rexresearch.com
GcMAF
[ Vitamin D binding protein
macrophage activating factor ]
Wikipedia
:Gc-MAF
YouTube : GcMAF: THE AMAZING ANSWER FOR YOUR
HEALTH
Drbradstreet.org : Dr Jeff Bradstreet
Michael Miller : Anti-vaccine doctor behind
‘dangerous’ autism therapy found dead. Family cries foul
YouTube : The real reason Holistic Doctors
are being killed and vanishing!
Richard Presser : Is this the real reason
Holistic Doctors are being killed and vanishing?
Informahealthcare.com : Vitamin D
binding protein
Kat Arney : “Cancer cured for good?” – Gc-MAF
and the miracle cure
RetractionWatch : Yet another study of
widely touted cancer “cure” retracted
Gcmaf.eu / YouTube : Cancer cells destroyed
by First Immune GcMAF
anticancerfund.org : Warning: GcMAF
has not been properly studied in clinical trials...
David M. Mosser : The many faces of macrophage
activation
Nobuto Yamamoto, et al. : Retracted:
Immunotherapy of HIV-infected patients with Gc protein-derived
macrophage activating factor (GcMAF)
Nobuto Yamamoto : Retracted: Immunotherapy
of metastatic breast cancer patients with vitamin D-binding
protein-derived macrophage activating factor
Efranat : Clinical Trials
Nobuto Yamamoto : Bio
Patents & Applications
Pharmocracy in
action :
https://en.wikipedia.org/wiki/Gc-MAF
Gc-MAF
Gc-MAF or Gc protein-derived macrophage activating factor is an
immunomodulatory protein that, by affecting the immune system, may
play a role in various diseases.[1]
In 2008 claims were made that Gc-MAF can provide a permanent cure
for cancer and HIV. These claims have been the subject of much
criticism[2] and are not supported by scientific evidence. The
papers supporting the claims have since been retracted by the
journals in which they were published.[3] Consumers have been
warned about illegal marketing of the substance over the
internet.[4]
Description
Biochemically, Gc-MAF results from sequential deglycosylation of
the vitamin D-binding protein (the Gc protein), which is naturally
promoted by lymphocytes (B and T cells).[1] The resulting protein
may be a macrophage activating factor (MAF).[1] MAFs are
lymphokines that control the expression of antigens on the surface
of macrophages, and one of their functions is to make macrophages
become cytotoxic to tumors.[5] Gc-MAF may play a role in various
diseases.[1]
Therapeutic claims
GcMAF has not been properly studied in clinical trials and its
laboratory results still need to be confirmed independently. So
far, all claims on the efficacy of this product have no solid
scientific basis. Its marketing is illegal; therefore there is no
controlled guarantee on the quality of the product for human
consumption sold over the internet.
Public warning issued by the Anticancer Fund[4]
Gc-MAF has been promoted as a cure for some cancers,[2] HIV[6] and
other diseases. The integrity of the research, conducted by Nobuto
Yamamoto and colleagues, that originally prompted claims regarding
cancer and HIV has been questioned.[2][4] Cancer Research UK has
warned the public about spurious claims of clinical benefits,
misleadingly based on reduced levels of the
alpha-N-acetylgalactosaminidase enzyme (also known as nagalase),
whose production might be increased in many cancers.[2] Nagalase
is an enzyme present in normal cells and its use to diagnose or
follow-up the diseases claimed to be cured by GcMAF has not been
validated. Nagalase deficiency, however, is associated to a rare
congenital metabolic disorder called Schindler/Kanzaki disease.
Three out of four of the original studies authored by Yamamoto
(published between 2007 and 2009) were retracted by the scientific
journals in which they were published in 2014, officially due to
irregularities in the way ethical approval was
granted.[7][8][6][9]
The Belgian Anticancer Fund has communicated serious concerns to
other journals that published studies on Gc-MAF by Yamamoto and
colleagues.[4] They have also warned consumers of illegal
marketing of Gc-MAF, over the internet and elsewhere.[4][10]
As of May 2014 there was one Phase I clinical trial registered to
evaluate Gc-MAF. This trial only aims to evaluate the safety of
this product for human consumption, efficacy is not yet being
studied. The product used in this trial is not available out of
the study and the companies commercializing Gc-MAF over the
internet are not involved. No results are yet available.[11]
1) Malik, Suneil; Fu, Lei; Juras, David James; Karmali, Mohamed;
Wong, Betty Y. L.; Gozdzik, Agnes; Cole, David E. C.
(January–February 2013). "Common variants of the vitamin D binding
protein gene and adverse health outcomes". Critical Reviews in
Clinical Laboratory Sciences 50 (1): 1–22.
http://informahealthcare.com/doi/full/10.3109/10408363.2012.750262
doi:10.3109/10408363.2012.750262. PMC 3613945. PMID 23427793.
2) Arney, Kat (3 December 2008). "'Cancer cured for good?' –
Gc-MAF and the miracle cure (revised 25 July 2014)". Cancer
Research UK.
http://informahealthcare.com/doi/full/10.3109/10408363.2012.750262
3) "Tracking retractions as a window into the scientific process
Yet another study of widely touted cancer “cure” retracted".
Retraction Watch.
http://retractionwatch.com/2014/10/10/yet-another-study-of-widely-touted-cancer-cure-retracted/
4) "GCMAF". Anticancer Fund. 24 July 2014.
http://www.anticancerfund.org/therapies/gcmaf
5) Mosser, David M. (February 2003). "The many faces of macrophage
activation". Journal of Leukocyte Biology 73 (2): 209–212.
https://en.wikipedia.org/wiki/Journal_of_Leukocyte_Biology
doi:10.1189/jlb.0602325. PMID 12554797.
6) Yamamoto, Nobuto; Ushijima, Naofumi; Koga, Yoshihiko (January
2009). " (Retracted): Immunotherapy of HIV-infected patients with
Gc protein-derived macrophage activating factor (GcMAF)". Journal
of Medical Virology 81 (1): 16–26.
https://en.wikipedia.org/wiki/Journal_of_Medical_Virology
doi:10.1002/jmv.21376. PMID 19031451
7) Yamamoto, Nobuto; Suyama, Hirofumi; Yamamoto, Nobuyuki;
Ushijima, Naofumi (15 January 2008). " (Retracted): Immunotherapy
of metastatic breast cancer patients with vitamin D-binding
protein-derived macrophage activating factor (GcMAF)".
International Journal of Cancer 122 (2): 461–467.
doi:10.1002/ijc.23107.
8) ) Yamamoto, N.; Suyama, H.; Nakazato, H.; Yamamoto, N.; Koga,
Y. (2014). "Retraction Note to: Immunotherapy of metastatic
colorectal cancer with vitamin D-binding protein-derived
macrophage-activating factor, GcMAF". Cancer Immunology,
Immunotherapy 63 (12): 1349. doi:10.1007/s00262-014-1616-x.
"Retraction". International Journal of Cancer 135 (6): 1509. 15
September 2014. doi:10.1002/ijc.29014.
9) "Retraction". International Journal of Cancer 135 (6):
1509. 15 September 2014.
doi:10.1002/ijc.29014
10) Emmanuel Borloz (19 June 2015). "Clinique privée sous enquête
pénale après cinq morts Enquête" (in French). 24 Heures.
http://www.24heures.ch/vaud-regions/lausanne-region/clinique-privee-vaudoise-enquete-penale-cinq-morts/story/12349881
11) "Safety Study of GcMAF (Globulin Component Macrophage
Activating Factor) in Subjects With Advanced Solid Tumors". U.S.
National Institutes of Health.
https://clinicaltrials.gov/ct2/show/NCT02052492?term=gcmaf&rank=1
https://www.youtube.com/watch?v=KqMohmjJ4mg
GcMAF: THE AMAZING ANSWER FOR YOUR HEALTH
Dr Jeff Bradstreet, MD, MD(H), FAAFP
Hope for complex health issues: (678) 288-9222 (NEW NUMBER)
Accepting New Patients
http://www.washingtonpost.com/news/morning-mix/wp/2015/06/29/anti-vaccine-doctor-behind-dangerous-autism-therapy-found-dead-family-cries-foul/
Anti-vaccine doctor behind ‘dangerous’
autism therapy found dead. Family cries foul
by
Michael E. Miller
https://www.youtube.com/watch?v=cALgIHETMDU&feature=youtu.be
Explosive: The real reason Holistic
Doctors are being killed and vanishing!
http://www.richardpresser.com/wordpress/is-this-the-real-reason-holistic-doctors-are-being-killed-and-vanishing/
July 24, 2015
Is this the real reason Holistic
Doctors are being killed and vanishing?
by
Richard Presser
Firstly, let me say that I do not have the knowledge to assess
this material from a scientific perspective, but it rings a lot of
bells for me, and there are some who read my posts who will know.
It is clear that several holistic doctors have died recently, and
another was announced today. Is there a link between some or all
of them?
It’s difficult to be certain, but when the breadcrumbs begin to
line up, a trail begins to emerge.
So let me cut to the chase of what this article says:
Human GcMAF, otherwise known as Vitamin D binding protein
macrophage activating factor, holds great promise in the treatment
of various illnesses including cancer, autism, chronic fatigue and
possibly Parkinson’s. Since 1990, 59 research papers have been
published on GcMAF, 20 of these pertaining to the treatment of
cancer. 46 of these papers can be accessed through the GcMAF web
site.
GcMAF is a vital part of our immune system which does not work
without it; and is part of our blood. GcMAF stimulates the
macrophage element of the immune system to destroy cancer cells.
It also blocks the supply of nutrients to cancer cells by stopping
blood vessel development to the site (anti-angiogenesis). Cancer
cells are weakened and starved, making them more vulnerable to
attack by the GcMAF stimulated macrophage system. Research has
shown macrophage activation and stopping diseased blood vessel
development can also help in various neurological diseases such as
Parkinson’s, Alzheimer’s, rheumatoid arthritis, inflammatory
conditions, and diabetic retinopathy…
…In the past months Dr. Bradstreet has become interested in
nagalese (also spelled nagalase in this document), which he
describes as an enzyme “produced by cancer cells and viruses.” He
thinks it unlikely that children with autism have undiagnosed
cancers, and thus suspicion falls on a viral etiology. Dr.
Bradstreet writes, “Viruses make the nagalese enzyme as part of
their attachment proteins. It serves to get the virus into the
cell and also decreases the body’s immune reaction to the
virus-thereby increasing the odds of viral survival.”
Further on Dr. Bradstreet writes, “It is reasonable and likely
that the nature of the immune dysfunction and the frequently
observed autoimmune problems in autism are mediated by persistent,
unresolved viral infections.” He claims to have tested
approximately 400 children with autism for the viral marker,
nagalese, and found that nearly 80% have significantly elevated
levels. He hopes to publish soon on this study and believes this
information “is one of the most important developments in the
clinical treatment of children on the spectrum that I have
experienced in the last 15 years.”…
…Nonetheless, his son’s case helped convince Bradstreet that
vaccines caused autism. He took his message to the highest levels
of government. Twice he testified about the supposed link between
vaccines and autism before the U.S. House of Representatives.
“He was a very happy, well connected child prior to his MMR at
approximately 12 months of age,” Bradstreet told representatives
in 2002, presenting copies of his son’s various tests. “Matthew
completely lost about 2 months after his MMR vaccine.”
From his clinic in Buford, Ga., Bradstreet treated patients from
around the world, many who sought him out online. Desperate
parents seeking answers for their children’s maladies would write
to him on his blog, begging him for help…
…In the case of autism, Dr. James Bradstreet has so far treated
1,100 patients with GcMAF with an 85% response rate. His results
show a bell curve response with 15% of the patients showing total
eradication of symptoms and 15% showing no response.
In addition, experimental and clinical evidence confirms that
GcMAF shows multiple powerful anti-cancer effects that have
significant therapeutical impact on most tumors including breast,
prostate, and kidney. GcMAF is created in the body by the release
of two sugar molecules from a GcProtein molecule…
…In conclusion, GcMAF restores the energetic balance in the cell.
