ADVFN Logo ADVFN

We could not find any results for:
Make sure your spelling is correct or try broadening your search.

Trending Now

Toplists

It looks like you aren't logged in.
Click the button below to log in and view your recent history.

Hot Features

Registration Strip Icon for pro Trade like a pro: Leverage real-time discussions and market-moving ideas to outperform.
Protide Pharmaceuticals Inc (GM)

Protide Pharmaceuticals Inc (GM) (PPMD)

0.22
0.00
( 0.00% )
Updated: 20:00:00

Unlock more advanced trading tools

Join ADVFN today

Key stats and details

Current Price
0.22
Bid
0.00
Ask
0.00
Volume
-
0.00 Day's Range 0.00
0.00 52 Week Range 0.00
Previous Close
0.22
Open
-
Last Trade
Last Trade Time
-
Average Volume (3m)
-
Financial Volume
-
VWAP
-

PPMD Latest News

No news to show yet.
PeriodChangeChange %OpenHighLowAvg. Daily VolVWAP
10000000CS
40000000CS
120000000CS
260000000CS
520000000CS
1560.18485250.03520.44950.035264200.13986356CS
2600.07551.7241379310.1450.480.021176300.136758CS

Market Movers

View all
  • Most Active
  • % Gainers
  • % Losers
SymbolPriceVol.
CAUDCollective Audience Inc
$ 0.751
(92.56%)
106.77M
DCPHDeciphera Pharmaceuticals Inc
$ 25.205
(72.05%)
36.92M
WKSPWorksport Ltd
$ 0.6778
(38.21%)
423.51k
HWHHWH International Inc
$ 2.495
(37.09%)
19.36M
CSSEChicken Soup for the Soul Entertainment Inc
$ 0.3247
(34.28%)
13.5M
CLVRClever Leaves Holdings Inc
$ 1.54
(-62.53%)
555.08k
ADXNAddex Therapeutics Ltd
$ 8.4179
(-47.71%)
97.75k
MFImF International Ltd
$ 1.63
(-30.04%)
2.2M
TSDDGraniteShares ETF Trust GraniteShares
$ 18.04
(-27.55%)
813.09k
TSLZT Rex 2X Inverse Tesla Daily Target ETF
$ 28.49
(-27.45%)
4.16M
TSLATesla Inc
$ 191.375
(13.72%)
113.95M
SINTSiNtx Technologies Inc
$ 0.0524
(29.70%)
110.6M
CAUDCollective Audience Inc
$ 0.751
(92.56%)
106.77M
SOFISoFi Technologies Inc
$ 7.035
(-10.61%)
100.32M
TSLLDirexion Daily TSLA Bull 2X Trust ETF
$ 9.125
(27.62%)
46.66M

PPMD Discussion

View Posts
Lazarus Lazarus 5 years ago
Website was updated within the last year

https://www.protidepharma.com/
👍️0
Lazarus Lazarus 6 years ago
This today:

Sold Out: Protide Pharmaceuticals Inc (PPMD)


Kitzinger Lautmann Capital Management Inc sold out the holdings in Protide Pharmaceuticals Inc. The sale prices were between $0.02 and $0.02, with an estimated average price of $0.02.

👍️0
Lazarus Lazarus 7 years ago
Taken from the FaceBook page of

Saint Basil of Ostrog Serbian Orthodox Church
February 9, 2015 ·


Congratulating and Supporting Small Business Friends of St. Basil of Ostrog Church - MiSPORT DRINK - Contemporary Hydration

MiSport Drinks are the new way to hydrate, naturally & safely. MiSport hydrates at the cellular level. Instead of drinking several bottles of other drinks, including water, you'll be satisfied faster and better with MiSport. MiSport replenishes, without having to saturate your system. MiSport Drinks contain no sucrose syrup or glucose-fructose syrup. Our product is sweetened with Stevia. Other products contain ester gum, an emulisfier made from wood and starch. MiSport Drinks are free and clear from these unappealing additives. New ideas that make a difference. Congratulations to Milo Polovina of MiSPORT DRINK. Milo and Gayle Polovina have three sons: Ben, Alec and Eli. Ben and Alec are attending U of M at Duluth. Eli attends Barrington High School and classes in St. Basil of Ostrog ChurchSchool as did Ben and Alec. Congratulations to the Polovina Family.

http://www.misportdrink.com/#!about-misport/cjg9

If you click the link iit will take you to the website for the sports drink that he is apparently the producer of --- an if you look at the MiSport address

220 Telser Road
Lake Zurich, IL 60047

you will notice that it is the same as Protide's.

-----------



👍️0
NathanH NathanH 8 years ago
Idk at least they are active and announced it.
👍️0
Lazarus Lazarus 8 years ago
is that a positive, negative, or neutral move?
👍️0
NathanH NathanH 8 years ago
They have a new address
Effective June 1st, 2016, our new address will be:

220 Telser Road, Lake Zurich
👍️0
Lebaneseproud Lebaneseproud 8 years ago
I did see that. I continue to believe there is hidden value here.
As a business man, I can tell you no company can keep its doors open as long as they have without making money. You can only do the smoke and mirror thing for so long. This company has been open for many years and they must be growing and making money to have survived.
👍️0
Lazarus Lazarus 8 years ago
Have you noticed that PPMD has a new product?

http://protidepharma.com/vaxmax

Also they have a new distributor

http://www.biotag.co.il/
👍️0
Lebaneseproud Lebaneseproud 9 years ago
Yes, I have been following this and regularly check for updates on the website as well. Still holding with no desire whatsoever to liquidate this position. I will hold another 10 to 20 years if need be and the Lord allows me to hang around that long.
👍️0
Lazarus Lazarus 9 years ago
While the shares slumber in penny purgatory...

thought I might mention a couple of things.

Notice on the PPMD website that the following page was added under
Products

The link at the bottom -- CIT Islet Cell Solutions

takes you HERE

There it says that only solutions from Protide and Mediatech are allowed in the formulation.

Mediatechs website ishere.

If you click the About tab you will see a link titled Mediatech Acquisition that explains that Mediatech was bought out by Corning (GLW) in 2011

READ ABOUT IT HERE

Anyway -- back to sleeper mode.
👍️0
Lazarus Lazarus 10 years ago
yep.... just commented here:

Its a SLEEPER.

👍️0
Lazarus Lazarus 10 years ago
yep.... :just commented here

Its a SLEEPER.

👍️0
Lebaneseproud Lebaneseproud 10 years ago
Looks like she woke up today. Nice paper spike on my account. Who knows, maybe its the beginning of new life here
👍️0
Lazarus Lazarus 12 years ago
you see that move up on BLFS?
👍️0
Lazarus Lazarus 12 years ago
yes, i expect that they are making money....

...its a pity they refuse to communicate with shareholders in any meaningful way.
👍️0
Lebaneseproud Lebaneseproud 12 years ago
Hi Lazarus,
Im sure you already saw this statement on the Protide website.

"Protide Pharmaceuticals, Inc. is a fully integrated, FDA registered medical device organization devoted to the discovery, development and commercialization of technologies and processes in clinical cell therapy, regenerative medicine, transfusion medicine, cell engineering,and transplantation.

With over 26 years of service, in over 60 countries, our organization continues as an entrepreneurial, science driven organization inspired by the patients, scientists and physicians we support."


..... They must be making some money if they can describe themselves as entepreneurial.


👍️0
kazcul kazcul 15 years ago
I added a few more this morning. I'm planning on adding 5000 more shares sometime early in 2011. If I add 5000 shares every 18 months I should have a about 200,000 shares by their next PR.
👍️0
Lazarus Lazarus 15 years ago
Excellent DD by chuuk on RB:

http://ragingbull.quote.com/mboard/boards.cgi?board=PPMD&read=3901

I just added 1500 shares.

Warning: this is not a shareholder friendly company
👍️0
Lazarus Lazarus 16 years ago
CHICAGO – There’s a fire sale going on in the U.S. at the moment.

As I sit here in New York while attending the annual NanoBusiness Alliance nanotechnology conference, the city is mobbed with foreign tourists who find the U.S. dirt cheap. Considering New York is probably the most expensive city in the U.S., you can imagine what the rest of the country must look like to these foreigners.

To get a little perspective, take a look at the five-year exchange difference from a number of different currencies around the world versus the dollar. Though I picked a five-year time period, a large change has taken place actually within the last 12 to 18 months. .....

entire article here:
http://www.midwestbusiness.com/news/viewnews.asp?newsletterID=19156

HE GOES ON TO SAY

.....As I started out the column and as my focus is life sciences companies, the price tag for doing business in the U.S. market – the largest market in the world with at least 45 percent market share – is getting cheaper and cheaper for foreign companies looking to set up shop here. In fact, the acquisition of an American company has rarely been cheaper.

👍️0
Lebaneseproud Lebaneseproud 16 years ago
Agree the environment in this sector will be much better with the new President. Companies like PPMD will be more agressively targeted for takeover or acquisition in a more friendly environment
👍️0
Lazarus Lazarus 16 years ago
Stem cells: A sure bet in the '08 race
McCain, Clinton, Obama all oppose Bush's limits on funding for human embryonic stem cells.

NEW YORK (CNNMoney.com) -- Whoever wins the White House, stem cell biotechs stand to reap the benefit from an incoming leader who is friendlier to stem cell researchers than President Bush, and that could lift stocks for the entire sector, experts say.

"Any candidate is going to have a better policy on stem cells than our current president," said Ren Benjamin, biotech analyst for Rodman & Renshaw. "If that's the case, then it will be good news not only for the companies working in the space, but for the space in general."

The three leading presidential candidates - Sen. John McCain, a Republican from Arizona, and Democratic Sens. Hillary Clinton of New York and Barak Obama of Illinois - have all come out in support of expanded federal funding of human embryonic stem cell research.

Stem cells taken from human embryos, created primarily through in vitro fertilization, are favored by many scientists for their ability to regenerate and to morph into different types of tissue. In theory, stem cell-based therapies could repair traumatic injuries like severed spines and brain damage, or reverse the affects of debilitating diseases including Alzheimer's and Parkinson's. So far, these types of therapies are in the earliest stages of experimentation.

President Bush created federal funding for human embryonic stem cell research back in 2001, but he limited support to only those cell lines that existed at the time in order to "not encourage the destruction of embryos." But these limits are likely to be lifted under a new regime.

"The next president can expand [funding] and remove the road blocks," said Stephen Brozak, biotech analyst for WBB Securities. "It removes the stigma from Bush."

Bush has twice vetoed legislative attempts to expand the funding, including those backed by McCain, Clinton and Obama. In reference to Bush's policies, Obama has said, "Stymieing embryonic stem cell research is a step in the wrong direction." Clinton has called for funding for "additional cell lines in order to pursue the promising avenues for research." McCain has said "stem cell research has the potential to give us a better understanding of deadly diseases and spinal cord injuries affecting millions of Americans."

Following his second veto in 2007, Bush said the legislation "would compel American taxpayers - for the first time in our history - to support the deliberate destruction of human embryos." Instead, the president touted the therapeutic potential of stem cells taken from adult tissue.

But proponents of embryonic stem cell research - including the presidential candidates - have emphasized that only those stem cells slated for destruction as medical waste would be used. In his support for stem cell research funding in 2006, McCain said the legislation provides funding only for "scientists who use embryos originally created for reproductive purposes" and those that are "now frozen or slated for destruction by in vitro fertilization clinics."

Many of the stem cell biotechs, like Aastrom (ASTM), Cytori Therapeutics (CYTX), Stemcells (STEM) and Osiris Therapeutics (OSIR), are buffered from the controversy because they use stem cells taken from adult tissue, such as organs, skin, or umbilical cords. But other biotechs, like Geron (GERN), Advanced Cell Technology, Novocell and Neuralstem (CUR), derive stem cells from human embryos. Many of these companies are showcasing their technologies at the 3rd annual Stem Cell Summit in New York on Tuesday.

Neuralstem, Geron and Advanced Cell Technology all plan to begin testing in humans this year. In theory, they have the most to gain from a president who's friendlier to the use of embryonic stem cells.

