on proliferation, integrin expression, adhesion, and migration in human glioma cells has been studied. gamma-tocopherol at 50 microM concentration exerted more inhibitory effect than alpha-tocopherol at the same concentration on glioma cell proliferation. Integrin alpha5 and beta1 protein levels were increased upon both alpha- and gamma-tocopherol treatments. In parallel, an increase in the alpha5beta1 heterodimer cell surface expression was observed. The tocopherols inhibited glioma cell-binding to fibronectin where gamma-tocopherol treatment induced glioma cell migration. Taken together, the data reported here are consistent with the notion that the inhibition of glioma cell proliferation induced by tocopherols may be mediated, at least in part, by an increase in integrin alpha5 and beta1 expression. Cell adhesion is also negatively affected by tocopherols, despite a small increase in the surface appearance of the alpha5beta1 heterodimer. Cell migration is stimulated by gamma-tocopherol. It is concluded that alpha5 and beta1 integrin expression and surface appearance are not sufficient to explain all the observations and that other integrins or in general other factors may be associated with these events.
Free Radic Biol Med. 2006 Aug 1;41(3):464-72.
Antiproliferative effects of tocopherols (vitamin E) on murine glioma C6 cells:
homologue-specific control of PKC/ERK and cyclin signaling.
Betti M(1), Minelli A, Canonico B, Castaldo P, Magi S, Aisa MC, Piroddi M, Di
Tomaso V, Galli F.
(1)Institute of Physiological Sciences, University of Urbino Carlo Bo, Urbino,
Italy.
Chemoprevention strategies for brain tumors (specifically gliomas) are few and
surprisingly poorly investigated. We have studied the effects of tocopherols
(TOCs; vitamin E) on proliferation and death processes of murine glioma C6 cells.
These vitamers showed different cell uptake and concentration- and time-dependent
inhibitory effects on cell growth that were significant at the lowest
concentrations tested (1-10 microM). However, the inhibitory potency of TOCs
seemed to reflect at least in part their actual cell concentrations at steady
state, with the order of magnitude gamma-TOC >or= alpha-TOC > delta-TOC
approximately or = beta-TOC. Moreover, for extracellular concentrations >or=10
microM, TOCs also showed a significant cytotoxic effects due mainly to necrosis,
while apoptosis was negligible. Gamma-TOC (the form showing preferential cell
uptake and lowest unspecific cytotoxicity) was the most effective inhibitor of
cell cycle progression (arrest in G0/G1 phase) leading to lowered expression of
cyclin E and cyclin-dependent kinases 2 and 4 and overexpression of p27 (specific
inhibitor of S-phase entering). According to these signals, activated ERK1/2 and
PKC upstream and Rb phosphorylation downstream were decreased. In conclusion,
within TOCs the gamma form exerts the most potent and specific control of cell
cycle progression in C6 cells (cytostatic effect). This suggests a
chemopreventive role of this form of vitamin E in gliomas.
Proc Soc Exp Biol Med. 1983 Nov;174(2):302-7.
Study on the specificity of alpha-tocopheryl (vitamin E) acid succinate effects
on melanoma, glioma and neuroblastoma cells in culture.
Rama BN, Prasad KN.
d- and dl-alpha-tocopheryl succinate inhibited growth and caused morphological
changes in mouse melanoma (B-16), mouse neuroblastoma (NBP2), and rat glioma
(C-6) cells in culture. To study whether the effects of alpha-tocopheryl (vitamin
E) succinate on tumor cells are mediated by antioxidant mechanisms, the effects
of lipid-soluble antioxidants, butylated hydroxyanisole (BHA) and butylated
hydroxytoluene (BHT) were compared with those of vitamin E succinate. Results
showed that these antioxidants produced alterations on the growth and morphology
of neuroblastoma, melanoma, and glioma cells which are similar to those produced
by vitamin E succinate; however, the extent of the effect depended upon the type
of antioxidant and the form of tumor cells. These data suggest that the effects
of vitamin E succinate on tumor cells may be mediated, in part, by antioxidant
mechanisms.
Neurol Med Chir (Tokyo). 1983 Jun;23(6):421-7.
[Effect of vitamin E (DL-alpha-tocopherol) on the growth of the rat glioma cells
in culture].
[Article in Japanese]
Kokunai T, Korosue K, Tamaki N, Matsumoto S.