Cancer cells driven by sugar metabolism become healthy oxygen
driven cells, so tumor cells no longer behave as parasitic
organisms. GcMAF stimulates macrophages to consume the cancer
cells and cells invaded by viruses. This stimulation of the immune
system and the anti-angiogenetic effect surrounding the tumor is
beneficial in cancer and several neurological disorders like
autism, chronic fatigue, Parkinson’s, and Alzheimer’s, and it is
available to the general public… (not any more – the company has
been shut down)
…After discussing her options the patient wanted to try GcMAF
therapy prior to considering more radiation or chemotherapy. After
6 weeks of GcMAF 100ng/week subcutaneous injections (much like a
shot of insulin) her repeat nagalase test returned at 2.10 (a 50%
reduction). All of her other tumor markers remain negative and she
is taking the dose of Vitamin D3 required to optimize her blood
levels (9000 iu/day). It is too soon for her PET to be repeated
but we will follow this soon to determine the course of the bone
metastasis. The nagalase test may be a more sensitive marker for
tumor burden than other more accepted blood tests. GcMAF given via
simple patient administered once weekly injections is clearly able
to reduce the nagalase level dramatically over a short period of
time. In previous published studies, nagalase response to GcMAF
was correlated with reduction and eventual elimination of cancer.
This is an encouragement to us all and I will keep you posted on
the patient’s progress.”…
…With compromised immune activation, increased nagalase cuts off
the conversion to GcMAF – result is a deglycosylated Gc protein
that cannot activate macrophages.
If you have increased nagalase, you have less GcMAF and your Gc
protein is not effectively transferred into GcMAF.
Nagalase is part of the gp120 enzyme in HIV. HERV’s or other
viruses active in cells may produce nagalase.
Several intestinal bacteria are producers of nagalase. Editor’s
Note: I found this connection to be quite interesting; the gut is
big.
Similar to HIV, CFS patients have many infections and reactivate
endogenous herpes viruses – EBV, CMV, HHV-6, HSV-1, as well as
Herpes 7.
Healthy controls have very low nagalase enzyme activity. Normal
people do have some, but it should be very low. There is a clear
difference in those with pathology…
What is Nagalase?
Nagalase is a protein made by all cancer cells and viruses (HIV,
hepatitis B, hepatitis C, influenza, herpes, Epstein-Barr virus,
and others). Its formal, official chemical name is
alpha-N-acetylgalactosaminidase, but this is such a
tongue-twisting mouthful of a moniker that we usually just call it
“Nagalase.” (Sometimes, when I want to impress friends with my
brilliance, I’ll say the entire word real fast:
“alpha-N-acetylgalactosaminidase.” I have found that it’s
important to practice beforehand if one doesn’t want to embarrass
oneself.)
Why is Nagalase important?
Nagalase causes immunodeficiency. Nagalase blocks production of
GcMAF, thus preventing the immune system from doing its job.
Without an active immune system, cancer and viral infections can
grow unchecked.
As an extremely sensitive marker for all cancers, Nagalase
provides a powerful system for early detection.
Serial Nagalase testing provides a reliable and accurate method
for tracking the results of any therapeutic regimen for cancer,
AIDS, or other chronic viral infection.
Nagalase proves that cancer cells break all the rules
Normal healthy cells cooperate with one another in a concerted
effort to further the good of all. Cancer cells refuse to play
ball. Their disdainful attitude toward the rest of our cellular
community is appalling. For example, these cellular scofflaws
ignore clear messages to stop growing and spreading and
encroaching on their neighbor’s space. How would you like it if
your neighbor moved his fence over into your backyard?
Of all the rules cancer cells break, none is more alarming than
the production of Nagalase, the evil enzyme that completely
hog-ties the immune system army’s ability to stop cancer cells.
Virus particles also make Nagalase. Their goal is the same as that
of the cancer cells: survival by incapacitating their number one
enemy: the immune system.
Nagalase precision
Like a stealth bomber, the Nagalase enzyme synthesized in and
released from a cancer cell or a virus particle pinpoints the
GcMAF production facilities on the surface of your T and B
lymphocytes and then wipes them out with an incredibly precise
bomb. How precise? Let me put it this way: Nagalase locates and
attacks one specific two-electron bond located at, and only at,
the 420th amino acid position on a huge protein molecule (DBP),
one of tens of thousands of proteins, each containing millions of
electrons. This is like selectively taking out a park bench in a
major city from six thousand miles away. More astonishing, if that
is possible, Nagalase never misses its target. There is no
collateral damage.
As you already know, GcMAF is a cell-signaling glycoprotein that
talks to macrophages, enabling them to rapidly find, attack, and
kill viruses and cancer cells. By activating macrophages, GcMAF
triggers a cascade that activates the entire immune system.
Blockage of GcMAF production by Nagalase brings all this wonderful
anti-cancer and anti-viral immune activity to a screeching halt,
allowing cancer and infections to spread.
What does Nagalase actually do? How does it destroy immune
functioning and deactivate macrophages?
Once synthesized and released into nearby tissue or into the
bloodstream, Nagalase, like that drill sergeant at boot camp,
shouts harsh commands at the vitamin D binding protein (DBP) that
is about to be turned into GcMAF. Nagalase demands that DBP not,
under any circumstances, attach itself to a specific sugar
molecule (galactosamine). If DBP has already grabbed (i.e.,
connected to, using a two-electron, “covalent” bond) a
galactosamine sugar molecule, it is commanded to immediately let
go. “Leave galactosamine alone, or you’ll be in big trouble!!!” is
the Nagalase sergeant’s command. We’ll probably never know whether
or not, on some deeper level, DBP knows that Nagalase’s motives
are dastardly—but it doesn’t really matter: DBP will definitely
always obey. Like the army private, the DBP literally has no
choice. Because of the way hierarchies work in cellular biology,
proteins must do the bidding of their enzymes. The enzymes, like
Nagalase, are the drill sergeant and the proteins, like DBP, are
the privates. That’s just the way it is. Obeying the drill
sergeant’s command means DBP can’t do its assigned task, that of
becoming GcMAF. It is rendered useless. For DBP, on a molecular
level, life no longer has meaning…
End of quotes. These quotes are not in the same order in the
document they are taken from.
So where does nagalase come from in infants?
Although this article does not spell it out, this video claims
that nagalase is INTENTIONALLY included in vaccines. Perhaps this
is one reason for the vaccine/autism correlation. It also fits
with the broader array of health issues flowing from vaccination.
There are many links to follow here for those interested; however,
it appears most if not all of the doctors who have recently “died”
were connected to treatment practices utilising GcMAF to defeat
the effects of nagalase.
I would appreciate feedback from those qualified to assess this.
To me, it has all the hallmarks of a bombshell.
https://www.youtube.com/watch?v=0v3IA2Hj1TA&feature=youtu.be
Dr.Bradstreet Search Warrant: the promising drug GcMAF was their
target!!
http://scienceblogs.com/insolence/files/2015/07/BradstreetSearchWarrant.pdf
http://informahealthcare.com/doi/full/10.3109/10408363.2012.750262
ABSTRACT
The vitamin D binding protein (DBP) is the major plasma carrier
for vitamin D and its metabolites, but it is also an actin
scavenger, and is the precursor to the immunomodulatory protein,
Gc-MAF. Two missense variants of the DBP gene – rs7041 encoding
Asp432Glu and rs4588 encoding Thr436Lys – change the amino acid
sequence and alter the protein function. They are common enough to
generate population-wide constitutive differences in vitamin D
status, based on assay of the serum metabolite, 25-hydroxyvitamin
D (25OHD). Whether these variants also influence the role of
vitamin D in an immunologic milieu is not known. However, the
issue is relevant, given the immunomodulatory effects of DBP and
the role of protracted innate immune-related inflammation in
response to tissue injury or repeated infection. Indeed, DBP and
vitamin D may jointly or independently contribute to a variety of
adverse health outcomes unrelated to classical notions of their
function in bone and mineral metabolism. This review summarizes
the reports to date of associations between DBP variants, and
various chronic and infectious diseases. The available information
leads us to conclude that DBP variants are a significant and
common genetic factor in some common disorders, and therefore, are
worthy of closer attention. In view of the heightened interest in
vitamin D as a public health target, well-designed studies that
look simultaneously at vitamin D and its carrier in relation to
genotypes and adverse health outcome should be encouraged.
http://scienceblog.cancerresearchuk.org/2008/12/03/cancer-cured-for-good-gc-maf-and-the-miracle-cure/
December 3, 2008
“Cancer cured for good?” – Gc-MAF and the miracle cure
by
Kat Arney
Note: This post has been updated as several research papers about
Gc-MAF have been retracted. We will continue to update this post
as more information becomes available. Last update 09/10/14
As an organisation dedicated to beating cancer, we have a
deep-rooted interest in any new research developments that could
lead to new, more effective treatments for the disease.
So when we received an enquiry from a supporter about an article
entitled “Cancer cured for good” by Bill Sardi and Timothy
Hubbell* we were intrigued. The article talks about research by
Nobuto Yamamoto in the US, looking at a protein called Gc-MAF (aka
GcMAF). His published studies appear to show that injections of
very small amounts of Gc-MAF can “cure” people with breast, bowel
and prostate cancer.
According to the article, “It works 100% of the time to eradicate
cancer completely, and cancer does not recur even years later.”
Could this be the so-called ‘cure for cancer’ that we’ve been
searching for all these years?
Sadly – as with so many things in life – if it sounds too good to
be true it probably is. Let’s explore a bit further.
What’s the idea behind it?
Dr Yamamoto studies the immune system – the highly complex network
of cells that helps to keep us healthy. The cells of the immune
system – white blood cells – fight bacterial and viral infections
because they can recognise and attack these ‘foreign’ invaders .
But they’re not so good at tackling cancer, since tumours grow
from our own cells and have clever mechanisms to ’cloak’ them from
immune attack.
Macrophages (meaning “big eaters” in Greek) are an important type
of white blood cell. They patrol the body, eating up foreign
invaders and dead cells. They also help to alert other immune
cells to the presence of infections.
Macrophages can be stirred into action by a small sugar-coated
protein (glycoprotein) called Gc-MAF, short for Gc Macrophage
Activating Factor, which is produced by the body. But the
production of Gc-MAF is blocked by an enzyme called Nagalase
(alpha-N-acetylgalactosaminidase), produced by many cancers. This
is one of the mechanisms that helps tumours evade the immune
system.
Yamamoto’s theory is that injecting cancer patients with Gc-MAF
should activate their macrophages to fight the cancer. He tested
it back in 1997 in a paper published in the journal Cancer
Research, showing that injecting Gc-MAF into mice transplanted
with cancer cells could improve their survival from around 16 days
to around 35.
But the treatment did not ‘cure’ the cancer, as the cancer cells
continued to multiply, eventually killing the mice.
Clinical trials
Fast-forward a few years, to the publication of three papers
detailing the results of clinical trials of Gc-MAF carried out by
Yamamoto, testing the treatment on patients with breast, bowel and
prostate cancer.
Note: The breast cancer paper (Yamamoto et al Int J Cancer 2008)
has now been retracted, due to various concerns with the work.
Read more on the RetractionWatch blog. [Updated KA 25/07/14] The
bowel cancer paper (Yamamoto et al Cancer Immunology Immunotherapy
2008) has also now been retracted. This letter details some of the
concerns about the work. [Updated KA 09/10/14]
The results appear to be startling – all the patients on the
trials are ‘cured’ of cancer. Surely this is an amazing
breakthrough?
Put bluntly, no it isn’t. There are significant scientific
problems with the trials. For a start, all the studies are very
small, involving fewer than twenty patients in each – rather than
the thousands needed to make the sort of claims mentioned above.
Next, all the patients involved had received standard treatment
for their cancer, including surgery, chemotherapy and/or
radiotherapy. This is a somewhat unorthodox design for a trial of
this kind, because it makes it very difficult to tell if any
successes are due to the new drug, or the more conventional
treatments.
On top of this, the researchers didn’t actually monitor the
progress of tumours in the patients, and provide no clinical
information about them. Instead they opt to measure levels of
Nagalase in the blood, rather than looking at any standard
established markers for cancer.
For example, in the case of the breast cancer patients, there is
no detail about their “TNM” (tumour, node, metastasis) status.
This is a standard measure of how far a patient’s cancer has
spread, and is used to calculate the likelihood that it will
return.
Furthermore, the researchers didn’t do any tests to show that
injected Gc-MAF was actually activating macrophages in the
patients’ blood, or even working in the way that they expect.
There is no information about levels of cytokines – the proteins
produced by immune cells when they are activated – or analysis of
the patients’ immune cells.
Perhaps most significantly, there are no controls – untreated
patients for comparison – and the studies only followed the
patients for a few years. We have no way of telling whether their
cancers were growing again, or had been successfully treated, and
whether this was due to Gc-MAF or the other treatment they had
received.
Given that 80 per cent of all women with breast cancer survive for
at least 5 years, an uncontrolled study showing that 16 women of
unknown TNM status survive for at least 4 years is no great
shakes, scientifically speaking.