But since investors don't often distinguish the nuances between the various stem cell companies, however controversial they may be, a rising tide may lift all boats.

"I think that all these names are going to do well going into the election," said Benjamin of Rodman & Renshaw. "The entire stem cell space is likely to benefit with the additional funding, because you're likely to see a spillover effect."

👍️0
Lazarus Lazarus 16 years ago
State Competition Heats Up For Stem-Cell Scientists

By Christine Vestal

Stateline.org Staff Writer
01/28/08

--------------------------------------------------------------------------------

Seven states — including Connecticut — are leading the world in political and financial support for embryonic stem-cell research.

Their goal: Attract the best stem-cell scientists from around the globe and become a hub for a multi-billion-dollar bioscience industry. So far, their plan appears to be working.

In the past two years, California, Connecticut, Illinois, Maryland, New Jersey, New York and Wisconsin have awarded some $230 million in grants — more than three times as much as the federal government spent on embryonic stem-cell studies in that time — and there has been no shortage of scientists seeking the money.

Three more states — Iowa, Massachusetts and Missouri — have affirmed the legality of the research in hopes of keeping or encouraging scientists to work within their borders.

But six others — Arkansas, Indiana, Louisiana, Michigan, North Dakota and South Dakota — now ban studies that result in the destruction of human embryos, and Arizona bars state funding for embryonic studies. These states have positions closer to those of Japan and most European countries.

Except in these states, work on embryonic stem cells is free to go on in the United States at places such as universities and private, nonprofit and corporate laboratories — as long as no federal money is involved. But states that want to be players in the nascent stem-cell arena are finding they must ante up with state financing and a science- friendly environment.

In 2005, Connecticut allotted $100 million and Illinois $10 million, adding another $5 million in 2006.

Maryland approved $15 million in 2006 and $23 million more in 2007, and New York invested $650 million in 2007.

Wisconsin Gov. Jim Doyle (D) created a $750 million building fund to construct a stem-cell research laboratory on the campus of the University of Wisconsin, Madison.

New Jersey in 2006 appropriated an additional $15 million in grant money, $9.5 million for administrative costs and $270 million to build five new research facilities.

While they didn’t pony up dollars, Missouri in 2006 and Iowa last year declared their state open for stem-cell business with measures legalizing work on embryos.

Massachusetts lawmakers in 2005 overrode a veto by then-Gov. Mitt Romney to ensure the legality of the research.

Among states seeding the fledgling science, California is the bellwether with a $3 billion fund of taxpayer dollars being spent to build worldclass research labs and lure leading stem-cell scientists to the sunny West Coast.

When all seven states’ investments are totaled, the commitment comes to nearly $5 billion over the next 10 years. Massachusetts could add another $1 billion.

“States that have chosen to fund the research are in an ideal position,” said Bernard Siegel, founder of the Genetics Policy Institute, a nonprofit stem-cell advocacy group.

“Scientists are energized by the new developments, and many of the best and brightest already are flocking to California and other states with generous grants and friendly science policies,” Siegel said.

Last year, 39 states considered more than 100 bills for and against the research, but only three laws were enacted, according to the National Conference of State Legislatures.
👍️0
Lazarus Lazarus 16 years ago
cold feet EVERYWHERE!

i picked up a few more but the dropped down below my buy price.

👍️0
Lebaneseproud Lebaneseproud 16 years ago
Somebody obviously got cold feet or impatient. Will be interesting to see if it continues monday or if it settle here. Might be tempted for some cheap pickens monday
👍️0
Lazarus Lazarus 16 years ago
i picked up a few as well and they took it down below my buy price. We are now in PENNY PURGATORY!
👍️0
kazcul kazcul 16 years ago
Probably my fault, I bought 5000 this morning and usually when I buy a stock it drops immediately.

Tom
👍️0
Lazarus Lazarus 16 years ago
some serious dumping going on!
👍️0
Lazarus Lazarus 16 years ago

2007 stem cell breakthrough is like turning lead into gold

It was the kind of breakthrough scientists had dreamed of for decades and its promise to help cure disease appears to be fast on the way to being realized.
Researchers in November announced they were able to turn the clock back on skin cells and transform them into stem cells, the mutable building blocks of organs and tissues.

Then just earlier this month a different team announced it had cured sickle cell anemia in mice using stem cells derived from adult mouse skin.

"This is truly the Holy Grail: To be able to take a few cells from a patient -- say a cheek swab or few skin cells -- and turn them into stem cells in the laboratory," said Robert Lanza, a stem cell pioneer at Advanced Cell Technology.

"This work represents a tremendous scientific milestone - the biological equivalent of the Wright Brothers' first airplane," he told AFP.

"It's bit like learning how to turn lead into gold."

Stem cells offer enormous potential for curing and treating disease because they can be transformed into any cell in the body and then hopefully used to replace damaged or diseased cells, tissues and organs.

But stem cell research has been highly controversial because -- until now -- viable embryos had to be destroyed to extract the stem cells.

US President George W. Bush has banned all federal funding for research on human embryonic stem cells and access to stem cells in other countries has also been restricted because of the difficulty in finding women willing to donate their eggs.

The new technique, while far from perfected, is so promising that the man who managed to clone the world's first sheep, Dolly, is giving up his work cloning embryos to focus on studying stem cells derived from skin cells.

"The fact that (the) introduction of a small number of proteins into adult human cells could produce cells that are equivalent to embryo stem cells takes us into an entirely new era of stem cell biology," said Ian Wilmut, the Scottish researcher who first created a viable clone by transferring a cell nucleus into a new embryo.

One of the greatest advantages of the new technique is its simplicity: it takes just four genes to turn the skin cell back into a stem cell.

This, unlike the complex and expensive process developed by Wilmut, can be done in a standard biological lab. And skin cells are much easier to harvest than embryos.

"It's an explosion of resources," said Konrad Hochedlinger, of the Harvard Stem Cell Institute.

Prior to this discovery, researchers who wanted to look at how diseases developed would usually have to study animals or organs harvested from cadavers because embryonic stem cells were so hard to use and access.


But with stem cells derived from skin, tissues and organs can be grown in a petri dish, making it easier for researchers to map the genetic structure of diseased cells, a process which could unlock a cure.

They could also allow researchers to do chemical screens to identify drugs which may cure or treat a disease, a process which could significantly speed up the process of bringing life-saving drugs to the market.

The use of skin cells will eventually allow doctors to create stem cells with a specific patient's genetic code, eliminating the risk that the body would reject transplanted tissues or organs.

Researchers have already shown this is possible when they cured sickle cell anemia in mice.

They used skin cells taken from the tails of sick mice, transformed them into stem cells, manipulated those stem cells into healthy bone marrow cells and then transplanted them into the sick mice.

And since the new cells came from the sick mice, there was also no need for dangerous immunosuppressant drugs to prevent rejection.

But leading stem cell researchers warned that the skin cells are not yet -- and might never be -- a substitute for embryonic stem cells.

"This new research is just the beginning -- we hardly understand how these cells work," said James Thomson of the University of Wisconsin at Madison, who led one of the two teams which made the simultaneous discoveries.

"It is not the time to abandon stem cell research," Thomson said, adding that embryonic stem cells will remain the "gold standard" by which other research is measured.

Further research is also needed to find a safer way to transform the skin cells and to make sure that the cells do not deteriorate over time.


👍️0
Lazarus Lazarus 16 years ago
Dolly creator Prof Ian Wilmut shuns cloning
By Roger Highfield, Science Editor
Last Updated: 6:30pm GMT 16/11/2007Page 1 of 3

The scientist who created Dolly the sheep, a breakthrough that provoked headlines around the world a decade ago, is to abandon the cloning technique he pioneered to create her.

Prof Ian Wilmut's decision to turn his back on "therapeutic cloning", just days after US researchers announced a breakthrough in the cloning of primates, will send shockwaves through the scientific establishment.


FULL STORY HERE: http://www.telegraph.co.uk/earth/main.jhtml;jsessionid=XFT2IYJUHGB4FQFIQMFSFF4AVCBQ0IV0?xml=/earth/2007/11/16/scidolly116.xml


👍️0
Lebaneseproud Lebaneseproud 16 years ago
PPMD will be acquired at some point and the shareholders will have to get paid
👍️0
Lazarus Lazarus 16 years ago
compare neostem financils to ppmds

http://yahoo.brand.edgar-online.com/fetchFilingFrameset.aspx?dcn=0001157523-07-008394&Type=HTML

share structure is similar - but NBS trading @ $2.25

someday....

poor typing due to hand surgery
👍️0
Lazarus Lazarus 16 years ago
i c :)
👍️0
Lebaneseproud Lebaneseproud 16 years ago
Thanks for that cut and paste Lazarus. Our other common holding THMG is perking up a bit.
👍️0
Lazarus Lazarus 17 years ago
This is a cut and paste from the RB board...

(courtesy chuuk333)

World Intellectual Property Organization

(Look down to the last couple of paragraphs)

Notice who produces the other product that didn't impair undifferentiated stem cell propagation? ROCHE!!!
So there are big names in this business-and hopefully are little charmers at PPMD will catch someones eye some day.
--------------------------------------------------------------------------------

(WO/2006/070370) STEM CELLS CULTURE SYSTEMSBiblio. Data
Description
Claims
National Phase
Notices
Documents
Note: OCR Text
Note: Text based on automatic Optical
Character Recognition processes. Please
use the PDF version for legal matters


STEM CELLS CULTURE SYSTEMS

FIELD OF THE INVENTION

The invention relates to stem cells (SC) in particularly to methods and systems for handling human embryonic stem cells (hESC).

LIST OF PRIOR ART

The following is a list of prior art, which is considered to be pertinent for describing the state of the art in the field of the invention.

(1) Thomson, J. A. et al. Embryonic stem cell lines derived from human blastocysts. Science 282, 1145-1147 (1998).

(2) Reubinoff, B.E., Pera, M.F., Fong, C.Y., Trounson, A. & Bongso, A. Embryonic stem cell lines from human blastocysts: somatic differentiation in vitro. Nat Biotechnol 18, 399-404 (2000)

(3) Amit, M. et al. Clonally derived human embryonic stem cell lines maintain pluripotency and proliferative potential for prolonged periods of culture.

Dev Biol 227, 271-278 (2000).

(4) Xu, C. et al. Feeder-free growth of undifferentiated human embryonic stem cells. Nat Biotechnol 19, 971-974 (2001).

(5) Amit, M. et al. Human feeder layers for human embryonic stem cells. Biol Reprod βS, 2150-2156 (2003).

(6) Richards, M., Fong, C.Y., Chan, W.K., Wong, P.C. & Bongso, A. Human feeders support prolonged undifferentiated growth of human inner cell masses and embryonic stem cells. Nat Biotechnol 20, 933-936 (2002).

(7) Cowan, CA. et al. Derivation of embryonic stem-cell lines from human blastocysts. N EnglJ Med 350, 1353-1356 (2004).

(8) Amit, M., Shariki, C, Margulets, V. & Itskovitz-Eldor, J. Feeder layer- and Serum-Free Culture of Human Embryonic Stem Cells. Biol Reprod 70(3):837-45 (2004).

(9) Pera, M.F. et al. Regulation of human embryonic stem cell differentiation by BMP-2 and its antagonist noggin. J Cell Sci 111, 1269-1280 (2004).

(10) GB2409208.

(11) WO 04/031343

(12) Xu, R.H., et al. Basic FGF and suppression of BMP signaling sustain undifferentiated proliferation of human ES cells. Nat Methods. 3, 164-5 (2005)

(13) Valuer L, et al. Activin/Nodal and FGF pathways cooperate to maintain pluripotency of human embryonic stem cells. J Cell Sci. 118, 4495-509 (2005)

BACKGROUND OF THE INVENTION Stem cells are immature, unspecialized cells that renew themselves for long periods through cell division. Under certain conditions, they can differentiate into mature, functional cells. Human embryonic stem cells (hESC) are derived from early surplus human blastocysts l' 2. Human ES cells are unique stem cells since they can self-renew infinitely in culture, and since they have a remarkable potential to develop into extraembryonic lineages as well as all somatic cells and tissues of the human body1' 2.

