Love Me Love My Phages -Bacteriophages - The New Antibiotics-Part1
                                                             
                                                 Love Your Phages
                                                    

                                         What Are Phages?

Phages (short form of Bacteriophages) are viruses that destroy many of the bacteria that create terrible harm in humans, animals and many of our wonderful plant species, and even our marvelous coral reefs. With the threat of global warming, there is also the fear that many of these bacteria borne diseases may increase their levels of fatal or disability creating effects.

History (From Wikipedia)

Since ancient times, there have been documented reports of river water having the ability to cure infectious diseases, such as leprosy. In 1896, Ernest Hanbury Hankin reported that something in the waters of the Ganges and Jumna rivers in India had marked antibacterial action against cholera and could pass through a very fine porcelain filter. In 1915, British bacteriologist Frederick Twort, superintendent of the Brown Institution of London, discovered a small agent that infected and killed bacteria. He considered the agent either 1) a stage in the life cycle of the bacteria, 2) an enzyme produced by the bacteria themselves or 3) a virus that grew on and destroyed the bacteria. Twort's work was interrupted by the onset of World War I and shortage of funding. Independently, French-Canadian microbiologist Félix d'Hérelle, working at the Pasteur Institute in Paris, announced on September 3, 1917 that he had discovered "an invisible, antagonistic microbe of the dysentery bacillus".

 For d’Hérelle, there was no question as to the nature of his discovery: "In a flash I had understood: what caused my clear spots was in fact an invisible microbe ... a virus parasitic on bacteria." D'Hérelle called the virus a bacteriophage or bacteria-eater (from the Greek phagein meaning to eat). He also recorded a dramatic account of a man suffering from dysentery who was restored to good health by the bacteriophages. In 1926 in the Pulitzer-prize winning novel Arrowsmith, Sinclair Lewis fictionalized the application of bacteriophages as a therapeutic agent. Also in the 1920s the Eliava Institute was opened in TbilisiGeorgia to research this new science and put it into practice. In 2006 the UK Ministry of Defence took responsibility for a G8-funded Global Partnership Priority Eliava Project as a retrospective study to explore the potential of bacteriophages for the 21st century. " (End of Wikipedia material)

The History of the Eliava Institute:

Eliava Institute itself is a fascinating story that I will leave for another article, but briefly The Institute was opened in TbilisiGeorgia in 1923, and was a bacteriology laboratory. Its founder, Prof. George Eliava, was not aware of bacteriophages until 1926. In that year he met Felix d'Herelle during a visit to the Pasteur Institute in Paris. There, Eliava was enthusiastic about the potential of phage in the curing of bacterial disease, and invited d'Herelle to visit his laboratory in Georgia. Sadly Professor Eliava was executed by the Stalin Regime, but his works lives on today and bacteriophage samples are supplied to many countries, including Australia.
Phage Therapy
(From Wikipedia) "Phages were discovered to be anti-bacterial agents and put to use as such soon after they were discovered, with varying success. However, antibiotics were discovered some years later and marketed widely, popular because of their broad spectrum; also easier to manufacture in bulk, store and prescribe. Hence development of phage therapy was largely abandoned in the West, but continued throughout 1940s in the former Soviet Union for treating bacterial infections, with widespread use including the soldiers in the Red Army - much of the literature being in Russian or Georgian, and unavailable for many years in the West. This has continued after the war, with widespread use continuing in Georgia and elsewhere in Eastern Europe. There is anecdotal evidence there, but no completed clinical trials in the US or Western Europe."
"Phage therapy has been attempted for the treatment of a variety of bacterial infections including: laryngitis, skin infections, dysentery, conjunctivitis, periodontitis, gingivitis, sinusitis, urinary tract infections and intestinal infections, burns, boils, etc.[2] - also poly-microbial biofilms on chronic wounds, ulcers and infected surgical sites." (End of Wikipedia)
 Where Do We Go From Here?
Hopefully, a very long way. As the effectiveness of current antibiotics are rapidly reduced, death rates will rise. (In Australian Hospitals, the death rate from "Golden Staph" are around 20% and rising. There is a form of virulent TB that has a death rate of 85%, which is frightening, but both are treatable with Phage Therapy. Exciting research continues.

 In GeorgiaPolandRussiaIsraelAustraliaUKFrance and other countries in Europe and the United States, just to name a few.
Gradually, I will pass on what I am learning, and we can all make sure that our Doctors, Nurses and other Health Specialists and Ministers of Health, and Hospitals will react quickly and save hundreds of thousands of lives.
Sadly, the 1,200 deaths from Cholera, recently in ZIMBABWE, were largely preventable., as it has been known for a very long time that Phage Therapy is very successful with this disease..
This Blog Site is dedicated to those who lost their lives in that epidemic and as a reminder that we must act as soon as we can, to especially insure that those suffering illnesses than can be prevented or treated by Phage Therapy, get this as soon as possible. And above all, in the Developing World, where antibiotics are often too expensive.
                                       ............................................................
Love Me Love My Phages




 George Eliava Institute -Tbilisi, Georgia



(From Wikipedia, the free encyclopedia)



“The Tbilisi Institute, now called the George Eliava Institute of Bacteriophage, Microbiology and Virology (IBMV) has been active since the 1930s in the field of phage therapy, which is used to combat microbial infection (cf. antibiotic-resistant strains).”


History



“The Institute was opened in Tbilisi, Georgia in 1923, and was a bacteriology laboratory. Its founder, Prof. George Eliava, was not aware of bacteriophages until 1926. In that year he met Felix d'Herelle during a visit to the Pasteur Institute in Paris. There, Eliava was enthusiastic about the potential of phage in the curing of bacterial disease, and invited d'Herelle to visit his laboratory in Georgia.”



“D'Herelle visited Tbilisi twice in 1934-35, and agreed to work with Prof. Eliava. It has been suggested the d'Herelle became enamoured of the communist idea. In 1934, Stalin invited d'Herelle to the Institute in Tbilisi; he accepted and worked there part-time for about a year - and even dedicated one of his books, "The Bacteriophage and the Phenomenon of Recovery," written and published in Tbilisi in 1935, to Stalin.”



“D'Herelle had planned to take up permanent residence in Tbilisi and had started to build a cottage on the grounds of the Institute (it would later house the KGB's Georgian headquarters).”



“However, the collaboration between the two scientists was not to be. Around the time d'Herelle was to take up residence, George Eliava became involved romantically with the woman that the head of Stalin's secret police, Lavrenti Beria, was also in love with. Eliava's fate was decided. He was executed and denounced as a supposed enemy of the people. D'Herelle fled from Tbilisi and, some believe, never returned. Another account states that he was in Paris at the time of Eliava's execution, and decided not to return. D'Herelle's book was also banned from distribution.”



“In spite of this development, the Institute did not change its practical specialization, and continued its activity in the field of bacteriophage research. In 1938, the Institute of Bacteriophage Research and the Institute of Microbiology & Epidemiology (founded separately in 1936) merged, and the Institute of Microbiology, Epidemiology and Bacteriophage was formed. It existed until 1951 and was authorized by the People's Commissary of Health of Georgia. After 1951, it came under the auspices of the All-Union Ministry of Health and was renamed The Institute of Vaccine and Sera.”



“Since its inception, the Institute was composed of a combination of industrial and scientific (research) departments. In 1988 the Institute was rearranged again and emerged as the Scientific Industrial Union "Bacteriophage" (SIU "Bacteriophage"). Around that time, its scientific portion was renamed the George Eliava Research Institute of Bacteriophage.”



“Based on the original intentions of D'Herelle and Eliava, the Bacteriophage Institute retained its leadership among other institutes of similar profile over the years. Teimuraz Chanishvili was the leader of the scientific part of the Institute for over 30 years, until his death in August 2007. His niece, Nino Chanishvili has since taken over operations.”


The institute behind the Iron Curtain



“The Institute in Tbilisi became a general Soviet institute for the development and production of bacteriophage drugs. Patients with serious infectious diseases came from all over the Soviet Union to receive treatments there, which were reportedly successful. Bacteriophages became a routine part of treatment in clinics and hospitals. Ointments for the skin, and pills, drops, and rinses consisting of phages were sold and are still sold at pharmacies throughout Eastern Europe at incredibly low prices.” “These therapies are very effective, completely harmless to humans, and are much cheaper than antibiotics. Further, much of this therapy is apparently available without a doctor's prescription.”



“As the world is well aware, the Soviet Union fell apart within 2 years after the fall of the Berlin Wall in 1989. In 1991, after the Republic of Georgia declined to join the Russian Federation and civil war broke out there, the Tbilisi facility was essentially ruined. The Eliava Institute's facilities were damaged and decades of research on bacteriophage nearly went down the drain. Thousands of bacteriophage samples identified over the years and catalogued in huge, refrigerated "libraries" suffered irreversible damage due to frequent electrical outages. Apparently, the Russians transferred some of the equipment to their territory and built plants for the production of bacteriophages in other locations. Clearly, they recognized the importance of the research and also that of continued bacteriophage therapy. The situation at the Eliava Institute continued to deteriorate until it was on the verge of closure.”


However, in 1997, a report on the Institute was broadcast by the BBC, sparking a flurry of media interest in the West. The headlines drew doctors and scientists to Tbilisi - and also, most importantly, energetic entrepreneurs from around the world who were determined to help save the Institute and its stocks and fully explore the potential of this "new" and highly effective therapy. Georgian scientists whose names were connected in some way to the Institute saw great opportunity, and some of them emigrated to the West to be part of joint projects. Some of the Institute's projects with the rest of the world can be seen on the website of the Georgian Academy of Sciences, the umbrella entity which now includes the Eliava Institute. The URL is http://www.eliava-institute.org/


Address



“The George Eliava Institute is located at:

3, Gotua Street

380060, Tbilisi

Georgia



Tel: +995 32 37 42 27 or +995 32 23 32 95 Fax: +995 32 99 91 53 or +995 32 22 19 6


External links



A few of the companies who are or have been connected to the Eliava Institute and/or its phage research are as follows:



















References


Nature Publishing Group: www.nature.com/naturebiotechnology. Volume 22, No. 1, Jan. 2004, Old Dogma, New Tricks - 21st Century Phage Therapy, by Karl Thiel.



“Proc. Natl. Acad. Sci. USA: Vol. 93, pp. 3167-3168, April 1996, Smaller fleas...ad infinitum: Therapeutic Bacteriophage Redux, by Joshua Lederberg, Raymond and Beverly Sackler Foundation Scholar at the Rockefeller University, 1230 York Avenue, New York, NY 10021.

The bactericidal action of the waters of the Jamuna and Ganga rivers on Cholera microbes, By M.E Hankin, Government Laboratory, Agra, India. Translated from the original article published in French, Ref. Ann. De l’ Inst. Pasteur 10.511 (1896).”


(END OF WIKIPEDIA MATERIAL)


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


Phage Therapy



Phage Therapy involves carefully chose and tested species of Bacteriophages, which are matched to the specific bacteria, which is causing the infection, inflammation, swelling or producing toxins, and causing other ill-effects.



Phage Therapy (which will be covered in succeeding pages over time), like antibiotics, can only tackle infections due to bacteria and not viruses. But when infected by a virus infection, we may become prone to deadly bacterial infections, which may actually kill us, rather than the virus. Secondary infections treated by Phage Therapy, can be reduced or eliminated, and allow our immune system to fight the virus.



Sadly, too often, people are killed or rendered disabled or even become chronically ill, because of the “double whammy” of a virus infection occurring, and then the person is hit by a serious bacterial infection. Our bodies have wonderful defence systems, but fighting a war on two or more fronts at the same time, may overwhelm us. 



There are many ways that Bacteriophages may help us, apart from direct application via injection, nasal spray or ointment. They can be used in bandages to reduce or prevent infections in burns case, or serious wounds, used to render catheters safe, used in special preparations on hospital staff clothing (effective for 3 weeks) or even used to render the operating room or recovery ward sterile and safe.



I will gradually explain these processes and also rendering food safe for pregnant women, babies or immune compromised people, against infections such as Listeria or e-coli based illnesses, and much, much more.


Love Me Love My Phages

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




  Love Me Love My Phages

Eat Me

The Soviet method for attacking infection that we can learn from.

By Daria Vaisman

Posted Tuesday, May 30, 2006, at 12:41 PM ET

From: Medical Examiner_Health and medicine explained.

(Illustration Above is stylized Bacteriophage)

In the 1920s and '30s, with diseases like dysentery and cholera running rampant, the discovery of bacteriophages was hailed as a breakthrough. Bacteriophages are viruses found virtually everywhere—from soil to seawater to your intestines—that kill specific, infection-causing bacteria. In the United States, the drug company Eli Lilly marketed phages for abscesses and respiratory infections. (Sinclair Lewis' Pulitzer-winning Arrowsmith is about a doctor who uses phages to prevent a diphtheria epidemic.) But by the 1940s, American scientists stopped working with phages for treatment because they no longer had reason to. Penicillin, discovered by the Scottish bacteriologist Alexander Fleming in 1928, had become widely available thanks to synthetic production and zapped infections without the expertise needed for finicky phages.

But now the equation has changed. Many kinds of bacteria have become antibiotic-resistant—prompting a few Western scientists, and patients, to travel to former Soviet Georgia to give bacteriophages for treatment a try. Phages have been used in the former Soviet Union for decades because scientists there had less access to antibiotics than their American and European counterparts did. Phages were a cheap alternative, and in Soviet clinical trials, they repeatedly stopped infections. Now in a bid for medical tourists, Georgia has opened a center in its capital, Tbilisi, which offers outpatient phage treatment to foreigners. In connection with the Eliava phage research institute, which Stalin helped set up in Tbilisi in 1923, the treatment center offers personalized cures for a host of infections the United States says it can no longer do anything about.

In 2000, the Centers for Disease Control, along with other federal agencies, warned that the world might soon return to a "pre-antibiotic era." Two million people each year now get hospital-borne bacterial infections, 1.4 million of them resistant to antibiotics and 90,000 of them lethal. One example is sepsis, the infection that sickened* Joan Didion's daughter, as Didion relates in The Year of Magical Thinking. New antibiotics are being discovered. But it takes 10 years and at least $800 million to bring an antibiotic to market, according to the Infectious Diseases Society of America. The big advantage that phages offer over antibiotics is that bacterial resistance is less of a problem. Unlike antibiotics, new phage batches can quickly be whipped up to take the place of phages to which bacteria become resistant.

The word phage comes from the Greek "to eat." A phage contains genetic material that gets injected into a virus's host. Whereas "bad" viruses infect healthy cells, phages target specific bacteria that then explode. At Eliava, phages are produced as a liquid that can be drunk or injected intravenously, as pills, or as phage-containing patches for wounds. Though few published articles in Western journals report positive clinical trials—most of the recent long-term research on phages comes out of the Soviet Union—some Western scientists say that phages are safe and that they work. "There is no evidence that phage is harmful in any way," says Nick Mann, a biology professor at the University of Warwick in England and co-director of phage R&D company Novolytics.

So, why do American patients need to go to all the way to Georgia for treatment? For starters, in their natural state phages are hard to patent, the route by which drug companies lock up future profits. The first company to spend millions of dollars to prove that a particular phage is safe could allow its competitors to capitalize on the results. As important is the difficulty of regulation. There are two ways that phages are currently used in the former Soviet Union, and both pose problems from the point of view of the Food and Drug Administration. At the Tbilisi phage center, phages are personalized: You send your bacterial sample to the lab, and it's either matched up with an existing phage or a phage is cultured just for you. In the United States, by contrast, drugs are mass produced, which makes it easier for the FDA to regulate them.

Phages are also sold over-the-counter in Georgia. People take the popular mixture piobacteriophage, for example, to fight off common infections including staph and strep. These phage mixtures are updated regularly so they can attack newly emerging bacterial strains. In the United States, the FDA would want the phages in each new concoction to be gene sequenced, because regulations require every component of a drug to be identified. To do so would entail prohibitively expensive and lengthy clinical trials.

In the early years of phage research in the United States, says former National Institutes of Health scientist Carl Merril, bacteriophages allegedly killed more people than they cured. Phages are culled from dirty, wet places—the first was found in the Ganges River—a recipe for infection unless you know what you're doing. And some kinds of phages—called lysogenic phages—are potentially dangerous, because they sometimes carry genes that cause bacteria to release toxins. So, there is reason for caution.

Despite the caveats, a number of phage biotechnology firms have recently opened up in the United States and also in countries like Canada and Israel. Phage biologists point out we know much more about phage biology now than when the viruses were first discovered. Methods of using phages for treatment, from distillation to identification, have improved significantly since then. Clinicians worldwide also report that patients using phages have had good recovery rates and minor or fleeting side effects. Evergreen State College professor Elizabeth Kutter, who collaborates closely with Eliava researchers in Georgia and heads an international phage conference each year, is working with others to find ways to commercialize phages that could sidestep some of the problems with patenting and regulation. Flu vaccine offers one instructive possibility. Like over-the-counter phages, the vaccine is updated regularly with the most recent strain of flu virus—without requiring FDA approval each time.

There are already multiple uses for phages without FDA approval. A promising area is American agriculture and livestock, which is regulated by the less stringent United States Department of Agriculture. Domestic scientists are looking at ways in which phages could kill bacteria before they cause infection (rather than fight an infection after it has begun). Alexander Sulakvelidze, an assistant medical professor at the University of Maryland and a co-founder of the phage R&D company Intralytix, awaits federal approval for a phage-based wash for meat and produce that protects against food poisoning. Vincent Fischetti, a professor at the Rockefeller Institute, is designing a phage-based enzyme solution that can be sprayed into the noses and mouths of hospital and nursing-home patients. Fischetti and researchers in Tbilisi are also experimenting with using phages to detect anthrax and cholera in the case of a terrorist attack.

Using phages to treat infections at home, on the other hand, for the moment seems unlikely. One company recently tried to open a phage center in Tijuana but was deterred by the Mexican government. Phages might be offered someday at clinics on Native American reservations, as a casino-like quirk of legislative autonomy. But for now, U.S. patients at a loss for options may decide that Tbilisi is close enough.

Correction, June 1, 2006: The original sentence stated that Didion's daughter died of sepsis. In fact, she got sick from sepsis but died later from another condition. Click here to return to the corrected sentence.

Love Me Love My Phages


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

Love Me Love My Phages 

Phage Therapy

Phage Therapy
 (From Wikipedia, the free encyclopedia)

Phage Therapy

Phage therapy is the therapeutic use of bacteriophages to treat pathogenic bacterial infections. Although extensively used and developed mainly in former Soviet Union countries for about 90 years, this method of therapy is still being tested elsewhere for treatment of a variety of bacterial and poly-microbial biofilm infections, and has not yet been approved in countries other than Georgia. Phage therapy has many potential applications in human medicine as well as dentistry, veterinary science, and agriculture.[1] If the target host of a phage therapy treatment is not an animal, however, then the term "biocontrol" (as in phage-mediated biocontrol of bacteria) is sometimes employed rather than "phage therapy".

An important theoretical benefit of phage therapy is that bacteriophages can be much more specific than more common drugs, so can be chosen to be harmless to not only the host organism (human, animal, or plant), but also other beneficial bacteria, such as gut flora, reducing the chances of opportunistic infections. They also have a high therapeutic index, that is, phage therapy gives rise to few if any side effects, as opposed to drugs, and does not stress the liver. Because phages replicate in vivo, a smaller effective dose can be used. On the other hand, this specificity is also a disadvantage: A phage will only kill a bacterium if it is a match to the specific strain. Thus, phage mixtures are often applied to improve the chances of success, or samples can be taken and an appropriate phage identified and grown.

Phages are currently being used therapeutically to treat bacterial infections that do not respond to conventional antibiotics, particularly in the country of Georgia.[2][3][4] They tend to be more successful than antibiotics where there is a biofilm covered by a polysaccharide layer, which antibiotics typically cannot penetrate.[citation needed] In the West, no therapies are currently authorized for use on humans, although phages for killing food poisoning bacteria (Listeria) are now in use.[5]

History

Following the discovery of bacteriophages by Frederick Twort and Felix d'Hérelle[6] in 1915 and 1917, phage therapy was immediately recognized by many to be a key way forward for the eradication of bacterial infections. A Georgian, George Eliava, was making similar discoveries. He travelled to the Pasteur Institute in Paris where he met d'Hérelle, and in 1926 he founded the Eliava Institute in Tbilisi, Georgia, devoted to the development of phage therapy.
In neighbouring countries including Russia, extensive research and development soon began in this field. In the USA during the 1940s, commercialization of phage therapy was undertaken by the large pharmaceutical company, Eli Lilly.