No Shinkei Geka. 1999 Feb;27(2):119-25.
[Proliferation inhibition of glioma cells by vitamin K2].
[Article in Japanese]
Sun L(1), Yoshii Y, Miyagi K, Ishida A.
(1)Department of Neurosurgery, School of Medicine, University of the Ryukyus,
Okinawa, Japan.
The antitumor effects of vitamin K2 were studied using three glioma cell lines:
C6 (rat glioma cell), RBR17T and T98G (human glioma cell). The antitumor effects
were estimated by count assay. The results was that vitamin K2 induced growth
inhibition in a dose-dependent manner. The RBR 17T cells exposed to vitamin K2
for 72 hours resulted in oligonucleosomal DNA fragmentation and formed a ladder
on agarose gel electrophoresis. Furthermore, the RBR17T cells exposed to vitamin
K2 for 24 hours were significantly accumulated in the G0G1 phase of the cell
cycle. Those results suggested that vitamin K2 can inhibit the proliferation of
cells through the induction of cell cycle arrest and apoptosis for tumor cells.
The combined treatment of vitamin K2 with ACNU or 5-FU or INF-beta or
1,25-dihydroxyvitamin D3 enhanced growth inhibition significantly. In conclusion,
vitamin K2 can be a useful drug for the treatment of glioma.

Acta Neurochir (Wien). 1995;133(3-4):184-90.
Effects of vitamin D and retinoic acid on human glioblastoma cell lines.
Magrassi L(1), Butti G, Pezzotta S, Infuso L, Milanesi G.
(1)Department of Surgery, University of Pavia, IRCCS Policlinico S. Mattco,
Italy.
The biological significance of vitamin D receptors expressed by glioblastoma and
other glial tumours is still unclear. In an effort to clarify this issue we
studied the effects of increasing concentrations of 25-dihydroxyvitamin D3 and
its metabolite 1 alpha,25-dihydroxyvitamin D3 on two human glioblastoma cell
lines. Both substances were capable of inducing a significant (> 50%) reduction
in growth of the two glioblastoma cell lines at dosages over 5 microM. When the
HU 70 cell line was treated by increasing dilutions of 25-dihydroxyvitamin D3
combined with 1 microM all trans-retinoic acid, significant inhibition was
apparent even after addition of 25-dihydroxyvitamin D3 in the nanomolar range.
Reduction of growth index was mainly due to induced cell death. Our results
provide in vitro evidence that vitamin D metabolites alone or in combination with
retinoids may be potentially useful agents in the differentiation therapy of
human malignant gliomas.
 

Travis

Member
Joined
Jul 14, 2016
Messages
3,189
So Leucine-restriction and not only Methionine-restriction is a way of mimicking fasting?

I have only very rudimentary understanding of the mTOR/AMPK pathway "dualism", a theme often variated in life-extender circles, but it seems to me that some aspects of this duality are vice-versa in the brain and the Rest of the body. What activates one pathway in the peripheria might activate the other in the CNS.
I think the brain initiates autophagy a bit later that most body locations because it has access to so much blood; I would imagine that it'd be exposed to leucine from autophagy elsewhere in the body for longer.

But other things also appear to interact with the mTOR pathway besides leucine and rapamycin, so it would be expected to respond a bit differently due to the different milieu inside of each cell (i.e. cAMP). Each cell in each specific location also has different corepressors in the nucleus, prevent inappropriate DNA from being transcribed. Remember, all cells were at one time identical and have all stemmed from the mitosis of just one cell (1 ⟶ ¹⁄₂ + ¹⁄₂ ⟶ ¹⁄₄ + ¹⁄₄ + ¹⁄₄ + ¹⁄₄)—the ova, which had needed to be differentiated into other cell types and kept that way. You'd think steroid hormones, peptide hormones, oxygen gradients, pH gradients, nitric oxide, calcium, and eventually blood pressure gradients would all contribute to this process. So the simple fact that corepressors exist strongly imply that some cells will respond differently to stimulus which would normally lead to the transcription of the DNA segment onto which said corepressor is attached. Besides have experimental confirmation, there is a philosophical need for such a thing; there needs to be nuclear factors which keep a differentiated cell expressing a unique genome while containing the entire DNA library in its nucleus.

Despite the difference between cell types, I have read of nothing more effective and reliable at influencing autolysis than leucine and rapamycin.
 
Last edited:

Travis

Member
Joined
Jul 14, 2016
Messages
3,189
He's talking about your "most people who eat meat are chubby". 97% of the population in the US eat meat. It's like saying most people who drink water are chubby. Or most people who use a phone are chubby.
You don't actually think I was actually trying to make an argument now? I was merely responding to one projected stereotype by intentionally projecting one of my own—just to make a point.

Nonetheless, I think you'll be able to find a strong correlation between body weight and certain foods even after adjusting for 'car ownership.' [?]
 
EMF Mitigation - Flush Niacin - Big 5 Minerals

Similar threads

Back
Top Bottom