Given the unique properties of hESC, they are expected to have far- reaching applications in the areas of basic scientific research, pharmacology, and regenerative medicine. Human ES cell lines can provide a powerful in vitro model for the study of the molecular and cellular biology of early human development, for functional genomics, drug screening, and discovery. They may serve for toxicology and teratogenicity high throughput screening. Since hESC can self-renew indefinitely and can differentiate into any cell type, they can serve

as a renewable, unlimited donor source of functionally mature differentiated cells or tissues for transplantation therapy. In addition, transplanted genetically- modified hESC can serve as vectors to carry and express genes in target organs in the course of gene therapy.

While the promise of hESC for basic scientific research pharmacology and regenerative medicine is remarkable, the exploitation of hESC for most applications depends upon further development. Improved control of the growth of undifferentiated hESC, the development of bulk feeder-free cultures of undifferentiated cells, the development of animal-free culture systems, and the development of methods and tools which direct the differentiation and generate pure cultures of mature functional cells of a specific type are required.

At present, few culture systems are most commonly used to propagate undifferentiated hESC1"4. In the initial culture system that was developed, undifferentiated hESC are cultured in serum-containing medium as colonies, upon a layer of fibroblast feeder cells (of mouse1' 2 or human origin5' π). It is possible to remove all animal products from this culture system and replace them with those from a human source6. It was found that in this system the cells are propagated as clumps on a low scale which does not allow cloning2.

An alternative culture system that was developed and used extensively is a serum-free system that includes the knockout (KO) medium supplemented with knockout serum replacement (KOSR) and FGF2. This system allows cloning of undifferentiated hESC, although at a low efficiency3. Undifferentiated cells are cultured as flat colonies and may be propagated as small clusters or single cells (by using trypsin7).''

Another alternative culture system for use in the proliferation of undifferentiated growth of hESC comprises a culture matrix comprising extracellular matrix (ECM) prepared from feeder cells and a conditioned medium being preconditioned by feeder cells. The suggested leading cells in the feeder

cells include primary mouse embryonic fibroblasts (PMEF) a mouse embryonic fibroblast cell line (MEF) murine foetal fibroblasts (MFF) human embryonic fibroblasts (HEF) human foetal muscle (HFM) human foetal skin cells (HFS) human adult skin cells, human foreskin fibroblasts (HFF)10 human adult Fallopian tubal epithelial cells (HAFT) or human marrow stromal cells (HMSC).

Undifferentiated propagation may be accomplished with the KO serum- free culture system without the use of feeders by plating and growing colonies on extracellular matrices (ECM) within a feeder-conditioned KO medium supplemented with KOSR and FGF24. Furthermore, it has been suggested that feeder conditioning may be replaced by substituting the medium with high concentrations of FGF2 and noggin12. Alternatively, feeder conditioning was replaced by transforming growth factor β 1 and human LIF (in addition to FGF2) and growing the cells on human fibronectin . In a recent publication, undifferentiated propagation of hESC colonies, in the absence of feeders' was reported with a chemically defined medium without serum replacer, supplemented with activin or nodal plus FGF2 .

A key limitation of hESC culture systems is that they do not allow the propagation of pure populations of undifferentiated stem cells and their use always involves some level of background differentiation. The stem cells most commonly follow a default pathway of differentiation into an epithelial cell type that grows either as a monolayer of flat squamous cells or form cystic structures. Most probably, this form of differentiation represents differentiation of hESC into extraembryonic endoderm9.

Spontaneous' differentiation of hESC into presumably extraembryonic lineages also interferes with the derivation of somatic differentiated cells. Under various differentiation-inducing conditions, such as in embryoid bodies (EB) suspension cultures, differentiation into cystic extraembryonic structures may be common or may predominate and limit differentiation into somatic lineages.

Control and elimination of the differentiation into extraembryonic lineages therefore, may be invaluable in the derivation of somatic lineages, in addition to its importance in maintaining the stem cells in an undifferentiated state. It has been recently demonstrated that under differentiation-inducing culture conditions, the bone morphogenetic protein (BMP) antagonist noggin can prevent extraembryonic differentiation of hESC and promote their differentiation into the neural lineage9.

SUMMARY OF THE INVENTION

In accordance with a first aspect, the present invention provides a cell culture comprising cells obtained from human umbilical cord tissue, the human umbilical cord derived cells being capable of maintaining stem cells (SC) in an undifferentiated state when co-cultured therewith. The human umbilical cord derived cells are preferably used as feeder cells in SC cultures.

The invention also provides a first culture system for maintenance of SC in an undifferentiated state, the culture system comprising feeder cells expanded from human umbilical cord cells, human embryonic fibroblast cells (HEF) and a combination of same. According to one preferred embodiment, the culture system comprises the human umbilical cord derived feeder cells of the invention.

Within this aspect of the invention there is also provided an undifferentiated pluripotent human embryonic SC culture obtained by incubating a cluster of cells from inside a blastocyst with the first culture system of the invention.

The invention also provides a method for maintaining SC in an undifferentiated state, the method comprising incubating said cells with a culture system comprising feeder cells expanded from human umbilical cord cells, human embryonic fibroblast cells (HEF) or a combination of same.

The use of feeder cells expanded from human umbilical cord derived cells, human embryonic fibroblast cells (HEF) and a combination of same for the preparation of a culture system for maintenance of SC in an undifferentiated state also forms part of the invention.

In accordance with a second aspect, the invention provides a further, second, culture system for inhibiting or preventing differentiation of SC to extraembryonic cells, the culture system comprising nicotinamide (NA) or a derivative of NA having an inhibitory effect on differentiation of stem cells to extraembryonic cells similar to that of NA. A human embryonic SC culture essentially free of extraembryonic cells is also provided in the context of this aspect of the invention, the SC culture being obtained by incubating a cluster of cells from inside a blastocyst with a culture system comprising said NA or derivative thereof.

In accordance with this second aspect, there is also provided a method for inhibiting or preventing differentiation of stem cells to extraembryonic cells, the method comprises incubating said stem cells in a culture system comprising NA or a derivative of NA having an inhibitory effect on differentiation of stem cells to extraembryonic cells similar to that of NA.

Further in accordance with this aspect of the invention there is provided the use of NA or a NA derivative having an inhibitory effect on differentiation of SC to extraembryonic cells similar to that of NA for the preparation of a culture system for inhibiting or preventing differentiation of SC to extraembryonic cells.

In yet a third aspect of the invention there is provided a further, third, culture system, a humanized culture system for maintenance of SC in an undifferentiated state, the culture system comprising an animal free basic stem cell culture medium and humanized serum replacement substitute.

In accordance with this aspect there is also provided an undifferentiated human embryonic SC culture obtained by incubating a cluster of cells from

inside a blastocyst with the humanized culture system comprising the animal free stem cell basic culture medium and a humanized serum replacement substitute.

In accordance with this aspect of the invention, there is also provided a method of maintaining stem cells in an undifferentiated state, the method comprises incubating said cells with a culture system comprising animal free stem cell basic culture medium and humanized serum replacement substitute.

In accordance with a fourth aspect of the invention there is provided a culture system for maintenance SC in an undifferentiated state, the culture system comprising Neurobasal™ medium.

Within this aspect there is also provided a culture of SC in an undifferentiated state, the SC culture being obtained by culturing a cluster of cells from inside a blastocyst with a culture system comprising Neurobasal™ medium.

In accordance with this aspect of the invention there is also provided a method for maintaining a culture of SC in an undifferentiated state, the method comprising incubating said cells with a culture system comprising Neurobasal™ medium as well as the use of Neurobasal medium for the preparation of a culture system for maintaining a suspension of stem cells in an undifferentiated state.

The SC may be maintained in the Neurobasal™-based culture system in the form of a suspension as well as in a monolayer (flat colonies). Preferably, the Neurobasal™ medium is supplemented with N2 supplement or an N2 like supplement as defined below.

Finally, there is provided in accordance with the invention a method of maintaining SC in an undifferentiated state comprising culturing SC with feeder cells expanded from human umbilical cord cells.

BRIEF DESCRIPTION OF THE DRAWINGS

In order to understand the invention and to see how it may be carried out in practice, a preferred embodiment will now be described, by way of non- limiting example only, with reference to the accompanying drawings, in which:

Figures 1A-1D - are phase contrast images of cord fibroblasts primary culture (Fig. IA), human embryonic fibroblasts (Fig. IB), fibroblasts derived from umbilical cord (Fig. 1C) and from foreskin (Fig. ID).

Figure 2A-2F - are immunofmorscent images of umbilical cord foreskin, and human embryonic fibroblasts stained by anti-vimentin antibody (Figs. 2A, 2C and 2E, respectively) and the corresponding DAPI nuclear counter staining (Figs. 2B, 2D and 2F) showing that the human feeders express vimentin.

Figure 3 — is a bar graph showing FACS analysis of the percentage of feeders expressing CD44 and that are immunoreactive with anti-fibroblast antibody indicating that a high percentage of the feeders that are derived from the three sources express CD44 and are immunoreactive with anti-fibroblast antibody.

Figures 4A-4B - are representative analysis of one metaphase plate of human embryonic fibroblasts (Fig. 4A) and foreskin (Fig. 4B) showing that the human feeders have a normal karyotype. Figure 5A-5C - are phase contrast images of colonies of undifferentiated hESC that are cultured on three types of human feeders, on umbilical cord derived feeders (Fig. 5A), human embryonic fibroblasts (Fig. 5B) and on foreskin derived feeders (Fig. 5C)

Figure 6A-6L are representative FACS histograms of marker expression by hESC cultured on the three feeder fibroblast types, including expression of

SSEA4, TRAl-60, TRA1-81 and SSEAl by hESC on cord derived feeders

(Figs. 6A-6D, respectively), by hESC on human embryonic fibroblasts (Fig. 6E-

6H5 respectively) or by hESC on foreskin (Fig. 6I-6L, respectively).

Figure 7A-7C - are immunofluorescent images of hES colonies expressing AP5 when cultured on foreskin derived feeder cells (Fig. 7A), on umbilical cord derived feeder cells (Fig. 7B) and on human embryonic fibroblast cells (Fig. 7C).

Figure 8A-8F - are immunofluorescent images (Figs. 8A, 8B and 8C) and the corresponding DAPI nuclear counter staining (Figs. 8D, 8E and 8F) of hESC colonies expressing Oct4 when cultured on human embryonic fibroblast cells (Figs. 8A and 8D, cultured for 6 weeks), on foreskin derived feeders (Figs. 8B and 8E, cultured for 1 week) and on umbilical cord derived feeder cells (Fig. 8C and 8F, cultured for 10 weeks).

Figure 9 - is a bar graph representing FACS analysis of the percentage of hESC cultured on two independent cord derived feeder cell lines (CORDl and CORD2), and expressing the indicated markers of undifferentiated pluripotent stem cells at early (1-5) and late (6-10) passage levels, showing that the percentage of hESC expressing these markers is stable during propagation, as determined after 5, 8, 4 and 9 weeks of culture (5 W, 8 W, 4W and 9W).

Figure 10 - is a bar graph representing FACS analysis of the percentage of hESC cultured on two independent foreskin-derived feeder cell lines (OR2 and OR4), and expressing the indicated markers of undifferentiated pluripotent stem cells at early (1-5) and late (6-10) passage levels, showing that the percentage of hESC expressing these markers is stable during propagation as determined after 3, 6, and 8 weeks of culture (3 W, 6W and 8W).

Figure 11 - is a bar graph representing FACS analysis of the percentage of hESC cultured on two independent human embryonic fibroblast feeder cell lines (HEFl and HEF2), and expressing the indicated markers of undifferentiated pluripotent stem cells at early (1-5) and late (6-10) passage levels, showing that the percentage of hESC expressing these markers is stable during propagation, as

determined after 2, 5 and 10 weeks (2W5 5 W and 10W).

Figure 12 - is a bar graph representing analysis of the percentage of hESC cultured on two independent cord derived feeder cell lines (CORDl and

CORD2), and expressing Oct 4 at early (1-5) and late (6-10) passage levels and which was found to be stable during propagation as determined after 2, 3, 7 and

10 weeks (2W, 3W5 7W and 10W).