Whilst knowledge was being accumulated regarding the biology of phages and how to use phage cocktails correctly, early uses of phage therapy were often unreliable. When antibiotics were discovered in 1941 and marketed widely in the USA and Europe, Western scientists mostly lost interest in further use and study of phage therapy for some time.[7]

Isolated from Western advances in antibiotic production in the 1940s, Russian scientists continued to develop already successful phage therapy to treat the wounds of soldiers in field hospitals. During World War II, the Soviet Union used bacteriophages to treat many soldiers infected with various bacterial diseases e.g. dysentery and gangrene. The success rate was as good as, if not better than any antibiotic.[citation needed] Russian researchers continued to develop and to refine their treatments and to publish their research and results. However, due to the scientific barriers of the Cold War, this knowledge was not translated and did not proliferate across the world.[8][9]

There is an extensive library and research center at the Eliava Institute in Tbilisi, Georgia. Phage therapy is today a widespread form of treatment in neighbouring countries. For 80 years Georgian doctors have been treating local people, including babies and newborns, with phages.

As a result of the development of antibiotic resistance since the 1950s and an advancement of scientific knowledge, there is renewed interest worldwide in the ability of phage therapy to eradicate bacterial infections and chronic polymicrobial biofilm, along with other strategies.
Phages have been explored as means to eliminate pathogens like Campylobacter in raw food[10] and Listeria in fresh food or to reduce food spoilage bacteria.[11] In agricultural practice phages were used to fight pathogens like Campylobacter, Escherichia and Salmonella in farm animals, Lactococcus and Vibrio pathogens in fish from aquaculture and Erwinia and Xanthomonas in plants of agricultural importance. The oldest use was, however, in human medicine. Phages were used against diarrheal diseases caused by E. coli, Shigella or Vibrio and against wound infections caused by facultative pathogens of the skin like staphylococci and streptococci. Recently the phage therapy approach has been applied to systemic and even intracellular infections and the addition of non-replicating phage and isolated phage enzymes like lysins to the antimicrobial arsenal. However, definitive proof for the efficiency of these phage approaches in the field or the hospital is only provided in a few cases.[11]

Some of the interest in the West can be traced back to 1994, when Soothill demonstrated (in an animal model) that the use of phages could improve the success of skin grafts by reducing the underlying Pseudomonas aeruginosa infection.[12] Recent studies have provided additional support for these findings.[13]

Recently, the use of phages as delivery mechanisms for traditional antibiotics has been proposed.[14][15] The use of phages to deliver antitumor agents has also been described, in preliminary in vitro experiments for cells in tissue culture.[16]





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


Potential Benefits

A potential benefit of phage therapy is freedom from the severe adverse effects of antibiotics. Also it would possibly be fast-acting, once the exact bacteria are identified and the phages administered. Another benefit of phage therapy is that although bacteria are able to develop resistance to phages the resistance might be easier to overcome.

Bacteriophages are often very specific, targeting only one or a few strains of bacteria.[17] Traditional antibiotics usually have more wide-ranging effect, killing both harmful bacteria and useful bacteria such as those facilitating food digestion. The specificity of bacteriophages might reduce the chance that useful bacteria are killed when fighting an infection.

Increasing evidence shows the ability of phages to travel to a required site — including the brain, where the blood brain barrier can be crossed — and multiply in the presence of an appropriate bacterial host, to combat infections such as meningitis. However the patient's immune system can, in some cases mount an immune response to the phage (2 out of 44 patients in a Polish trial[18]). This might possibly be therapeutically significant.

Development and production is faster than antibiotics, on condition that the required recognition molecules are known.[citation needed]

Research groups in the West are engineering a broader spectrum phage and also target MRSA treatments in a variety of forms - including impregnated wound dressings, preventative treatment for burn victims, phage-impregnated sutures. Enzobiotics are a new development at Rockefeller University that create enzymes from phage. These show potential for preventing secondary bacterial infections e.g. pneumonia developing with patients suffering from flu, otitis etc..[citation needed]

Some bacteria such as multiply resistant Klebsiella pneumoniae have no non toxic antibiotics available, and yet killing of the bacteria via intraperitoneal, intravenous or intranasal of phages in vivo has been shown to work in laboratory tests.[19]

Application

Collection

In its simplest form, phage treatment works by collecting local samples of water likely to contain high quantities of bacteria and bacteriophages, for example effluent outlets, sewage and other sources.[2] They can also be extracted from corpses. The samples are taken and applied to the bacteria that are to be destroyed which have been cultured on growth medium.
The bacteria usually die, and the mixture is centrifuged. The phages collect on the top of the mixture and can be drawn off.

The phage solutions are then tested to see which ones show growth suppression effects (lysogeny) and/or destruction (lysis) of the target bacteria. The phage showing lysis are then amplified on cultures of the target bacteria, passed through a filter to remove all but the phages, then distributed.

Treatment

Phages are "bacterium specific" and it is therefore necessary in many cases to take a swab from the patient and culture it prior to treatment. Occasionally, isolation of therapeutic phages can typically require a few months to complete, but clinics generally keep supplies of phage cocktails for the most common bacterial strains in a geographical area.

Phages in practice are applied orally, topically on infected wounds or spread onto surfaces, or used during surgical procedures. Injection is rarely used, avoiding any risks of trace chemical contaminants that may be present from the bacteria amplification stage,and recognizing that the immune system naturally fights against viruses introduced into the bloodstream or lymphatic system.

The direct human use of phage might possibly be safe; suggestively, in August 2006, the United States Food and Drug Administration approved spraying meat with phages. Although this initially raised concerns since without mandatory labeling consumers won't be aware that meat and poultry products have been treated with the spray,[20] it confirms to the public that, for example, phages against Listeria are generally recognized as safe (GRAS status) within the worldwide scientific community and opens the way for other phages to also be recognized as having GRAS status.
Phage therapy has been attempted for the treatment of a variety of bacterial infections including: laryngitis, skin infections, dysentery, conjunctivitis, periodontitis, gingivitis, sinusitis, urinary tract infections and intestinal infections, burns, boils, etc.[2] - also poly-microbial biofilms on chronic wounds, ulcers and infected surgical sites.[citation needed]

In 2007, Phase 2a clinical trials have been reported at the Royal National Throat, Nose and Ear Hospital, London for Pseudomonas aeruginosa infections (otitis).[21].[22][23] Documentation of the Phase-1 and Phase-2a study is not available at present.

Phase 1 clinical trials are underway in the South West Regional Wound Care Center, Lubbock, Texas for an approved cocktail of phages against bacteria, including P. aeruginosa, Staphylococcus aureus and Escherichia coli (better known as E. coli).[citation needed]
Reviews of phage therapy indicate that more clinical and microbiological research is needed to meet current standards.[24]

Distribution

Phages can usually be freeze dried and turned into pills without materially impacting efficacy.[2] In pill form temperature stability up to 55 C, and shelf lives of 14 months have been shown.[citation needed] Other forms of administration can include application in liquid form. These vials are usually best kept refrigerated.[citation needed]
Oral administration works better when an antacid is included, as this increases the number of phages surviving passage through the stomach.[citation needed]
Topical administration often involves application to gauzes that are laid on the area to be treated.[citation needed]

Obstacles

General

The host specificity of phage therapy may make it necessary for clinics to make different cocktails for treatment of the same infection or disease because the bacterial components of such diseases may differ from region to region or even person to person. Such a process would make it difficult for large scale production of phage therapy. Additionally, patent issues (specifically on living organisms) may complicate distribution for pharmaceutical companies wishing to have exclusive rights over their "invention"; making it unlikely that a for-profit corporation will invest capital in the widespread application of this technology.

In addition, due to the specificity of individual phages, for a high chance of success, a mixture of phages is often applied. This means that 'banks' containing many different phages are needed to be kept and regularly updated with new phages, which makes regulatory testing for safety harder and more expensive.


Some bacteria, for example Clostridium and Mycobacterium, have no known therapeutic phages available as yet.

To work, the virus has to reach the site of the bacteria, and viruses do not necessarily reach the same places that antibiotics can reach.

Funding for phage therapy research and clinical trials is generally insufficient and difficult to obtain, since it is a lengthy and complex process to patent bacteriophage products. Scientists comment that 'the biggest hurdle is regulatory', whereas an official view is that individual phages would need proof individually because it would be too complicated to do as a combination, with many variables. Due to the specificity of phages, phage therapy would be most effective with a cocktail injection, which are generally rejected by the FDA. Researchers and observers predict that for phage therapy to be successful the FDA must change its regulatory stance on combination drug cocktails.[25] Public awareness and education about phage therapy are generally limited to scientific or independent research rather than mainstream media.[26]

The negative public perception of viruses may also play a role in the reluctance to embrace phage therapy.[27]

Safety

Phage therapy is generally considered safe. As with antibiotic therapy and other methods of countering bacterial infections, endotoxins are released by the bacteria as they are destroyed within the patient (Herxheimer reaction). This can cause symptoms of fever, or in extreme cases toxic shock (a problem also seen with antibiotics) is possible.[28] Janakiraman Ramachandran, a former president of AstraZeneca India who 2 years ago launched GangaGen Inc., a phage-therapy start-up in Bangalore,[29] argues that this complication can be avoided in those types of infection where this reaction is likely to occur by using genetically engineered bacteriophages; which have had their gene responsible for producing endolysin removed. Without this gene the host bacterium still dies but remains intact because the lysis is disabled. On the other hand this modification stops the exponential growth of phages, so one administered phage means one dead bacterial cell.[4] Eventually these dead cells are consumed by the normal house cleaning duties of the phagocytes, which utilise enzymes to break the whole bacterium and its contents down into its harmless sub-units of proteins, polysaccharides and lipids.[30]

Care has to be taken in manufacture that the phage medium is free of bacterial fragments and endotoxins from the production process.

Lysogenic bacteriophages are not generally used therapeutically. This group can act as a way for bacteria to exchange DNA, and this can help spread antibiotic resistance or even, theoretically, can make the bacteria pathogenic (see Cholera).

The lytic bacteriophages available for phage therapy are best kept refrigerated but discarded if the pale yellow clear liquid goes cloudy.

Cultural references


See also

References

  1. ^ McAuliffe et al. "The New Phage Biology: From Genomics to Applications" (introduction) in Mc Grath, S. and van Sinderen, D. (eds.) Bacteriophage: Genetics and Molecular Biology Caister Academic Press ISBN 978-1-904455-14-1.reprint
  2. ^ a b c d BBC Horizon: Phage - The Virus that Cures
  3. ^ Parfitt T (2005). "Georgia: an unlikely stronghold for bacteriophage therapy". Lancet 365 (9478): 2166–7. doi:10.1016/S0140-6736(05)66759-1. PMID 15986542.
  4. ^ a b Thiel, Karl (January 2004). "Old dogma, new tricks—21st Century phage therapy". Nature Biotechnology (London UK: Nature Publishing Group) 22 (1): 31–36. doi:10.1038/nbt0104-31. ISSN 1087-0156. http://www.nature.com/nbt/journal/v22/n1/full/nbt0104-31.html. Retrieved on 15 December 2007.
  5. ^ Pirisi A (2000). "Phage therapy—advantages over antibiotics?". Lancet 356 (9239): 1418. doi:10.1016/S0140-6736(05)74059-9. PMID 11052592.
  6. ^ Shasha SM, Sharon N, Inbar M (2004). "[Bacteriophages as antibacterial agents]" (in Hebrew). Harefuah 143 (2): 121–5, 166. PMID 15143702.
  7. ^ Hanlon GW (2007). "Bacteriophages: an appraisal of their role in the treatment of bacterial infections". Int. J. Antimicrob. Agents 30 (2): 118–28. doi:10.1016/j.ijantimicag.2007.04.006. PMID 17566713. http://linkinghub.elsevier.com/retrieve/pii/S0924-8579(07)00203-8.
  8. ^ "Stalin's Forgotten Cure" Science (magazine) 25 October 2002 v.298 [www.sciencemag.org]reprint
  9. ^ Summers WC (2001). "Bacteriophage therapy". Annu. Rev. Microbiol. 55: 437–51. doi:10.1146/annurev.micro.55.1.437. PMID 11544363.
  10. ^ Mangen MJ, Havelaar AH, Poppe KP, de Wit GA (2007). "Cost-utility analysis to control Campylobacter on chicken meat: dealing with data limitations". Risk Anal. 27 (4): 815–30. doi:10.1111/j.1539-6924.2007.00925.x. PMID 17958494. http://www.blackwell-synergy.com/openurl?genre=article&sid=nlm:pubmed&issn=0272-4332&date=2007&volume=27&issue=4&spage=815.
  11. ^ a b Mc Grath S and van Sinderen D (editors). (2007). Bacteriophage: Genetics and Molecular Biology (1st ed. ed.). Caister Academic Press. ISBN 978-1-904455-14-1 . http://www.horizonpress.com/phage.
  12. ^ Soothill JS (1994). "Bacteriophage prevents destruction of skin grafts by Pseudomonas aeruginosa". Burns 20 (3): 209–11. doi:10.1016/0305-4179(94)90184-8. PMID 8054131.
  13. ^ McVay CS, Velásquez M, Fralick JA (2007). "Phage therapy of Pseudomonas aeruginosa infection in a mouse burn wound model". Antimicrob. Agents Chemother. 51 (6): 1934–8. doi:10.1128/AAC.01028-06. PMID 17387151. http://aac.asm.org/cgi/pmidlookup?view=long&pmid=17387151.
  14. ^ Yacoby I, Bar H, Benhar I (2007). "Targeted drug-carrying bacteriophages as antibacterial nanomedicines". Antimicrob. Agents Chemother. 51 (6): 2156–63. doi:10.1128/AAC.00163-07. PMID 17404004. http://aac.asm.org/cgi/pmidlookup?view=long&pmid=17404004.
  15. ^ Yacoby I, Shamis M, Bar H, Shabat D, Benhar I (2006). "Targeting antibacterial agents by using drug-carrying filamentous bacteriophages". Antimicrob. Agents Chemother. 50 (6): 2087–97. doi:10.1128/AAC.00169-06. PMID 16723570. http://aac.asm.org/cgi/pmidlookup?view=long&pmid=16723570.
  16. ^ Bar H, Yacoby I, Benhar I (2008). "Killing cancer cells by targeted drug-carrying phage nanomedicines". BMC Biotechnol. 8: 37. doi:10.1186/1472-6750-8-37. PMID 18387177. http://www.biomedcentral.com/1472-6750/8/37.
  17. ^ Duckworth DH, Gulig PA (2002). "Bacteriophages: potential treatment for bacterial infections". BioDrugs 16 (1): 57–62. PMID 11909002.
  18. ^ "Non-antibiotic therapies for infectious diseases." by Christine F Carson, and Thomas V Riley Communicable Diseases Intelligence Volume 27 Supplement - May 2003 Australian Dept of health website
  19. ^ [1]
  20. ^ http://www.forbes.com/business/healthcare/feeds/ap/2006/08/18/ap2959720.html
  21. ^ "Press & News". Retrieved on 2007-12-13.
  22. ^ "biocontrol.ltd.uk". Retrieved on 2007-12-13.
  23. ^ "biocontrol-ltd.com". Retrieved on 2008-04-30.
  24. ^ " "Phage therapy: the Escherichia coli experience" by Harald Brüssow in Microbiology (2005) v. 151, p.2133-2140. publisher site
  25. ^ Thiel K (2004). "Old dogma, new tricks—21st Century phage therapy". Nat. Biotechnol. 22 (1): 31–6. doi:10.1038/nbt0104-31. PMID 14704699.
  26. ^ Brüssow, H 2007. Phage Therapy: The Western Perspective. in S. McGrath and D. van Sinderen (eds.) Bacteriophage: Genetics and Molecular Biology, Caister Academic Press, Norfolk, UK. ISBN 978-1-904455-14-1
  27. ^ Verbeken G, De Vos D, Vaneechoutte M, Merabishvili M, Zizi M, Pirnay JP (2007). "European regulatory conundrum of phage therapy". Future Microbiol 2 (5): 485–91. doi:10.2217/17460913.2.5.485. PMID 17927471. http://www.futuremedicine.com/doi/abs/10.2217/17460913.2.5.485.
  28. ^ Evergreen PHAGE THERAPY: BACTERIOPHAGES AS ANTIBIOTICS
  29. ^ Stone, Richard. "Stalin's Forgotten Cure." Science Online 282 (25 October 2002).
  30. ^ Fox, Stuart Ira (1999). Human Physiology -6th ed.. McGraw-Hill. pp. : 50,55,448,449. ISBN 0-697-34191-7. http://www.mhhe.com.
  31. ^ Summers WC (1991). "On the origins of the science in Arrowsmith: Paul de Kruif, Felix d'Herelle, and phage". J Hist Med Allied Sci 46 (3): 315–32. doi:10.1093/jhmas/46.3.315. PMID 1918921.
  32. ^ "Phage Findings - TIME". Retrieved on 2007-12-13.
  33. ^ "SparkNotes: Arrowsmith: Chapters 31–33". Retrieved on 2007-12-13.

 
Illustrations below are Frederick Twort and Felix d'Hérelle


                                            Love Me Love My Phages


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


Page 1                 
  Love Me Love My Phages

 Cholera epidemics and the role bacteriophages may play in ending them

 Cholera epidemics (caused by the bacterium Vibrio cholerae) cause widespread illness and death in developing countries. The Ganges Delta region of Bangladesh and India, for example, suffers two cholera epidemics each year. ICDDR,B researchers and Harvard Medical School have therefore been working to identify what factors trigger and end these seasonal epidemics. Results suggest that bacteriophages (viruses that attack bacteria) may play a key role.

"Researchers will use their improved understanding of the interactions among hosts,
V. cholerae, and cholera-killing bacteriophages to identify new ways of preventing
cholera epidemics
"
 During a three-year study of patients in ICDDR,B’s Dhaka hospital, researchers confirmed that the number of cholera patients (which varied seasonally) often coincided with the presence of disease-causing V. cholerae strains in water samples. They also showed that, during epidemic-free periods, water supplies typically contained cholera-killing phages but no viable bacteria.

Importantly, researchers also found that the phage peak in water samples coincided with a rise in the number of phages found in the excrement of cholera patients. So, it seems that the epidemics are ended by phages which amplify in people with cholera—these then attack the cholera bacteria once they are excreted and enter water supplies. This may well explain why the seasonal cholera epidemics that occur in Bangladesh are self-limiting.

Before the development of modern antimicrobials, phage therapy was considered a feasible option for the treatment of bacterial infection. The study team will therefore use their improved understanding of the interactions among hosts,
V. cholerae,and cholera phages to identify new ways of preventing cholera epidemics


Page 1                 


Phage in the Time of Cholera
Joshua S. Weitz
, and Hyman Hartman

Department of Ecology and Evolutionary Biology,
Princeton University, Princeton, NJ 08544.
Email: jsweitz@princeton.edu
*
Corresponding author
2
Center for Biomedical Engineering,
Massachusetts Institute of Technology, Cambridge, MA 02139.
Email: hhartman@mit.edu
January 27, 2006
Bacteriophage (bacterial viruses) were heralded as revolutionary therapeutic agents soon
after the discovery by Félix d’Herelle in 1917 of an “invisible microbe”
capable of lysing bacteria
1
. Bacteriophage appeared to be efficient killers of their bacterial hosts – 
we now know that their life history is far more complex than first
assumed
2
– and so the effort to use phage as curatives or prophylaxis spread
quickly to research institutes in Europe, North America, and Asia
3
. d’Herelle
himself spearheaded many of these efforts, the most famous of which was the
initiation of an extensive campaign to use phage in the treatment and prevention of
cholera in colonial India. The authors of one such study conclude by noting that
“the results establish sufficient probability in favour of a significant effect of the
administration of bacteriophage to form a basis of practical policy in the treatment


and prevention of cholera in villages”

4

. The early hopes never fulfilled

expectations, for both clinical and political reasons

3

, and the eventual development

of broad spectrum antibiotics provided a more reliable, effective means of

controlling bacterial infections. The rise of antibiotic resistance has, in turn,

revived interest in bacteriophage therapy despite concerns and uncertainties as to

its effectiveness

5

. We consider here an alternative approach to modern

bacteriophage therapy, by revisiting the idea of inoculating bacteriophage directly

into the environment.