Further problems
Another telling point is the type of journal in which the research
was published. If this research was truly groundbreaking, and
pointed the way to a cure for cancer, then the research would
likely be found in top-tier ’high-impact’ medical journals
journals like The Lancet, The New England Journal of Medicine or
the Journal of the American Medical Association.
And finally, virtually all the references in the papers are to
other papers published by Yamamoto and his team. If Gc-MAF was
indeed a promising candidate for a successful cancer treatment,
you’d expect plenty of other research to show the same thing.
Scientists are usually quick to spot promising, emerging fields of
research and jump on the bandwagon.
The poor quality of scientific papers supporting GcMAF is
discussed here on the Scholarly Open Access blog. [Updated KA
25/07/14]
Is there hope?
Although this particular approach isn’t all it’s hyped up to be,
harnessing the power of immune system could be a very potent way
to treat cancer. We’ve blogged many times already on high-quality
research into immunotherapy (for example here, here, here and
here)
And many Cancer Research UK-funded scientists are also working in
this field. For example, Professor Fran Balkwill and her team are
working on ways to trick macrophages and other immune cells into
attacking cancer cells.
In 2014, researchers in Israel started a small-scale early-stage
clinical trial looking at the dosage and safety of GcMAF in cancer
patients. The full protocol and further information are available
on the Clinicaltrials.gov register. [Updated KA 25/07/14]
To sum up
The advent of the internet has led to a wild proliferation of
stories of ‘miracle cures’ for cancer – virtually all of which are
based on shaky (or zero) science.
Some companies are selling Gc-MAF for use by cancer patients. This
treatment is not approved or licensed in the UK for treating
cancer or any 0ther disease. Given that there is no solid
scientific evidence to show that the treatment is safe or
effective, we would not recommend that cancer patients use it.
[Updated KA 25/07/14]
Cancer is an extremely complex disease. In fact, it is more than
200 distinct diseases, each requiring different treatment. And the
success of treatment depends on many things, including the genetic
make-up of the tumour, the stage of diagnosis, and how aggressive
the cancer is.
To suggest that there is a ‘magic bullet’ that cures all cancers
is simplistic in the extreme.
http://retractionwatch.com/2014/10/10/yet-another-study-of-widely-touted-cancer-cure-retracted/
Retraction Watch
Yet another study of widely touted
cancer “cure” retracted
A third study of GcMAF, a protein being used to treat a variety of
conditions from AIDS to autism to cancer, all without the blessing
of health agencies, has been retracted.
Here’s the notice in Cancer Immunology, Immunotherapy for
“Immunotherapy of metastatic colorectal cancer with vitamin
D-binding protein-derived macrophage-activating factor, GcMAF:
"This article has been retracted by the Journal’s Editors-in-Chief
in conjunction with the Publisher (Springer) due to irregularities
in the Institutional Review Board documentation."
The paper has been cited 28 times, according to Thomson
Scientific’s Web of Knowledge.
As we wrote in another post about Nobutu Yamamoto’s work,
we’ve noticed that retractions for IRB documentation
problems are often a bit like jailing Al Capone on tax evasion:
They’re the easiest charges to prove, but they’re likely the least
of a study’s problems.
A website, GcMAF.eu, continues to hawk the results of treatment:
The results from all the diseases we list are astonishing, but in
late stage cancer the clinics achieve an average of 25% tumour
reduction per week. (We get that reduction with pancreatic cancer
too.)
The other two retractions for Yamamoto were in the International
Journal of Cancer and the Journal of Medical Virology.
https://gcmaf.se/
Here is a time lapse video of the 8th assay we do in our
laboratory - our GcMAF activates macrophages that eat cancers
cells. We are probably the only people in the world with this
technology.
Watch the video to see what happens to cancer cells when GcMAF is
added without macrophages. This again is a world first, and again
it has been done in our laboratory. Within just 4 weeks a research
abstract paper on our results has already been accepted for
publication at this year’s Immunology Conference in California.
https://youtu.be/D1WZrnCcH24
https://www.youtube.com/watch?t=119&v=D1WZrnCcH24
Cancer cells destroyed by First Immune
GcMAF (gcmaf.eu)
http://www.anticancerfund.org/therapies/gcmaf
Warning: GcMAF has not been properly studied in clinical trials
and its laboratory results still need to be confirmed
independently. So far, all claims on the efficacy of this product
have no solid scientific basis. Its marketing is illegal; and the
activities of Immuno Biotech Ltd. (www.gcmaf.se formerly
www.gcmaf.eu) formerly with contact address in Brussels are
currently under investigation by different European regulatory
authorities. The factory where GcMAF was being manufactured in the
UK, was closed because it was found that the product was not being
produced under Good Manufacturing Practice (GMP) standards. There
were concerns on the sterility of the products and the equipment
used to produce it. The blood plasma used to manufacture this
product should not be injected to humans nor should it be used to
produce drugs. Not only did UK authorities shut down the GcMAF
factory, but importation of the product has also been banned to
guarantee the wellbeing of patients.
Immuno Biotech Ltd. opened three centers (www.immunocentre.eu),
one in Switzerland and two purportedly in Germany and The
Netherlands. The Swiss center was closed down by Swiss authorities
earlier this year.
In contrast to the statements made by David Noakes from Immuno
Biotech Ltd., GcMAF needs to be investigated in randomized
clinical trials and it is currently being investigated in a
registered clinical trial that complies with the established
guidelines for the first time. This trial is a phase I carried out
by a company independent of Noakes’s operation (NCT02052492).
GcMAF is a protein claimed to cure cancer and other ailments, but
no proper clinical studies have been ever performed to confirm
this claim.
Three clinical studies, by Nobuto Yamamoto et al., are referred as
the initial evidence that GcMAF can cure cancer. However, after a
thorough review of this work and discussion with experts, many
flaws have been identified. Specially the use of an invalid
endpoint to evaluate treatment response: the measurement of an
enzyme in blood called Nagalase. After months of trying to
get additional information on the patients and scientists involved
in this research we came to the conclusion that these data should
not be relied on since there are important issues in the
methodology and procedures. The same group has also presented
their results to different scientific conferences and we could
confirm that one co-author’s participation was denied by the
person himself, while we could not contact others besides Nobuto
Yamamoto. The editors of the journals where Yamamoto et al.
published their cancer-related articles were informed on the
irregularities and flaws on this research. The articles claiming
that GcMAF is effective to treat breast and colorectal cancer have
been retracted, as reported here and here. Unfortunately due to
lack of interest from the Editorial Board of the Translational
Oncology Journal on discussing the flaws and irregularities in the
prostate cancer article, it is still part of the official
literature.
Other groups have reported their results treating cancer patients
with GcMAF, but they used the same invalid methods to measure the
response to treatment than Yamamoto, Nagalase test in blood. In
recent articles they based their conclusions on ultrasound
imaging, which is specifically advised against by the RECIST
criteria as a way to measure tumor response. These groups
are also involved in the illegal marketing of GcMAF. So far, we
could not find any evidence that their products are being
manufactured according to Good Manufacturing Practices (GMP)
guidelines and have been properly tested for safety for human
consumption according to the Good Clinical Practices (GCP)
guidelines.
http://www.jleukbio.org/content/73/2/209.short
The many faces of macrophage
activation
by
David M. Mosser
INTRODUCTION
It used to be easy. In the old days (~8 years ago), activated
macrophages were simply defined as cells that secreted
inflammatory mediators and killed intracellular pathogens. Things
are becoming progressively more complicated in the world of
leukocyte biology. Activated macrophages may be a more
heterogenous group of cells than originally appreciated, with
different physiologies and performing distinct immunological
functions. The first hint of this heterogeneity came with the
characterization of the “alternatively activated macrophage” [1].
The exposure of macrophages to interleukin (IL)-4 or
glucocorticoids induced a population of cells that up-regulated
certain phagocytic receptors but failed to produce nitrogen
radicals [2] and as a result, were relatively poor at killing
intracellular pathogens. Recent studies have shown that these
alternatively activated cells produce several components involved
in the synthesis of the extracellular matrix (ECM) [3], suggesting
their primary role may be involved in tissue repair rather than
microbial killing. It turns out that the name alternatively
activated macrophage may be unfortunate for a few reasons. First,
although these cells express some markers of activation, they have
not been exposed to the classical, activating stimuli,
interferon-? (IFN-?) and lipopolysaccharide (LPS). Second, and
more importantly, the name implies that this is the only other way
to activate a macrophage. Recent studies suggest that this may not
be the case. Exposure of macrophages to classical activating
signals in the presence of immunoglobulin G (IgG) immune complexes
induced the production of a cell type that was fundamentally
different from the classically activated macrophage. These cells
generated large amounts of IL-10 and as a result, were potent
inhibitors of acute inflammatory responses to bacterial endotoxin
[4]. These activated macrophages have been called type 2-activated
macrophages [5] because of their ability to induce T helper cell …
http://onlinelibrary.wiley.com/doi/10.1002/jmv.21376/abstract
Retracted: Immunotherapy of
HIV-infected patients with Gc protein-derived macrophage
activating factor (GcMAF)
Nobuto Yamamoto, Naofumi Ushijima, Yoshihiko Koga
Abstract
The above article, published online on 21 Nov 2008 Wiley Online
Library (wileyonlinelibrary.com), has been retracted by agreement
between Dr. Ari Zuckerman, Editor-in-Chief, Journal of Medical
Virology and Wiley Periodicals, Inc. due to irregularities in the
documentation for institutional review board approval.
http://onlinelibrary.wiley.com/doi/10.1002/ijc.23107/abstract
Retracted: Immunotherapy of metastatic
breast cancer patients with vitamin D-binding
protein-derived macrophage activating factor (GcMAF)
Nobuto Yamamoto, Hirofumi Suyama, Nobuyuki Yamamoto, Naofumi
Ushijima
Abstract
The above article from the International Journal of Cancer,
published online on 12 October 2007 in Wiley Online Library and in
Volume 122, Issue 2, pp 461–467, has been retracted by agreement
between the journal Editor-in-Chief Peter Lichter and Wiley
Periodicals, Inc. due to irregularities in the documentation for
institutional review board approval.
[ Excerpt ]
https://clinicaltrials.gov/ct2/show/NCT02052492?term=gcmaf&rank=1
Safety Study of GcMAF (Globulin
Component Macrophage Activating Factor) in Subjects With
Advanced Solid Tumors
Activated macrophages, present in excess during natural
inflammatory responses, bear the potential to kill and eradicate
cancer cells. Macrophage activation has been demonstrated to
require the serum-borne vitamin D binding protein (known as Gc
protein), as well as B and T lymphocytes. However, in various
cancer patients the Gc protein is deglycosylated by serum
a-N-acetyl galactosaminidase (Nagalase) secreted from cancer
cells. This deglycosylated Gc protein, lacking the
N-acetylgalactosamine monosaccharide, cannot be converted to its
form of Macrophage Activating Factor, leading to immunosuppression
rather than Macrophage activation against cancer cells.
Efranat has developed cancer immunotherapy based on Macrophage
Activating Factor produced from natural Gc protein extracted from
FDA approved healthy human plasma.
In this phase I study, the treatment is given as Intramuscular,
once-weekly injection of GcMAF, for two courses, while each course
is comprised of 4 injections.
The investigational treatment is expected to enhance immune
response, thereby, improve patient's well being, quality of life
and disease control.
Primary objectives:
To determine the safety and tolerability of GcMAF and to define
the Maximal Tolerated Dose (MTD)
To identify the Dose Limiting Toxicity (DLT) of GcMAF
Secondary objectives:
To determine the 'Recommended Phase 2 Dose' (RP2D) based on MTD
data, immunological and pharmacodynamic markers
.
To explore the preliminary efficacy of GcMAF in advanced solid
tumors, considering the 'Immune-related Response Criteria' (irRC),
the 'Response Evaluation Criteria in Solid Tumors' (RECIST) and
blood levels of tumor-related markers known to reflect tumor
burden.
Exploratory objectives:
To assess levels of immune-related factors in peripheral blood,
reflecting induced immunological activities.
To immunohistochemically assess and compare tumor derived tissue
samples Pre and post treatment. To analyze the infiltration of
different population of cells into the tumor bed...
Detailed Description:
Part 1:
Eligible subjects will be assigned, successively in order of
accrual, to one of the three cohorts, to receive intramuscular
(IM) injections of GcMAF, once weekly for two courses of
treatment. Each course will consist of 4 injections with one week
intervals (total: 8 weeks of treatment).