Figure 13 - is a bar graph representing analysis of the percentage of hESC cultured on two independent foreskin-derived feeder cell lines (OR2 and OR4), and expressing Oct 4 at early (1-5) and late (6-10) passage levels and which was found to be stable during propagation as determined after 1, 2, 5 and 9 weeks

(IW, 2W5 5W and 9W).

Figure 14 - is a bar graph representing analysis of the percentage of hESC cultured on two independent human embryonic fibroblast cell lines (HEFGl and

HEFG2), and expressing Oct 4 at early (1-5) and late (6-10) passage levels and which was found to be stable during propagation as determined after 1 and 6 weeks (IW and 6W).

Figures 15A-15I are immunofluorescent images of EBs-derived differentiated cells expressing β-tubulin (Figs. 15A5 15D and 15G)5 AFP (Figs. 15B5 15E and 15H)5 desmin (Figs. 15C and 151), or muscle-actin (m- actin, Fig. 15F) when cultured on cord-derived feeders (Figs. 15A-15C); on human embryonic fibroblasts (Figs. 15D-15F); and on foreskin derived feeders (Figs. 15G-15I).

Figure 16 - is a bar graph showing the effect of bFGF at the indicated concentration on the number of cells that were harvested per flask at the time of the culture split as shown.

Figure 17A-17D - are phase contrast images of cord-derived feeders, showing the effect of bFGF on their morphology after prolonged propagation in the presence of serum without bFGF-supplementation (Fig. 17A) or with the two

indicated concentrations of bFGF supplementations (Fig. 17B and Fig. 17C), FACS analysis of the percentage of feeders expressing CD44 and that are immunoreactive with anti-fibroblast antibody (Anti-fib ab) is also shown (Fig. 17D). Analysis was performed at passage 10 in the presence of serum, and at passage 17 when the medium was supplemented with 5ng/ml and 10 ng/ml of bFGF.

Figure 18A-18F - are phase contrast images (Fig. 18A-18C) and immunofluorescent images (Fig. 18D-18F) of hESC colonies cultured on cord- derived fibroblasts that were propagated for 17 passages in the presence (Figs. 18B, 18C, 18E and 18F) or absence (Figs. 18A and 18D) of bFGF. The cord-derived fibroblasts supported undifferentiated proliferation of the hESCs as determined by the expression of alkaline phosphatase by the hESC (Fig. 18D-18F) and the expression of stem cell markers by a high percentage of the hESCs (FACS analysis, Fig. 18G). Figure 19 - is a phase contrast micrograph of hESC cultured on HEF feeder layer in Cellgro medium supplemented with 1% TCH showing that hESC retain the morphology of undifferentiated pluripotent stem cells when TCH is used as the serum replacement supplement..

Figure 20 - is a bar graph showing the percentage of SSEA-4 expressing on hESC, when cultured on a foreskin or HEF feeder layers, being similar when the KO DMEM was supplemented with KO SR5 2% TCH, or 2% Nutridoma and showing that Nutridoma-CS is as effective as TCH in supporting undifferentiated propagation of hESC.

Figure 21 - is a phase contrast micrograph of hESC colonies cultured within NBN2 showing that hESCS retain the morphology of undifferentiated cells when colonies are cultivated on human feeders in NBN2.

Figure 22A-22D - are dark field micrographs of small transparent clusters of cells that develop 7 days after transfer of undifferentiated hESCs into

suspension culture within NBN2 medium (Fig. 22A) and after 3 weeks in suspension culture within NBN2, indirect immunofluorescent analysis showed that the majority of hESCs express SSEA4 (Fig. 22B) and Oct4 (Fig. 22D). Nuclei of cells in D are counterstained with DAPI in (Fig. 22C).

Figure 23A-23B - are bar graphs showing the percentage of SSEA-4+ cells (Fig. 23A) and total number of cell/well (Fig. 23B) as analyzed after 3 weeks suspension culture of equal initial numbers of hESC in NBN2 medium + FGF2 supplemented with various combinations of ECM components and factors.

Figure 24A-24D - are dark field micrographs of EBs that were cultured for 4 weeks in the presence and absence of NA (the culture medium included 10% FCS). EBs with typical cystic structures (cystic EBs) developed in the absence of NA (Figs. 24A and 24B), while in the presence of NA, cystic formation was not observed and the EBs were comprised of tightly packed cells (Figs. 24C and 24D). Figures 25A-25P - are dark field micrographs of EBs that were cultured for 2-5 weeks in chemically-defined medium (NBN2) in the presence or absence of NA and retinoic acid as indicated. EBs with typical cystic structures (cystic EBs) developed in the absence of NA (Figs. 25A-25D, i.e. upper panel). In the presence of NA, cystic formation was not observed, the EBs were comprised of tightly packed cells and were significantly larger (Figs. 25E-25H, i.e. second panel). In the presence of RA, the EBs were smaller and included multiple cysts (Figs. 25I-25L, i.e. third panel). NA blocked the effects of RA (Figs. 25M-25P, i.e. lower panel).

Figure 26 - is a RT-PCR analysis demonstrating that the expression of the endodermal marker α-fetoprotein is suppressed within EBs that differentiated in the presence of NA in comparison to control EBs that were cultured in the absence of NA. After 4 weeks of differentiation, the effect of NA was more prominent in comparison to the effect after 2 weeks.

Figures 27A-27D - are immunocytochemical studies demonstrating suppressed expression of α-fetoprotein (AFP) and cytokeratin-8 (CK) in EBs that differentiated in the presence of NA. Following 4 weeks of differentiation in the presence of NA, only a few cells in sections of EBs were immunoreactive with anti-α-fetoprotein (Fig. 27A) and cytokeratin-8 (Fig. 27B). Cells that expressed α-fetoprotein (Fig. 27C) and cytokeratin-8 (Fig. 27D) were abundant within sections of control EBs that differentiated in the absence of NA.

Figure 28A-28D - are dark field micrographs of EBs differentiating in the presence of NA showing that the percentage of EBs that included clusters of differentiated cells expressing melanin increased with time (Figs. 28 A, 28B, 28C and 28D representing results after 2, 4, 6 and 12, respectively).

Figure 29 - is a real time PCR analysis of EBs differentiated for 6 weeks in the presence or absence of NA, demonstrating the induction of expression of RPE markers by NA. Figures 30A-30H -_are images showing that melanin-expressing cells that were generated in the presence of NA had morphological characteristics that are typical of RPE cells. Specifically shown are dark field micrograph (Fig. 30A), phase contrast image (Fig. 30B) and indirect immunofluorescent stainings of RPE cells markers including ZO-I (Fig. 30C), Pax6 (Fig. 30D), MITF (Fig. 30E), CRALBP (Fig. 30F), Bestrophin (Fig. 30G) and RPE65 (Fig. 30H).

Figure 31 - is RT-PCR analysis showing the expression of chordin-like 1 by cells within EBs that were developed in the presence of NA.

Figure 32A-C - are H&E and fluorescent images demonstrating the survival of transplanted hESC-derived RPE cells and their integration within the host RPE layer of cells. An H&E image showing the survival of an intra-vitreal graft, 4 weeks after transplantation into the eye of a mature RCS rat (Fig 32A). The graft includes melanin expressing cells (dark pigmented cells). Indirect immunofluorescent staining demonstrates that the cells within the graft express

GFP (white spots), confirming their human identity (Figure 32B). Integration of transplanted hESC-derived RPE cells (pigmented cells marked with arrows) in the albino rat RPE layer is also demonstrated (Figure 32C). Pigmented cells were not observed in the RPE layer of control non transplanted eyes.

DETAILED DESCRIPTION OF EXEMPLARY EMBODIMENTS

The invention is described in the following detailed description with reference to cell cultures and culture systems for handling stem cells, preferably human embryonic stem cells. It should be noted that in addition to the cell cultures and culture systems discussed in detailed hereinbelow, also encompassed within the present invention are uses of specific components described with reference to the culture system in the preparation of such culture systems as well as to methods of use of the culture system in handling stem cells cultures and methods of preparing culture cells.

As used in the specification and claims, the forms "a", "an" and "the" include singular as well as plural references unless the context clearly dictates otherwise. For example, the term "a culture system" includes one or more culture systems.

As used herein, the term "or" means one or a combination of two or more of the listed choices Further, as used herein, the term "comprising" is intended to mean that the methods or composition includes the recited elements, but not excluding others. Similarly, "consisting essentially of is used to define methods and systems that include the recited elements but exclude other elements that may have an essential significance on the functionality of the culture systems of the inventions. For example, a culture system consisting essentially of a basic medium, medium supplements and feeder cells will not include or include only insignificant amounts (amounts that will have an insignificant effect on the

propagation and differentiation of cells in the culture system) of other substances that have an effect on cells in a culture. Also, a composition consisting essentially of the elements as defined herein would not exclude trace contaminants from the isolation and purification method. "Consisting of shall mean excluding more than trace elements of other elements. Embodiments defined by each of these transition terms are within the scope of this invention.

Further, all numerical values, e.g., concentration or dose or ranges thereof, are approximations which are varied (+) or (-) by up to 20%, at times by up to 10% of from the stated values. It is to be understood, even if not always explicitly stated that all numerical designations are preceded by the term "about". It also is to be understood, although not always explicitly stated, that the reagents described herein are merely exemplary and that equivalents of such are known in the art.

In its broadest sense, the present invention concerns culture cells, systems and methods for use of same in culturing of stem cells. As used herein, the term "stem cells" refers to cells which are capable of differentiating into other cell types having a particular, specialized function (i.e., "fully differentiated" cells) or self renewing and remaining in an undifferentiated pluripotential state as detailed below.

As used herein, the term "cell" refers to a single cell as well as to a population of (i.e. more than one) cells. The population may be a pure population comprising one cell type. Alternatively, the population may comprise more than one cell type. Furthermore, as used herein, the term "cell culture" refers to any in vitro culture of cells. Included within this term are continuous cell lines (e.g. with an immortal phenόtype), primary cell cultures, finite cell lines (e.g., non- transformed cells), and any other cell population maintained in vitro.

As used herein, the teπn "primary cell" is a cell which is directly obtained from a tissue, or organ of an animal, including a human, in the absence of culture. Typically, though not necessarily, a primary cell is capable of undergoing ten or

fewer passages in vitro before senescence and/or cessation of proliferation. In contrast, a "cultured cell" is a cell which has been maintained and/or propagated in vitro for ten or more passages

Non-limiting examples of stem cells are hematopoietic stem cells obtained from bone marrow tissue of an individual at any age or from cord blood of a newborn individual, embryonic stem (ES) cells obtained from the embryonic tissue formed after gestation (e.g., blastocyst), or embryonic germ (EG) cells obtained from the genital tissue of a fetus any time during gestation, preferably before 10 weeks of gestation. Preferred stem cells according to the present invention are human stem cells, more preferably, hESC.

Stem cells can be obtained using well-known cell-culture methods. For example, hESC can be isolated from human blastocysts. Human blastocysts are typically obtained from human in vivo preimplantation embryos or from in vitro fertilized (IVF) embryos. Alternatively, a single cell human embryo can be expanded to the blastocyst stage. For the isolation of human ES cells the zona pellucida is removed from the blastocyst and the inner cell mass (ICM) is isolated by immunosurgery, in which the trophectoderm cells are lysed and removed from the intact ICM by gentle pipetting. The ICM is then plated in a tissue culture flask containing the appropriate medium which enables its outgrowth. Following 9 to 15 days, the ICM derived outgrowth is dissociated into clumps either by a mechanical dissociation or by an enzymatic degradation and the cells are then re- plated on a fresh tissue culture medium. Colonies demonstrating undifferentiated morphology are individually selected by micropipette, mechanically dissociated into clumps, and re-plated. Resulting ES cells are then routinely split every 1-2 weeks. For further details on methods of preparation human ES cells see Thomson et al., [U.S. Pat. No. 5,843,780; Science 282:1145, 1998; Curr. Top. Dev. Biol. 38:133, 1998; Proc. Natl. Acad. Sci. USA 92: 7844, 1995]; as well as Bongso et al, [Hum Reprod 4: 706, 1989]; and Gardner et al., [Fertil. Steril. 69:84, 1998].