Most tests, theories, and proposals to implement bacteriophage therapy

regard the human body as the potential site for intervention

6,7

. But for many

bacterial diseases affecting human health, the pool of infecting bacteria comes

from water, soils, food, and other host organisms; some of these potential sources

of infection do not possess a complex immune system capable of selectively

eliminating foreign agents. In contrast to agricultural settings where environmental

application of phage as biocontrol is already being considered

8

, we believe there

exists an as yet overlooked opportunity to reduce the severity, extent, and

persistence of some bacterial epidemics by developing ecological-based cures for

human disease.

A suitable target disease is cholera. Recent studies have demonstrated a

significant correlation between the increase in density of cholera-specific phage

and the decrease in density of Vibrio cholerae (in both water sources and fecal

matter from infected patients)

9,10

. The reasons are apparently simple: presence of

V. cholerae provides an opportunity for the spread and increase of phage which

leads to decreasing host density, which in turns leads to the washout/death of

phage. A comprehensive description of cholera disease dynamics involves many




factors including environmental seasonality

11

, long-distance dispersal mediated by

alternative hosts

12

, as well as life-history modalities that enable V. cholerae to

respond to stressful conditions

13

. Without diminishing the importance of these and

other factors, in the case of cholera it is apparent that phage and bacteria go

through alternating boom-and-bust cycles. What are the practical steps of

intervention so as to minimize the likelihood of devastating epidemic booms of V.

cholerae?

Briefly, the peak of phage lags behind the peak of bacteria. Growing up O1

and/or O139 serogroup-specific phage in the lab, therapy by the flask as it were,

and then adding phage to at-risk water sources may augment the ability of phage to

keep pace with the dynamics of its host and suppress the spread of an epidemic. In

a sense, we are suggesting altering the “natural course”

10

of host-phage population

dynamics with an artificial injection of phage. The utility and effectiveness of any

such ecological inoculation depend on careful balancing of environmental

connectivity of infected areas, risks to human populations, as well as the life-

history and parameterization of the biocontrol agent themselves. Ultimately,

limiting and/or eliminating an undesirable bacterial population constitutes a

problem in coevolutionary biological control. Recent theoretical work on

coevolutionary dynamics of bacteria and bacteriophage in simple aquatic

environments demonstrates that coevolution-induced outcomes, e.g. eradication of

phage and host, sequential strain replacement, or host-phage diversification,

depend on characterizing (and possibly manipulating) rates of mutagenesis, host

growth rate and strain-specific adsorption rates, and host-range characteristics of

mutants

14

. However, the ecology of natural environments is far more complex.

Likely sites for intervention include sources of drinking water, wells, and sewage




systems so as to minimize the flow of bacterial agents into water used for drinking

and bathing. Assessments of the lifetime of phage in local habitats would be

necessary as conditions (e.g., temperature, salinity, pH) change over the course of

intervention. In addition, the ecohydrology of the affected region may be

important, as intervention strategies will depend on whether disease outbreaks are

localized to isolated sites, linked to seasonal flooding, or occur along riverine

corridors.

These concerns notwithstanding, cholera-specific phage are already found

in natural environments and there exists strong evidence to suggest that their

presence leads to the decline of cholera epidemics

10

. The risks associated with

ecological bacteriophage therapy should be mitigated by the use of virulent, in

contrast to temperate, strains of phage. In this regard, the previously identified lytic

phages JSF1 and/or JSF5 specific to V. cholerae serogroup O1 seem ideal

candidates for initial studies

10

. If the origins of seasonal cholera epidemics are

harbored within environmental pools, then efforts should be made to seek out the

most effective means of adding bacteriophage to eliminate the incubation and

growth of V. cholerae populations when they are at their most vulnerable.

Diminishing the density of V. cholerae would also be important to impeding the

spread of disease, since the infectious dose is generally considered to be on the

order of 10

4

bacterial cells. Thus far, the spread of cholera has been mitigated by

improvements in water quality, low-cost preventative measures in at-risk regions,

e.g., filtering water through sari cloth

15

, as well as by improvements in post-

infection treatment, e.g., single-dose antibiotic therapy

16

, though the global cholera

pandemic has not abated. Bacteriophage could become an additional tool in the

public health struggle against cholera. The initiation of controlled experiments that




incorporate recent advances in the genetics and evolutionary ecology of phage may

offer hope that d’Herelle’s early mission to eradicate cholera in the Indian

subcontinent need not have been in vain.

References

[1] d’Herelle, F. Sur un microbe invisible antagoniste des bacilles dysentèriques.

Cr. R. Acad. Sci. Paris 165 (1917).

[2] Weinbauer, M. Ecology of prokaryotic viruses. FEMS Microbiology Reviews

28, 127–81 (2004).

[3] Summers, W. Félix d’Herelle and the Origins of Molecular Biology (Yale

University Press, 1999).

[4] Morison, J., Rice, E. & Pal Choudhury, B. Bacteriophage in the treatment and

prevention of cholera. Indian Journal of Medical Research 21, 790–907 (1934).

[5] Thiel, K. Old dogma, new tricks – 21st Century phage therapy. Nature

Biotechnology 22, 31–6 (2004).

[6] Merril, C., Scholl, D. & Adhya, S. The prospect for bacteriophage therapy in

Western medicine. Nature Reviews Drug Discovery 6, 489– 97 (2003).

[7] Levin, B. & Bull, J. Population and evolutionary dynamics of phage therapy.

Nature Reviews Microbiology 2, 166–73 (2004).

[8] Goodridge, L. & Abedon, S. Bacteriophage biocontrol and bioprocessing:

application of phage therapy to industry. SIM News 53, 254–62 (2003).

[9] Faruque, S. et al. Seasonal epidemics of cholera inversely correlate with the

prevalence of environmental cholera phages. Proceedings of the National Academy

of Sciences, USA 102, 1702–7 (2005).




[10] Faruque, S. et al. Self-limiting nature of aseasonal cholera epidemics: Role of

host-mediated amplification of phage. Proceedings of the National Academy of

Sciences, USA 102, 6119–24 (2005).

[11] Koelle, K., Rodó, Pascual, M., Yunus, M. & Mostafa, G. Refractory periods

and climate forcing in cholera dynamics. Nature 436, 696–700 (2005).

[12] Colwell, R. Global climate and infectious disease: the cholera paradigm.

Science 274, 2025–31 (1996).

[13] Roszak, D. & Colwell, R. Survival strategies of bacteria in the natural

environment. Microbiology Reviews 51, 365–79 (1987).

[14] Weitz, J., Hartman, H. & Levin, S. Coevolutionary arms races between

bacteria and bacteriophage. Proceedings of the National Academy of Sciences,

USA 102, 9535–40 (2005).

[15] Colwell, R. et al. Reduction of cholera in Bangladeshi villages by simple

filtration. Proceedings of the National Academy of Sciences, USA 100, 1051–1055

(2003).

[16] Saha, D. et al. Single-dose ciproflaxacin versus 12-dose erythromycin for

childhood cholera: a randomised controlled trial. The Lancet DOI:10.1016/S0140–

6736(05)67290–X (2005).





                            Love Me Love My Phages
              ......................................................................................... 
                    The Cocktail That Cures

                                        THE HINDU

                                                  

Online edition of India's National Newspaper

      
Sunday, November 26, 2000

HOW far away are we from a return to time when people die from a sore throat? At a press conference on May 23, 1997, scientists finally acknowledged the arrival of untreatable bacteria they had feared for years - bacteria that resist antibiotics. Drugs which have kept us safe for 50 years were beginning to fail, they said. 

Today, superbugs look triumphant and this is a serious situation. Over the last five years, scientists have clearly seen a change in their ability to tackle what should have been easily treatable infections, because bacteria are developing the ability to resist antibiotics. And the more antibiotics we use, the more resistant bacteria become. 

Every year, more than five million people die from infections that do not respond to antibiotics. Things are going to get worse. Staphylococcus, one of the most dangerous bacteria, now has only one antibiotic to keep it in check - Vancomycin. 

This year, Japanese doctors saw the world's first case of infection with Vancomycin-resistant staphylococcus - a baby boy in hospital for major heart surgery. When antibiotics failed, doctors had to pour strong disinfectants directly into the wound on his chest. It was quite shocking because the outcome of that infection was quite hideous. The patient suffered a lot. 

Terrible bacteria will inevitably spread, and when they do, being in hospital even for minor surgery, or a hip operation or to have a baby, could be lethal. If even the smallest wound becomes infected, bacteria would most probably kill you. It would be very hard to conduct major surgery that we have got used to. Transplants, cancer chemotherapy, are all dependent on the ability to kill off bacteria which may infect patients. We will lose all that. 

It is hard to understand why no new drugs have been developed to save mankind. Pharmaceutical companies should have come to grips with the situation. But they thought that that was not profitable a decade ago. Now, it is too late. 

Even today, drug companies do not promise a new class of antibiotics for at least 10 years and they may never discover one. They have already exhausted traditional chemistry and computer drug design. Today, they analyse the genes of the bacteria, hoping to find new strategies. But the frightening truth is that no fundamentally new antibiotic has been discovered for more than 30 years. 

One begins to worry that we are indeed moving into what some people have called the post-antibiotic era, where bacteria are supreme. 

But there is a major remedy that kills even the most resistant bacteria. 

Unknown to the rest of the world, in a small country in the heart of the Caucasus Mountains, south of Moscow and north of Turkey, scientists in the Republic of Georgia may have the answer against superbugs. 

In the central hospital in the Republic of Georgia, a former Soviet State, patients recover like elsewhere. They are weak and vulnerable and would probably die if they caught an infection. 

The doctors here know that there is a problem. It has become the breeding ground for particularly nasty bacteria, which they suspect are resistant to all the antibiotics they have. They are taking samples from every surface in the ward to know exactly what they are up against. Some of the samples have traces of staphylococcus strains. 

Anywhere else in the world, this would be a death sentence. But here in Georgia, the doctors are not too worried because they have an answer - a unique medicine that still works on antibiotic-resistant infections. The same amazing potion is used to treat both patients and wards. It has a remarkable effect on bacteria. 

Two flasks contain bacteria. A few drops of the Georgian medicine have been added to one and it has a magical effect. The bacteria have all been killed. This astonishing effect is caused by something we usually fear - a virus, and one that comes from sewage. Says Dr. Teimuraz Chanishvili, Institute of Bacteriophage, Tbilisi, "This happened 51 years ago. It was the first experiment I did here. I took the culture of bacteria and added sewage to it, just ordinary sewage water from the drains. I first saw this a long time ago, when I was still a student. We were all very enthusiastic about it." 

The first to spot what happened was an irascible French-Canadian called Felix d'Herelle. In 1917, he suggested that the viruses which killed bacteria in the bottles could be used to treat disease. An ardent Communist, d'Herelle was enticed to Russia by Stalin, who wanted this magic medicine for his army. And in the Finnish war of the 1930's, and even in World War II, it was used to protect the Red Army from the dysentery and gangrene that plagued the battlefield. 

With Stalin's blessing, d'Herelle founded an institute in Tbilisi, the capital of Georgia, that was dedicated to the study of these magical viruses and the way they cure infections. D'Herelle named these healing viruses as bacteriophage, which means "bacteria eaters". There is a phage to kill every kind of bacteria. Just as bacteria attack people, these tiny viruses attack them, and they are found wherever bacteria thrive, most often in sewage. 

Phage have an extraordinary structure. Their bulk is a head in which their genes are stored. They have six legs or filaments, which attach themselves to the bacteria and a tail that works like a hypodermic syringe to infect it with their genes. Inside the bacteria, the phage viruses grow and multiply. Sometimes as many as 5,000 grow in a single cell. 

When the new phage burst out, they kill the bacteria and then each goes on to find another victim. Each phage only grows on a particular kind of bacteria. That is why when we talk of Staphylococcus phage, it is known that it reproduces only on Staphylococcus. 

But being specific makes phage tricky to work with. Over the years, the institute has trained specialists to find phage, grow them and turn them into medicine. In the 1970s when antibiotic- resistant bacteria became a serious problem in Soviet hospitals, phage became the saviours. 

A decree was issued that all bacteria resistant to antibiotics and local phages must be sent to the institute, where a new preparation was made. It was very difficult to organise, but everything was done under one roof. The centralised Soviet system was ideal for the labour-intensive work needed to make phage effective. Together, Communism, phage and the institute thrived. 

Phage medicine had its heyday in the 1980s. It was manufactured in factories across the Soviet Union and the Tbilisi Institute. At that time, Teimuraz Chanishvili and Amiran Meipariani ran the institute. 

Remembering those days they say, "We used to inject phage into one, two, three, four vats. There were 500 litres in each vat, and remember we had to take orders on top of that. We produced tablets and bottles. We made phages for the Soviet Union. They were not only for dysentery but typhoid and salmonella too. Most were for intestinal infections. Phages are our daily bread. We have devoted our lives to them. The prospects for phage are tremendous. There is no question that phage medicine can be extremely effective." 

An old woman recalls how phage helped cure her son, "My son became ill when he was young. They checked his throat and nose and found Staphylococcus. I went to the institute and they gave him phage and cured him. Phage medicine is a wonderful thing. It works against dysentery. They even give you phage in an enema." 

Today, when people in Georgia get an infection, they take antibiotics at times. Often the doctors prescribe phage. There is a pharmacy on the grounds of the Tbilisi Institute. Everyday, people who choose to use phage, rather than antibiotics, go there to have their infections diagnosed and to pick up prescriptions for medicines, tablets and creams made of phage. 

They rub the healing viruses into their wounds, drink them for a bad stomach, or swirl a solution in their months to cure a gum infection. A woman developed gangrene in a wound on her thumb. The doctors cut away most of the infected tissue, the rest they treated with phage. If the resistant bacteria rife in the hospital got into this woman's wound, it would have caused fatal blood poisoning. Phage does not work well in the bloodstream. 

Being a virus, it is fought off by the immune system. But the surgeons make sure infection does not take hold in the first place, by using phage to sterilise the room and equipment as well as the wound. The doctor in attendance says, "We think that the phage that was used during the first operation helped the wound to be in such good condition." Phage works wonders in Georgia and given the chance, it could do the same everywhere." 

A unique library of phage medicine exists in Tblisi. It is a national treasure, built up over 50 years when problem bacteria was sent here from all across the Soviet Union. Phages were found to fight every new infection and then they and the bacteria they killed were stored separately, for future research. It is the biggest collection of phage medicine in the world. 

Phage therapy has been so successful in the Soviet Union for so long, that it is hard to understand why people in the West have never even heard of it. To scientists like Dr. Chanishvili, it seems incredible that this medicine, once used daily right across the Soviet Union, has been ignored by the West. But the reasons are woven tightly into history. 

In Britain today, very few people know anything about phage therapy, but those that do are clear about how it became discredited in the West in the 1930s. 

Some of the claims made for some of these bacterial phages which are isolated were, quite frankly, barmy. There was one commercial preparation, for example, called Enterofagos, which supposedly had miraculous powers against both herpes infections and eczema. 

These claims were not fraudulent. In fact, a lack of understanding plagued most phage work done around the world in the early days. In India in 1935, even the British army tried it out on the local population during a cholera epidemic. 

It has to be said that some of the clinical trials that were carried out were of exceedingly poor quality. For example, in some of the early work on choleraphage in the 1920s and the 1930s, there were no control groups. So it was impossible to see whether the phage had worked. 

And some of the trials consisted merely in pouring bacteriophage down drinking wells in a village and see whether it had any effect with no understanding of dosage or the mechanisms whereby bacteria produced the diseases. 

Some of the problems with early studies though is the best phages for the job were not selected. They did not check whether the organism was sensitive to the phage. By 1941, phage was still thought to be too unreliable to be useful by researchers in the West. W. When powerful antibiotics arrived a few years later, it was quickly forgotten. Meanwhile, unknown to the outside world, the Georgian scientists went on working. 

Research that proves the worth of phage medicine was published by the Georgian scientists, but only in the Soviet Union

And even today, it is ignored because of a strange phenomenon that is true across the world of science. The articles were published either in Russian or in Georgian. Thus, language has proved to be the final barrier. So, perhaps the real problem was not because the science was bad, but because the findings could not be read. And so the West could not get acquainted with it. 

Modern medicine faces a crisis as new strains of antibiotic- resistant bacteria threaten advanced treatments and intensive care. But there is an unlikely saviour - a virus derived from sewage that can kill bacteria. To learn more about this unlikely saviour watch "Vital Breakthroughs", Sunday, from 7 to 8 p.m., on the Discovery Health Block, only on Discovery Channel. 
Information and picture courtesy: Discovery Channel

Love Me Love My Phages
                                 ...................................................... 

 Phage Therapy in Agriculture and Animal Husbandry 

It may surprise many people just how important that the study of Bacteriophages and the implementation of Phage Therapy in the growing of crops, and in looking after animals or even fish, as like all living things they take the same risks as we do, and can infect us, or ev en wipe out civilisations with huge crop failures.

The following article deals  the following important issues, among many others.


1)      Phage to control plant diseases such as bacterial spot on tomatoes and Erwinia infections of fruit trees (fire blight) and root crops (soft rot).

2)      Phage to treat animal disease such as E. coli respiratory infections in chickens, furunculosis (Aeromonas salmonicida) in fish, and mastitis in cattle.

3)      Phage to control human food-borne pathogens, such as Salmonella in fowl and Escherichia coli O157:H7 in cattle, and Listeria during food processing.

Phage Therapy: new methods for the potential eradication of E. coli O157 in livestock.

http://www.adsa.org/discover/7thDISCOVERProg4-8-03_files/INTERPRETIVE%20SUMMARIES/BrabbanADSAphageabstract.htm

Andrew Brabban1, Raul Raya1, Todd Callaway2, and Elizabeth Kutter1. 1The Evergreen State College, Olympia WA., and 2USDA Agricultural Station, College Station, TX. 


Since the advent of antibiotics, both the health care and agriculture sectors have relied heavily on them to control bacterial pathogens. However, increasing levels of antibiotic resistance have reduced the efficacy of many current therapies, prompting legislation that has reduced the use of antibiotics in animals. This has led researchers to seek fresh ideas. Bacteriophage therapy is one “old” idea undergoing a renaissance, with the potential to resolve the antibiotic predicament we find ourselves in today. Lytic bacteriophages are viruses that attach to specific bacterial surface receptors, inject their DNA, and express genes that lead to the synthesis of new phages. The process ends with the programmed lysis (death) of the host and the release of dozens or hundreds of new phages. The use of phages as antimicrobial agents has a number of advantages over other methods. Phages are highly specific allowing for the removal of the targeted microorganisms from a mixed population. Unlike antibiotics that decay over time and distance, phage numbers actually increase working their way deeper into pockets of infestation. Further phage are living entities that adapt and evolve. As phage can also pass from host to host, they have the potential to establish an infectious cure. Interest in agricultural applications is now rapidly expanding in 3 major areas;


1)      Phage to control plant diseases such as bacterial spot on tomatoes and Erwinia infections of fruit trees (fire blight) and root crops (soft rot).


2)      Phage to treat animal disease such as E. coli respiratory infections in chickens, furunculosis (Aeromonas salmonicida) in fish, and mastitis in cattle.


3)      Phage to control human food-borne pathogens, such as Salmonella in fowl and Escherichia coli O157:H7 in cattle, and Listeria during food processing.