Dose escalation will only proceed in the absence of dose-limiting
toxicity (DLT) during course 1. For this purpose, each cohort will
only begin its first course of GcMAF when the cohort preceding it
has successfully completed its first 4-week treatment course
without any signs of DLT. During this first course, should 1/3
patients experience DLT, dose escalation for the next cohort will
not be authorized; the next cohort will receive the same dose as
the one preceding it. If 2 patients or more of all patients
treated with a given dose develop DLT, dose escalation will be
halted and no more patients will be treated at the DLT dose. The
value of MTD will be defined as the GcMAF dose below the dose at
which DLT was seen for at least 2 subjects. Upon determination of
the MTD, an additional cohort will be opened (confirmatory cohort)
and treated with two courses of that dose.
Part 2:
Once MTD is established, or, when the last cohort completes its
first course of treatment without an established MTD, an
additional cohort will be opened in order to treat up to
additional subjects with a selected dose of GcMAF already
confirmed to be safe in part 1 of the study.
Continued treatment after completion of course 2 will be as per
the investigator's discretion. Patient follow-up will continue for
12 months after start of treatment (Day 1). Patients for whom
progressive disease (PD) is observed prior to completion of those
two courses will be followed up only until PD...
Nobuto Yamamoto
Prof Nobuto Yamamoto is the pioneer and discoverer of the GcMAF
glycoprotein and has conducted over 30 years of extensive research
on its anti-cancer traits. Dr. Yamamoto has been a visiting
Scientist at Fox Chase Cancer Center in Philadelphia where he
studied the genetic evolution of bacterial viruses (1959-1961) He
has been a scientist at Biological Standards NIH (1962-1964) and
became Chief of Virology and Genetics of The Fels Institute at the
Temple University School of Medicine in Philadelphia where he
served from 1964 up till 1980 and again from 1990 till 1994. In
1980 Dr. Yamamoto was appointed Professor of Microbiology and
Immunology at Hahnemann University School of Medicine,
Philadelphia. (1980-1990) In this role he continued his research
on immunology which emphasized mechanism of macrophage activation
and discovered GcMAF.In 1994 he founded the Socrates Institute for
Therapeutic Immunology, Philadelphia (1994 to date) where he
continues to study the therapeutic efficacy of GcMAF for a variety
of cancers and HIV. And in 2009 became one of the founders and
board member of Efranat.
[ NOTE : The following article touts the work of Dr
Burzynski re: Antineoplastons vs Cancer. Dr B. has however brought
much of his problem upon himself because ( according to the FDA )
of his :
Enrollment of subjects into antineoplaston study protocols prior
to the protocol-specified interval following prior chemotherapy
and/or radiation therapy.
Failure to report all serious adverse events (SAEs) and adverse
events (AEs) to the agency and/or IRB.
Failure to follow proper informed consent procedures.
Failure to maintain adequate drug accountability records.
Discrepancies between case report forms and source documents.
Failure to keep a copy of the study protocol and informed consent
form.
Failure to receive and/or require progress reports from the
principal investigator for the study.
Failure to receive and/or require a final report from the
principal investigator for the study prior to removal from the
IRB’s active list of studies.
Failure to assure that FDA approval was obtained by the principal
investigator for the study prior to the treatment of a patient
under a special exception.
Approval of special exceptions via expedited review.
The IRB approved research without determining that the following
criteria were met: That risks to subjects were minimized and that
risks to subjects were reasonable in relation to anticipated
benefits, if any, to subjects, and the importance of the knowledge
that may be expected to result.
The IRB failed to prepare, maintain, and follow written procedures
for conducting its initial and continuing review of research.
The IRB failed to ensure that informed consent would be sought
from each prospective subject or the subject’s legally authorized
representative.
The IRB failed to ensure that no member participated in the
initial or continuing review of a project in which the member had
a conflicting interest.
The IRB failed to conduct continuing reviews.
And :
The IRB failed to follow FDA regulations regarding expedited
review procedures [21 CFR 56.110(b)].
The IRB approved research without determining that the following
criteria were met: risks to subjects were minimized [21 CFR
56.111(a)(1)]; risks to subjects were reasonable in relation to
anticipated benefits, if any, to subjects, and the importance of
the knowledge that may be expected to result [21 CFR
56.111(a)(2)].
The IRB failed to determine at the time of initial review that
studies involving children are in compliance with 21 CFR part 50,
subpart D, Additional Safeguards for Children in Clinical
Investigations [21 CFR 56.109(h)]. This is a repeat violation from
our 2010 inspection.
The IRB failed to prepare, maintain, and follow written procedures
and maintain adequate documentation governing the functions and
operations of the IRB [21 CFR 56.108(a), 21 CFR 56.108(b), and 21
CFR 56.115(a)(6)].
For more information -- http://www.rexresearch.com/burzynski/burzynski.htm
]
[ Excerpt ]
http://anh-europe.org/news/how-maverick-cancer-treatments-are-suppressed-by-the-mainstream
How Maverick Cancer Treatments are
Suppressed by the Mainstream
Gc-MAF: antineoplastons mark II?
...Back in 1993, Nobuto Yamamoto, then working at Temple
University School of Medicine in Philadelphia, PA, USA, first
described a remarkable molecule. His paper reported the conversion
of vitamin D3 binding protein (DBP, known in humans as Gc) into a
potent macrophage-activating factor (MAF), known as Gc-MAF.
Macrophages are a key part of the human immune system with two
roles: to engulf and destroy pathogens and cellular debris, and to
recruit other immune cells to respond to the pathogen.
Macrophages are crucial to both innate, or non-specific, immunity
and adaptive, or specific, immunity. Under normal circumstances,
Gc-MAF is upregulated when the immune system detects a threat,
‘activating’ macrophages so that they single-mindedly pursue
pathogens. Cancer cells, a prime target of macrophages, are clever
little critters that secrete an enzyme –
alpha-N-acetylgalactosaminidase or nagalase – that inactivates
Gc-MAF, thus preventing macrophages from becoming activated and
protecting cancer cells. Administration of Gc-MAF is proposed to
bypass nagalase, stimulating macrophages to become activated and
attack tumour cells. In other words, it’s a potent
immunomodulator, rather like antineoplastons.
So why haven’t most of us heard of it?
Unlike antineoplastons, however, Gc-MAF hasn’t had the benefit of
a single patent owner – as a natural molecule, it cannot be
patented without being modified – with the will and resources to
push it under the noses of the public and health authorities. Dr
Yamamoto has run small human trials in breast, prostate and
colorectal cancers, with promising results. However, he is by no
means alone, as David Noakes is at pains to point out: “There’s
better research than Dr Yamamoto’s out there these days, and it’s
all listed on our website,” he says.
David Noakes might just be the person to bring Gc-MAF into the
mainstream. He’s the CEO of Immuno Biotech Ltd. and spokesperson
for First Immune Gc-MAF, a project he describes as, “PhD and BSc
biochemists and biomedical scientists...with external doctors,
oncologists and scientists who kindly provide advice, committed to
bringing some of the increasing number of published but relatively
unused medical cures to as many people as we can.” At the
moment, Noakes and his colleagues are supplying Gc-MAF to 30
countries where it is legal, via a network of “around 300”
doctors. Their Gc-MAF is made to extremely high standards, and is
being used in ongoing clinical research by Noakes’ collaborators
and others. Their ultimate goal is to, “Build the case that GcMAF
is effective for various illnesses, which will help to make it
available to the public”.
An uphill struggle
Noakes has no illusions about the struggle he faces, as he
explained to ANH-Intl. “Doctors, cancer doctors especially, know
what they’re up against in countries like the US and UK. In all of
the US, there’s only one doctor – Jeffrey Bradstreet, MD – who’s
so far been prepared to put his head above the parapet about
Gc-MAF. Many more take a cautious but pragmatic attitude; they
might say to their patients, “Get it, but please don’t tell me!””
Getting nowhere in Guernsey
Recently, Noakes asked the Guernsey authorities whether they would
allow First Immune Gc-MAF to produce the product in peace, in
their own laboratory, hoping to take advantage of the island’s
historically liberal attitude and exemption from EU laws. “We even
offered to treat Guernsey kids with autism or cancer with Gc-MAF,
for free – and anyone with infectious diseases for one-third of
the normal cost. But they said no!” he reports in disgust.
“Guernsey has even declared Gc-MAF an illegal drug, unlike
anywhere else in the world. It’s come to something when Dubai is a
better environment for the production and distribution of a
potentially life-saving treatment than Guernsey, the UK or USA,
but there you go.”...
PATENTS & PATENT APPLICATIONS
WO2014199373
COMPOSITIONS COMPRISING GC- MACROPHAGE ACTIVATING FACTOR AND
USES THEREOF
The present invention relates to stable pharmaceutical
compositions comprising Gc macrophage activating factor (GcMAF).
The present invention relates in particular to storage-stable
pharmaceutical compositions comprising GcMAF and at least one
pharmaceutically acceptable surfactant and/or a synthetic
water-soluble polymer having surface activity and uses thereof for
treating diseases associated with macrophage activation.
RU2198218
METHOD OF VITAMIN D3-BINDING PROTEIN (PROTEIN GC)
CLONING...
FIELD: molecular biology, protein engineering, medicine.
SUBSTANCE: invention relates to strong factors of macrophages
activation. Vitamin D3-binding protein (Gc-protein) and its small
domain (about 1/5 of Gc-peptide) that is known as domain III also)
is cloned using baculovirus vector. The cloned Gc-protein and
cloned peptide domain III (Cd) are treated with immobilized
beta-galactosidase and sialidase and factors of activation of
macrophages GcMAFc and CdMAF, respectively, are prepared. These
cloned factors of activation of macrophages and GcMAF can be used
as adjuvants in immunization and vaccination. Invention allows to
treat sicknesses, for example, osteopetrosis. EFFECT: improved
method of cloning and preparing, valuable medicinal properties.
US2014030215 / JP2014511857 /
CN103547280 / WO2012137199
Macrophage activating factor for pharmaceutical
compositions
The present invention relates to pharmaceutical compositions
comprising macrophage activating factor (MAF) and method of
producing same, particularly to MAF compositions essentially
devoid of glycosidase enzymes. The compositions of the present
invention and pharmaceutical compositions comprising same are
particularly suitable for intravenous administration. Thus
according to one aspect, the present invention provides a
composition comprising,Gc protein-derived macrophage activating
factor (GcMAF), wherein the composition is essentially devoid of
glycosidase enzymes.
Vitamin D-based complexes for use as supplements
GB2515347
Vitamin D-based complexes for use as supplements
Vitamin D based preparations are disclosed comprising a complex of
de-glycosylated vitamin D-binding protein (also called GcMAF),
vitamin D3 or its analogues, and at least on unsaturated fatty
acid. The preparations may include an aqueous alcoholic saline
solvent, and the unsaturated fatty acid can be oleic acid or
eicosapentaenoic acid. Orally administrable compositions
comprising the aforementioned preparations encapsulated in
liposomes are also disclosed, along with methods for the
production of the aforementioned preparations
US2013129670
MACROPHAGE ACTIVATING FACTOR FOR USE IN THE TREATMENT OF
CHRONIC FATIGUE SYNDROME (CFS) AND CFS-RELATED DISEASES AND
DISORDERS
The present invention relates to Macrophage Activating Factors
such as GcMAF and compositions thereof, for use in the treatment
of a patient suffering from CFS/ME and/or XMRV infection.
US2011123591
Tumoricidal, bactericidal, or viricidal macrophage activation
The activation of macrophages and methods for treating cancer,
bacterial pathogens and viral pathogens are disclosed. In
particular, Gc protein is converted to Gc-macrophage activating
factor (GcMAF), in vivo or ex vivo. The GcMAF activates
macrophages which can then target cancer cells, bacterial
pathogens and/or viral pathogens. Alternatively, macrophages are
activated by contacting them, in vivo or ex vivo, with GcMAF.
Optionally, nagalase is inactivated in a patient receiving the
present macrophage activating treatment by contacting the
patient's blood with a Nagalase-binding ligand immobilized on an
inert medium.
WO2012029954
PROCESS FOR PRODUCTION OF NOVEL DEGALACTOSYLATED GC GLOBULIN
This invention provides a Gc globulin derivative that can be
easily produced from Gc globulin and can be used as GcMAF. This
invention also provides degalactosylated Gc globulin obtained by
processing Gc globulin with -galactosidase.
WO2016162867
COMBINATION THERAPY OF MACROPHAGE ACTIVATING FACTOR AND
PD-1 SIGNALING INHIBITORS
Inventor: SHAHAR MICHAL, et al.