Commercially available stem cells can be also be used in accordance with the invention. Human ES cells can be purchased from the NIH human embryonic stem cells registry. Non-limiting examples of commercially available embryonic stem cell lines are BGOl, BG02, BG03, BG04, CY12, CY30, CY92, CYlO, TE03 and TE32.

Potential applications of hESC are far ranging and include drug discovery and testing, generation of cells, tissues and organs for use in transplantation, production of biomolecules, testing the toxicity and/or teratogenicity of compounds and facilitating the study of developmental and other biological processes. For example, diseases presently expected to be treatable by therapeutic transplantation of hESC or hESC derived cells include Parkinson's disease, cardiac infarcts, juvenile-onset diabetes mellitus, and leukemia [Gearhart J. Science 282: 1061-1062, 1998; Rossant and Nagy, Nature Biotech. Yl: 23-24, 1999]. There are, however, significant hurdles to the practical exploitation of hESC. To maintain hESC in an undifferentiated pluripotential state, the cells are usually cultured on feeder cells. The feeder cells can secrete factors needed for stem cell self-renewal and proliferation, while at the same time, inhibit their differentiation. Commonly used feeder cells includes a primary mouse embryonic fibroblast (PMEF), a mouse embryonic fibroblast (MEF), a murine fetal fibroblast (MFF), a human embryonic fibroblast (HEF), a human fetal muscle cell (HFM), a human fetal skin cell (HFS), a human adult skin cell, a human foreskin fibroblast '(HFF), a human adult fallopian tubal epithelial cell (HAFT) and a human marrow stromal cells (hMSCs).

As used herein, the term "undifferentiated pluripotential hES cells" or

"hESC" refers to human precursor cells that have the ability to form any adult cell. Such cells are true cell lines in that they (i) are capable of indefinite

proliferation in vitro in an undifferentiated state; and (ii) are capable of differentiation to derivatives of all three embryonic germ layers (endoderm, mesoderm, and ectoderm) even after prolonged culture. Human ES cells are derived from fertilized embryos that are less than one week old.

Pluripotent SC present at their surface or express biological markers which are used to identify pluripotent SC as well as to verify that the cells in the culture are maintained in an undifferentiated state [Thomson JA et al. Embryonic Stem Cell Lines Derived from Human Blastocysts Science 282(5391):1145 - 1147 (1998)]. A" non-limiting list of such cell markers comprise stage-specific embryonic antigen such as SSEA-3, SSEA-4; antibodies to specific extracellular matrix molecule which are synthesized by undifferentiated pluripotent SC, such as TRA-1-60, TRA- 1-81 and GCTM-2; elevated expression of alkaline phosphatase which is associated with undifferentiated pluripotent SC; transcription factors unique to pluripotent SC and which are essential for establishment and maintenance of undifferentiated SC, such as , OCT-4 and Genesis [Carpenter, m.k., Rosier, E., Rao M.S., Characterization and Differentiation of Human Embryonic Stem Cells. Cloning and Stem Cells 5, 79- 88, 2003].

While widely used, human SC cultures based on murine derived feeder cells, are less desired. Non-species specific feeder cell technology reduces the value of stem cell cultures due to the foreign nature of the source of the feeder cell. For example, such non-species specific feeder cells contain both foreign cells and foreign growth factors. Further, it is believed that the use of non-species specific feeder cells in combination with different but desirable cultured cells cannot provide the optimum growth conditions as species specific derived feeder cells or conditioned media. The issue of cross-species contamination is particularly relevant to agricultural animals, endangered species, laboratory animals, non-human primate cells, and hESC. It has been shown that hESC are contaminated by foreign molecules when cultured with mouse-derived feeders

(Martin, M.J., et al., Human embryonic stem cells express an immunogenic nonhuman sialic acid. Nat Med. 2005; 11: 228-32). Contamination of hESC by mouse derived molecules/pathogens may interfere with their exploitation as a model for basic research and raises concerns as to their use in transplantation therapy. Still further, non-human feeder cell technology reduces the value of human derived SC cultures, as, for example, such non-human feeder cells contain both non-human cells and non-human growth factors. Also, it is believe that the use of non-human feeder cells in combination with human cultured cells cannot provide the optimum growth conditions as human derived feeder cells. Thus, the present invention provides, in accordance with a first of its aspects, a cell culture derived from human umbilical cord tissue, preferably excluding hematopoietic tissue, and being capable of maintaining SC in an undifferentiated state when co-cultured therewith. These feeder cells are obtained from culturing, preferably in an animal free culture system, of cells taken from umbilical cord tissue under conditions which allow the cells to propagate/expand and isolating the thereby propagated cells. The cells in the culture are essentially fibroblast cells and are preferably used as feeders in stem cell culture systems.

The cell cultures (either the feeder cells or the SC cultures) in accordance with the invention may be a fresh cell culture, cryopresrved culture as well as cryopreserved and thawed cells.

As used herein, the term "derived" which may be used interchangeably with the term "obtained" when used in the context of cell formation denotes the development of a new cell line from another cell line. For example, human embryonic cord derived cells denote, in accordance with one embodiment of the invention, fibroblast cells originating from embryonic cord tissue, which under suitable condition propagate into a fibroblast cell line.

Further, as used herein, the term "feeder cells" which is known interchangeably with "feeders" denotes any type of cells which may be used as a

substratum for other cells attachment and growth in a culture system. Feeder cells are typically used to allow growth and survival of single undifferentiated stem cells. The Feeder cells provide conditions that maintain cell proliferation, inhibit cell differentiation and preserve pluripotency. Specifically, the feeder cells are cells that secrete factors needed for stem cell proliferation, while inhibit their differentiation. Methods of preparing feeder cells are well known in the art (see, for example, U.S. patent Pub. No. 20030143736). Generally, the feeder cells may be fibroblasts or other types of cells, and the cells are inactivated by large- dose radiation before use, such as γ-ray, or by drugs, such as mitomycin C. After the inactivation process, the surviving cells lost the capability to proliferate, but retained their physiological functions, such as metabolism and synthesis of growth factors.

As indicated above, the feeder cells are derived (expanded) from umbilical cord tissue. Umbilical cord tissue may be obtained in the course of vaginal delivery. However, a major advantage of using umbilical cord tissue is that it may be obtained during elective cesarean section in a sterile environment of an operating theater. Moreover, the umbilical cord is obtained from the sterile environment of the amniotic sac and has not been exposed to any external contagious agents prior to donation. The sterile nature of umbilical cord donation allows the derivation of feeders from the umbilical cord tissue without the use of antibiotics or anti-fungal drugs. Avoiding the use of anti-bacterial and anti-fungal drugs is an advantage since these drugs may interfere with the growth of cells in culture, alter the results of basic science studies and most importantly may induce allergic reactions in, recipients of cells that were cultured in the presence of these drugs. Derivation of feeders from other human primary tissues such as foreskin or aborted fetuses are done under significant less sterile conditions. The foreskin is exposed to bacteria that colonize the genital area and it may be disinfected but not sterilized. Aborted fetuses are also exposed to potential contamination by vaginal and genital flora during dilatation and curettage.

An additional advantage of umbilical cord as opposed to foreskin or human fetal tissues is that a significant volume of blood may be sampled from the umbilical cord, tested for contagious agents and archived. This is not possible with foreskin tissues donated by newborn babies or with aborted fetuses. Lastly, umbilical cord is routinely discarded and its donation is not associated with emotional or moral constrains, while donation of fetal tissues raises ethical concerns and is not morally accepted by many.

In this connection there is thus also provided a method for preparing umbilical cord derived feeder cells, the method comprising isolating umbilical cord cells from umbilical cord tissue and culturing said umbilical cord cells in a culture medium including serum, thereby preparing said human umbilical cord feeder cells. The umbilical cord cells may be isolated from the umbilical cord tissue by mincing the tissue and affixing the umbilical cord to a wall, such as a wall of a flask, and allowing the cells to incubate undisturbed for a number of weeks until fibroblast cells begin to migrate out of the minced umbilical cord tissue.

The umbilical cord tissue may be obtained from healthy pregnant women undergoing elective Cesarean sections at term.

In accordance with the invention there is also provided a culture system for maintaining stem cells (SC) in an undifferentiated state, the culture system comprising feeder cells selected from cells obtained from human umbilical cord tissue (excluding cells obtained from umbilical cord blood), human embryonic fibroblast cells (HEF) or a combination of same. The culture system according to this aspect of the invention is term herein the "human derived feeder cell aspect of the invention".

As used herein with respect to all aspects of the invention, the terms "maintenance" means continued survival of a cell or population of cells, at times, with an increase in numbers of cells. "Proliferation", "propagation" ,

" expansion" and "growth", which are used interchangeably, refer to such an increase in cell number. According to one embodiment, when referring to maintenance of hESC on feeder cells, this term refers to a continuous survival of the cells for at least 10 weeks.

The culture systems in accordance with the invention are preferably for enabling maintenance of a population of stem cells when cultured on feeder cells, and at time, propagation of same, for a prolonged period of time, the period of time being at least 10 weeks.

When the feeder cells are derived from human umbilical cord tissue, the feeder cells are essentially fibroblast cells. The term "essentially fibroblast cells" denotes that the feeder cells comprise in its majority fibroblasts, i.e. at least 70 of the cells in the feeder cell population are fibroblast, preferably 85%, a most preferably all the cells, i.e. essentially 100% of the feeder cells are fibroblasts.

In accordance with one embodiment, the feeder cells are provided in a form of a monolayer coated culture dish to which a nutrient medium is added along with the culture cells. As used herein, the terms "monolayer", "monolayer culture" and "monolayer cell culture" refer to cells that have adhered to a substrate and grow as a layer that is one cell in thickness. Monolayer cells may be grown in any format, including but not limited to flasks, tubes, coverslips (e. g., shell vials), roller bottles, etc. Monolayer cells may also be grown attached to microcarriers, including but not limited to beads. At times, the term monolayer also includes growth of cells as flat colonies.

The term "culture system" denotes a combination of elements, such as an extracellular matrix (ECM) and a culture (nutrient) medium which together provide suitable conditions that support SC growth. The conditions are such that SC can proceed through the cell cycle, grow and divide. Preferably, the conditions are such which enable growth of human stem cells, preferably, hESC. Further, the culture system provides conditions that permit the embryonic stem

cells to stably proliferate in the culture system for at least 10 weeks. The nutrient medium may contain any of the following appropriate combinations: a basic medium (a cell culture medium usually comprising a defined base solution, which includes salts, sugars and amino acids) as well as serum or serum replacement, and other exogenously added factors. It is not intended that the term "culture medium" or "nutrient medium" be limited to any particular culture medium. For example, it is intended that the definition encompass outgrowth as well as maintenance media. In accordance with the human derived feeder cell aspect of the invention, the culture system also comprises the feeder cells. However, the feeder cells may be substituted with components derived from feeder cells or other known and acceptable substitutes thereof, e.g. when referring to other culture systems disclosed herein.

In accordance with one embodiment, the culture system is employed for maintaining hESC in an undifferentiated pluripotential state, as evidenced in the following non-limiting examples by the expression of proteins such as SSEA-4, TRA- 1-60, OCT-4, APase, but not SSEA-I. Methods of preparing culture systems for culturing hESC are well known in the art [see, for example, Reubinoff Be. et. al., Nat. Biotechnol. 18:399-404, 2002; Richards, M. et al, Nat. Biotechnol. 20:933-936, 2002]. A hESC medium may typically contain 80% Dulbecco's Modified Eagles

Medium (DMEM), 20% defined Fetal Calf Serum, 1% L-Glutamine, 0.5% penicillin/streptomycin, 1% non-essential amino acids, 1% Insulin-Transferrin- Selenium G supplement and 1 mM β-mercaptoethanol.