Today I will talk about our most recent efforts to control Escherichia coli O157:H7; a pathogen that can produce severe diarrhea, kidney damage and death in humans. In the U.S., >70,000 illnesses and 60 deaths are reported yearly with most cases traced back to livestock contamination - drinking water, milk, produce or ground beef.. Livestock show no signs of illness and the levels are generally low, making contaminated animals hard to identify. We have developed a promising way of reducing O157:H7 concentrations in livestock through bacteriophages CEV1, CEV2 and CEV3. Phage CEV1 was isolated from the feces of sheep resistant to gastrointestinal colonization by O157:H7 strains that had previously been used as standard gut colonizers. Phages CEV1 and CEV2 infect >90% of the O157:H7 strains tested, and a small number other strains. Preliminary trials of CEV1 and CEV2 to evaluate their use as a potential management strategy in vitro and in vivo have been highly successful. In model systems reflecting the cow/sheep gut, CEV1 eliminated the two virulent 0157:H7 strains in 11 days. Initial animal studies have shown the phage have great promise as an orally administered treatment; treated sheep showed a substantial reduction in intestinal levels of O157:H7 in with a single dose of CEV1 within 2 days. Further reduction was seen with a cocktail of CEV1 and CEV2, as well as in those sheep naturally carrying phage CEV2. These results indicate the protective effect of bacteriophages against E. coli O157:H7. We hope this approach will significantly contribute to the important job of creating a safer human food supply for the 21st Century.


Dr. Andrew D. Brabban, Lab I, The Evergreen State College, Olympia, WA 98503. Tel: 360-867-6157, Fax: 360-867-6791. brabbana@evergreen.edu, http://academic.evergreen.edu/b/brabbana/ http://www.evergreen.edu/phage/home.htmL

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

Archivum Immunologiae et Therapiae Experimentalis, 1999, 47, 267–274
PL ISSN 0004-069X

 

 Review
Phage Therapy: Past History and Future Prospects


RICHARD M. CARLTON

Exponential Biotherapies, Inc., 150 Main Street, Port Washington, NY 11050, USA
  • Abstract. Bacterial viruses (bacteriophages, also called “phages”) can be robust antibacterial agents in vitro. However, their use as therapeutic agents, during a number of trials from the1920s to the 1950s, was greatly handicapped by a number of factors. In part, there were certain limitations inherent in phage physiology (e. g. narrow host range, and rapid clearance from the body); in part there were technological limitations in the era (e.g. lysogeny not yet discovered); but the greatest limitation was the highly inadequate scientific methodologies used by practitioners at the time (e.g., their failure to conduct placebo-controlled studies, to remove endotoxins from the preparations, and to re-confirm phage viability after adding sterilizing agents to the preparations). In recent years, well-controlled animal models have demonstrated that phages can rescue animals from a variety of fatal infections, while non-controlled clinical reports published in Eastern Europe have shown that phages can be effective in treating drug-resistant infections in humans. This encouraging data, combined with the fact that drug-resistant bacteria have become a global crisis, have created a window of opportunity for phage therapy to be tested anew, this time using modern technologies and placebo-controlled designs. If successful, it can be used as a stand-alone therapy when bacteria are fully resistant to antibiotics, and as a valuable adjunct to antibiotics when the bacteria are still susceptible.                     -------------------------------------------------------------------------
Phage Therapy - Past History and Future Prospects
       
A number of reviews provide details on phage therapy’s ascent and decline in the historical era 1–3, 13, 14.

We will summarize some of the more salient features of this history.

Phages were discovered in 1915 by British microbiologist Felix Twort, and, independently in 1917, by French-Canadian microbiologist Felix d’Hérelle. Twort did not pursue his discovery, whereas d’Hérelle systematically investigated the nature of bacteriophages and explored their ability to function as therapeutic agents 5, 6.

D’Hérelle received a fair measure of fame for his discovery. He was appointed Professor of Protobiology at Yale University Medical Center, and was also on the staff of the Pasteur Institute. In 1931 he gave a series of monthly lectures on phage therapy to the New YorkAcademy of Medicine. He established phage therapy centers in several countries, including the U. S., France, and Soviet Georgia. A fictionalized account of his work was depicted in rrowsmith, the Pulitzer-prize winning novel by Sinclair Lewis.

There are many attributes of phages (see Table 1) that would tend to favor a positive outcome in therapy. Despite these attributes of phages, there were so many problems with the way phage therapy was practiced in the historical era that, by the time antibiotics were introduced in mid-century, it was already in sharp decline in the West. The investigators who developed antibiotics did not make the kinds of mistakes exhibited by the early phage investigators.
Key problems with phage therapy, and how the problems can be overcome

Problem 1. Host range

The issue. Phages tend to have a relatively narrow host range, posing certain disadvantages. A disadvantage is that one should administer only those phage strains shown to be strongly lytic for the bacterial strain infecting the given patient. If the patient’s condition is too critical to take the time required for this matching, then one should use a grouping (a panel) of phages, where each of the phages therein has a broad-enough host range that most strains of the bacterial target are likely to be targeted. In his lectures to the New York Academy of Medicine in 1931, d’Hérelle cited the reports of other colleagues whose initial trials used phages “off the shelf” (without being shown to be virulent for the bacteria infecting the patient) and had negative outcomes, but who did match the phage to the bacteria in subsequent trials and obtained positive outcomes.

The solution. 1) Screen the bacteria infecting a given patient against a panel of phages, to ensure that one of the phage strains will be lytic (analogous to the “culture and sensitivity test” that physicians should perform; and 2) develop “multivalent” phages that lyse all or most of the bacterial strains within a given species of pathogen.
..........................................................................................................................

Table 1   - Attributes of phages that tend to favor a therapeutic response              
  
The Issue :  Fate of the  drug   molecule
Limitations of Antibiotics : Metabolic destruction of the molecule, as it works
Advantages of phages
Exponential growth in numbers, so that the “ drug”  makes more of itself at the site of infection, where it is needed
                                    .....................
The Issue : Concentration of the "drug ”  required to kill a given bacterium within the spectrum
Limitations of antibiotics:  Numerous molecules of the antibiotic are needed to  kill  a  given           bacterium.  During  initiation  of     therapy (and between doses), the sub-lethal dose that bacteria “see'' affords them the opportunity to express resistance genes
Advantages of phages:  “All or nothing”  effect: one phage particle is sufficient  to  kill  a  given  bacterium
                             ...........................
The issue:  Ability to overcome bacterial resistance

Limitations of antibiotics: Antibiotics are fixed, immutable chemicals that cannot  adapt  to  a  bacterial  mutation  and therefore become obsolete. Bacteria that have resisted them can pass along the resistance trait within and between species

Advantages of phages:  Phages are “living”  organisms that undergo mutations, some of which can overcome bacterial  mutations.  E.  g.,  mutated  phage  tail fibers can allow binding to a mutant bacterial receptor, or mutated phage DNA can escape cleavage by mutant bacterial endonucleases
                                         ....................
The issue:  Spread of bacterial resistance
Limitations of antibiotics: The antibiotics in use tend to be broad spectrum, thereby provoking resistance in several species and genera of bacteria (in addition to the one targeted}
Advantages of phages:  Although there are some exceptions, phages tend not to cross species boundaries. Thus even though the targeted bacterial species may become resistant to the phage, it is unlikely that other species will.
Numerous molecules of the antibiotic are needed to kill a given bacterium. During initiation of
therapy (and between doses), the sub-lethal dose that bacteria “see” affords them the opportunity to express resistance genes. Antibiotics are fixed, immutable chemicals that cannot adapt to a bacterial mutation and therefore become obsolete. Bacteria that have resisted them can pass along the resistance trait within and between species.

The antibiotics in use tend to be broad spectrum, thereby provoking resistance in several species and genera of bacteria (in addition to the one targeted) . Exponential growth in numbers, so that the “drug” makes more of itself at the site of infection, where it is needed.

     “All or nothing” effect: one phage particle is sufficient to kill a given bacterium. Phages are “living” organisms that undergo mutations, some of which can overcome bacterial mutations. E. g., mutated phage tail fibers can allow binding to a mutant bacterial receptor, or mutated phage DNA can escape cleavage by mutant bacterial endonucleases Although there are some exceptions, phages tend not to cross species boundaries. Thus even though the targeted bacterial species may
become resistant to the phage, it is unlikely that other species will."

268 R. M. Carlton: Phage Therapy in the Past and Future
....................................................................................................................
  
Problem 2. Bacterial debris present in the phage preparations

The issue. Injection of even minute amounts of endotoxin and other bacterial debris can be fatal to patients. Unfortunately, many of the phage preparations used by practitioners in the historical era were crude lysates. When these preparations were injected i.v., i.p., and in some cases even intrathecally, any beneficial effect of the phages would likely have been counteracted by illness and deaths resulting from the endotoxin.

The solution. Modern technology allows density centrifugation, banding, and other methods of purification.

Problem 3. Attempts to remove host bacteria from therapeutic preparations

The issue: In order to ensure that phage preparations would not contain live bacteria, some early investigators added mercurials and/or oxidizing agents, while others heated them. It is now known that such agents and procedures will denature or otherwise inactivate the phage coat proteins. These investigators did not check for continued viability of the phages. The false-negative results of such studies were the unintended (but inevitable) consequence of such practices.

The solution: Sterile filtration. If chemical agents must be used, reiterate the preparation over time to ensure that the phage remain viable.

  Problem 4. Rapid clearance of phages

The issue. In fairness to phage investigators in the historical era, at the time it was not an accepted practice, in any discipline, to conduct pharmacokinetic studies. However, had the early phage investigators conducted such studies, they would have discovered that bacteriophages (being foreign proteins) tend to be rapidly cleared from the circulation. This clearance problem was first documented by Merril and his colleagues in 1973 who injected high titers of phage lambda into non-immune germ-free mice. They discovered that the phages were rapidly cleared by the spleen, liver and other filtering organs of the reticulo-endothelial system (RES)7. 

This was a seminal observation, given Gunther Stent’s widely-accepted statement that one of the principal reasons phages had failed as a therapeutic was their supposed inactivation by pre-existing antibodies to them. However, any clearance of the phages from the bloodstream of the germ-free animals used by Merril and his group (ref.7) would not be due to antibodies, since those animals had never previously been exposed to bacteria or bacteriophages (and so would not have antibodies). Moreover, the phages in Merril’s experiment remained viable in the spleens of these animals over a period of several days, indicating that they were neither neutralized by antibody nor engulfed by macrophages. Rather, they appeared to have been passively entrapped in (sequestered by) these filtering organs. Such trapped phages would be unavailable to reach bacteria.

The solution. The author of this review collaborated with investigators at the U.S. National Institutes of Health (MERRIL et al.11) in the development of a method to isolate and amplify phage strains that are cleared at a slower rate. We reasoned that in all species of phage, minor variations in coat proteins might be present that would enable some variants to be less easily recognized by the RES organs and to thereby remain in the circulation for longer periods of time than the “average” wild-type phage. In this “serial passage” method, the wild-type preparation is injected into an animal, and then blood samples are taken at progressively longer time points. Any phages found in the blood sample are grown to high titer and reinjected. Through iterative rounds of passage, one can amplify the long-circulating strains being isolated. U.S. and PCT patents have been granted on this method. 
For coliphage lambda as well as for salmonella phage P22, phage variants were isolated in this manner that were much longer-circulating than the wild-type. For example, for every 100 000 particles of the wild-type lambda used at baseline, only one particle remained in circulation at 18 h; whereas for the long-circulating phage mutant isolated at the 8th round of serial passage, for every 100 000 injected, at 18 h 62 500 particles remained in circulation. For each moment of time, far more of these long-circulating phages are propagating exponentially, as compared to the situation for the wild-type phages.

As predicted, these long-circulating phages were far superior to the wild-types from which they were derived, in terms of rescuing animals from an otherwise-fatal fulminant bacteremia: 1) with no treatment, all animals were dead within 48 h; 2) treatment with the wild-type phages prevented death, but the animals became critically ill (a human with such degrees of illness would be in the intensive care unit); and 3) in contrast, with administration of the long-circulating phage strain, the only sign of illness seen was mild lethargy. These results were published in the Proceedings of the National Academy of Sciences (ref. 11), and were accompanied by a Commentary by Nobel laureate Dr. JOSHUA LEDERBERG 8.


R. M. Carlton: Phage Therapy in the Past and Future  P 269

We have elucidated the molecular basis of the mutation in lambda that reduced its rate of clearance: a single point mutation, an A to G transition, had occurred in the gene encoding the major head protein E.This mutation substituted a basic amino acid (lysine) for an acidic one (glutamic acid), causing a double charge shift readily seen on 2D gel electrophoresis.Computer modeling predicted that the mutation occurred in a loop of the E protein that sticks out into space and that therefore may interact with the external environment. A double charge shift in this region ofa protein that is highly represented on the surface of the virion could conceivably alter the phage’s interaction
with the microcirculation of the spleen, in such a waythat the mutant phage is less easily entrapped than thewild-type.

Problem 5. Lysogeny

The issue. It was not until the late 1950s that Lwoff demonstrated the ability of some phage genomes to integrate into the bacterial chromosome as “prophages.”After a period of time (up to days or weeks, or longer), such prophages can enter the lytic cycle, and will thus appear as plaques on a bacterial lawn. It is likely that some phage therapy trials in the historic era had a negative outcome due to the inadvertent use of phage strains that, being lysogens, could not provide the rapid lysis and exponential growth in numbers that are needed for full efficacy.

The solution. Use only phages that are lytic; sequence phages that are strong candidates for clinical trials, looking for (among other things) homologies to known genes of lysogeny.

Problem 6. Anti-phage antibodies

The issue.   There are reports in the literature20 that neutralizing antibodies appear a few weeks after administering phages to humans or animals. Given the time lag, antibodies would not seem likely to interfere with an acute treatment lasting a week or so. However, in chronic treatment, or in treatment of a recurrence of the same bacterial infection, the neutralizing antibodies might prevent some proportion of the administered dose of phages from being able to adhere to the bacterial target.

The solution. In treating chronic or recurrent infections it may be possible to administer a higher dose of phage, to compensate for those that are rendered non-viable by interaction with neutralizing antibodies. In any case, the types and titers of antibodies that develop should be systematically studied in humans. 

Problem 7. Failure to establish scientific proof of efficacy 

In scholarly reviews of comparative styles of research, Dutch historian TON VAN HELVOORT24 has discussed d’Hérelle’s systematic failure to conduct double-blind studies. As van Helvoort pointed out, while it is true that ethical problems are faced by anyone who has to administer placebo to some patients (in order to prove efficacy), nevertheless the investigators who later tested antibiotics did conduct double--blind, placebo-controlled trials. Van Helvoort points out that, even when using phages to treat an epidemic of diarrhea in poultry on a French farm, d’Hérelle failed to use a placebo on half the flock (a situation where ethical considerations would not have been an issue). As a consequence, all reports of phage therapy’s successes in the historical era were anecdotal. No systematic proof was available to demonstrate that the results were reliable and repeatable.

Problem 8. The scientific style of phage investigators in the historical era, D’Hérelle’s failure to conduct placebo-controlled studies, even on chickens, is an important example of his style. This story is a notable example of the negative impact an investigator’s personality can have on the outcome of a discovery, and d’Hérelle’s style contrasts sharply to the strongly positive influence that other scientists (such as Pasteur) have had on the outcomes of their discoveries. Whereas Pasteur excelled at conceiving of definitive experiments, and was persuasive in style, d’Hérelle failed to conduct definitive experiments, and was antagonistic rather than persuasive. For example, d’Hérelle maintained to the end that phages are the sole mechanism of defense against bacterial infection.

 While he may have been correct in his view that epidemics can sometimes be checked by the spontaneous appearance of a lytic strain of phage, nevertheless he was incorrect in categorically dismissing the discoveries of Nobel laureates Metchnikoff and Ehrlich, who had shown that cellular elements (white blood cells) and humoral elements (antibodies and complement) constitute the innate host defenses against infection. D’Hérelle was afforded many opportunities to integrate his discovery with those of Metchnikoff and Ehrlich, but refused to the end (see below). 
In addition to the damage he was doing to himself and his cause with this adamance, d’Hérelle was attacked by Nobel laureate Jules Bordet (for whom Bordetella pertussis was named), who had an intense dislike not just for d’Hérelle’s science but also for the man himself. Bordet used his considerable influence to discredit D’HÉRELLE 5.

D’Hérelle retreated from attacks by Bordet and others, and moved to Soviet Georgia in the 1930s (see ref. 13). An ardent communist, he dedicated the last of his published treatises to Josef Stalin. He was in Paris at the outbreak of World War II, refused to offer his skills with phage therapy to the Germans*, and spent the occupation years in prison. By the time of the liberation his health had been broken. He was invited to a post-War international scientific symposium, where colleagues made a last effort to see if they could help him bridge the gulf. He persisted in his belief that phages were the body’s sole mechanism of defense against bacteria (“Ce n’est que la phage…”), and he died in isolation in 1949. Surely the prospects of phage therapy in the historical era would have been better served if d’Hérelle had possessed some of the personality traits and scientific style of Pasteur.
 
      * The push of the German army into the region of Georgia was intended not only to capture the region’s oil  wells, but also to obtain the collection of phages manufactured at the Eliava-d’Hérelle Institute in Tblisi. That institute was providing phages to the Russian army, to control dysentery, Staphylococcus aureus infections ofwounds, and other bacterial problems associated with war

Animal Models of Phage Therapy

From the 1950s to the 1980s there was little published on the subject of phage therapy. Then papers began to appear demonstrating the utility of phage therapy in animal models. For example, phages were shown to be effective in rescuing rats from fatal systemic infections (induced with E. coli)14 in rescuing calves and lambs from fatal diarrhea (induced with E. coli)15, 16, in rescuing chicks from fatal diarrhea (induced with S. typhimurium) 4, and in preventing destruction of skin grafts in burned rabbits by Pseudomonas aeruginosa18. As mentioned above MERRIL et al.11 demonstrated in 1996 that mice with fulminant E. coli bacteremia could be rescued by phages, and that long-circulating phage variants were superior to the wild-types (see below). In one of those studies cited, Smith and Huggins (ref. 6) demonstrated that, in rats inoculated with a lethal intramuscular dose of E. coli, a single injection of a phage preparation was more effective than multiple injections of antibiotics (chloramphenicol, tetracycline, etc.). This work was replicated in 1997 by LEVIN and BULL9, who used mathematical modeling in a population dynamics approach to study the titers of phages and bacteria in the animals. The investigators concluded that the reason a single injection of phage was
superior to multiple injections of antibiotics was that the phages grew exponentially in number, overwhelming the bacteria present.

Current Status of Human Phage Therapy Efforts

Poland. Phage therapy is practiced in Poland, albeit on a small scale. In the mid-1980s a series of papers was published by a group led by the late Prof. S. S´lopek  and his colleagues, including Dr. M. Mulczyk and Dr. B. Weber-Da˛browska, working at the L. Hirszfeld Institute of Immunology and Experimental Therapy (a branch of the Polish Academy of Sciences). These papers20–23 reported on 550 cases of suppurative bacterial infections (empyemas, peritonitis, osteomyelitis, etc.) in humans. Most of the cases were chronic; most were resistant to all available antibiotics; and most had not been referred for this form of therapy until all else had failed, meaning that it was often quite late in thedisease progression. The bacterial pathogens targeted included Staphylococcus aureus, Pseudomonas aeruginosa, Klebsiella pneumoniae and E. coli. The phages used by these investigators are reported to have cured approximately 90% of the cases.

The criteria of cure were cessation of suppuration and,where applicable, complete closure of wounds/fistulae (many of which had been draining for months). These investigators administer phages orally, because they are aware of the hazards of administering them parenterally (not all of the bacterial debris has been removed). They pre-treat the patients with antacids and gelatin in order to protect the phages from destruction by gastric acidity. These same investigators have published evidence that phages administered orally to humans in this manner do in fact reach the bloodstream 26.

The Polish investigators have been rigorous in   matching the phages to the bacterial strain infecting the given patients. Their practice, as stated in the published reports, is to culture the bacteria during the course of treatment, so that the occurrence of a mutant resisting the phage can be countered by switching to a different phage strain. The group also has panels of multivalent
phages available, for use in fulminant infections (such as septicemia with acute respiratory distress syndrome) where time is insufficient to classify the offending bacteria or to match phages to bacteria.