The present invention relates to methods for treating cancer or
infectious diseases comprising administering to a subject an agent
that reduces or inhibits signal transduction mediated by PD-1 in
combination with Gc protein derived macrophage activating factor
(GcMAF). Particularly, the present invention provides methods of
treating cancer comprising administering to a subject an anti-PD-1
antibody and GcMAF.
US2011123591
TUMORICIDAL BACTERICIDAL OR VIRICIDAL
MACROPHAGE ACTIVATION
Inventor: KNEZEVICH CHARLES, et al.
The activation of macrophages and methods for treating cancer
bacterial pathogens and viral pathogens are disclosed. In
particular Gc protein is converted to Gc-macrophage activating
factor (GcMAF) in vivo or ex vivo. The GcMAF activates macrophages
which can then target cancer cells bacterial pathogens and/or
viral pathogens. Alternatively macrophages are activated by
contacting them in vivo or ex vivo with GcMAF. Optionally nagalase
is inactivated in a patient receiving the present macrophage
activating treatment by contacting the patient’s blood with a
Nagalase-binding ligand immobilized on an inert medium.
US2015361151
MACROPHAGE ACTIVATING FACTOR FOR PHARMACEUTICAL COMPOSITIONS
Inventor(s): YAMAMOTO NOBUTO
The present invention relates to pharmaceutical compositions
comprising macrophage activating factor (MAF) and method of
producing same, particularly to MAF compositions essentially
devoid of glycosidase enzymes. The compositions of the present
invention and pharmaceutical compositions comprising same are
particularly suitable for intravenous administration.
US 5712104 / US6410269
Preparation of potent macrophage activating factors derived
from cloned vitamin D binding protein...
Nobuto Yamamoto
Vitamin D-binding protein (Gc protein) and its small domain
(approximately [1/5] of the Gc peptide also known as domain III)
were cloned via a baculovirus vector. The cloned Gc protein and
the cloned domain (Cd) peptide were treated with immobilized
beta-galactosidase and sialidase to yield macrophage activating
factors, GcMAFc and CdMAF, respectively. These cloned macrophage
activating factors and GcMAF are to be used for therapy of cancer,
HIV-infection and osteopetrosis, and may also be used as adjuvants
for immunization and vaccination.
FIELD OF THE INVENTION
[0001] This invention relates to potent macrophage activating
factors, prepared by oligosaccharide digestion of the cloned
vitamin D binding protein (Gc protein) and the cloned Gc protein
domain III, and the use of these macrophage activating factors for
various cancer, HIV-infection and osteopetrosis, and as adjuvants
for immunization and vaccination.
TABLE OF TERMS
[0002] Gc protein Vitamin D3-binding protein
[0003] MAF macrophage activating factor
[0004] GcMAF Gc protein-derived macrophage activating protein
[0005] GcMAFc cloned Gc protein-derived macrophage activating
factor
[0006] Gc domain III domain III region of Gc protein
[0007] CdMAF cloned domain III-derived macrophage activating
factor
SUMMARY OF THE INVENTION
[0008] Vitamin D-binding protein (Gc protein) and its small domain
(approximately [1/5] of the Gc peptide also known as domain III)
were cloned via a baculovirus vector. The cloned Gc protein and
the cloned domain (Cd) peptide were treated with immobilized
[beta]-galactosidase and sialidase to yield macrophage activating
factors, GcMAFc and CdMAF, respectively. These cloned macrophage
activating factors and GcMAF are to be used for therapy of cancer,
HIV-infection and osteopetrosis, and may also be used as adjuvants
for immunization and vaccination.
DESCRIPTION OF THE DRAWINGS
[0009] Other objects and many attendant features of this invention
will become readily appreciated as the same becomes better
understood by reference to the following detailed description when
considered in connection with the accompanying drawings wherein:
[0010] FIG. 1a is a schematic illustration of the formation of
macrophage activating factor (MAF).
[0011] FIG. 1b is a schematic illustration of the
deglycosylation of Gc protein in a cancer or HIV-infected
patient's blood stream.
[0012] FIG. 2 shows the correlation between plasma
[alpha]-N-acetylgalactosaminidase activity and tumor burden
(total cell counts) in the peritoneal cavity of Ehrlich ascites
tumor.
[0013] FIG. 3 shows the amino acid sequence of cloned GcMAF
which is SEQ ID NO:1 which is the entire cloned Gc protein.
[0014] FIG. 4 shows the construction of the DNA fragment
encoding the leader sequence of EcoRi fragment E1 and domain III
regions of the Gc protein; A, the entire cDNA for Gc protein; B,
the construct to be inserted into the non-fusion vector; the
shaded area indicates the compressed regions of about 1,000 base
pairs (bp).
[0015] FIG. 5 shows the 89 amino acid sequence, SEQ ID
NO:2, of the cloned domain III (CdMAF1), using the non-fusion
vector.
[0016] FIG. 6 shows the baculovirus fusion vector for
cloning the domain III of Gc protein.
[0017] FIG. 7 shows the 94 amino acid sequence, SEQ ID
NO:3, of the cloned domain III (CdMAF2), using the fusion
vector.
[0018] FIG. 8A shows the therapeutic effect of GcMAF in
accordance with the present invention on adult persons suffering
from prostate cancer.
[0019] FIG. 8B shows the therapeutic effect of GcMAF in
accordance with the present invention on adult persons suffering
from breast cancer.
[0020] FIG. 8C shows the therapeutic effect of GcMAF in
accordance with the present invention on adult persons suffering
from colon cancer.
[0021] FIG. 8D shows the therapeutic effect of GcMAF in
accordance with the present invention on adult persons suffering
from leukemia.
BACKGROUND OF THE INVENTION
[0022] A. Inflammatory Response Results in Activation of
Macrophages
[0023] Inflammation results in the activation of macrophages.
Inflamed lesions release lysophospholipids. The administration
into mice of small doses (5-20 [mu]g/mouse) of
lysophosphatidylcholine (lyso-Pc) and other lysophospholipids
induced a greatly enhanced phagocytic and superoxide generating
capacity of macrophages (Ngwenya and Yamamoto, Proc. Soc. Exp.
Biol. Med. 193:118, 1990; Yamamoto et al, Inf. Imm. 61:5388, 1993;
Yamamoto et al., Inflammation. 18:311, 1994).
[0024] This macrophage activation requires participation of B and
T lymphocytes and serum vitamin D binding protein (DBP; human DBP
is known as Gc protein). In vitro activation of mouse peritoneal
macrophages by lyso-Pc requires the step-wise modification of Gc
protein by 3-galactosidase of Iyso-Pc-treated B cells and
sialidase of T cells to generate the macrophage activating factor
(MAF), a protein with N-acetylgalactosamine as the remaining sugar
moiety (FIG. 1a (Yamamoto et al., Proc. Natl. Acad. Sci. USA.
88:8539, 1991; Yamamoto et al., J. Immunol. 151:2794, 1993;
Naraparaju and Yamamoto, Immunol. Lett. 43:143, 1994). Thus, Gc
protein is a precursor for MAF.
[0025] Incubation of Gc protein with immobilized
[beta]-galactosidase and sialidase generates a remarkably high
titered MAF (GcMAF) (Yamamoto et al., Proc. Natl. Acad. Sci. USA.
88:8539, 1991; Yamamoto et al., J. Immunol. 151:2794, 1993;
Naraparaju and Yamamoto, Immunol. Lett. 43:143, 1994; U.S. Pat.
No. 51,177,002). Administration of a minute amount (10 pg/mouse;
100 ng/human) of GcMAF resulted in greatly enhanced phagocytic and
super-oxide generating capacities of macrophages.
[0026] When peripheral blood monocytesimacrophages (designated as
macrophages hereafter) of 258 cancer patients bearing various
types of cancer were treated in vitro with 100 pg GcMAF/ml,
macrophages of all cancer patients were activated for phagocytic
and superoxide generating capacity. This observation indicates
that cancer patient macrophages are capable of being activated.
However, the MAF precursor activity of plasma Gc protein was lost
or reduced in approximately 70% of this cancer patient population.
Loss of the MAF precursor activity prevents generation of MAF.
Therefore, macrophage activation cannot develop in certain cancer
patients. Since macrophage activation is the first step in the
immune development cascade, such cancer patients become
immunosuppressed. This may explain at least in part why cancer
patients die from overwhelming infection. Lost or reduced
precursor activity of Gc protein was found to be due to
deglycosylation of plasma Gc protein by
[alpha]-N-acetylgalactosaminidase detected in cancer patient blood
stream. Deglycosylated Gc protein cannot be converted to MAF (FIG.
1b.
[0027] Similarly, when peripheral blood macrophages of 160
HIV-infected/AIDS patients were treated in vitro with 100 pg
GcMAF/ml, macrophages of all patients were activated for
phagocytic and superoxide generating capacity. However, the MAF
precursor activity of plasma Gc protein was low in approximately
35% of the HIV-infected patient population. As in cancer patients,
these patients' plasma Gc protein is deglycosylated by
[alpha]-N-acetylgalactosaminidase detected in HIV-infected
patients.
[0028] Both cancer and HIV-infected patients having severely
decreased precursor activity of plasma Gc protein carried large
amounts of [alpha]-N-acetylgalactosaminidase while patients having
moderately decreased precursor activity had moderate levels of
plasma [alpha]-N-acetylgalactosaminidase activities. Patients with
high precursor activity, including asymptomatic HIV-infected
patients, had low but significant levels of plasma
[alpha]-N-acetylgalactosaminidase activity. Since a large amount
(260 [mu]g/ml) of Gc protein exists in the blood stream, a low
level of the enzyme does not affect the precursor activity.
Nevertheless, [alpha]-N-acetylgalactosaminidase activity was found
in plasmas of all cancer and HIV-infected patients and had an
inverse correlation with the precursor activity of their plasma Gc
protein (Yamamoto et al., AIDS Res. Human Ret. 11:1373, 1995).
Thus, increase in patient plasma [alpha]-N-acetylgalactosaminidase
activity is responsible for decrease in the precursor activity of
plasma Gc protein. These observations lead us to propose that
plasma [alpha]-N-acetylgalactosaminidase plays a role in
immunosuppression in cancer and HIV-infected/AIDS patients.
[0029] B. The Origin of Immunosuppression
[0030] The source of the plasma [alpha]-N-acetylgalactosaminidase
in cancer patients appeared to be cancerous cells. High
[alpha]-N-acetylgalactosaminidase activities were detected in
tumor tissue homogenates of various organs, including eleven
different tumor tissues including 4 lung, 3 breast, 3 colon and 1
cervix tumors, though the [alpha]-N-acetylgalactosaminidase
activity varied from 15.9 to 50.8 nmoles/mg/min. Surgical removal
of malignant lesions in human cancer results in subtle decrease of
plasma [alpha]-N-acetylgalactosaminidase activity with concomitant
increase in the precursor activity, particularly if malignant
cells are localized.
[0031] In a preclinical mouse tumor model, BALB/c mice were
transplanted with 5*10<5 >Ehrlich ascites tumor cells/mice
into peritoneal cavity and analyzed for serum
[alpha]-N-acetylgalactosaminidase activity. When plasma enzyme
level were measured as transplanted Ehrlich ascites tumor grew in
mouse peritoneal cavity, the enzyme activity was directly
proportional to tumor burden as shown in FIG. 2. This was also
confirmed with nude mouse transplanted with KB cells (human oral
squamous cell carcinoma cell line). Serum
[alpha]-N-acetylgalactosaminidase activity increased as tumor size
(measured by weight) of the solid tumor increased. Thus, I have
been using plasma [alpha]-N-acetylgalactosaminidase activity as a
prognostic index to monitor the progress of therapy.
[0032] Radiation therapy of human cancer decreased plasma
[alpha]-N-acetylgalactosaminidase activity with a concomitant
increase of precursor activity. This implies that radiation
therapy decreases the number of cancerous cells capable of
secreting [alpha]-N-acetylgalactosaminidase. These results also
confirmed that plasma [alpha]-N-acetylgalactosaminidase activity
has an inverse correlation with the MAF precursor activity of Gc
protein. Even after surgical removal of tumor lesions in cancer
patients, most post-operative patients carried significant amounts
of [alpha]-N-acetylgalactosaminidase activity in their blood
stream. The remnant cancerous lesions in these post-operative
patients cannot be detectable by any other procedures, e.g.,
X-ray, scintigraphy, etc. I have been using this most sensitive
enzyme assay as prognostic index during the course of GcMAF
therapy for treating cancer.