In an animal free culture system, which provides a pathogen-free environment for the growth of ES cells, the cultures rely on human feeder layers supplemented with human serum or serum replacement suitable for the growth of human stem cells. The feeder cells may be any suitable cells from human source as known in the art or the isolated umbilical cord derived feeder cells of the

invention; the stem cells medium DMEM (used as the basic media) may be replaced with KO DMEM (Gibco, or equivalent), X- Vivo 10 (Biowhittaker, Maryland, or equivalent) or Cellgro Stem Cell Growth Medium (CellGenix, Freiburg, Germany, or equivalent); the FCS may be replaced with humanized serum replacement substitute, such as TCH (Protide Pharmaceuticals, St. Paul, MN, or equivalent) or Nutridoma-CS (Roche, Germany, or equivalent). Since the animal free system provides a pathogen free environment, reducing agents such as β-mercaptoethanol and antibacterial agents such as penicillin/streptomycin) may be eliminated. In the context of the human derived feeder cell culture system aspect of the invention there is also provided a method for maintaining stem cells in an undifferentiated state, the method comprises incubating (co-culturing) said cells with a culture system comprising feeder cells selected from human umbilical cord tissue derived cells, human embryonic fibroblast cells (HEF) or a combination of same.

According to one embodiment, the stem cells are incubated in a culture system where the feeder cells are preferably provided as a layer of cells, preferably a mono-layer, formed on a base of culture dish. The culture system is then provided with a growth environment, typically, an environment in which cells of interest will proliferate in vitro. Temperatures of 370C and 5% CO2 in air are generally adopted.

In cultures of undifferentiated hESCs there is always some level of background extraembryonic differentiation. Further, in currently-used systems for the cultivation of undifferentiated hESCs, or for induction of their differentiation towards somatic lineages, three is tendency of hESCs to differentiate towards extraembryonic lineages. In addition, upon induction of differentiation, the default pathway of differentiation towards extraembryonic lineages may predominate, and limit differentiation into desired somatic lineages.

Thus, the invention also provides a culture system for inhibiting or preventing differentiation of stem cells towards extraembryonic lineages (to extraembryonic cells). The culture system in accordance with this aspect of the invention comprise NA (NA) or a derivative of NA having an inhibitory effect on differentiation of stem cells towards extraembryonic lineages (to extraembryonic cells) similar to that of NA. This aspect of the invention is referred to herein as the "nicotinamide aspect of the invention".

NA is a form of Vitamin B3 that may preserve and improve beta cell function. NA is essential for growth and conversion of foods to energy and it has been used in diabetes treatment and prevention. It has now been found that NA is capable of inhibiting, preferably, preventing differentiation of embryonic stem cells towards extraembryonic lineages (to extraembryonic cells).

The term "derivative of nicotinamide" as used herein denotes a compound which is a chemical modification of the natural NA.


Nicotinamide (NA)

The chemical modification may include substitution on the pyridine ring of the basic NA structure (via the carbon or nitrogen member of the ring), via the nitrogen or the oxygen atoms of the amide moiety, as well as deletion or replacement of a group, e.g. to form a thiobenzamide analog of NA, all of which being as appreciated by those versed in organic chemistry. The derivative in the context of the invention also includes the nucleoside derivative of NA (e.g. nicotinamide adenine). A variety of NA derivatives are described, some also in

connection with an inhibitory activity of the PDE4 enzyme [WO03068233; WO02060875; GB2327675A], or as VEGF-receptor tyrosine kinase inhibitors [WO01/55114]. For example, the process of preparing 4-aryl-nicotinamide derivatives are described in WO05014549A. The NA derivatives in the context of the invention are compound determined to have an inhibitory effect, preferably preventative effect, on differentiation of stem cells to extraembryonic lineages (extraembryonic cells), similar to that of NA.

The effect of NA may be the result of inhibition of poly (ADP-ribose) polymerase (PARP). Therefore the effect of NA may be also achieved by treating the cells with other PARP inhibitors such as 3-aminobenzmide, PJ-34 or 1, 5- dihydroxyisoquinoline. These other PARP inhibitors are also included in the context of the term "modification of NA". Yet further, the effect of NA may also be attributed to the inhibition of SIRT protein deacetylase. Therefore its effect may be also obtained by other SIRT inhibitors such as splitomicin and sirtinol, which are thus, also included in the context of the term term "modification of NA".

In accordance with the NA aspect of the invention, the stem cells may be as described above, i.e. they may be stem cells from any source, but are preferably human stem cells, further preferably, human embryonic stem cells. As used herein "inhibition of extraembryonic differentiation" used synonymy with the term "prevention of extraembryonic differentiation" denotes the maintainance as well as the expansion of embryonic stem cell in a cell culture and that the resulting cell culture is essentially free of extraembryonic cells or membranes. The term "essentially free" is used to exclude extraembryonic cells that may have an essential significance on the functionality of the stem or somatic cells in the culture or that the amount of the extraembryonic cells in the cell culture is insignificant (an amount that will have an insignificant effect on the propagation and differentiation of cells in the culture system).

It is well appreciated that if extraembryonic differentiation is essentially eliminated, a key challenge is to further direct differentiation into a specific somatic lineage and into a specific type of cell. It has now been found that supplementation of a culture medium with NA can prevent the default differentiation of hESCs towards extraembryonic lineages. It may also direct the differentiation towards specific somatic lineage such as but not limited to neural differentiation. The examples provided herein show differentiation to neural precursor cells.

Proliferation and differentiation of embryonic stem cells into insulin- producing cells in the presence of NA was suggested by Vaca P .et al,

[Transplant Proc. 35(5):2021-3 2003] Specifically, it was shown that while proliferation within EBs with or without supplementation of the medium with

NA is similar (Figure IA in Vaca P .et al.), insulin content is increased in cells that differentiate in the presence of NA. Nevertheless it is unclear whether the increased insulin content was not related to increased uptake of insulin from the medium.

It has now been found that NA effectively induced differentiation of stem cells into somatic cells. Specifically, albeit, not exclusively, NA was shown to induce differentiation to neural cells, and within the neural lineage, NA treatment was found to promote differentiation towards retinal pigmented epithelial (RPE) cells. The use of RPE cells in transplantation has already been described [Haruta, M. et al, In vitro and in vivo characterization of pigment epithelial cells differentiated from primate embryonic stem cells. Invest Ophthalmol Vis Sci, 45:1020-1025 (2004)]. Thus, it is to be understood that the RPE cells obtained in accordance with the present invention have various therapeutic applications. One such application includes transplantation of such cells in the eye to replenish malfunctioning or degenerated RPE cells in retinal degenerations. Genetically modified RPE cells may serve as a vector to carry and express genes in the retina after transplantation. Other applications may be the use of hESC-derived RPE

cells as an in vitro model for the development of new drugs to promote RPE survival and function. hESC-derived RPE cells may serve for high throughput screening for compounds that are toxic, trophic, induce differentiation proliferation and survival of RPE cells. They may be used to uncover mechanisms, new genes, soluble or membrane-bound factors that are important for the development, differentiation, maintenance, survival and function of photoreceptor cells.

The culture system in the NA aspect of the invention comprises standard elements of culture media, as defined above combined with NA. The concentration of NA in the medium may vary, however, will preferably be in a concentration range between about ImM to about 2OmM, more preferably at a concentration of about 1OmM.

In the context of this aspect of the invention there is also provided a method for inhibiting or preventing differentiation of stem cells towards extraembryonic lineages (to extraembryonic cells), the method comprises incubating said stem cells in a culture system comprising NA or a derivative of

NA as defined above.

It should be noted that in the context of the present invention the NA based culture systems was also effective for increasing the survival of SC in the culture system. According to one embodiment, cells survived in the culture for at least 12 weeks.

It should also be noted that the NA based culture system of the invention was effective to induces an increase in number of cells within embryoid bodies (EB) cultured therein. For induction of somatic differentiation, the stem cells in accordance with the NA aspect of the invention are preferably grown as free floating clusters in a suspension. As used herein, the terms "suspension" and "suspension culture" refer to cells that survive and proliferate without being attached to a substrate.

A further aspect of the invention concerns the use of serum (e.g., fetal bovine serum (FBS)), in SC cultures. It has already been established that serum is a major source of undefined differentiation factors and thus tends to promote ES cell differentiation. Other problems are also" associated with serum. Lot-to-lot variation is often observed and some lots of serum have been found to be toxic to cells [Robertson, EJ., ed., Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, IRL Press, Oxford, UK (1987)]. Moreover, serum may be contaminated with infectious agents such as mycoplasma, bacteriophage, and viruses. Finally, because serum is an undefined and variable component of any medium, the use of serum prevents the true definition and elucidation of the nutritional and hormonal requirements of the cultured cells.
?????????????????????????????????????????????????????????????????????
+++++++++++++++++++++++++++++++++++++++++++++++++++++++++++++++++++++
It has now been found that the use of two well known and commercially available humanized serum replacement (SR) substitutes, which have not been used hitherto in human embryonic stem cell technology, i.e. TCH™ (Protide Pharmaceuticals, St. Paul, MN, or equivalent) and Nutridoma-CS (Roche, Germany, or equivalent) used as serum replacement substitute, did not impair the undifferentiated propagation of the stem cells in the culture system.
+++++++++++++++++++++++++++++++++++++++++++++++++++++++++++++++++++++
??????????????????????????????????????????????????????????????????????
Thus, according to a further aspect, the invention also provide a humanized culture system for maintenance of stem cells (SC) in an undifferentiated state, the humanized culture system comprising animal free stem cell basic medium and a humanized serum replacement substitute. This aspect of the invention is referred to as the "humanized serum free culture system of the invention".

In accordance with one embodiment, the humanized culture system comprises a serum free basic medium as known to those versed in the art of stem cells (i.e. a medium which is free of animal origin and is suitable for growth of stem cells), selected from Cellgro Stem Cell Growth Medium, KO DMEM,

Neurobasal™, or X-Vivo 10.

In accordance with another embodiment, the humanized culture system comprises a serum replacement substituent selected from TCH™, Nutridoma-CS or combination of same.

In accordance with yet another embodiment, when said serum free basic medium is Neurobasal™, the culture system further comprises N2 supplement [GIBCO® Cell Culture] or a modified N2 supplement, the modification rendering the medium supplement suitable for use with stem cells. It is noted that the standard and commercially available N2 supplement comprises insulin, transferrin, progesterone, putrascine, selenite. The specific composition of N2 supplement as published by StemCell Technologies Inc (Product Information Sheet, revised on December 2002) includes 2.5mg/mL rh insulin, lOmg/mL human transferring (which may be iron-poor or iron-saturated), 0.52μg/mL sodium selenite, 1.61mg/mL putrascine, 0.63μg/mL progesterone, all in phosphate buffered saline. Nonetheless, modifications of the standard N2 supplement for stem cells maintenance are readily envisaged by those versed in the art. For example, medium for the propagation of ESC-derived neural stem cells is supplemented with modified N2 [Conti, L, et al, Niche-independent symmetrical self-renewal of a mammalian tissue stem cell. PLoS Biol. 9:e283 (2005)]. TCH™ is a completely biochemically defined serum replacement developed primarily for human cells and production of cell-secreted proteins. TCH™ may be purchased from Protide Pharmaceuticals (MN, USA) as well as from BM Biomedicals (CA, USA).

👍️0
Lazarus Lazarus 17 years ago
New liver without transplant, from bone marrow stem cells
27 Sep 2007, 0048 hrs IST,Kounteya Sinha,TNN

NEW DELHI: There's finally some hope for patients across the globe needing an urgent liver transplant but are unable to find a compatible donor organ.

Scientists have, for the first time, found that stem cells, taken from the patients' bone marrow and injected into the diseased liver, can keep them alive until donor organs become available. Miraculously, the cells can also support liver function, until the organ is able to regenerate itself, eliminating the need for a transplant at all.

The demand for liver donors is very high and many patients die waiting for one or are taken off because their condition deteriorates to the extent that they would not survive the operation when their turn finally arrives. A longstanding goal in hepatology has been to achieve suppression of liver cell death until regeneration could occur.


In a series of animal studies, Massachusetts General Hospital researchers successfully treated rats with liver diseases by manipulating their immune response. If this procedure can be repeated in humans, it could potentially reduce the number of donor organs used in urgent transplant procedures, thereby increasing the number available for patients on waiting lists.