The group now has statistics on the treatment of approximately 1 300 cases. The overall cure rate across the spectrum of pathogens and sites of infection is approximately 86% (personal communication from Dr. B. Weber-Dabrowska). A criticism of the work by Slopek’s group is that - the absence of placebo controls means the power of suggestion cannot be definitively ruled-out. It is clear that the difficulties of that nation’s economy over recent decades has denied the investigators the financial resources needed to enroll matched cohorts in a placebo arm of a clinical trial. While the criticism is valid, and absolute proof of principle can be obtained only through placebo-controlled trials, nevertheless the usefulness of the data is improved by the detailed statistical accounting of the percentages of complete, partial and nil response. One of the factors that enables this author to find the data from Poland more believable (even in the absence of double-blind proof) is that in conditions such as emphysema where phage efficacy might be somewhat impeded, the group’s statistics show that the success rate is considerably lower than for other conditions where such impediments do not obtain*.

The Republic of Georgia.

 The work started in Tblisi in the 1930s by d’Hérelle and his Georgian colleague, Eliava, continues to this day. In the 1970s, under the direction of Dr. Teimuraz Chanishvili, the Eliava-d’Hérelle Institute had a large staff manufacturing considerable quantities of phage preparations per year, primarily for the control of dysentery in the troops of the Soviet Army. This group has anecdotal evidence of the efficacy of phage therapy. They report, for example, that in certain adult and pediatric hospitals it is routine for their phage preparations to be administered topically on surgical incisions. Given the lack of statistical analysis, there is little to be said other than the anecdotal reports are encouraging that phage therapy can be useful.

Multidrug-Resistant (MDR) Bacteria Have Created a Need for Phage Therapy

Several species of bacteria have become resistant to most antibiotics, with some strains being resistant to all antibiotics. One example is vancomycin-resistant Enterococcus faecium (VRE), a low-virulence pathogen that now frequently causes fatal bacteremias due to complete resistance 2. 

Another example is vancomycin intermediate-resistant Staphylococcus aureus (VISA), strains of which have recently emerged in three nations (Japan, U.S. and Scotland), and are known to have killed 4 patients to date. Such strains spread throughout Japanese hospitals within a year of their first appearance.

Unfortunately, it has been demonstrated that some hospital strains of methicillin-resistant S. aureus (MRSA) that are widespread have become vancomycin resistant upon exposure of the patients to vancomycin  1, 2. 

Experts predict that S. aureus will progress to become completely resistant to vancomycin (the antibiotic of last resort for most strains of this pathogen), and that when this occurs, millions of people will die each year from infections that had until recently been fairly easy to control. Based on such developments and impending developments with pathogens such as MRSA and VRE, opinion leaders have been warning that we are entering the “Post-Antibiotic Era”.

While pharmaceutical companies are developing new antibiotics to counter the trend, it has been shown that half a century of global antibiotic abuse has equipped the surviving bacteria with “supergenes” that enable them to quickly resist new classes of antibiotics, even those to which they have never been exposed 1.

Examples of the “supergenes” are mutations that 1) enable bacteria to pump out several classes of antibiotics (through an efficient efflux pump), or that 2) alter the antibiotic binding sites on ribosomal subunits, so that several different classes of antibiotics can no longer inhibit those subunits. As a consequence, in recent years, by the time newer antibiotics have gone through
clinical trials and have reached the market, 20% or more of clinical isolates in the hospitals are already resistant to them at the time of regulatory approval, and within a few more years the majority of strains are resistant. 

Future Prospects for Phage Therapy 

Infectious disease experts have warned that there is now a compelling need to develop totally new classes of antibacterial agents, ones that cannot be resisted by the same genes that render bacteria resistant to antibiotics.

Phage therapy represents such a “new” class. We believe that the impediments cited above (bacterial debris in the preparations, rapid clearance in the body, etc.) can be overcome, freeing up the phages so that their attributes (such as exponential growth, and the ability to mutate against resistant bacteria) can be used to great advantage.

There are 3 additional attributes of phages that should be noted: Host specificity. While the host specificity is somewhat of a drawback (requiring a match up of phage to bacterial target, and/or the development of highly multi-valent phages), it also offers the great advantage that the phages will not kill other species of bacteria. 

* Conditions where phage efficacy is predicted to be reduced would include 1) hypoxic sites, where bacterial replication is slower and therefore phage replication is reduced; and 2) chronic obstructive pulmonary disease, where high acidity and proteases would be expected to inactivate some percentage of the phages. 

Thus, e.g., phage therapy is not likely to kill off the healthy flora of the intestines, lungs or urogenital tract, and it is therefore unlikely to provoke the illnesses and deaths seen when antibiotics cause overgrowth of pathogens (such as Clostridia difficile and Candida albicans).

Genetic engineering. It is possible to genetically engineer phages to express new traits of potential value. In so doing, scientists will have to deal with the legitimate concerns of regulatory agencies concerning recombinant organisms. The regulatory obstacles may be well worth the price, given the powerful engineering tools that are currently available.

Ideal candidates for co-therapy with antibiotics. If a given bacterium acquires resistance to a phage (e.g. by a mutation in the receptor site or in the endonuclease enzymes), that mutation is not likely to “teach” the bacterium to resist the antibiotics (which do not target those structures). Similarly, if a given bacterium acquires resistance to an antibiotic (e. g. by a mutation in the reflux pump or in the ribosomal subunits), that mutation is not likely to “teach” the bacterium to resist the phage (which does not target those structures). Thus, if the bacterium is exposed to both agents, the odds are remote that any resistance genes it starts to express (or acquires anew) will enable it to survive. There are reports that bacteria tend to mutate against antibiotics once in every 106 divisions, while they tend to mutate against phages once in every 107 divisions. 

Therefore the odds of a given bacterium mutating against a phage and an antibiotic at the same time would be the product of 106×107, meaning it would likely take 1013 bacterial divisions for such a double mutation to occur. Given that low probability, the co-administration of phages and antibiotics may help prevent the emergence of bacterial resistance to antibiotics, thereby greatly prolonging their clinical usefulness (and vice versa). Just as multiple classes of anti-HIV medications are administered to AIDS patients, to prevent the emergence of resistant strains of that virus, so it is that co-therapy with phages and antibiotics may also prove to be of great clinical value.

Conclusion 

Multidrug-resistant bacteria have opened a second window for phage therapy. Modern innovations, combined with careful scientific methodology, can enhance mankind’s ability to make it work this time around. Phage therapy can then serve as a stand-alone therapy for infections that are fully resistant. It will also then be able to serve as a co-therapeutic agent for infections that are still susceptible to antibiotics, by helping to prevent the emergence of bacterial mutants against either agent.

References  

1. ACKERMANN H. -W. and DUBOW M. (1987): Viruses of prokaryotes
I: General properties of bacteriophages (chapter 7). Practical
applications of bacteriophages. CRC Press, Boca Raton,
Florida.
2. ALISKY J. et al. (1998): Bacteriophages show promise as antimicrobial
agents. J. Infect., 36, 5–15.
3. BARROW P. A. and SOOTHILL J. S. (1997): Bacteriophage therapy
and prophylaxis: rediscovery and renewed assessment of
the potential. Trends Microbiol., 5, 268–271.
4. BERCHIERI A. et al. (1991): The activity in the chicken alimentary
tract of bacteriophages lytic for Salmonella typhimurium.
Res. Microbiol., 142, 541–549.
5. D’HÉRELLE F. (1917): Sur un microbe invisible antagoniste des
bac. dysentériques. Crit. Rev. Acad. Sci. Paris, 165, 373.
6. D’HÉRELLE F. (1922): The bacteriophage: its role in immunity.
Williams and Wilkens Co. /Waverly Press, Baltimore, USA.
7. GEIER M., FRIGG M. E. and MERRIL C. (1973): Fate of bacteriophage
lambda in non-immune germ-free mice. Nature, 246,
221–222.
8. LEDERBERG J. (1996): Commentary. Proc. Natl. Acad. Sci.
USA, 93, 3167–3168.
9. LEVIN B. and BULL J. J. (1996): Phage therapy revisited: the
population biology of a bacterial infection and its treatment
with bacteriophage and antibiotics. Am. Naturalist, 147, 881–898.
10. LEVY S. (1992): The antibiotic paradox. Plenum Press, New
York.
11. MERRIL C. et al. (1996): Long-circulating bacteriophage as
antibacterial agents. Proc. Natl. Acad. Sci. USA, 93, 3188–
2192.
12. MURRAY B. (1998): Diversity among multidrug-resistant Enterococci.
Emerging Infect. Dis., 4, 37–47.
13. SHRAYER D. (1996): Felix d’Hérelle in Russia. Bull. Inst. Pasteur,
94, 91–96.
14. SMITH H. W. and HUGGINS R. B. (1982): Successful treatment
of experimental E. coli infections in mice using phage: its general
superiority over antibiotics. J. Gen. Microbiol., 128, 307–318.
15. SMITH H. W. and HUGGINS R. B. (1983): Effectiveness of
phages in treating experimental E. coli diarrhoea in calves, piglets
and lambs. J. Gen. Microbiol., 129, 2659–2675.
16. SMITH H. W. and HUGGINS R. B. (1987): The control of experimental
E. coli diarrhea in calves by means of bacteriophage.
J. Gen. Microbiol., 133, 1111–1126.
17. SMITH T. et al. (1999): Emergence of vancomycin resistance in
Staphylococcus aureus. N. Engl. J. Med., 340, 493–501.
18. SOOTHILL J. S. (1992): Treatment of experimental infections of
mice with bacteriophages. Med. Microbiol., 37, 258–261.
19. SUMMERS W. C. (1998): D’Hérelle. Yale University Press (in
press).
20. S´LOPEK S. and KUCHAREWICZ-KRUKOWSKA A. (1987): Immunogenic
effect of bacteriophage in patients subjected to phage
therapy. Arch. Immunol. Ther. Exp., 35, 553–561.
21. S´LOPEK S., KUCHAREWICZ-KRUKOWSKA A., WEBER-DA˛BROWSKA
B. and DA˛BROWSKI M. (1985): Results of bacteriophage
treatment of suppurative bacterial infections. IV. Evaluation of
R. M. Carlton: Phage Therapy in the Past and Future 273
the results obtained in 370 cases. Arch. Immunol. Ther. Exp.,
33, 219–240.
22. S´LOPEK S., KUCHAREWICZ-KRUKOWSKA A., WEBER-DA˛BROWSKA
B. and DA˛BROWSKI M. (1985): Results of bacteriophage
treatment of suppurative bacterial infections VI. Analysis of
treatment of suppurative staphylococcal infections. Arch. Immunol.
Ther. Exp., 33, 261–273.
23. S´LOPEK S., WEBER-DA˛BROWSKA B., DA˛BROWSKI M. and KUCHAREWICZ-
KRUKOWSKA A. (1987): Results of bacteriophage
treatment of suppurative bacterial infections in the years 1981–
1986. Arch. Immunol. Ther. Exp., 35, 569–583.
24. VAN HELVOORT T. (1992): Bacteriological and physiological
research styles in the early controversy on the nature of the
bacteriophage phenomenon. Med. Hist., 36, 243–270.
25. WALDVOGEL F. (1999): New resistance in Staphylococcus
aureus. N. Engl. J. Med., 340, 556–557.
26. WEBER-DA˛BROWSKA B., DA˛BROWSKI M. and S´ LOPEK S.
(1987): Studies on bacteriophage penetration in patients subjected
to phage therapy. Arch. Immunol. Ther. Exp., 35, 563–568.
Received in May 1999
Accepted in June 1999


274 R. M. Carlton: Phage Therapy in the Past and Future 

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

 Bdellovibrio, a Predatory Bacteria that has Future Potential in Fighting the Diseases That Kill Us.
 Dr. Liz Sockett. Liz

 

Bdellovibrio are natural, tiny predatory bacteria that have other bacteria as their main food. Their strategy is to break into bacterial cells, close up the hole behind them suck their guts out! Having dissolved the inside of
 
Chris - What has the genetic information told you, and how can they be used in fighting infection?

Liz - We found that Bdellovibrio have lots of genes that make bacteria-dissolving enzymes. They also secrete a juice that breaks down chromosomes. These will both kill bacteria. Bdellovibrio don't look for any specific target sites on their bacteria prey, so there is no way for the bacteria to hide. Unfortunately, Bdellovibrio can't get into MRSA, but they can get into many others. We hope to use them on things like burns and leg ulcers, although more testing needs to be carried out. We might even be able to take genes and put them in the bacteria so they can attack MRSA.

Chris - What happens when the infection has cleared up?

Liz - If the Bdellovibrio burst out of the dead bacterium and can't find any more food, they just die. This makes it a self-terminating therapy that leaves no residue. They are likely to be best for wound infections, as eating them will kill good and bad stomach bacteria, and they also end up going down the loo [with unforeseen consequences]. 

December 2004
                         .............................................

              
https://www.nature.com/articles/nrmicro959


Opinion

Bdellovibrio as therapeutic agents: a predatory renaissance?

  • Nature Reviews Microbiology volume 2, pages 669675 (2004)
  • doi:10.1038/nrmicro959
  • Download Citation
Published:
Credit: Dr Andrew Lovering, University of Birmingham

In the fight against antimicrobial resistance, scientists discover how bacterial predators evolved to kill other bacteria without harming themselves.
  • Bacteria-killing bacteria (“predatory bacteria”) may assist humans in fighting pathogens in the post-antibiotic era
  • How predatory bacteria function has been little understood to date
  • Predators have been found to produce a protein “antidote” that protects them from their own weapons
  • Self-protection technique allows one bacterium to destroy others
  • The findings offer clues to how bacterial predation may have first evolved
  • Understanding how these predators attack bacteria could provide new ways of combatting antimicrobial resistance
A joint study by the labs of Dr Andrew Lovering and Prof Liz Sockett, at the Universities of Birmingham and Nottingham, has shown how predatory bacteria protect themselves from the weapons they use in their bacterial killing pathway. The research, published in Nature Communications, offers insights into early steps in the evolution of bacterial predators and will help to inform new ways of combatting antimicrobial resistance.

A useful predatory bacterium called Bdellovibrio bacteriovorus eats other bacteria (including important pathogens of humans, animals and crops). It attacks them from inside out using enzymes (called DD-endopeptidases) that first loosen the cell walls of prey bacteria and then cause them to round up like a pufferfish, providing space as a temporary home for the predator. However, Bdellovibrio also have similar cell walls so why don’t they fall victim of their own attack?
The project, funded by the Biotechnology and Biological Sciences Research Council (BBSRC), found that the bacterium uses an ankyrin-type protein called Bd3460 as a shield. It binds to the tip of the enzyme weapons, nullifying their action until they are safely secreted out of the Bdellovibrio and into the prey bacteria.

Dr. Andrew Lovering and Ian Cadby at the University of Birmingham determined the structure of the ankyrin protein using X-ray crystallography and found that that it attaches to two DD-endopeptidase weapons to temporarily deactivate them.

“When I first showed this to Liz, she hit the nail on the head by describing it as a decorative “quiff” on top of the endopeptidase” said Dr Lovering. “This covers up the active site of the enzymes that are used to cut cell walls and offers protection to the Bdellovibrio until these weapons are excreted into the prey.”

Carey Lambert, Rob Till and Prof Liz Sockett at the University of Nottingham confirmed the antidote protein’s use when the gene responsible for its production was deleted.
Prof Liz Sockett: “When the bd3460 gene responsible for antidote production was deleted, the Bdellovibrio had no way of protecting itself from its own weapons. When it attacked harmful bacteria with its cell-wall-damaging enzymes it also felt the effects.

“The Bdellovibrio bacteria lacking the bd3460 gene tried to invade the bacteria but suddenly rounded up like pufferfish and couldn’t complete the invasion – the fatter predator cell could not enter the prey cell.”

This is the first paper to discover a ‘self-protection’ protein in predatory bacteria.

Prof Liz Sockett added, “Most bacteria are not predatory and so understanding these mechanisms gives us a glimpse of how predation evolved. In this case it seems that the bd3460 gene was transferred into ancestors of Bdellovibrio, probably when they were beginning to develop as predators.”

Commenting on the potential impact of the study, Dr Andrew Lovering added: “If we are to use Bdellovibrio as a therapeutic in the future, we need to understand the mechanisms underpinning prey killing and be sure that any self-protective genes couldn’t be acquired by pathogens, causing resistance. Brilliantly, Liz and Carey have demonstrated this did not happen with the bd3460 antidote protein, and Ian and I showed how the mechanism works on predator enzymes only – this is a great inter-university collaboration.”

Notes to editor

Contact

Chris Melvin, 01793 414 694, Chris.melvin@bbsrc.ac.uk
BBSRC is one of the UK Research Councils. The Research Councils, led by the Medical research Council, are working on a joint Antimicrobial Resistance (AMR) Programme to find new ways to kill infectious pathogenic bacteria that are drug-resistant. Bdellovibrio is a natural predator and understanding the mechanisms it requires for successful predation provides vital knowledge towards these aims.

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



https://en.wikipedia.org/wiki/Bdellovibrio



Bdellovibrio

From Wikipedia, the free encyclopedia


Bdellovibrio
Slice from electron cryotomogram of Bdellovibrio bacteriovorus cell.jpg
Central slice through a cryotomogram of an intact Bdellovibrio bacteriovorus cell. Scale bar 200 nm
Scientific classification
Kingdom: Bacteria
Phylum: Proteobacteria
Class: Deltaproteobacteria
Order: Bdellovibrionales
Family: Bdellovibrionaceae
Genus: Bdellovibrio
Species: B. bacteriovorus
Binomial name
Bdellovibrio bacteriovorus
Stolp & Starr 1963

Appearance

Under a light microscope, host dependent Bdellovibrio appears to be a comma-shaped motile rod that is about 0.3–0.5 by 0.5–1.4 µm in size with a barely discernible flagellum. Bdellovibrio show up as a growing clear plaque in an E. coli lawn.
Another notable feature of Bdellovibrio is the sheath that covers its flagellum. This is a rare characteristic among bacteria. Flagellar motility stops after Bdellovibrio penetrates its prey, and the flagella is shed.

Host independent Bdellovibrio appear amorphous, and larger than their predatory phase.

Culture conditions

B. bacteriovorus seems to be pretty ubiquitous in nature and manmade habitats. They have been found in soil samples, rhizosphere of plant roots, rivers, oceans, sewage, intestines and feces of birds and mammals, and even in oyster shells and the gills of crabs.[4] B. bacteriovorus are able to thrive in almost any habitat, the general requirements are that there needs to be oxygen and some other Gram-negative bacteria present in its environment. Its optimal temperature is between 28-30C, making B. bacteriovorus a mesophile. Bdellovibrio is grown in the laboratory in its stationary HI (host indepdent) phase at 29°C on yeast peptone broth agar. Host dependent (predatory) cultures are grown with a population of E. coli S-17 at 29 °C for 16hrs.[2] They may also be cultured using YPSC(yeast extract, peptone, sodium acetate, calcium chloride) overlays or prey lysates.[citation needed]

Life cycle and parasitism

 Bdellovibrio cells can swim as fast as 160 µm/s, or over 100 times their length per second. It swims using a single sheathed polar flagellum with a characteristic dampened filament waveform. Bdellovibrio attacks other Gram-negative bacteria by attaching itself to the prey cell's outer membrane and peptidoglycan layer, after which it creates a small hole in the outer membrane. The Bdellovibrio cell then enters the host periplasmic space. It remains reversibly attached to it for a short "recognition" period. After the recognition period, it becomes irreversibly attached via the pole opposite the flagellum. Once inside the periplasm, the Bdellovibrio cell seals the membrane hole and converts the host cell to a spherical morphology, this is due to secretion of L,D transpeptidases which breaks the peptidoglycan apart, and therefore causes the cell to become amorphous. The two-cell complex formed is called a bdelloplast. The Bdellovibrio cell uses hydrolytic enzymes to break down the host cell molecules, which it uses to grow filamentously. When the host cell nutrients are exhausted, the filament septates to form progeny Bdellovibrios. The progeny become motile before they lyse the host cell and are released into the environment. The entire life cycle takes three to four hours, and produces an average of 3–6 progeny cells from a single E. coli, or up to 90 from larger prey such as filamentous E. coli.[6]

Targets of Bdellovibrio species, including Vibrio vulnificus, may undergo co-infection by Bdellovibrio and bacteriophage.[7] Although the Bdellovibrio rounding of prey is thought to be evolved to reduce co-infection of multiple Bdellovibrio, larger prey that do not round may be infected by multiple Bdello's.