[0033] HIV-infected cells appeared to secrete
[alpha]-N-acetylgalactosaminidase. When peripheral blood
mononuclear cells (PBMC) of HIV-infected patients were cultured
and treated with mitomycin as a provirus inducing agent (Sato et
al., Arch. Virol. 54:333, 1977), [alpha]-N-acetylgalactosaminidase
was secreted into culture media. These results led us to suggest
that [alpha]-N-acetylgalactosaminidase is a virus coded product.
In fact, HIV-envelope protein gp120 appears to carry the
[alpha]-N-acetylgalactosaminidase activity.
[0034] C. A Defect in Macrophage Activation Cascade Manifests
Osteopetrosis
[0035] An inflammation-primed macrophage activation cascade has
been defined as a major process leading to the production of
macrophage activating factor. Activation of other phagocytes such
as osteoclasts shares the macrophage activation cascade (Yamamoto
et al., J. Immunol. 152:5100, 1994). Thus, a defect in the
macrophage activation cascade results in lack of activation in
osteoclasts.
[0036] Autosomal recessive osteopetrosis is characterized by an
excess accumulation of bone throughout the skeleton as a result of
dysfunctional osteoclasts, resulting in reduced bone resorption
(Marks, Clin. Orthop. 189:239, 1984). In animal models of
osteopetrosis, depending on the degree of osteoclast dysfunction,
marrow cavity development and tooth eruption are either delayed or
more commonly absent (Marks, Am. J. Med. Genet. 34:43, 1989). In
human infantile osteopetrosis, death occurs within the first
decade of life usually overwhelming infection (Reeves, Pediatrics.
64:202,1979), indicating immunosuppression. Accumulated evidence
suggests that deficient or dysfunctional osteoclasts in
osteopetrotic animals are often accompanied by deficiencies or
dysfunctions of macrophages. The studies of the present inventor
on the activation of both osteoclasts and macrophages in the
osteopetrotic mutations revealed that osteoclasts and macrophages
can be activated by a common signaling factor, the macrophage
activating factor and that a defect in [beta]-galactosidase of B
cells incapacitates the generation process of macrophage
activating factor (Yamamoto et al., J. Immunol. 152:5100, 1994).
Since GcMAF and its cloned derivatives bypass the function of
lymphocytes and Gc protein and act directly on macrophages and
osteoclasts, administration of these factors into osteopetrotic
hosts should rectify the bone disorder. In fact the present
inventor has recently found that four administrations of purified
cloned human macrophage activating factor (GcMAFc) (100 pg/week)
to the p mutant mice beginning at birth for four weeks resulted in
the activation of both macrophages and -osteoclasts and subsequent
resorption of the excess skeletal matrix.
[0037] D. Therapeutic Application of GcMAF and its Cloned
Derivatives on Cancer
[0038] Despite defects in the macrophage activation cascade in
cancer, HIV-infected and osteopetrotic patients, GcMAF bypasses
the functions of lymphocytes and Gc protein and acts directly on
macrophages (or osteoclasts) for activation. Macrophages have a
potential to eliminate cancerous cells and HIV-infected cells when
activated. When cancer patients were treated with 100 ng
GcMAF/patient weekly for several months, GcMAF showed remarkable
curative effects on a variety of human cancer indiscriminately.
[0039] Instead of obtaining of GcMAF from human blood source, it
can be obtained from the cloned Gc protein or its small domain
responsible for macrophage activation. The cloning Gc protein
require an eukaryotic vector/host capable of the glycosylation of
the cloned products. The Gc protein having a molecular weight of
52,000 and 458 amino acid residues) is a multi-functional protein
and carries three distinct domains (Cooke and Haddad, Endocrine
Rev., 10:294,1989).
[0040] Domain I interacts with vitamin D while domain III
interacts with actin (Haddad et al., Biochem., 31:7174, 1992).
Chemically and proteolytically fragmented Gc enabled me to
indicate that the smallest domain, domain ill, contains an
essential peptide for macrophage activation. Accordingly, I cloned
both Gc protein and the entire domain III peptide, by the use of a
baculovirus vector and an insect host, and treated them with the
immobilized [beta]-galactosidase and sialidase to yield potent
macrophage activating factors, designated GcMAFc and CdMAF,
respectively. Like GcMAF, these cloned GcMAFc and CdMAF appear to
have curative effects on cancer.
[0041] E. A Potent Adjuvant Activity of GcMAF for Immunization
with Antigens or Vaccines
[0042] Macrophages are antigen presenting cells. Macrophages
activated by GcMAF rapidly phagocytize target antigens or cells
and presented the processed antigens to antibody producing cells.
I observed a rapid development of a large amount of antibody
secreting cells immediately (1 to 4 days) after inoculation of
small amount of GcMAF (100 pg/mouse) and sheep erythrocytes
(SRBC). This finding indicates that GcMAF and its cloned
derivatives, GcMAFc and CdMAF, should serve as potent adjuvants
for immunization and vaccination.
DESCRIPTION OF THE METHODS FOR GENE CLONING FOR MACROPHAGE
ACTIVATING FACTORS
[0043] A. Cloning of the cDNA of Gc Protein into an Insect
Virus.
[0044] A full length cDNA encoding the human Gc protein was
isolated from a human liver cDNA library in bacteriophage
[lambda]gt11 (Clontech, Palo Alto, Calif.) by the use of pico
Blue(TM) immunoscreening kit available from Stratagene of La
Jolla, Calif. The baculoviral expression system in the insect
cells takes advantages of several facts about the polyhedron
protein: (a) it is expressed to very high levels in infected cells
where it constitutes more than half of the total cellular protein
late in the infection cycle; (b) it is nonessential for infection
or replication of the virus, meaning that the recombinant virus
does not require any helper function; (c) viruses lacking
polyhedron gene have distinct plaque morphology from viruses
containing the cloned gene; and d) unlike bacterial cells, the
insect cell efficiently glycosylate the cloned gene products.
[0045] One of the beauties of this expression system is a visual
screen allowing recombinant viruses to be distinguished and
quantified. The polyhedron protein is produced at very high levels
in the nuclei of infected cells late in the viral infection cycle.
Accumulated polyhedron protein forms occlusion bodies that also
contain embedded virus particles. These occlusion bodies, up to 15
[mu]m in size, are highly refractile, giving them a bright shiny
appearance that is readily visualized under a light microscope.
Cells infected with recombinant viruses lack occlusion bodies. To
distinguish recombinant virus from wild-type virus, the
transfection supernatant (recombinant containing virus lysate) is
plaqued onto a monolayer of insect cells. Plaques are then
screened under a light microscope for the presence (indicative of
wild-type virus) or absence (indicative of recombinant virus) of
occlusion bodies.
[0046] Unlike bacterial expression systems, the baculovirus-based
system is an eukaryotic expression system and thus uses many of
the protein modification, processing such as glycosylation, and
transport reactions present in higher eukaryotic cells. In
addition, the baculoviral expression system uses a
helper-independent virus that can be propagated to high titers in
insect cells adapted for growth in suspension cultures, making it
possible to obtain large amounts of recombinant protein with
relative ease. The majority of the overproduced protein remains
soluble in insect cells by contrast with the insoluble proteins
often obtained from bacteria. Furthermore, the viral genome is
large (130 kbp) and thus can accommodate large segments of foreign
DNA. Finally, baculoviruses are noninfectious to vertebrates, and
their promoters have been shown to be inactive in mammalian cells
(Carbonell et al., J. Virol. 56:153, 1985), which gives them a
possible advantage over other systems when expressing oncogenes or
potentially toxic proteins.
[0047] 1) Choice of Baculoviral Vector.
[0048] All available baculoviral vectors are pUC-based and confer
ampicillin resistance. Each contains the polyhedron gene promoter,
variable lengths of polyhedron coding sequence, and insertion
site(s) for cloning the foreign gene of interest flanked by viral
sequences that lie 5' to the promoter and 3' to the foreign gene
insert. These flanking sequences facilitate homologous
recombination between the vector and wild-type baculoviral DNA
(Ausubel et al., Current Protocols in Mol. Biol. 1990). The major
consideration when choosing the appropriate baculoviral expression
vector is whether to express the recombinant as a fusion or
non-fusion protein. Since glycosylation of Gc peptide requires a
leader signal sequence for transfer of the peptide into the
endoplasmic reticulum, the cDNA containing initiation codon (-16
Met) through the leader sequence to the +1 amino acid (leu) of the
native Gc protein should be introduced to non-fusion vector with a
polylinker carrying the EcoRI site, pLV1393 (Invitrogen, San
Diego, Calif.).
[0049] During partial digestion of the cDNA for Go protein in
[lambda]gt11 with EcoRI enzyme, a full length Gc cDNA with EcoRi
termini was isolated electrophoretically, mixed with EcoRI-cut
pVL1393, and ligated with T4 ligase. This construct in correct
orientation should express the entire Gc peptide, a total of 458
amino acids (FIG. 3). To obtain the correct construction,
competent E. coli HB101 cells were transformed with pVL vector and
selected for transformants on Luria broth agar plates containing
ampicillin (LB/ampicillin plates). The DNA was prepared for the
sequencing procedure to determine which colony contains the insert
or gene with proper reading orientation, by first searching for
the 3' poly A stretch. The clones with 3' ply A (from the poly A
tail of mRNA) were then sequenced from the 5' end to confirm the
correct orientation of the full length DNA for the Gc peptide.
[0050] 2) Co-transfection of Insect Cells with the Cloned
Plasmid DNA and Wild-type Viral DNA
[0051] A monolayer (2.5*10<6 >cells in each of 25-cm<2
>flasks) of Spodoptera frugiperda (Sf9) cells was
co-transfected with a cloned plasmid (vector) DNA (2 [mu]g) and a
wild-type (AcMNPV) baculoviral DNA (10 [mu]g) in 950 [mu]l
transfection buffer (Ausubel et al., In Curr Protocols in Mol.
Biol. 1990). When the cells were cultured for 4 or 5 days, the
transfection supernatant contained recombinant viruses.
[0052] 3) Identification of Recombinant Baculovirus
[0053] The co-transfection lysates were diluted 10<4>,
10<5 >or 10<6 >and plated on Sf9 cells for cultivation
for 4 to 6 days. After the plaques were well formed, plaques
containing occlusion-negative cells were identified at a frequency
of 1.3%. Several putative recombinant viral plaques were isolated
and twice re-plaqued for purification. Pure recombinant viral
plaque clones were isolated.
[0054] B. Analysis of Protein of Interest from Recombinant
Baculovirus
[0055] 1) Preparation of Recombinant Virus Lysate
[0056] An insect cell Sf9 monolayer (2.5*10<6 >cells per
25-cm<2 >flask) was infected with a recombinant virus clone
and cultured in 5 ml GIBCO serum-free medium (from GIBCO
Biochemicals, Rockville, Md.) or medium supplemented with 0.1% egg
albumin to avoid contamination of serum bovine vitamin D binding
protein. The culture flasks were incubated at 27[deg.] C. and
monitored daily for signs of infection. After 4 to 5 days, the
cells were harvested by gently dislodging them from the flask and
the cells and culture medium were transferred to centrifuge tubes
and centrifuged for 10 min at 1000* g, 4[deg.] C. To maximize
infection for recombinant protein production, Sf9 cells were grown
in a 100-ml spinner suspension culture flask with 50 ml complete
medium up to about 2*10<6 >cells/ml. The cells were
harvested, centrifuged at 1000* g for 10 min and re-suspended in
10 to 20 ml serum-free medium containing recombinant virus at a
multiplicity of infection (MOI) of 10. After 1 hour of incubation
at room temperature, the infected cells were transferred to a
200-ml spinner flask containing 100 ml serum-free medium and
incubated for 40 hr. More than 40% of the protein secreted was the
protein of interest. The protein in the supernatant was isolated.
[0057] 2) Qualitative Estimation of the Protein of Interest
[0058] Coomassie Blue staining of the SDS-polyacrylamide gel,
loading 20 to 40 [mu]g total cell protein per lane, was to
estimate quantity of expressed protein. Because the samples
contain cellular proteins, the recombinant protein was readily
detected by comparison with uninfected cellular proteins.
[0059] 3) Enzymatic Conversion of the Cloned Gc Protein to
Macrophage Activating Factor (GcMAFc).
[0060] The cloned Gc protein (2 [mu]g) with a molecular weight of
52,000 and 458 amino acid residues (FIG. 3) was isolated by
electroeluter and treated with immobilized [beta]-galactosidase
and sialidase. The resultant cloned macrophage activating factor
(GcMAFc) was added to mouse and human macrophages and assayed for
phagocytic and superoxide generating capacity. Incubation of
macrophages with 10 pg GcMAFc/ml for 3 hours resulted in a 5-fold
increased phagocytic and a 15-fold increase in the superoxide
generating capacity of macrophages.