For example in India, 15,000 people require a liver transplant every year. Shockingly, only 150 get lucky. India requires 22,000 donors annually. Doctors say they get only 50 donors a year.

Eminent liver transplant specialist Dr A K Soin from Sir Gangaram Hospital said, "This therapy can be promising for patients with acute liver failure in whom the effect of liver failure can prove fatal before the patient's liver has had a chance to regenerate. The technique can be an important bridge to liver recovery. If it can support liver function for a few weeks, then it will give time for the patient's liver to regenerate."

The researchers used mesenchymal stem cells (MSCs) — cells from the bone marrow that develop into tissues supporting blood cell development in the marrow cavity. Previous research has shown that MSCs are able to inhibit several immune system activities, apparently by putting a break on the movement of immune cells to areas of damage. The researchers tested several ways of using the cells to treat rats with liver failure. Simply transplanting MSCs into the animals' livers did not work. Two subsequent methods of delivering molecules secreted by cells lessened inflammation within the liver and halted cell death.

kounteya.sinha@timesgroup.com�
👍️0
Lazarus Lazarus 17 years ago
Male Testicles Are Potential Fountain of Stem Cell Youth
By Brandon Keim September 20, 2007 | 1:10:17 PMCategories: Biotechnology, Stem Cell Research

Adult stem cells taken from testicles could be a source for everything from blood vessels and heart tissue to new brain cells, report Howard Hughes Medical Institute researchers.

Unsurprisingly, the combination (stem cells + testes!) caught the attention of journalists. Australia's ABC News did a nice job of pointing out that the findings, made in mice, have a long way to go before helping people, and that women's ovaries might provide equally adaptable adult stem cells. Scientific American noted that similar findings were made earlier in the year, so at least this isn't a one-off.

The BBC hinted that men would be reluctant because extracting the cells would be "very painful," but didn't say how it's done; apparently it's like getting a biopsy, which I'd imagine is rather less painful than, say, heart disease or dementia. The New York Post covered it briefly -- mostly, I suspect, to let their headline writers have some fun. (The result: "New Ballgame for Stem Cells".)

But the best story might have come from the Globe and Mail. They gave some edifying but gruesome detail -- teeth and hair are sometimes found in testicular tumors, so these cells really can grow -- and didn't skimp over the fact that the researchers don't actually understand how their experiment worked.

None of the writers mentioned the poetic justice of men someday regenerating their whole bodies from their nuts, but I'm sure they all thought of it.

👍️0
Lazarus Lazarus 17 years ago
BioLife Solutions' CryoStor(TM) Adopted by Tissue Regeneration Therapeutics for Preservation Protocol for New, Non-Embryonic Source of Stem Cells
Monday September 17, 6:00 am ET
Umbilical Cord Tissue Provides Building-Block Cells for Muscle, Bone, Connective Tissues


BOTHELL, Wash., Sept. 17 /PRNewswire-FirstCall/ -- BioLife Solutions Inc. (OTC Bulletin Board: BLFS - News), a leading developer and marketer of proprietary hypothermic storage and cryopreservation media products for cells, tissues, and organs, today announced that privately held Tissue Regeneration Therapeutics Inc. (TRT) has adopted and recommends BioLife's CryoStor as part of its standard preservation protocol for mesenchymal stem cells derived from umbilical cord tissue.

TRT holds patents and intellectual property rights to a process of removing and storing stem cells from umbilical cord tissue. Mesenchymal stem cells are the progenitor cells for muscle, bone, and connective tissues. Published uses of mesenchymal cells in cell therapy include treatment of auto-immune and inflammatory diseases, cancer, heart disease, and tissue engineering.

John E. Davies, President of TRT, Professor of Biomaterials and Biomedical Engineering at the University of Toronto, and inventor of the HUCPVC (Human Umbilical Cord PeriVascular Cell) technology, commented: "As with all cell-based therapies, post-preservation yield and viability are critically important and directly related to clinical efficacy and the overall success of product development, commercial production, and the cost of delivering the therapy to the patient. Our use of CryoStor was driven by a need for a high performance, serum free, fully defined, GMP compliant preservation media. CryoStor uses only USP or highest-available grade components. All of these safety and quality factors led to our decision to adopt this new enabling technology."

BioLife Chairman and CEO Mike Rice remarked: "We are very pleased to be able to provide a key component that substantially improves the preservation and utility of umbilical cord tissue derived stem cells. As a result, TRT and its partners around the world can now offer parents not only a new source of stem cells but also a more viable means of preserving and storing stem cells for the potential treatment of numerous diseases. TRT's adoption of CryoStor represents further validation of the benefits of CryoStor and proof of growing customer acceptance of all our proprietary preservation media products by the cell therapy market."

About Tissue Regeneration Therapeutics

TRT is a private, Canadian life sciences company that exclusively licensed HUCPVC technology from the University of Toronto. TRT's business model involves licensing HUCPVC technology to national Cord Blood Banks seeking to offer new families a mesenchymal stem cell product in addition to their existing cord blood storage. In June 2006, TRT exclusively licensed rights for the Canadian market to CReATe Cord Blood Bank in Toronto, who market HUCPVCs as Peristem(TM). In June 2007, TRT licensed exclusive USA rights for HUCPVC technology for familial banking to Stem Cell Authority, Akron OH. TRT's preclinical and clinical development program is designed to define patient benefits for auto-immune and inflammatory diseases. For more information please visit http://www.verypowerfulbiology.com.

About BioLife Solutions

BioLife Solutions develops, manufactures and markets patented hypothermic storage and cryopreservation solutions for cells, tissues, and organs. The Company's proprietary HypoThermosol® and CryoStor(TM) platform of solutions are marketed to academic and commercial organizations involved in cell therapy, tissue engineering, cord blood banking, drug discovery, and toxicology testing. BioLife's products are serum-free and protein-free, fully defined, and are formulated to reduce or prevent preservation-induced, delayed-onset cell damage and death. BioLife's enabling technology provides academic and clinical researchers significant improvements in post-thaw cell, tissue, and organ viability and function. For more information please visit http://www.biolifesolutions.com.

This news release contains forward-looking statements as that term is defined in the Private Securities Litigation Reform Act of 1995. These forward-looking statements include any statements that relate to the intent, belief, plans or expectations of the Company or its management, or that are not a statement of historical fact. Any forward-looking statements in this news release are based on current expectations and beliefs and are subject to numerous risks and uncertainties that could cause actual results to differ materially. Some of the specific factors that could cause BioLife Solutions' actual results to differ materially are discussed in the Company's recent filings with the Securities and Exchange Commission. BioLife Solutions disclaims any obligation to update any forward-looking statements as a result of developments occurring after the date of this press release.


Media Relations: Investor Relations:
Len Hall Matt Clawson
Allen & Caron Inc. Allen & Caron Inc.
(949) 474-4300 (949) 474-4300
len@allencaron.com matt@allencaron.com
👍️0
lefty18 lefty18 17 years ago
So I take it you don't agree with Lowman's $30.00 price target on CTUM?
👍️0
Lazarus Lazarus 17 years ago
i have been looking at so many turdy biotechs...


that make PPMD look like a flipping steal at these prices.

here's one i just reviewed:

http://pinksheets.com/edgar/GetFilingHtml?FilingID=5198638

note that with its 35 million+ shares OS the market cap is over 24 million and they have zero revenues.

Lazarus - microcap miner and flea trainer extraordinaire
👍️0
Lazarus Lazarus 17 years ago
Life Sciences Funding: Has the Private Equity Boom Peaked?

7/23/2007


CHICAGO – In April 2007, I wrote about the $29 billion KKR deal with First Data as a springboard for discussion on life sciences innovation, the relative shift of capital from early stage to later-stage deals and pharmaceutical research pipelines in general.

In that column, I wrote:

There is also another price to be paid for an economy that is tilted in favor of debt rather than equity. Roughly speaking, debt is a conservative financing instrument. While not inimical to innovation, it is not the currency that lubricates revolutions.

As catalyzed by low interest rates and cheap debt capital, there have been rumors that the private equity boom has peaked. The lead article in the Economist earlier in July headlined “The Trouble With Private Equity” points exactly to that:

It is also possible that the weather is turning and the debt that powers private equity’s siege engines is starting to become harder to scrape together. It may not happen this month – perhaps not even this year – but sooner or later, the private-equity boom will come to an end.

This phenomenon – the inverse relationship between private equity volume and interest rates – has also been the subject of other financial commentators as well. See “Fallout From End of Low-Interest Rates Likely to Be Widespread in U.S.” in the International Herald Tribune, “The Money Binge” in the New York Times and “The End of Easy Money” in Time magazine.

Though this is just a brief column, I will refer to some quantitative research. I would like to acknowledge my gratitude to Manoj Jain of Pipal Research. He leads that Chicago-based business intelligence and research outsourcing firm.

What Happens When Interest Rates Rise?

The private equity boom has potentially crowded out earlier-stage investments. One could also argue that higher interest rates and a slowing of the private equity train could potentially allow capital to flow toward earlier-stage, riskier projects as investors seek to maximize their return on capital.

Because of the importance of this to the life sciences and to the global economy more broadly, this deserves a closer (albeit simplified) view.

At a high level, inexpensive debt capital (e.g. low interest rates) means that very high returns on capital (e.g. high risk) is not necessary to obtain a relatively good return. Lower-risk investments (e.g. established companies with sustained revenues) are sufficient targets for investment. Likewise, the positive cash flow enables debt-leveraged capital.

As outlined in the KKR and First Data column, this means in real terms that investors are more likely to allocate their capital toward more efficient credit card processing (e.g. First Data) than to a risky, no-revenue, high-potential return biotech or medical tech start-up. There are other structural issues including:

# the increasing size of deals
# the cost of due diligence
# the preferential tax treatment of private equity investments

VC Booms When Interest Rates Increase

Below is a graph of 10-year U.S. Department of Treasury yields (from the U.S. Federal Reserve) from 1962 to 2006. I’ve annotated the data with several comments.

There are several items to note:

1. 1978 (when interest rates were rising to 8.41 percent) was the first big year for venture capital.

2. As interest rates peaked to nearly 14 percent during the late 1970s and early 1980s, venture capital experienced a relative boom. Investors had to take risks in order to get some return on capital during the nefarious stagflation years.

3. As interest rates went to a relatively low level (though still not as low as present times), investors switched to private equity. During the late 1980s, this caused the well-known leveraged buyout (LBO) boom.

4. With the rise of dot-coms, the growth in venture capital was somewhat anomalous. Still, it should be noted that as the Fed began to raise interest in the late 1990s in response to Alan Greenspan’s “irrational exuberance” speech in 1996.

This may have fueled even more riskier, early stage investments as the dot-com boom continued its advance.

5. The low interest rates following the dot-com bust and the Sept. 11, 2001 attacks have sparked the private equity boom. The slight uptick in interest rates (4.80 percent in 2006) should be noted.

During the last private equity boom, statistics bear out the dichotomy between venture capital funding and private equity investment.

From 2005 to 2006, VC funding in the U.S. increased 12 percent from $23.5 billion to $26.4 billion. During the comparable period, private equity investment increased 220 percent from about $130 billion to $415 billion, according to Thomson Financial and Pipal Research.

Implications For Life Sciences

So what are the implications for life sciences? First of all, life sciences VC funding as a proportion of overall VC funding has been markedly increasing. Life sciences (biotech and medical devices together) accounted for 36 percent of total first-quarter 2007 VC dollars.

Medical device investing skyrocketed to an all-time high with $1.08 billion going into 96 deals. This was a 60 percent increase over fourth-quarter 2006 dollars. Biotech was the largest sector with $1.0 billion actually displacing software, which has traditionally been the largest sector, according to the NVCA and Pipal Research.

I believe higher interest rates will serve to stimulate investors to allocate funds toward higher potential return, higher risk investments. This means venture capital will benefit just like during the late 1970s and potentially the latter part of the 1990s.

If we combine the premise that life sciences funding is intrinsically increasing and venture capital will see a relative influx in investment flows, then the future is bright for life sciences funding in the foreseeable future.