Genomics

The genome of Bdellovibrio bacteriovorus HD100 was sequenced in 2004.[8] The HD100 genome is 3782950 nucleotides long, larger than expected given its small size.[9]
                
                                   ''''''''''''''''

References
 1.      Rittenberg SC, Shilo M (April 1970). "Early host damage in the infection cycle of Bdellovibrio bacteriovorus". Journal of Bacteriology. 102 (1): 149–60. PMID 4908670.
2.    Hobley L, Lerner TR, Williams LE, Lambert C, Till R, Milner DS, et al. (November 2012). "Genome analysis of a simultaneously predatory and prey-independent, novel Bdellovibrio bacteriovorus from the River Tiber, supports in silico predictions of both ancient and recent lateral gene transfer from diverse bacteria". BMC Genomics. 13: 670. doi:10.1186/1471-2164-13-670. PMC 3539863?Freely accessible. PMID 23181807.
3.    Koval SF, Hynes SH, Flannagan RS, Pasternak Z, Davidov Y, Jurkevitch E (January 2013). "Bdellovibrio exovorus sp. nov., a novel predator of Caulobacter crescentus". International Journal of Systematic and Evolutionary Microbiology. 63 (Pt 1): 146–51. doi:10.1099/ijs.0.039701-0. PMID 22368169.
4.  Shemesh, Y (2003). "Small eats big: ecology and diversity of Bdellovibrio and like organisms, and their dynamics in predator-prey interactions". Agronomie. 23: 433–439.
Madigan MT (2011-01-07). Brock Biology of Microorganisms: Global Edition. Pearson Education. ISBN 978-0-321-73551-5

5. Madigan MT (2011-01-07). Brock Biology of Microorganisms: Global Edition. Pearson Education. ISBN 978-0-321-73551-5.


6  Strauch E, Beck S, Appel B (2007). "Bdellovibrio and Like Organisms: Potential Sources for New Biochemicals and Therapeutic Agents?". Predatory Prokaryotes. Microbiology Monographs. 4. p. 131. doi:10.1007/7171_2006_055. ISBN 978-3-540-38577-6.
Chen H, Williams HN (2012). "Sharing of prey: coinfection of a bacterium by a virus and a prokaryotic predator". mBio. 3 (2): e00051–12. doi:10.1128/mBio.00051-12. PMC 3345577?Freely accessible. PMID 22511350.


7   Chen H, Williams HN (2012). "Sharing of prey: coinfection of a bacterium by a virus and a prokaryotic predator". mBio. 3 (2): e00051–12. doi:10.1128/mBio.00051-12. PMC 3345577?Freely accessible. PMID 22511350.

8. Rendulic S, Jagtap P, Rosinus A, Eppinger M, Baar C, Lanz C, Keller H, Lambert C, Evans KJ, Goesmann A, Meyer F, Sockett RE, Schuster SC (January 2004). "A predator unmasked: life cycle of Bdellovibrio bacteriovorus from a genomic perspective". Science. 303 (5658): 689–92. doi:10.1126/science.1093027. PMID 14752164.

9.  Tudor JJ, McCann MP (2007). "Genomic Analysis and Molecular Biology of Predatory Prokaryotes". Predatory Prokaryotes. Microbiology Monographs. 4. p. 153. doi:10.1007/7171_056. ISBN 978-3-540-38577-6.
                               .......................................................

https://www.bmj.com/content/329/74

The tabloid fixation on superbugs

BMJ 2004; 329 doi: https://doi.org/10.1136/bmj.329.7465.578 (Published 02 September 2004) Cite this as: BMJ 2004;329:578


  1. Peter Wilson, consultant microbiologist (peter.wilson@uclh.org)
    Author affiliations
“Superbug crisis worse than feared,” “Superbug kills 22 in one hospital in a year,” “Our squalid hospitals: no wonder the MRSA superbug is so rife,” “We find 80 times danger level of MRSA in hospital.” The media in the United Kingdom have developed a fascination for methicillin resistant Staphylococcus aureus recently. The New York Times was moved to observe that “newspapers around the country have been clamoring to find victims and to publish their sordid stories.” Most articles centre on poor hand hygiene of staff and the state of cleanliness of the hospitals, illustrating the problem with some unfortunate patient's story. Some even include cases of methicillin sensitive S aureus (MSSA), particularly if it happens to involve a minor celebrity.


There is little doubt that MRSA infection can be difficult to treat and may spread easily to some patients. Infections prolong stays in hospital and can increase mortality. The number of lawsuits citing MRSA infection is increasing exponentially. However, MRSA has been a constant problem in many UK hospitals since 1993, so why has attention become so intense now?


Part of the coverage stemmed from the publication in mid-July of the National Audit Office's report on hospital acquired infection, which noted disappointing progress since its last report on the subject four years ago. Lack of mandatory surveillance was a major problem. High bed occupancy was common practice in order to meet performance targets but made separation of elective and emergency patients difficult. The first page of the report gave a map of Europe, derived from a 2002 survey, that showed that the United Kingdom had the highest proportion of methicillin resistance in S aureus causing bacteraemia. S aureus bacteraemia had increased by 8% since 2001 and MRSA bacteraemia by 5%, representing 7647 cases a year.



Embedded Image

A few days ahead of the report the health secretary tried to soften its impact by releasing an outline of his intended measures to combat MRSA. These included installing hotline phones by patients' beds to alert cleaning staff, publication of infection rates, flying in experts to advise, and asking patients to police the hand hygiene of hospital staff. Some of these ideas attracted angry letters from UK experts and patients' groups. Then, in response to an MP's question, the Office of National Statistics released crude data on numbers of deaths in hospital that were possibly related to MRSA, which promptly appeared in the press as a league table. However, the figures took no account of the underlying medical conditions, mix of patients, or the number of patients admitted already carrying the organism. Furthermore, inclusion of MRSA on a certificate very much depends on the interests and diligence of the doctor. Private healthcare companies reported very low rates of MRSA bacteraemia, probably because they have a high proportion of single rooms, so that contact and airborne transmission is reduced. However, NHS consultants observed that the most susceptible patients are usually managed in NHS hospitals and that the capital and staffing costs to provide similar accommodation in NHS hospitals would be immense.


To that extent the coverage was understandable, if lurid at times. However, throughout the early months of this year the main focus had been on poor standards of hospital cleanliness, with MRSA as the benchmark. Undercover reporters were sent to examine hospitals. Bloodstained walls, overflowing clinical waste bins, and a culture of laziness in cleaning staff were frequent complaints, although no analysis was done to establish which failings would be likely to cause infections in patients. Hospital trusts said their buildings were old, cramped, and often had little storage, all complaints familiar to hospital staff. But hospitals need to be kept clean whatever their condition. More investment was needed in providing cleaning services and in refurbishment. Although use of alcohol hand rub was increasing, compliance with hand hygiene remained poor and was worse when staffing levels were low.


More worrying was a string of reports on the activities of an unaccredited laboratory conducting clandestine environmental sampling in hospital entrances, shops, public transport, and food shops. MRSA was apparently found in a high proportion of sites, resulting in sensational headlines and more newspaper sales. Even ticket machines and escalators in train stations were said to have traces of MRSA greatly exceeding “danger level,” though the reports failed to define what that might be. When some institutions conducted their own tests and found no evidence of MRSA anywhere, the same papers did not question the performance and validation of their own tests or ask for independent review. Publication of scientific results without peer review or opportunity for scientific debate has long been one of the less attractive pastimes of some of the press. A peer reviewed study from St Thomas' Hospital did appear at this time showing contamination of the hospital environment by MRSA, but in the wards and patient rooms, and it did not examine communal areas.



Embedded Image

The tabloids love MRSA
Credit: KARI LOUNATMAA/SPL

MRSA is well adapted to causing disease in hospital patients, particularly those with wounds. Infections with MRSA can be more difficult to treat than infections with MSSA but are hardly a new phenomenon. Britain has a particular problem with the organism and needs to move away from a low cost, high turnover ethic in the NHS to an ethic of investing in and allowing time for cleaning. Staffing levels need to rise to allow individuals to practise better hand hygiene. Hospital architects should be trained in infection control. MRSA does contaminate the local environment of hospital patients who carry the organism and may sometimes be further spread by hand after contact with these surfaces. However, even if MRSA exists further afield in the community it is at a low level, and there is no evidence that it causes additional infections. Raising public awareness can be helpful, but the creation of a climate of fear among patients entering hospital is more likely to increase newspaper sales than to provide a solution. Clearly a part of the press considers MRSA to be a useful political stick. Objective reports such as that of the National Audit Office are more helpful to microbiologists at the front line than sensational stories based on questionable results.
View Abstract

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

https://search.proquest.com/openview/17812


https://www.medscape.com/viewarticle/554935

Resistant 'Superbugs' Create Need for Novel Antibiotics

Teri Capriotti, DO, MSN, CRNP

A wide number of "smart" bacteria in the environment have "learned" how to resist the arsenal of antibiotics. Decades of overuse and misuse of antibiotics have caused this crisis: overzealous antibiotic prescribing by clinicians, excess use of antibacterial household products by the public, and widespread use of antibiotics in livestock. Antibiotics are ubiquitous within the environment. Bacteria are highly adaptable organisms which have an extraordinary ability to mutate in response to their environmental conditions. The widespread use of antibiotics has provided the conditions needed for bacteria to mutate genetically and develop resistance to these drugs.

How Bacteria Become ‘Superbugs'

Each time a new antibiotic is introduced and used widely, a small number of bacterial organisms decipher how to resist the drug's bactericidal effects. The bacteria that survive the effects of antibiotics are the most adaptable organisms which develop genome mutations or resistance genes. These genetically changed bacteria multiply to produce a population of antibiotic-resistant organisms. The resistant bacteria also can transfer their newly acquired resistance genes to other species of bacteria through the process of conjugation (a reproductive interaction). As bacteria reproduce and transfer resistance to other bacterial species, new strains develop which can resist the effects of existing antibiotics. Lethal and contagious, these antibiotic-resistant organisms have been termed superbugs. Staphylococci, enterococci, and pneumococci have proven the ability to develop superbug status (Rybak, 2004).

Methicillin-resistant staphylococcus aureus (MRSA) is probably the best known superbug. First observed in 1960, MRSA continued to increase slowly in the bacterial population, until clinicians realized its significant virulence in the 1980s-1990s. MRSA was once eradicated reliably by vancomycin (VancocinAE), an antibiotic of the glycopeptide class. With time, the MRSA bacteria became resistant to vancomycin and vancomycin-resistant staphylococcus aureus (VRSA) soon arose.


To add to the challenge, MRSA infection, once confined to clinical settings as a nosocomial problem, recently became a community-acquired infection. Its spread into the population at large has required a dual classification for MRSA infection: either community-associated MRSA (CA-MRSA) or health care-associated MRSA (HA-MRSA). CA-MRSA infection has involved the skin and soft tissue most commonly, with cases increasing worldwide. However, a specific strain of CA-MRSA has been the cause of a severe necrotizing pneumonia in otherwise healthy individuals. This virulent strain of MRSA has been under intense investigation since the first cases were diagnosed in 2002 (Francis et al., 2004).

The increasing incidence of community-acquired MRSA infection has created an urgent need for antibiotics with unique mechanisms of action. Most of the current available antibiotics are chemical modifications of existing agents with similar mechanisms. According to a study of emergency room patients in the Los Angeles area, skin and soft tissue infections caused by CA-MRSA have increased in incidence from 29% in 2001 to 64% in 2004 (Moran, Amii, Abrahamian, & Talan, 2005). Although nosocomial MRSA infections have been challenging clinicians since the early 1990s, CA-MRSA infection is a more ominous threat to the population as a whole and is increasing rapidly in incidence.

In 2003, 60% of infections due to S. aureus in intensive care patients were resistant to methicillin (National Nosocomial In fections Surveillance, 2004). HA-MRSA frequently causes bacteremia, sepsis, or pneumonia in clinical settings. HA-MRSA has been responsible for surgical site and wound infection, ventilator-associated pneumonia, and central line bacteremia.

In addition, MRSA has been successful in transmitting resistance genes to a completely different species of bacteria, enterococcus faecalis. MRSA has transmitted its gene for vancomycin resistance to enterococcus. This has led to the strain identified as vancomycin-resistant enterococcus (VRE) or glycopeptide-resistant enterococcus (GRE). VRE has created its own set of treatment challenges for clinicians (Pfeltz & Wilkinson, 2004).

Which Bacteria Are Called ‘Superbugs'?

In the 1990s, MRSA, VRSA, and VRE were the major superbugs which required clinical attention and pharmacological ingenuity. Sta phylococcus aureus and enterococcus were the glycopeptide-resistant gram-positive cocci of major concern. Some strains of staphylococcus, completely resistant to vancomycin, were called vancomycin-resistant staphylococcus aureus (VRSA). Some strains had some susceptibility to vancomycin and were called vancomycin intermediately susceptible staphylococcus aureus (VISA) or glycopeptide intermediately susceptible staph ylococcus aureus (GISA). This terminology also was used for enterococci: VRE and GRE (Pfeltz & Wilkinson, 2004; Shah, 2005).

As attention has been on staphylococcus and enterococcus for the past decade, another bacterium, streptococcus pneumoniae (also called pneumococcus), has been developing resistance. It classically has been a major cause of community-acquired infections, such as upper respiratory infections, bronchitis, pneumonia, otitis media, pharyngitis, and meningitis. Al though the bacterium was once eradicated easily with penicillin, significant antibiotic resistance has now become a major problem in strains of pneumococcus (Centers for Disease Control [CDC], 2003; Whitney et al., 2000).

Strains of S. pneumoniae have developed resistance to penicillin, trimethoprimsulfmeth oxazole (BactrimAE), macrolides (for example, azithromycin [Zith ro maxAE]), tetracyclines (for ex ample., minocycline [MinocinAE]), and fluoroquinolones (for example, ciprofloxacin [CiproAE]) (Hoff man-Roberts, Bab cock, & Mitro poulous, 2005; Karchmer, 2004). In 2002, the CDC reported that 34% of all S. pneumoniae infections were resistant to at least one antibiotic and 17% were resistant to three or more antibiotics (CDC, 2003). Drug-resistant streptococcus pneumoniae (DRSP) or penicillin-resistant streptococcus pneumoniae (PRSP) became the newest superbug of concern in 2002.


The trend of rising bacterial resistance continues to challenge health care providers. Antibiotic resistance has now become worthy of concern in pseudomonas aeruginosa, acinetobacter baumannii, and group A beta hemolytic streptococcus (GABHS, also called streptococcus pyogenes). These bacteria have not posed a major threat yet, but are anticipated to be the next highly resistant superbugs (Navon-Venezia, Ben-Ami, & Carmeli, 2005).

The health care literature uses a number of acronyms to describe the significant antibiotic-resistant bacteria which exist within the community and clinical settings. Nurses should be familiar with the terminology in Table 1 .

Antibiotics Developed to Combat MRSA, VRSA, and VRE

Glycopeptides

Recent worldwide emergence of CA-MRSA has prompted the development of several new antibiotics (see Table 2 ). In the past, as vancomycin became less effective against MRSA, clinicians turned to another glycopeptide, teico planin (Targ ocidAE). Glyco pepti des exert their bactericidal action by interfering with synthesis of the bacterial cell wall and RNA. However, due to the similarity of the glycopeptides, bacteria quickly developed resistance to teicoplanin. The short-lived success of teicoplanin as an alternative to vancomycin motivated pharmacologic investigators to develop different antibiotic approaches.

Streptogramins

The failure of glycopeptide antibiotics led to the creation of a new class of drug, the streptogramins, a combination of quini pristin and dalfopristin. Quinupristin/ dalfopristin (SynercidAE) became the drug of choice for MRSA nosocomial infections by the year 2000. It exerts bactericidal activity by interfering with protein synthesis at the bacterial ribosome, which differs from the glycopeptide strategy. The dalfopristin component interferes with early phases of bacterial protein synthesis and quinipristin interferes with the late phase of protein synthesis. The two act synergistically to hinder the synthesis of bacterial proteins (Medical Eco nomics, 2005).


However, Synercid has a major drawback. It is an intravenous medication requiring slow infusion within a large volume of fluid, and this administration method makes it impractical for the outpatient setting (Shah, 2005). Its limitation to parenteral administration was a particular disadvantage in light of the increasing incidence of community-acquired MRSA infections. It also has caused disabling myopathy as a side effect in some treated individuals. These drawbracks heightened the need for an oral form of the drug and further antibiotic development for resistant bacteria.

The oral streptogramin pristinamycin has been used in Europe in combination with doxycycline to combat MRSA infections. In a small trial of 53 patients, 74% (39 patients) were effectively cured of MRSA infection. More extensive clinical investigations with larger population groups are needed to evaluate this drug combination further (Dancer, Robb, Crawford, & Morrison, 2003). It is not available presently in the United States.

Newer Antibiotics with Promise For MRSA, VRSA, and VRE

Second generation glycopeptides: Dalbavancin and Orita vancin

With the first generation glycopeptides (vancomycin and teicoplanin) as prototypes, the second generation glycopeptides have been synthesized. Dalba vancin and oritavancin have shown bactericidal activity against MRSA, VRSA, VRE, and DRSP and are undergoing the final stages of clinical trials (Bradley, 2005; Hoffman-Roberts et al., 2005; Van Bambeke, Van Laetham, Cour valin, & Tulkens, 2004).

In clinical trials, dalbavancin has shown efficacy against ser ious gram-positive hospital infections, particularly MRSA and teicoplanin-resistant strains. Due to the drug's long half-life of 9-12 days, a dalbavancin treatment regimen consists of a once-a-week intravenous dose. The pharmacokinetics of this drug allow an intermittent dosing schedule which could be suitable for home intravenous therapy. A once-a-week dose schedule could obviate the need for continuous intravenous lines and decrease risk of iatrogenic local or bloodstream infections (Mushtaq, Warner, Johnson, & Livermore, 2004).


Oritavancin, like dalbavancin, is under investigation in the late phases of clinical trials. It ex hibits potent antimicrobial activity against vancomycin-resistant sta phylococcus aureus and enterococcus species. It has a long half-life of approximately 6-10 days, allowing for an infrequent dosing schedule. Oritavancin has dem onstrated efficacy in the treatment of complicated skin and soft tissue infections (Coyle & Rybak, 2001). Few studies elucidate the side effects and tolerability of dalbavancin and oritavancin. When available for patient use, both these drugs should be limited to treatment of only multi-drug resistant gram-positive bacterial infections.

Daptomycin (CubicinAE)

This cyclic lipopeptide represents a new class of antibiotic. This drug exhibits concentration-dependent bactericidal activity against resistant gram-positive bacteria by inhibiting protein synthesis, and DNA and RNA synthesis. It is bactericidal against staphylococcus aureus including MRSA, staphylococcus epidermidis (in cluding methicillin-resistant S. epidermidis [MRSE]), enterococcus including VRE, penicillin-resistant S. pneumoniae, and S. pyogenes (also called group A beta hemo lytic streptococcus [GABHS]). Daptomycin has been effective for the treatment of skin and soft tissue infections. However, it has not shown efficacy in pneumonia (Hirsch, 2004).

The drug was approved by the Food and Drug Admin istration (FDA) in 2003 for intravenous administration only. Dap tomycin should be administered at a dosage of 4 mg/kg in 0.9% sodium chloride over a 30-minute period once every 24 hours for 7-14 days. It is not compatible with dextrose intravenous solutions. Eliminated primarily by the kidney, the drug should have dosage modification for patients with renal impairment. It has not been tested sufficiently in children, or pregnant or nursing women. Currently daptomycin is undergoing investigation for treatment of bacteremia and endocarditis (Pham, 2003).


Antibiotics Developed to Combat DRSP

Linezolid (ZyvoxAE), an oxazolidinone, is an antibiotic specifically developed for resistant bacterial infections. Linezolid interferes with bacterial protein synthesis at the ribosome. However, it is only bacteriostatic for staphylococcus and enterococcus. Investigators have found it has bactericidal activity against DRSP, and gram-negative and anaerobic bacterial infections (Anderegg, Sader, Fritsche, Ross, & Jones, 2005; Medical Ec onomics, 2005). Line zolid is recommended for pneumonia caused by drug-resistant strep tococcus pneumoniae, complicated skin and soft tissue infections, and diabetic foot infections. Available in oral and parenteral forms, it can be taken with or without food. Possible adverse effects include myelosuppression, pseu do mem branous colitis, and lactic acidosis. Also some drug-drug interactions are particularly significant.