[0061] C. Subcloning of a Domain Required for Macrophage
Activation
[0062] I. Cloning Procedure I: Non-fusion Vector.
[0063] 1) Cloning the Domain Responsible for Macrophage
Activation (CdMAF)
[0064] The entire cDNA sequence for Gc protein in [lambda]gt11,
including 76 bp of the upstream 5' flanking region and 204 bp of
the 3' flanking stretch, was fragmented by EcoRi to yield four
restriction fragments designated E1, 120; E2, 314; E3, 482; and
E4, 748 bp, respectively. Each was cloned into the EcoRI site of
the plasmid pSP65 from Promega (Madison, Wis.) by the method of
Cooke and David (J. Clin. Invest., 76 2420, 1985). Although I
found that a region less than one half of the domain III was found
to be responsible for macrophage activation, small segments less
than 40 amino acid residues cannot be expressed in the insect
cells. Moreover, short peptides are rapidly degraded by proteases
in human plasma, and thus are not clinically useful. Accordingly,
the entire domain III (approximately 80 amino acid residues)
should be subcloned into an insect virus where I anticipate the
efficient production and glycosylation of the peptide in the
infected cells.
[0065] 2) Subcloning cDNA Fragment into the Polyhedron Gene of
Baculovirus.
[0066] Since the glycosylation of a peptide requires a leader
signal sequence for transfer of the peptide into the endoplasmic
reticulum, the DNA segment of E1 containing the initiation codon
(-16 Met) through the leader sequence to the +1 amino acid (Leu)
of the native Gc protein should be introduced into the vector.
Because this segment carries the initiation codon for the Gc
protein, non-fusion vector, pVL1393 (Invitrogen, San Diego,
Calif.) was used. A segment containing the initiation codon-leader
sequence of the cDNA clone E1 and a segment coding for 85
C-terminal amino acids (the entire domain III plus 3' non-coding
stretch) of the cDNA clone E4 were ligated together and cloned
into the EcoRI site of the insect virus pVL vector. To achieve
this construct, both E1 and E4 DNA were fragmented with HaeIII to
yield two fragments each; E1hl (87 bp), E1hs (33 bp) and E4hs (298
bp), E4hl (450 bp), respectively. Both the larger fragments E1hl
and E4hl were isolated electrophoretically, mixed with EcoRI-cut
pVL, and ligated with T4 ligase, as shown in FIG. 4. This
construct in correct orientation should express the entire domain
III, a total of 89 amino acids, including the 4 amino acids of
E1hl, also referred to herein as CdMAF1 as shown in FIG. 5. To
obtain the correct construction, competent E. coli HB101 cells are
transformed with pVL vector and selected for transformants on
LB/ampicillin plates. DNA was prepared for sequencing procedures
to determine which colony contains the construct with proper
reading orientation by first searching for the 3' poly dA stretch.
Those clones with 3' poly dA (from the poly A tail of mRNA) were
then sequenced from the 5' end to confirm correct orientation of
the E1hl fragment. I found that the vector contains the entire
construct (domain III) in the correct orientation.
[0067] 3) Isolation of Recombinant Baculovirus, Purification of
the Cloned Domain Peptide (Cd) and Enzymatic Generation of the
Cloned Macrophage Activating Factor (CdMAF)
[0068] Monolayers (2.5*10<6 >cells in each of 25-cm<2
>flasks) of Spodoptera frugiperda (Sf9) cells were
co-transfected with cloned plasmid DNA (2 [mu]g) and wild-type
(AcMNPV) baculoviral DNA (10 [mu]g) in 950 [mu]l transfection
buffer. Recombinant baculovirus plaques were isolated and used for
production of the Gc domain III peptide in insect cells. This
cloned domain with a molecular weight (MW) of 10,000 and 89 amino
acids as shown in FIG. 5, was purified electrophoretically. Two
[mu]g of the cloned domain (Cd) peptide was treated with
immobilized [beta]3-galactosidase and sialidase to yield a cloned
macrophage activating factor, designated as CdMAF1.
[0069] II. Cloning Procedure II: Fusion Vector.
[0070] 1) Cloning the Domain Responsible for Macrophage
Activation (CdMAF)
[0071] A baculovirus fusion vector, pPbac vector (Stratagene, La
Jolla, Calif.), contains human placental alkaline phosphatase
secretory signal sequences that direct the nascent cloned peptide
chain toward the secretory pathway of the cells leading to
secretion into culture media. The signal sequence is cleaved off
by signal-sequence peptidase as the nascent cloned peptide is
channeled toward the secretory pathway of the host insect cells
leading to secretion of the cloned domain (Cd) peptide. FIG. 6
depicts that the vector carries the stuffer fragment for gene
substitution and lacZ gene for identification of the gene
insertion.
[0072] The stuffer fragment of pPbac vector was excised by
digesting the vector DNA with restriction enzymes Smal and BamHI
and was removed by electroelution. The E4 cDNA fragment of the Gc
protein was digested with HaeIII and BamHI, yielding a fragment
practically the same as E4hl (see FIG. 4). This fragment was mixed
with the above pPbac vector and ligated with T4 ligase. This
strategy not only fixes the orientation of ligation but also fuses
the fragment with the reading frame. The E. coli DH5aF' cells were
transformed with the reaction mixture. The cloned DNA insert was
isolated from a number of colonies after digestion with HaeIII and
BamHI. The insert was confirmed by sequencing. The sequence
confirmed the correct orientation.
[0073] 2) Isolation of Recombinant Baculovirus by Transfection
of Sf9 Insect Cells with Wild Type Baculovirus and the Cloned
DNA Insert.
[0074] For transfection of insect cells (Spodoptera frugiperda,
Sf9), linear wild type (AcMNPV) baculoviral DNA and insectin
liposomes (Invitrogen, San Diego, Calif.) have been used.
Liposome-mediated transfection of insect cells is the most
efficient transfection method available. For transfection to a
monolayer of Sf9 cells (2*10<6>) in a 60 mm dish a mixture
of the following was gently added:
[0075] 3 [mu]g cloned plasmid DNA
[0076] 10 [mu]l linear wild type baculovirus (AcMNPV) DNA (0.1
[mu]g/[mu]l)
[0077] 1 ml medium
[0078] 29 [mu]l insectin liposomes
[0079] The dishes were incubated at room temperature for 4 hours
with slow rocking. After transfection, the 1 ml of medium was
added and incubated at 27[deg.] C. in a humidified environment for
48 hours. The resultant transfection lysate was plaque assayed.
Purification of recombinant virus, isolation of the cloned domain
peptide (Cd) and enzymatic generation of the cloned macrophage
activating factor designated CdMAF2 were described in the Cloning
Procedure I. This CdMAF is composed of 94 amino acid residues as
shown in FIG. 7, including 9 amino acids from the fusion vector
and is referred to herein as CdMAF2. Although CdMAF2 has five
amino acids more than the CdMAF1 peptide derived from the
non-fusion vector, they exhibited the same biological activities.
Supporting Observations
[0080] A. Effects of Cloned Macrophage Activating Factors,
GcMAFc and CdMAF on Cultured Phagocytes (Macrophages and
Osteoclasts).
[0081] The three hour treatment of human macrophages and
osteoclasts with picogram quantities (pg) of the cloned macrophage
activating factors, GcMAFc and CdMAF, resulted in a greatly
enhanced superoxide generating capacity of the phagocytes as shown
in Table 1. The levels of the phagocyte activation are similar to
those of macrophage activation by GcMAF (Yamamoto et al., AIDS
Res. Human Ret. 11:1373, 1995).
TABLE 1
Activation of phagocytes by in vitro treatment with GcMAF and
its cloned derivatives.
nmole of superoxide produced/min/10<6 >phagocytes
Conc. Mouse peritoneal Human
pg/ml Human macrophages* macrophages osteoclasts
GcMAFc
0 0.07 0.06 0.03
10 3.20 3.46 2.56
100 5.18 5.08 4.22
CdMAF
0 0.01 0.02 0.08
10 2.96 2.87 2.43
100 4.26 4.53 4.09
*Peripheral blood monocytes/macrophages of cancer patients.
Similar results were also observed when those of HIV-infected
patients were used.
[0082] B. Activation of Mouse Peritoneal Macrophages by
Administration of Cloned Macrophage Activating Factors, GcMAFc and
CdMAF.
[0083] One day post-administration of a picogram quantity (10 and
100 pg/mouse) of GcMAFc or CdMAF to BALB/c mice, peritoneal
macrophages were isolated and assayed for superoxide generating
capacity. As shown in Table 2, the macrophages were efficiently
activated. These results are similar to those of macrophage
activation with GcMAF (Naraparaju and Yamamoto, Immunol. Lett.
43:143, 1994; Yamamoto et al., AIDS Res. Human Ret. 11:1373,1995).
TABLE 2
Activation of mouse peritoneal macrophages by administration of
cloned GcMAF derivatives.
Dosage nmole of superoxide produced/min/10<6
>phagocytes
pg/mouse Mouse peritoneal
macrophages
0 0.05
10 3.18
100 5.23
CdMAF
0 0.03
10 2.54
100 4.23
[0084] C. Therapeutic Effects of GcMAF, GcMAFc or CdMAF on Tumor
Bearing Mice and Osteopetrotic Mice.
[0085] 1) Therapeutic Effects of GcMAF, GcMAFc or CdMAF on Ehrlich
Ascites Tumor Bearing Mice.
[0086] When BALB/c mice were administered with GcMAF, GcMAFc or
CdMAF (100 pg/mouse) and received 10<5 >Ehrlich ascites
tumor cells/mouse, they survived for at least 5 weeks. All the
control mice received only the ascites tumor and died in
approximately 14 days. When mice were administered with an
additional 100 pg GcMAF/mouse 4 days post-transplantation, the
tumor cells were completely eliminated (Table 3).
[0087] When mice were transplanted with 10<5 >Ehrlich
ascites tumor cells/mouse and treated twice with GcMAF, GcMAFc or
CdMAF (100 pg/mouse) at 4 days and 8 days post-transplantation,
all treated mouse groups survived over 65 days while the untreated
8 mouse groups all died at approximately 13 days (Groups 4 through
9 of Table 3).
TABLE 3
Therapeutic effects of GcMAF and cloned derivatives on mice
bearing Ehrlich ascites tumor.
No. of Post-transplantation No. of mice
Group mice treatment survived/period
Group 1.
6 mice untreated control 6 mice/13 ± 3 days
10 mice day 0 100 pg GcMAF/mouse 10 mice/36 ± 7 days
Group 2.
6 mice untreated control 6 mice/14 ± 4 days
10 mice day 0 100 pg GcMAFc/mouse 10 mice/35 ± 6 days
Group 3.
6 mice untreated control 6 mice/14 ± 5 days
10 mice day 0 100 pg CdMAF/mouse 10 mice/34 ± 3 days
Group 4.
8 mice untreated control 8 mice/15 ± 5 days
12 mice day 0 100 pg GcMAF/mouse
day 4 100 pg GcMAF/mouse 12 mice/>65 days
Group 5.
8 mice untreated control 8 mice/14 ± 5 days
12 mice day 0 100 pg GcMAFc/mouse
day 4 100 pg
GcMAFc/mouse 12 mice/>65 days
Group 6.
8 mice untreated
control 8 mice/14 ± 5 days
12 mice day 0 100 pg CdMAF/mouse
day 4 100 pg
CdMAF/mouse 12 mice/>65 days
Group 7.
8 mice untreated
control 8 mice/14 ± 4 days
8 mice day 4 100 pg
GcMAF/mouse
day 8 100 pg
GcMAF/mouse 8 mice/>65 days
Group 8.
8 mice untreated
control 8 mice/13 ± 3 days
8 mice day 4 100 pg
GcMAFc/mouse
day 8 100 pg
GcMAFc/mouse 8 mice/>65 days
Group 9.
8 mice untreated
control 8 mice/13 ± 5 days
8 mice day 4 100 pg
CdMAF/mouse
day 8 100 pg
CdMAF/mouse 8 mice/>65 days
[0088] With respect to the results of Table 3, GcMAF was
administered intraperitoneally for Groups 1 through 6, and GcMAF
was administered intramuscularly (systemically) for Groups 7
through 9; mice in all groups received 105 tumor cells/mouse.