_______________________________________________________________
Dr. Ogan Gurel is chairman of the Aesis Research Group, which provides forward-looking information and research services to the health-care and life sciences investment community. Gurel was previously CEO of Duravest, a publicly traded Chicago investment company that initiates and develops next-generation medical technologies. Previous to Duravest, he was a vice president and medical director at Sg2, a health-care intelligence think tank and consultancy serving hospitals and health systems. He can be e-mailed at ogan@midwestbusiness.com.
👍️0
Lazarus Lazarus 17 years ago
is this patent of any significance???

http://www.patentgenius.com/patent/7157273.html

Date Issued: January 2, 2007
Application: 10/311,406
Filed: April 17, 2002

if you do a "find on page" for Nephrigen, one of PPMD's trademarked products, you will find:

.times.10.sup.7 PFU of adenovirus was added to a medium (Nephrigen; Celox (Minnesota, USA)) containing 9.5.times.10.sup.5 of 293 cells and the admixture was allowed to stand for one hour to infect the cells with the virus.

Lazarus

Nephrigen not mentioned here: http://www.protidepharma.com/products/celox.aspx

but it is mentioned in the SEC filings under Celox Labs

👍️0
Lazarus Lazarus 17 years ago
The Future is Bright For Life Sciences in State of Illinois

CHICAGO – In 2006, Chicago was the home of the successful BIO 2006 conference. I wrote this in a follow-up column: “It was quite an event. More than 19,000 attendees – anywhere from Bill Clinton to biotech graduate students – teemed throughout the cavernous halls of McCormick Place.”

The strong support of many of the large Midwestern drug and device companies was a factor in the success of this conference. Very important, of course, was Chicago Mayor Richard Daley’s personal encouragement (with both his enthusiasm as well as dollars) along with a push from Illinois Department of Commerce & Economic Opportunity (DCEO) director Jack Lavin.

The conference also had the goal of sparking a number of initiatives to put Chicago and the Midwest on the map with respect to the growing life sciences industry.

With Abbott Laboratories, Baxter, Takeda and many other major companies, Chicago is no lightweight. Still, there has been the impression that many promising ideas would leave the area for Boston, San Francisco or San Diego when it has come to start-ups and other engines of innovation and growth.

With a fabulous infrastructure, vibrant economic growth, top-notch professional services, a strong financial community and one of the world’s leading centers of life sciences university research, there are all the reasons for start-up and development-stage companies to stay or even come to the Chicago area.

Both city and state leaders recognize that the life sciences sector – with an aging population, innumerable unmet needs in medicine and increasing globalization of pharmaceuticals and medical technology – represents a strong part of commerce for the future. That means lots of meaningful and high-paying jobs.

The 2006 column also focused on one particular aspect of BIO 2006: the rising importance of convergent or combination medical technologies.

By definition, these are technologies that straddle both the device and drug worlds and also incorporate aspects of IT and nanotechnology. In the diagram below, areas of overlap represent potential convergent medical technology applications. The “device” sector is indicated here as “surgical tech”:

While the paradigm for convergent medical technologies (CMT) has been the drug-eluting stent (DES), other areas have also seen recent applications such as implantable insulin delivery pumps and programmable intracardiac defibrillators. However, it is not so easy to develop CMT as there are significant cultural and regulatory differences between the biopharma and device sectors.

So what does this have to do with the Midwest? While there have been a number of significant initiatives pushing biotech forward, the 2006 column touched on how Chicago is especially well poised to help integrate and cross this cultural divide.

Not only are we seeing a renaissance of life sciences in the Chicago area, but more specifically, I would predict that interdisciplinary areas such as CMT (including nanotech and smart devices) will especially find Chicago a congenial area to move forward.

Just one example of the many initiatives that have been spawned from BIO 2006 is the recently unveiled iBIO PROPEL project. iBIO is the Illinois Biotechnology Industry Organization. Its mission is to strengthen the leadership position of Illinois as a globally recognized life sciences center.

The PROPEL project is an entrepreneurship coaching program designed to facilitate and accelerate the development of management at life sciences start-ups in Illinois. I attended the PROPEL kickoff event last Wednesday, which was written up in the Chicago Tribune.

The day was notable not only because of the importance of the program but also the fact that the kickoff itself brought a remarkable assembly of nearly all the top industry, academic and government leaders together in one room.

While it wasn’t as raucous as the seventh-inning stretch at Wrigley Field, you could sense the excitement and enthusiasm in the air. Ultimately it is people and the commitment of people that will be critical to moving PROPEL and innumerable other such initiatives forward.

I had the chance to speak with iBIO President David Miller after the event. We both agreed that the outlook for life sciences in Illinois is truly promising.

“Prospects are strong for the entire state because of the range of applications under development [in Illinois],” Miller said. “What’s new – and the reason I’m so confident – is that we have engineered a phenomenal level of cooperation among the three primary sectors: public, private and education/research.”

Kudos to Miller and to Mayor Daley, Jack Lavin and many others who have helped to bring this spirit of collaboration to reality.

That speaks directly to why Chicago and Illinois are perfectly poised to be the world leader in next-generation convergent medical technologies as well as a major player in life sciences. Progress in our increasingly interconnected technologies can only come through collaboration. We are seeing that in spades in Chicago.

The next mega BIO conference (dare we say “Biopalooza”?) is scheduled to return to Chicago as BIO 2010. That fact alone speaks for itself. It’ll be interesting to see how Chicago and Illinois fit into the growing life sciences landscape at that time.



--------------------------------------------------------------------------------

Dr. Ogan Gurel is chairman of the Aesis Group, which provides consulting services to companies and investment firms in the life sciences and health care sectors. He is also chief medical officer of BlueBob Analytics and an adjunct associate professor of bioengineering at the University of Illinois at Chicago. Gurel was previously CEO of Duravest, which is a publicly traded Chicago investment firm that develops next-generation medical technologies. He was previously a vice president and medical director at Sg2. Gurel can be e-mailed at ogan@midwestbusiness.com.

👍️0
Lazarus Lazarus 17 years ago
WASHINGTON (AP) - Pushing back against the Democratic-led Congress, President Bush intends to veto a bill Wednesday that would have eased restraints on federally funded embryonic stem cell research—work that supporters say holds promise for fighting disease.
At the same time, Bush will issue an executive order directing the Health and Human Services Department to promote research into cells that, like human embryonic stem cells, also hold the potential of regenerating into different types of cells that could help treat illness.

White House spokesman Tony Fratto said Tuesday that Bush would outline an initiative that could make federal funding available for research on additional "pluripotent" stem cells—ones that can give rise to any kind of cell in the body except those required to develop a fetus.

The president has accused majority Democrats of recycling an old measure that he already vetoed and argued that the bill would mean American taxpayers would—for the first time—be compelled to support the deliberate destruction of human embryos.

"The president supports and encourages stem cell research—including using embryonic lines—as long as it does not involve creating, harming or destroying embryos," Fratto said. "That is an ethical line that should not be crossed."

Democrats made the legislation a top priority when they took control of the House and Senate in January, but they don't have enough votes to override Bush's decision.

Senate Majority Leader Harry Reid appealed to Bush on Tuesday not to veto the bill. He said the measure acknowledges the ethical issues at stake and offers even stronger research guidelines than exist under the president's current policy.

House Speaker Nancy Pelosi used Bush's veto threat as a reason to send out an e-mail letter soliciting contributions to the Democratic Congressional Campaign Committee to help elect more Democrats.

"By vetoing a bill that expands stem cell research, the president will say `no' to the more than 70 percent of Americans who support it, `no' to our Democratic Congress' fight for progress, and `no' to saving lives and to potential cures for diseases such as diabetes and Parkinson's," Pelosi wrote. "He will say `no' to hope."

In light of the veto, Sen. Norm Coleman, R-Minn., who planned to be at the White House event, sought support for a stem cell bill he is sponsoring. It has passed the Senate but has not yet been taken up by the House.

"My stem cell bill, which passed the Senate with broad bipartisan support, offers a clear alternative for our colleagues in the House to significantly expand federally funded stem cell research, while ensuring no taxpayer dollars are used for the destruction of human embryos," Coleman said.

Coleman urged Democrats who favored the bill Bush was to veto to get behind his legislation.

"Those who support the stem cell research bill ... are at a definitive crossroads," he said. "Do they seek to advance lifesaving research for millions of Americans suffering from serious disease or do they, in fact, prefer to keep stem cell research at a political stalemate? "

This will be the third veto of Bush's presidency. His first occurred last year when he rejected legislation to allow funding of additional lines of embryonic stem cells—a measure that passed over the objections of Republicans then in control. Earlier this year, he vetoed legislation that would have set timetables for U.S. troop withdrawals from Iraq.

Opponents of the latest stem cell measure insisted that the use of embryonic stem cells was the wrong approach on moral grounds—and possibly not even the most promising one scientifically. They cite breakthroughs involving medical research conducted with adult stem cells, umbilical cord blood and amniotic fluid, none of which involve the destruction of a human embryo.

The science aside, the issue has weighty political implications.

Public opinion polls show strong support for the research, and it could return as an issue in the 2008 elections.

Democratic presidential candidate Hillary Rodham Clinton appeared in Hanover, N.H., this week with a child who has diabetes and a paralyzed 23-year-old to urge Bush not to veto the bill. Last month, the issue was a topic at a debate with Republican presidential hopefuls in California.

The bill Bush is vetoing passed Congress on June 7, drawing the support of 210 House Democrats and 37 Republicans. That was 35 votes fewer than needed to override a veto. The Senate cleared the bill earlier by a margin that was one vote shy of the two-thirds needed to overcome Bush's objections.

According to the National Institutes of Health Web site, scientists were first able to conduct research with embryonic stem cells in 1998. There were no federal funds for the work until Bush announced on Aug. 9, 2001, that his administration would make the funds available for lines of cells that already were in existence.




👍️0
Lazarus Lazarus 17 years ago
article on biotech:


http://www.midwestbusiness.com/news/viewnews.asp?newsletterID=17320
👍️0
Lazarus Lazarus 17 years ago
some stocks are worth owning just for the intrigue...


i tend to be drawn to [and often rewarded] to stocks like PPMD that have no following and management that not only doesnt hype the stock --- but rather even seem to be deliberately suppressing the stock price.


Lazarus - the ever-speculating
👍️0
Lazarus Lazarus 17 years ago
from the last Q:

The Company continues to sell its products on a direct basis to customers in the United States as well as to seventeen different countries around the world. In addition the Company has formed the following distribution avenues:

The Company has a nonexclusive worldwide distribution agreement with MP Biomedicals, Inc., Costa Mesa, CA. Under the agreement, MP Biomedicals is marketing Celox’ TCMTM, TCH™, TM-235™ serum replacement products as well as Cellvation™. MP Biomedicals manufactures and markets medical and biotechnology products worldwide.

In 1997, the Company began providing its proprietary products to Sigma Chemical Company (NASDAQ:SIAL), St. Louis, MO. under a private label distribution agreement for worldwide distribution.

The Company’s Celox product line is distributed in Japan through Funakoshi Co., LTD, a well-established Japanese distributor.

The Company also has distribution of its STEMSOLTM and DMSO/Dextran products in Europe and the Pacific Rim through various non-exclusive agreements with local distributors.

👍️0
Lebaneseproud Lebaneseproud 17 years ago
Well someone is still enjoying STEMSOL as of Nov. 2006

https://secure-celltherapy.malachite-mgmt.com/imisl/source/Communities/viewDiscussionTopic.cfm?section=&CmtyId=2&FrmId=2&TopicId=29
👍️0
Lebaneseproud Lebaneseproud 17 years ago
Its comforting to know our public companies product is still being distributed through MPBioproducts

http://www.mpbio.com/advanced_search_result.php?depth=nested&keywords=tcm&submit=Search
👍️0
Lebaneseproud Lebaneseproud 17 years ago
Im not interested in any contest either.
I thought you were insinuating i was the seller when you replied to one of my earlier posts of mine saying i was adding. I'll be here with you when this one pops. This one intrigues me very much
👍️0
Lebaneseproud Lebaneseproud 17 years ago
You are correct about THMG as well. Have owned it for quite some time and have no desire to sell.
👍️0

Your Recent History

Delayed Upgrade Clock