Repeated studies have shown that linezolid is superior to vancomycin in treating MRSA infections, particularly ventilator-associated pneumonia and surgical site infections (Conte, Golden, & Kipps, 2002; Weigelt et al., 2005; Wunderink, Cammarata, Oli phant, & Kollef, 2003). Linezolid has out-performed glycopeptides in both HA-MRSA and CA-MRSA infections. Unfortunately, increasing reports of resistance and treatment failures have been associated with linezolid since 2005 (Shah, 2005).

Telithromycin (KetekAE ) is a ketolide antibiotic which blocks bacterial protein synthesis. Structural derivatives of macrolide antibiotics, ketolides are erythromycin-type drugs. Telithro mycin has been designed uniquely to combat DRSP. It is indicated for treating upper and lower respiratory infections, such as acute sinusitis, chronic bronchitis, and community-acquired pneumonia (Medical Economics, 2005). Strep tococcus pneumoniae, haemo philus influenzae, streptococcus pyogenes, and moxarella cat arrhalis are susceptible to tel ithromycin. It also is active against some of the atypical respiratory pathogens such as chlamydia pneumoniae, legionella pneumophila, and mycoplasma pneumoniae (Skerret & Stratton, 2004). However, it does not cover MRSA, GRE, or any enteric gram-negative bacteria (Shah, 2005).


Telithromycin is available as an oral agent and can be taken without regard for meals. Total recommended dosage is 800 mg per day administered as 400 mg in the morning and evening. It is well absorbed and penetrates respiratory tissues rapidly. An 800 mg daily dose is recommended for 5 days for chronic bronchitis or sinusitis. For community-acquired pneumonia, 800 mg should be administered daily for 7-10 days (Aventis Pharma ceuticals, 2004).

Because telithromycin is metabolized mainly by the liver, dosage adjustment may be necessary in patients with liver impairment. Caution must be used when telithromycin is co-administered with other hepatically metabolized drugs. Hepatic dysfunction can occur and may become apparent by elevated liver enzymes, hepatitis, or jaundice. Particular attention is needed when ketoconazole (NizoralAE), itraconazole (SporanoxAE), anti-lipidemic stat ins, midazolam (VersedAE), and cisapride (PropulsidAE) are administered concomitantly with tel ithromycin because these drugs have direct hepatic effects. Concomitant administration of rifampin (RifadinAE) should be avoided. Phenobarbital (BarbitaAE, SolfotonAE, LuminalAE), phenytoin (DilantinAE), and carbamazepine (TegretolAE) will cause sub-therapeutic levels of telithromycin. Digitalis (DigoxinAE), theophylline (TheolairAE, TheodurAE, Bronk odylAE, Slo-bidAE), metaprolol (Lo pressorAE, ToprolAE), and oral contraceptive levels may be affected by telithromycin.

Prolongation of the QT interval on the electrocardiogram has been observed in patients on teli thromycin. This can increase risk of ventricular arrhythmias. Therefore, patients with risk of cardiac dysrhythmia, bradycardia, or potential for hypokalemia or hypomagnesemia should not take telithromycin. It also is contraindicated in patients with myasthenia gravis (Medical Economics, 2005).

The most common side effects reported are diarrhea, nausea, vomiting, headache, dizziness, and persistent unpleasant taste. Antibiotic-associated pseu do membranous colitis due to clostridium overgrowth in the bowel is a potential side effect of all potent antibiotics. This condition should be considered in all patients with persistent diarrhea. Transient vision disturbances, such as blurriness and diplopia, also have been reported, usually after the first or second dose. A problem with focusing from near to far has been described as a side effect by some recipients of telithromycin. Visual disturbances prohibit the patient from driving, operating machinery, or engaging in any potentially hazardous activity. Also, this drug should be used with caution in pregnant or nursing women only if benefit outweighs risks. The drug's safety has not been investigated thoroughly in children. Dosage of teli thromycin has not been established for patients with severe renal impairment (Aventis Phar maceuticals, 2004; Medical Economics, 2005).

Ceftobiprole medocaril is an injectable anti-MRSA cephalo sporin antibiotic in development. Cephalo sporins are related closely to penicillins and act by destroying the bacterial cell wall. This new cephalosporin is in phase III clinical trials for complicated skin and soft tissue infections as well as nosocomial pneumonia caused by resistant strains of MRSA, enterococci, and S. pneumoniae. The Ceftobiprole in Hospital-acquired Pulmonary In fections (CHOPIN) study is a randomized controlled trial which began in 2005. It is focusing on patients who develop ventilator-associated pneumonia (Basilea Pharma ceutica J & JPRD, 2005).

 New Pneumococcal Conjugate Vaccine 

Due to rising antibiotic resistance, pneumococcal infections are becoming increasingly difficult to treat with antibacterial agents. A vaccine is the best strategy to prevent pneumococcal infection in susceptible individuals. There are now two types of pneumococcal vaccine: pneumococcal polysaccharide vaccine (PneumovaxAE, Pnu-ImmuneAE) and pneumococcal conjugate vaccine (PrevnarAE). Pneumococcal polysaccharide vaccine has been available since 1977 and has been recommended approximately every 5-10 years for older adults and certain at-risk populations. However, this vaccine has not been effective in children under 2 years of age. In February 2000, the FDA approved another form of this vaccine, the pneumococcal conjugate vaccine (PrevnarAE). The pneumococcal conjugate vaccine can prevent invasive pneumococcal disease, which is the cause of serious, often fatal, forms of pneumococcal infection, such as meningitis, pneumonia, and bacteremia (CDC, 2005).

In the past, the pneumococcal polysaccharide vaccine was formulated based on certain epidemic serotypes of S. pneumoniae. With the emergence of resistant strains of the organism, a change in the vaccine has been necessary. The pneumococcal conjugate vaccine has been developed which can impart immunity against drug resistant strains of S. pneumoniae. Ex panded use of the newly formulated conjugate pneumococcal vaccine can provide protection against approximately 80% of resistant pneumococcal strains. This form of the vaccine is recommended for children under 2 years of age and other at-risk patient populations. Individuals with risk factors for pneumococcal infection can receive both forms of the vaccine (CDC, 2003).

Conclusion

Superbug resistance is escalating within the clinical setting and community at large. In novative antibiotic strategies are still lacking within the pharmaceutical industry to keep pace with the growing resistance, with a glaring absence of any novel class of antibacterial drug in the United States for decades. Most new antibiotics are chemical modifications of existing drugs and are quickly outsmarted by the bacteria in the environment. Clinicians are challenged by some strains of bacteria which are resistant to essentially all available antimicrobial agents. New antibiotics must be used with precision after the infectious organism is identified by culture and sensitivity testing. Using the exact antibiotic which specifically targets the identified organism is a key strategy to limit bacterial resistance.

In addition, health care pro viders must use all precautions to prevent spread of infection. Infection control procedures are essential. Health care pro viders need education regarding organism-specific guidelines. Ag gressive hand hygiene, use of gloves and gowns, patient isolation, and dedicated patient equipment are some of the recommended strategies. Im muni zation of susceptible individuals with pneumococcal conjugate vaccine can stave off serious drug-resistant pneumonia. Nurses can find succinct organism-specific MRSA and VRE precautions online (http://info.med.yale.edu/ynhh/infection/guidelines).Information about the pneumococcal conjugate vaccine can be obtained from the CDC (http://www.cdc.gov/nip).

Nurses are responsible for administering the new antibiotics to combat infections caused by superbugs. Increasing numbers of new agents are available and in clinical trials. All of the agents have different pharmacokinetics, routes of administration, and potential side effects. In addition to implementing infection control procedures, nurses will need to review specific prescribing information of new antibiotics. All health care providers, particularly in medical-surgical and critical care settings, need educational programs which disseminate information about new antibiotics and reinforce infection control procedures.



REFERENCES:
  1. Anderegg, T.R., Sader, H.S., Fritsche, T.R., Ross, J.E., & Jones, R.N. (2005). Trends in linezolid susceptibility patterns: Report from the 2002-2003 worldwide Zyvox Annual Appraisal of Potency and Spectrum (ZAAPS) Program. International Journal of Antimicrobial Agents, 26(1), 13-21.
  2. Aventis Pharmaceuticals. (2004). Prescribing information for Ketek (telithromycin) tablets. Retrieved October 6, 2005, from http://www.aventispharma-us.com
  3. Basilea Pharmaceutica J & JPRD. Ceftobiprole medocaril: Injectable anti-MRSA cephalosporin antibiotic. Retrieved October 4, 2005, from http://www.drugdevelopment-technology.com
  4. Bradley, J.S. (2005). Newer antistaphylococcal agents. Current Opinion in Pediatrics, 17(1), 71-77.
  5. Centers for Disease Control and Prevention (CDC). National Center of Infectious Diseases. Division of Bacterial and Mycotic Diseases. (2003). Fact sheet: Drug-resistant streptococcus pneumoniae disease. Retrieved August 20, 2005, from http://www. cdc.gov/ncidod/dbmd/diseaseinfo
  6. Centers for Disease Control and Prevention (CDC). (2005). Direct and indirect effects of routine vaccination of children with 7-valent pneumococcal conjugate vaccine on incidence of invasive pneumococcal disease - United States, 1998-2003. MMWR Morbidity and Mortality Weekly Report, 54(36), 893-897.
  7. Conte, J.E., Golden, J.A., & Kipps, J. (2002). Intrapulmonary pharmacokinetics of linezolid. Antimicrobial Agents and Chemotherapy, 46, 1475-1480.
  8. Coyle, E.A., & Rybak, M.J. (2001). Activity of oritavancin (LY333328), an investigational glycopeptide, compared to that of vancomycin against multidrug-resistant streptococcus pneumoniae in an in-vitro pharmacodynamic model. Antimicrobial Agents and Chemotherapy, 45(3), 706-709.
  9. Dancer, S.J., Robb, A., Crawford, A., & Morrison, D. (2003). Oral streptogramin in the management of patients with methicillin-resistant Staphylococcus aureus (MRSA) infections. Journal of Antimicrobial Chemo therapy, 51, 731-735.
  10. FDA Approves Tygacil, First-in-Class Antibiotic. (2005). Retrieved October 1, 2005, from http://www.wyeth.com/news
  11. Francis, J.S., Doherty, M.C., Lopatin, U., Johnston, C.P., Sinha, G., Ross, T., et al. (2004). Severe community-onset pneumonia in healthy adults caused by methicillin-resistant staphylococcus aureus carrying the Panton-Valentine leukocidin gene. Clinical Infectious Disease, 40(1), 100-107.
  12. Hirsch, A. (2004). Daptomycin (Cubicin): A new treatment option for gram positive infections. Retrieved October 1, 2005, from http://www.clevelandclinicmeded.com
  13. Hoffman-Roberts, H.L., Babcock, E., & Mitropoulous, I.F. (2005). Invest igational new drugs for the treatment of resistant pneumococcal infections. Expert Opinion in Investigational Drugs, 14(8), 973-995.
  14. Karchmer, A.W. (2004). Increased antibiotic resistance in respiratory tract pathogens: PROTEKT US – an update. Clinical Infectious Disease, 39(Suppl. 3), S142-S150.
  15. Medical Economics. (2005). Physician's desk reference. Montvale, NJ: Medical Economics.
  16. Moran, G.J., Amii, R.N., Abrahamian, F.M., & Talan, D.A. (2005). Methicillin-resistant Staphylococcus aureus in community acquired skin infections. Emerging Infectious Diseases, 11(6), 928-930.
  17. Mushtaq, S., Warner, M., Johnson, A.P., & Livermore, D.M. (2004). Activity of dalbavancin against staphylococci and streptococci, assessed by BSAC and NCCLS agar and dilution methods. Journal of Antimicrobial Chemo therapy, 54(3), 617-620.
  18. , D. (2005). Tigecycline: Clinical evidence and formulary positioning. International Journal of Antimicrobial Agents, 25(3), 185-192.
  19. Navon-Venezia, S., Ben-Ami, R., & Carmeli, Y. (2005). Update on Pseudomonas aeruginosa and Acinetobacter baumannii infections in the healthcare setting. Current Opinions in Infectious Disease, 18(4), 306-313.
  20. National Nosocomial Infections Surveillance. (2004). System report, data summary from January 1992 through June 2004, issued October 2004. American Journal of Infection Control, 32, 470-485.
  21. Pfeltz, R.F., & Wilkinson, B.J. (2004). The escalating challenge of vancomycin resistance in Staphylococcus aureus. Current Drug Targets of Infectious Disorders, 4(4), 273-294.
  22. Pham, P.A. (2003). FDA approves daptomycin, a new cyclic lipopeptide antibiotic, for the treatment of resistant gram positive organisms. Retrieved October 1, 2005, from http://www.cubistpharmaceut icals.com
  23. Rybak, M.J. (2004). Resistance to antimicrobial agents: An update. Pharmacotherapy, 24(12 Pt 2), 203S- 215S.
  24. Shah, P.M. (2005). The need for new therapeutic agents: What is the pipeline? Clinical Microbiological Infections, 11(Suppl. 3), 36-42.
  25. Skerret, S., & Stratton, C. (2004). New antibiotics useful in primary care. Retrieved August 11, 2005, from http://www.jaapa.com/issues/ j20040601/articles/antibiotics.
  26. Smith, K.L., McCabe, S.M., & Aeschlimann, J.R. (2005). Tigecycline: A novel glycylcycline antibiotic. Retrieved October 1, 2005, from http://www. formularyjournal.com
  27. Van Bambeke, F., Van Laetham, Y., Courvalin, P., & Tulkens, P.M. ( 2004). Glycopeptide antibiotics: From conventional molecules to new derivatives. Drugs, 64(9), 913-936.
  28. Weigelt, J., Itani, K., Stevens, D., Lau, W., Dryden, M., & Knirsch, C. (2005). Linezolid versus vancomycin in treatment of complicated skin and soft tissue infections. Antimicrobial Agents and Chemotherapy, 49, 2260-2266.
  29. Whitney, C.G., Farley, M.M., Hadler, J., Harrison, L.H., Lexau, C., Reingold, A., et al. (2000). Increasing prevalence of multi-drug resistant Streptococcus pneumoniae in the United States. New England Journal of Medicine, 343(26), 1917-1924.
  30. Wunderink, R.G., Cammarata, S.K., Oliphant, T.H., & Kollef, M.H. (2003). Continuation of a randomized, double blind, multicenter study of linezolid versus vancomycin in the treatment of patients with nosocomial pneumonia. Clinical Therapeutics, 25, 980-992.



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




Biocontrol Ltd (UK) presents data from First Superbug Clinical Trial

22 February 2008


Technology to defeat bacterial infections shows positive results

The use of natural controls against serious bacterial infection and superbugs is being led by a UK company.  Scientists at Biocontrol Ltd have today presented at the Bacteriophage 2008 meeting in Hertfordshire a first look at the results of their initial Phase II clinical trial.  This is the first fully-regulated clinical trial to test whether phage therapy really works and it shows positive results for Biocontrol’s innovative treatments, which attack and destroy previously untreatable bacterial infections. 

Set up in 1997, Biocontrol has been developing the clinical use of bacteriophages – literally “eaters of bacteria” – that attack dangerous infection-causing bacteria.  First discovered in the early 20th century, bacteriophages, or phages, are naturally occurring viruses that attack and destroy harmful bacteria.  They are highly specialised, usually attacking only specific strains of a single species of bacteria. 

Biocontrol’s clinical trial has been testing a treatment against the Pseudomonas aeruginosa bacteria – which is highly and increasingly resistant to traditional antibiotics and is a potential killer, especially when it infects the lungs – a future target for the Company.  

Pseudomonas aeruginosa is also a major cause of ear infections, including the outer ear infection known as “swimmer’s ear.”  Over a period of 17 months, from July 2006 to November 2007, a double-blind Phase II clinical trial took place at a specialist London hospital involving 24 patients with chronic ear infections that were not responding to antibiotic treatments.  Results reported by both the patients who received Biocontrol’s phage treatment and the medical staff treating them, showed improvements.  This amounted to a mean 50% reduction in symptoms, compared to a mean of only 20% in the control group who did not receive phages.

Among the most striking of the trial’s findings were the bacterial counts, analysed from samples taken from patients’ ears at periods of one, three and six weeks after the treatments were applied.  In the test group of patients, the mean count of Pseudomonas aeruginosa bacteria present dropped by an average of around 80% by week three, and stayed there.  By contrast, in the control group, the mean levels of bacteria showed a small increase over the same period.

Biocontrol’s founder and Chief Scientific Officer, Dr David Harper, commented: “This is the first fully-regulated double-blind clinical trial of the efficacy of a phage treatment, and is exactly what is needed to find out whether such a treatment really works.  The fact that patients seem to have been getting better is the most exciting thing.  Obviously a major part of the trial was monitoring to ensure that the treatments were safe – and there were no reportable safety events throughout, another very positive result.

“Originally designed for 40 patients, the trial was halted after 24, as patients seemed to be getting better.  The Company, working with the clinicians in charge, has decided to move on with bringing such a potentially important treatment to the market as quickly as possible.  We are also planning to offer phage treatment to the patients who did not receive it during the trial.”

Commenting on the successful outcome, the clinical director of the trials, Professor Tony Wright, Consultant ENT surgeon at the Royal National Throat, Nose and Ear Hospital in London, said: “All the patients had long histories of ear problems that had failed to respond to all sorts of treatments including topical and oral antibiotics, and sometimes even surgery.  Some had had problems for more than 40 years.” 

The next step for Biocontrol is to obtain permission for the pivotal Phase III clinical trial for the treatment that has now completed the Phase II trial.

Dr Harper said: “We want to get the Phase III trial for the ear treatment up and running as soon as possible.  In addition, Pseudomonas aeruginosa bacteria are particularly dangerous when infecting the lungs of patients with cystic fibrosis.  80% of adult CF sufferers have chronic lung infections and it is the major cause of death in these patients.  We are currently working on the development of an aerosol treatment that we hope will also produce a positive outcome in trials that we plan to start in late next year.  


“We are also fundraising now to be able to take these trials forward, and are considering an IPO in 2009 to secure the levels of investment needed to take our products to market.”

Biocontrol is based in London, and as part of ongoing expansion, the Company has recently opened laboratories in Nottingham, close to the planned centre for next year’s trial.  It has taken space in the BioCity complex – the leading centre for bioscience and healthcare sector companies in the East Midlands.

Welcoming Biocontrol, BioCity Nottingham’s Chief Executive Officer, Glenn Crocker, said: “We are delighted that Biocontrol has joined us at BioCity Nottingham.  It is exactly the sort of fast growing company, developing truly innovative healthcare products that we were established to attract.  Everyone is aware of the problems dealing with bacterial infections and Biocontrol’s products are increasingly being shown to be an effective solution.”

Regulatory note: The information contained in this release relates to the outcomes of an initial (Phase 1/2) clinical trial.  The experimental treatment referred to will require successful completion of additional, large scale trials before being the subject of application for marketing approvals by appropriate regulatory bodies.  It will not be available for the general treatment of patients until such a process is completed.


                              

Company Overview

Biocontrol Limited develops biological agents for controlling bacterial infection with pseudomonas aeruginosa in United Kingdom. Its bacteriophage therapeutics are used to treat pseudomonas aeruginosa, ear infections, cystic fibrosis, hospital-acquired infections, and burns. The company was incorporated in 1997 and is based in London, United Kingdom. As of January 7, 2011, Biocontrol Limited operates as a subsidiary of AmpliPhi Biosciences Corporation.
    ..........................................................................................................................................

Tackling Problems Such As Sleeping Sickness With Bacteriophages

Although it is not always possible to directly treat many serious infections, such as Sleeping Sickness (which Doctors Without Borders warns may threaten as many as 60 Million people), with Phage Therapy, but as the following article shows there is the possibility of interfering with the life cycle of the offending insects where a symbiotic bacteria is an essential part of their life cycle. It opens up  fascinating potential for research in this area.
 

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

Tuesday, January 6, 2009


SMART: SCOTLAND Supporting Scottish Innovation Case Studies

IN-PHAGE LTD

Winner of SMART Award



It can take up to 5 years to get a new pesticide to market.