[0089] 2) Therapeutic Effects of GcMAF and Cloned GcMAF
Derivatives (GcMAFc and CdMAF) On Osteopetrotic Mice.
[0090] Administration of GcMAFc or CdMAF to new born litters of
osteopetrotic op/op mouse was performed by the weekly injection of
100 picograms for four weeks beginning from a day after birth.
Mice were sacrificed at 28 days. The tibiae were removed from the
treated and untreated control mice, longitudinally bisected, and
examined under a dissecting microscope to measure the size of the
bone marrow cavity. The cavity size was expressed as a percentage
of the distance between the epiphyseal plates of the tibia. The
untreated mouse group formed bone marrow with 30% of the total
length of tibia. The treated mouse group experienced a 20%
increased bone marrow formation over that of the untreated mouse
group. This increased bone marrow cavity formation is an
indication of osteoclast activation and increased osteoclastic
bone resorption.
[0091] D. Therapeutic Effects of GcMAF, GcMAFc and CdMAF on
Human Cancer and Virus Infected Patients.
[0092] 1. Cancer Patients: Therapeutic Effect of GcMAF on
Prostate, Breast and Colon Cancer and Adult Leukemia Patients.
[0093] The administration of GcMAF (100 and 500 ng/human) to
healthy volunteers resulted in the greatly enhanced activation of
macrophages as measured by the 7-fold enhanced phagocytic capacity
and the 15-fold superoxide generating capacity of macrophages. The
administration of GcMAF showed no signs of any side effects to the
recipients. Administration of various doses (100 pg to 10
ng/mouse) to a number of mice produced neither ill effects nor
histological changes in various organs including liver, lung,
kidney, spleen, brain, etc. When patients with various types of
cancer were treated with GcMAF (100 ng/week), remarkable curative
effects on various types of cancer were observed. The therapeutic
efficacy of GcMAF on patients bearing various types of cancers was
assessed by tumor specific serum [alpha]-N-acetylgalactosaminidase
activity because the serum enzyme level is proportional to the
total amount of cancerous cells (tumor burden). Curative effects
of GcMAF on prostate, breast and colon cancer and leukemia are
illustrated in FIGS. 8A to 8D. After 25 weekly administrations of
100 ng GcMAF the majority (>90%) of prostate and breast cancer
patients exhibited insignificantly low levels of the serum enzyme.
A similar result was also observed after 35 GcMAF administrations
to colon cancer patients. Similar curative effects of GcMAF on
lung, liver, stomach, brain, bladder, kidney, uterus, ovarian,
larynx, esophagus, oral and skin cancers were observed. Thus,
GcMAF appeared to be effective on a variety of cancers
indiscriminately. However, GcMAF showed no evidence of side
effects in patients after more than 6 months of therapy. This was
also confirmed by blood cell counts profile, liver and kidney
functions, etc.
[0094] When GcMAFc (100 ng/week) and CdMAF (100 ng/week) were
administered to two prostate cancer patients each, curative
effects similar to those of GcMAF were observed.
[0095] 2. Virus Infected Patients
[0096] Treatment of peripheral blood macrophages of
HIV-infected/AIDS patients with 100 pg GcMAF/ml resulted in a
greatly enhanced macrophage activation (Yamamoto et al., AIDS Res.
Human Ret. 11:1373, 1995). HIV-infected patients carry anti-HIV
antibodies. HIV-infected cells express the viral antigens on the
cell surface. Thus, macrophages have a potential to eliminate the
infected cells via Fc-receptor mediated cell-killing/ingestion
when activated.
[0097] Similarly, treatment of peripheral blood macrophages of
patents chronically infected with Epstein-Barr virus (EBV) and
with herpes zoster with 100 ng GcMAF/ml resulted in a greatly
enhanced macrophage activation. Like HIV, EBV infects lymphocytes
(B cells). Since these enveloped viruses code for
[alpha]-N-acetylgalactosaminidase and infected cells secrete it
into blood stream. Thus this enzyme activity in patient sera can
be used as a prognostic index during therapy. After approximately
25 administrations of GcMAF (100 ng/week) to patients chronically
infected with EBV and with herpes zoster, the enzyme activity
decreased to that of healthy control levels. When GcMAFc (100
ng/week) and CdMAF (100 ng/week) were administered to EBV-infected
patients, curative effects similar to those of GcMAF were
observed.
[0098] E. Adjuvant Activities of GcMAF, GcMAFc and CdMAF for
Immunization and Vaccinations.
[0099] 1. Rapid Increase of the Number of Antibody
Secreting Cells (PFC) in Mice after Administration of GcMAF and
Sheep Erythrocytes.
[0100] BALB/c mice were inoculated with SRBC 6 hours after the
intraperitoneal administration of 50 pg GcMAF/mouse. At various
intervals (1-5 days) after immunization, IgM-antibody secreting
cells in the spleen were determined using the Jerne plaque assay
(Jerne et al., Cell-bound antibodies, Wistar Institute Press,
1963). One day post-administration of GcMAF and SRBC produced
1.35*10<4 >PFC/spleen. Two days after administration of
GcMAF and SRBC, the number of antibody secreting cells had
increased to 8.2*10<4 >PFC/spleen. By the 4th day the number
of antibody secreting cells reached the maximal level (about
23.6*10<4 >PFC/spleen), as shown in Table 4. In contrast,
mice that received an injection of SRBC alone produced about
3.8*10<4 >PFC/spleen, 4 days after SRBC-injection.
[0101] To ascertain the dose response, mice were injected with
SRBC 6 hours after administration of various doses of GcMAF
ranging from 1 to 50 pg/mouse. On the 4th day post-administration
of GcMAF and SRBC, the number of antibody secreting cells per
spleen was determined by the Jerne plaque assay. On the 4th day
post-administration there was a commensurate increase in the
number of plaque forming cells as the concentration of GcMAF was
increased above 1 pg per mouse. At a GcMAF dose of 5, 10 and 50
pg/mouse, I detected 12.6*10<4>, 20.2*10<4 >and
24.3*10<4 >PFC/spleen, respectively.
TABLE 4
Time course studies on development of cells secreting antibody
against sheep erythrocytes (SRBC) in BALB/c mice after
administration of GcMAF and SRBC
After SRBC
immunization Antibody secreting cells/spleen (*10<4>)
(days) SRBC only GcMAF + SRBC
1 0.01 ± 0.002 1.35 ± 0.21
2 0.08 ± 0.02 8.28 ± 1.26
3 1.18 ± 0.42 14.42 ± 2.32
4 3.86 ± 0.95 23.68 ± 6.05
5 2.15 ± 0.63 18.63 ± 3.43
<a>Mice were inoculated with SRBC (10<8 >cells)
6 hr after administration of GcMAF (50 pg/mouse). The number of
plaques (IgM secreting cells) was quantified microscopically on
various days post-SRBC injection. The number of plaque-forming
cells (PFC) per spleen is expressed as the mean value of
triplicate assays ± SEM.
[0102] Without further elaboration the foregoing will so fully
illustrate my invention that others may, by applying current or
future knowledge, adapt the same for use under various conditions
of service.
REFERENCES CITED
[0103] The following references are cited and their entire text is
incorporated fully herein as are all references set forth above in
the specification.
U.S. Patent Documents
[0104] U.S. Pat. Nos. 5,177,001, 5,177,002 and 5,326,749
(Yamamoto).
Other Publications
[0105] 1. Jerne, N. K., Nordin, A. A. and Henry, C., The agar
plaque technique for recognizing antibody producing cells. In Amos
and Koprowski (eds). Cell-bound Antibody. Wistar Institute Press,
Philadelphia, PA (1963).
[0106] 2. Sato, M., Tanaka, H., Yamada, T. and Yamamoto, N.,
Persistent infection of BHK/WI-2 cells with rubella virus and
characterization of rubella variants. Arch. Virology 54:333-343
(1977).
[0107] 3. Reeves, J. D., August, C. S., Humbert, J. R., Weston, W.
L, Host defense in infantile osteopetrosis. Pediatrics.
64:202-(1979).
[0108] 4. Marks, S. C., Jr., Congenital osteopetrotic mutations as
probes of the origin, structure and function of osteoclasts. Clin.
Orthop. 189:239-(1984).
[0109] 5. Carbonell, L. F., Klowden, M. J. and Miller, L. K.,
Baculovirus-mediated expression of bacterial genes in dipteran and
mammalian cells. J. Virol. 56:153-160 (1985).
[0110] 6. Ngwenya, B. Z., and Yamamoto, N., Activation of
peritoneal macrophages by lysophosphatidylcholine. Biochem.
Biophys. Acta 839: 9-15 (1985).
[0111] 7. Cooke, N. E. and Haddad, J. G., Vitamin D binding
protein (Gc-globulin). Endocrine Rev. 10:294-307 (1989).
[0112] 8. Marks, S. C., Jr., Osteoclast biology: Lessons from
mammalian mutations. Am. J. Med. Genet. 34:43-54 (1989).
[0113] 9. Ngwenya, B. Z. and Yamamoto, N., Contribution of
lysophosphatidylcholine treated nonadherent cells to mechanism of
macrophage stimulation. Proc. Soc. Exp. Biol. Med. 193:118-124
(1990).
[0114] 10. Ausubel, F. A., Brent, R., Kingston, R. E., Moore, D.
D., Seidman, J. G., Smith, J. A. and Struhl, K. (eds.), Expression
of proteins in insect cells using baculoviral vectors. Current
Protocols in Molecular Biology. Sections 16.8.1-16.11.7. Greene
Publishing and Wiley-Interscience, New York (1990).
[0115] 11. Yagi, F., Eckhardt, A. E. and Goldstein I. J.,
Glycosidases of Ehrlich ascites tumor cells and ascitic
fluid-purification and substrate specificity of
[alpha]-N-acetylgalactosaminidase and [alpha]-galactosidase:
comparison with coffee bean [alpha]-galactosidase. Arch. Biochem.
Biophys. 280:61-67 (1990).
[0116] 12. Yamamoto, N. and Homma, S., Vitamin D3 binding protein
(group-specific component, Gc) is a precursor for the macrophage
activating signal from lysophosphatidylcholine-treated
lymphocytes. Proc. Natl. Acad. Sci. USA. 88:8539-8543 (1991).
[0117] 13. Cooke, N. E. and David, E. V., Serum vitamin D-binding
protein is a third member of the albumin and alpha-fetoprotein
gene family. J. Clin. Invest. 76:2420-2424 (1985).
[0118] 14. Haddad, J. G., Hu, Y. Z., Kowalski, M. A., Laramore,
C., Ray, K., Robzyk, P. and Cooke, N. E., Identification of the
sterol- and actin-binding domains of plasma vitamin D binding
protein (Gc-globulin). Biochemistry 31:71747181 (1992).
[0119] 15. Yamamoto, N. and Kumashiro, R., Conversion of vitamin
D3 binding protein (Group-specific component) to a macrophage
activating factor by stepwise action of [beta]-galactosidase of B
cells and sialidase of T cells. J. Immunol. 151:27-94-2902 (1993).
[0120] 16. Homma, S., Yamamoto, M. and Yamamoto, N., Vitamin D
binding protein (group-specific component, Gc) is the sole serum
protein required for macrophage activation after treatment of
peritoneal cells with lysophosphatidylcholine. Immunol. Cell Biol.
71:249-257 (1993).
[0121] 17. Yamamoto, N., Kumashiro, R., Yamamoto, M., Willett, N.
P. and Undsay, D. D., Regulation of inflammation-primed activation
of macrophages by two serum factors, vitamin D3-binding protein
and albumin. Inf. Imm. 61:5388-5391 (1993).
[0122] 18. Yamamoto, N., Lindsay, D. D., Naraparaju, V. R.,
Irelalnd, R. A. and Popoff, S. M., A defect in the
inflammation-primed macrophage activation cascade in osteopetrotic
(op) rats. J. Immunol. 152:5100-5107 (1994).
[0123] 19. Yamamoto, N., Willett, N. P. and Lindsay, D. D.,
Participation of serum proteins in the inflammation-primed
activation of macrophages. Inflammation. 18:311-322 (1994).
[0124] 20. Naraparaju, V. R. and Yamamoto, N., Roles of
[beta]-galactosidase of B lymphocytes and sialidase of T
lymphocytes in inflammation-primed activation of macrophages.
Immunol. Lett. 43:143-148 (1994).
[0125] 21. Yamamoto, N., Naraparaju, V. R. and Srinivasula, S. M.,
Structural modification of serum vitamin D3-binding protein and
immunosuppression in HIV-infected patients. AIDS Res. Human Ret.
11:1373-1378 (1995).