The SMART Award effectively kick-started this process, allowing a full-time researcher to be employed in the laboratory whilst the founders concentrated on developing the commercial side of the business.'



Drs Alison Blackwell and Sue Welburn were both members of staff at the University of Edinburgh's Centre for Tropical Veterinary Medicine when they set up In-Phage Ltd with seed corn funding from the Edinburgh Technology Fund. Both are entomologists but working at different ends of the spectrum; Alison on insect ecology, behaviour and control, whilst Sue is a molecular entomologist, with particular interest in tsetse fly transmitted sleeping sickness. Between them, a window of opportunity was identified for novel methods of insect control: many existing insect control products are rapidly becoming redundant, due to increasingly restrictive legislation (relating to health and safety issues), in addition to growing resistance of some major pests to traditional control products. Furthermore, there is an increasing customer demand for environmentally-friendly, non-chemical means of insect control.



It was decided to try and target the very life-support mechanism of large numbers of insect pests. Insects which live in specialised niches (e.g. blood-, plant- and cellulose feeders) often lack essential nutrients in their diets, which instead are provided by symbiotic bacteria living in or close to their gut. Start-up funds from Edinburgh Technology Fund enabled us to confirm the importance of these bacteria and that their removal (with antibiotics) resulted in disruption of insect growth, survival and reproduction. Clearly, antibiotic treatment of insects would not be a viable method of control, which would require a different line of attack.



'Phage therapy' is growing in importance in both human and veterinary medicine to combat antibiotic-resistant infections. 'Phage' (or bacteriophage) are bacteria-specific viruses which destroy bacteria by injecting their own DNA into the bacterial cell, which is then instructed to produce new phage particles. The bacterial cell bursts within 30 minutes of infection, releasing new phage to infect further bacterial cells. Our aim was to try and develop this technique with insect bacterial symbionts to effect control. Hence, a SMART:SCOTLAND Award was applied for to initiate the R&D which would help us with this aim.



Using the SMART Award



The SMART Award allowed In-Phage to explore their ideas regarding the use of bacteriophage in insect control. It can take up to five years to get a new pesticide to market due to the regulatory issues which have to be attended to

The SMART Award effectively kick-started this process, allowing a full-time researcher to be employed in the laboratory, whilst the Founders concentrated on developing the commercial side of the business, including investigating the market in more detail and protecting the intellectual property associated with the technology.



By the end of the project, we had investigated a range of both specific and generalist bacteriophage for in vitro activity against insect symbiont cultures, in addition to in vivo assays of active bacteriophage with target insects, assessing the effects on symbionts, insect survival and reproduction. Key pest targets were identified through both our market research and discussions with significant players in the marketplace. These include the ubiquitous cockroach and the house dust mite, which is the major cause of childhood asthma.



The Effect of the SMART Award



The SMART Award enabled us to make significant progress in assessing the potential for applying 'phage therapy' techniques to insect control, raising a number of important technical issues regarding specificity and delivery mechanism which we hope to explore in a SPUR application. The endorsement from SMART also helped raise the profile of In-Phage within the businesses community, putting us in touch with a number of invaluable contacts. It has also enabled us to speak directly to the pest control industry, who largely have given very good feedback, seeing the potential of the technology. This has been a massive boost to our confidence of taking an academic concept through the stages required to get it out into industry.



Through a detailed examination of the pest-control market during the project, we have been able to revise our business model, deciding to concentrate on developing our platform bacteriophage technology for key markets, with the aim of gaining revenue through licensing deals. In addition, we have begun to create a revenue stream from contract research and consultancy, drawing on a wealth of research experience which lies within the company. These contracts are allowing us to build relationships with international pest-control companies, which may eventually become licensing partners for our core bacteriophage technology.



The business now employs two full-time researchers and several consultants and is being headed by Alison, who is currently supported through a prestigious Scottish Enterprise/Royal Society of Edinburgh 'Enterprise Fellowship'.



In-Phage Ltd

Veterinary Centre

Roslin

Midlothian

EH25 9RG

 Contact: Dr Alison Blackwell

Chief Executive Officer

Tel: 0131 650 6266 or 07876 495737

Fax: 0870 458 3844

E-mail: ablackwell@in-phage.com

www.in-phage.com

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

The MRSA (methicillin-resistant Staphylococcus Aureus) Problem


Dr Mark Farrington Chris - We are told that MRSA kills 5000 people per year. What is MRSA and where did it come from?


Mark - MRSA, or Methicillin Resistant Staphylococcus aureus, is a special variety of a very common bacterium. We come into contact with it every day, and a third of us have Staphylococcus aureus up our noses all the time! MRSA is just a small group of this bacterium that has become resistant to a common type of antibiotics. The bacteria itself isn't particularly nasty; it is just more resistant to treatment, which has implications when trying to find ways of preventing and treating infection. The problems Staphylococcus would cause on a day to day basis are wide ranging, and are one of the commonest causes of infection in normal healthy people. When we have a cut, it is usually our own Staphylococcus that infects us. The MRSA strain was first found early 1960s, which was the same time that the antibiotic methycillin was introduced. The reason there were a few isolates of the resistant strain so early on is probably due to methicillin being related to beta-lactin. Beta-lactin is a natural antibiotic produced by fungi; probably used as defence against bacteria. Therefore, Staphylococcus has been in contact with the antibiotic for a long time, allowing a few resistant strains to evolve. It is only since people have been repeatedly prescribed antibiotics that problems have arisen. The non-resistant strains die out, leaving the resistant strains behind.



Chris - When did MRSA become a big problem in hospitals?



Mark - it actually took quite a long time. For many years, the bacteria were only found occasionally, and only slightly more often in people in hospital than people not in hospital. MRSA started to appear in clusters of people in nearby beds, but the outbreaks were still small. The problem has increased as the resistant bacteria has been passed from patient to patient. The most significant method of transmission is via the hands of healthcare workers in general, not just doctors. If a patient has MRSA on their skin and receives care from a member of staff who forgets to wash their hands, the next patient they treat will have the bacteria passed onto them. It is important to distinguish between being colonised by bacteria and being infected by bacteria.

Studies have shown that people visiting hospital acquire a range of bacteria different from those they came in with. However, they do not develop a similar range of infections: having MRSA on your skin does not necessarily mean that you will become ill.



December 2004

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

PHAGE THERAPY

The Use of Bacteriophages Against Infections

by Amiran Meipariani, PhD, Academician

Felix d'Herelle the discoverer of bacteriophages, was of the opinion that all bacteriophages belong to the same biological origin, but this was disproved in later research.

It was discovered that there are different groups of special bacterial viruses, which are widely spread throughout nature. The bacteriophages coexist in the same environments as where microbes are reproducing. The bacteriophages can be found everywhere in human and animal intestines, in running water, in the soil and in the cell of microbes.

The research of the bacteriophage phenomena has exceptional importance in the history of microbiology. The simplicity of their cultivation, short generations, exact accountability, helped to clarify not only the structure of viruses, but also the relationship between bacteria and viral particles.

Using an electron microscope in the process of research definitely enriched our knowledge about the nature of bacteriophages. It was discovered that the majority of bacteriophages have the shape of spermatozoids. It has a head, which contains nucleic acid and a tail. Some phages have a very short tails or no tail at all.

According to their different structures, the bacteriophages belong to different morphological groups. Bacteriophages and viruses contain nucleic acid and protein. The majority of phages contain deoxyribonucleic acids, but some contain ribonucleic acid. The bacteriophages are more resistant to physical and chemical factors than human viruses. Phages are inactivated at high temperatures (60 to 70 degrees Celsius), but are resistant to low temperatures for long periods of time. Bacteriophages are also very sensitive to acids.

There is an interdependency between bacteriophage and microbe cells. Their interaction usually ends with the destruction of the microbial cell. For example, if we have an infection called absorption, the bacteriophage is not reproducing within the cells and the microbe cells are kept alive. In some cases, when the microbe cell in infected by the bacteriophage, lysogenic conditions are produced, as the bacteriophage's genome integrates itself with the microbe's genome. The microbial cells will lyse and die. 

The relationship between the bacteriophage cells and the microbe cells has four stages of development:

Adsorbability: There are the receptors on the wall of the bacterial cell, which attract certain bacteriophages. It is possible to adhere hundreds of bacteriophages on the wall of one bacterium, although one bacteriophage is enough for destruction of the microbe.

The adsorbed bacteriophage transfers its own nucleic acid into the microbial cell. The mechanism of this transfer can happen in different ways, but it is not yet fully understood. 

Then biosynthesis of bacteriophage starts, nucleic acid is transferred into the microbe and bacteriophage proteins are being produced inside the microbe. The proteins will late! be used to assemble new bacteriophage.

This is a process of morphogenesis of bacteriophage, filling of the bacteriophage with nucleic acid and complete its development
.
The bacteriophage is separating from the microbial cell. This is the process of destruction of the microbe and the release of new bacteriophages. The bacteriophage is self-reproducing only in a suitable microbe cell and reproduces individual bacteriophages such as: dysentery bacteriophages, which cause destruction of dysentery microbes, and staphylococcus and streptococcus bacteriophages, which are destroying the staphylococcus and streptococcus.

It has been almost 60 years since bacteriophage were discovered and used as a therapy agent in medical practice. Since the discovery of the phage and up to present, research of phage as a therapeutic agent has been a difficult and controversial process. Some scientists were in doubt of the bacteriophage and its use in medical practice. Dr. d'Herelle and his scientists proved that their doubts had no foundation.

When we are talking about the use of bacteriophages, we have to remember those who believed in phage-therapy. Their names are: Eliava, Mikaladze, Tsulukidze, Antadze, Kvitaishvili, Maslokovets, Kazarnovskaia, Krestovnikova, I Makashvili, Viskovski, Fisher, Pokrovskaia, Karpov, and Timakov. Their scientific researches saved hundreds of lives. The process of phage-therapy can be divided into several periods.

Part of the scientific community believed that the discovery of bacteriophage would help doctors to treat infectious diseases successfully. At that time, the bacteriophage-therapy was a powerful agent against dysentery, typhoid, sepsis and cholera. During the 1920's, there were enough materials about the spread of phages in the environment and its biological nature. The bacteriophages were discovered to fight against all infectious diseases. The development of sulfonamides and antibiotics significantly reduced the interest towards phage- therapy. Theoretical research of nature and reproduction of bacteriophage took a very important place in biological science during 1945-1955.

The bacteriophage became a unique model in research for molecular biology. Research of bacteriophages helped to clarify the exact role of deoxyribonucleic acid in heredity and also other important questions in molecular biology. For example, the blue print of DNA-RNA-protein, discovery of informational RNA (ribonucleic acid), research of genetic coding, etc. The research in theoretical biology diminished the role of bacteriophages in treatments against infectious diseases. The importance of theoretical research of bacteriophages put the practical use into second place. At the same time, successful use of antibiotics completely isolated bacteriophage-therapy. Later it was discovered that antibiotics had some disadvantages, like high mutation rates of microbes toward resistance to the antibiotics. The irrational use of antibiotics created different kinds of allergies, lack of intestinal bacteria (especially among children), etc.

Pathology of intestinal infections is 30% and up, among all the infections in the human population. By statistical information of the International Health Organization, almost 500 million people have intestinal infection annually. According to this information, the preventive medicine in very important in therapy of intestinal infections. The scientific development of bacteriophage is closely connected with its practical use.

F. d'Herelle successful experiment motivated the scientists to consider the phage-therapy a very important agent as a treatment of dysentery. There were enough materials to consider the bacteriophage the right preventative agent to treat intestinal infection, dysentery and typhoid sepsis.

Analyzing all those materials about phages against dysentery and typhoid sepsis, there were some mistakes about dosage of medication, time of use, and the most important part -- the quality of medication, and specification of phage. The bacteriophage against dysentery can be used in two ways: first – the bacteriophage as a curative agent and second the bacteriophage as a preventative agent. 

The specific treatment of dysentery by the bacteriophage depends not only on its ability to destroy the microbes, but also when and how many times it is used. The bacteriophage must be specific, polyvalent with a wide spectrum of activity. It is very important to give this medication to the patient in the beginning stage of intestinal infection, before any serious pathological changes take place.

The research has proved, that when bacteriophage is used in the first stage of infections (during the first 5-6 days) 64% of the patients after 24 hours of administering the drug showed a clear clinical improvement; and after 48 hours, a complete cure. Not only clinical recovery shows the value of bacteriophage therapy, but also bacteriological sanitation.

The treatment of chronic dysentery with phage was not perfect. The doctors tried to create better and more effective drugs. One of the drugs developed was well-adapted bacteriophage with wide spectrum. It was administered in the complex therapy of dysentery.

In many cases, the results were more effective than treatments with antibiotics and sulfonamides. During the chronic form of dysentery, microbes from patients were treated with specially adapted bacteriophages. Children with chronic dysentery were given bacteriophage 2-3 times a day (5-15 milliliters) during 3 days. The period of recovery decreased from 1 year to 3-6 months. The treatment with other drugs takes at least 9 months to show similar results.

The clinical experience shows that desirable therapeutic results can also be reached using bacteriophage in large quantities. In case of late treatment, it is recommended to increase the dosage and quantity of drugs. The research by the noted Georgian scientist, Professor Kvitaishvili, shows that bacteriophage treatment can be more effective than antibiotics and sulfonamides in case of dysentery. The right calculations of time and specific qualities have to be taken into consideration. There is no doubt that bacteriophage is one of the strongest biological agents and rational use can advance the fight against dysentery.

Today, antibiotics are used to treat tuberculosis, cholera, typhoid, sepsis, etc. In spite of all successful use of antibiotics, lately it has been discovered that some microbes are resistant to them. It can be hereditary or caused by mutation. In 1940, Abraham and Chase found penicillinase as a neutralizer of antibiotic - penicillin. Years of research showed that the percentage of microbe resistance to antibiotics grows not only in dysentery microbes but in other microbes as well.

The Japanese scientist --Watanabe, performed some very interesting research regarding antibiotic resistance of microbes. He was mentioning that antibiotic treatment in medical practice gave a big hope to doctors, that all the infectious diseases would be cured, but it did not happen. The cause was the resistance of microbes to the antibiotic and sulfondmides. This phenomenon is called "Infectious Resistance." The mechanism of the resistance is based on genetic analysis and proved to be hereditary.

"Infectious Resistance" is quite dangerous, because it might involve all kinds of microbes and create complex difficulties, especially in treatments of intestinal infection. Irrational use of antibiotics caused lots of unwanted results, such as lack of bacteria, when micro-flora of the intestines is destroyed, allergy syndrome, etc. That is why it is very interesting to quote Watanabe, "If we will not stop irrational use of antibiotics and other synthetic drugs, we will end up back in the time when medicine had not yet discovered antibiotics."

The last 10 years of research confirmed once more that use of antibiotics especially against dysentery, created resistant forms of microbes in the human body and environment. With growth of resistance other biological changes occurring in microbes such as: fermenting activities, antigen, virulent, reaction toward physical and chemical factors. The study of these changes of microbes from the use of antibiotics has not only theoretical, but also practical importance which is connected to treatments with antibiotics and antibacterial drugs, such as bacteriophage. The research shows that antibiotics do not inactivate the bacteriophage.

Based on these factors, combined treatment using bacteriophages and antibiotics was initiated. Once again, the research proved effective use of bacteriophage, for acute and chronic dysentery. The combined therapy proved to be effective, when the patient first is given bacteriophage, then antibiotic. The result of therapy is much better when started in the beginning stage of infection.

It has to be noted that bacteriophages are safe and do not have side effects.

For acute forms of dysentery, among children, specific bacteriophage of dysentery was used. The patients were given the drug every day during 7 days. After therapy, the result was positive. There was 78% of clinical and bacterial recovery, which means it was not necessary to give patients the antibiotics. For intestinal infection, caused by pathogenic microbes, they used combined therapy, after antibiotics they used bacteriophages against Proteus and intestinal bacillus. After therapy 67.7% of patients were recovered and 29.7% of patents improved, 2.5% had no result.

The results of dysentery treatment proved effectiveness of preventative use of the drug. The length of circulation period of bacteriophage was established along with its effect on the micro-flora of intestine, the time of receiving the drug, intervals and its dosage.

The research proved the effectiveness of bacteriophage. The children up to 3 years old were given 3 milligrams of drugs; the adults took 5 milligrams of drug 3 times a day with 7 -day intervals. Increased dosage up to 15 milligrams of drugs and shorter intervals (from 6 to 3 days) gave even better results.

Use of bacteriophage as a preventative agent against the dysentery was proven, when people who were in contact with sick patients had been given bacteriophage. As a result, most of them were not infected by dysentery. The majority of scientists are saying that the bacteriophage is most effective as a curative and preventative agent against dysentery. It is safe and without side effects.

The combined drug use against the intestinal infections among children contained bacteriophages to fight dysentery, intestinal bacillus and Proteus microbes. The drug was given to children during an epidemic season. As a result of phagetherapy, the level of intestinal infections decreased 9 times, with 5 day intervals, because increased concentration of phages in the intestines created the right conditions to resist the microbes (which cause disease).

Antidysentery bacteriophage was used not only as a curative and preventative agent, but also as an agent of gave good results in the second stage of treatment. Bacillus was a positive second stage of therapy. Each stage of therapy meant use of the drug once for 4 days.

The department of children's intensive care in Leningrad Medical Institute proved, that during pneumonia, contaminated wounds, burns, sepsis, the main cause of infection is pseudomonas (an acute pusforming microbe). For this preventative therapy, they used bacteriophage, created in the Tbilisi Institute of Vaccine. During 5 days, children were given 10-30 milligrams of drug daily.

After therapy, tile bacterial research proved that 83.6% of cases reproduction of the microbes stopped without any side effects.

The effectiveness of bacteriophage increased when the phage-cultures came from an infected human body. Finding the therapeutic effect of specific phage was possible with bacterial analysis - with controlled temperature, index of peripheral blood, and wound condition. After therapy, 66.6% of the result was good and effective. The result was better when the phage was used directly in the wound area.

The effect was less favorable, when phage was used in deeper wounds. The time factor is very important. Early therapy gives better results. Today, phage-therapy of typhoid sepsis belongs to the past. Antibiotic therapy completely replaced phage-therapy of typhoid sepsis, but phage-therapy, as a preventative agent is still very important.

We use phage-therapy against typhoid sepsis with the same success, as when we used it against dysentery infection. We had good clinical result using the bacteriophage during the explosion of typhoid sepsis. After therapy, the level of infection decreases, and later disappears. After recovery from typhoid sepsis, some patients are still carrying the bacteria, which creates potential spreading of the microbe.

The fight against "germcarriers," and their sanitation demands systematic observation and research. After scientific experiments on animals, researchers concluded that bacteriophage for fighting typhoid sepsis is acting not only as an agent to lyse microbes, but also as an immunogene drug. Using Salmonella. typhimurium bacteriophage in massive quantities during anti-epidemic period as a prevention reduced the number of cases, during the period of one month. Compared to the previous year, the number of illnesses decreased six times.

The percentage of sanitation in controlled areas was 4.4 and 9% in treated group. The same positive result was given, when bacteriophage was used against typhoid sepsis explosion. The results were interesting, when bacteriophage was inserted to the animal by internal application. It was discovered that you could find the phage in the blood after 2-9 days, in mesenteric gland and spleen after 2-10 days, in the lever after 3-8 days, in the fecal content 4-10 days.

The process of cleansing the body from typhoid sepsis microbe takes 1-9 days. At the .same time, phagocytic process becomes very active. It has practical importance to know how long bacteriophage stays in the human body, when periodically taken. By experiment it was proved, that during a 3-hour period, bacteriophage completely spread throughout the body. Later findings have sporadic nature and it slowly disappear. Increased intake of dosage (10-15 milligrams) stabilizes the time of spreading and cleansing period.

The bacteriophage stays longer in the reticuloendothelial system, especially in the spleen and in the large intestine. In the blood system, phage can be found in 30 minutes after intake. Intensive reproduction of bacteriophages from children's intestines starts in the first 3 days. The increased dosage and interval between intakes of the drug makes circulation and reproducing period much longer. The scientists think that higher concentration of phage in the human body increases effectiveness of the drug.

For the purpose of sanitation of chronic bacterial-carriers during the typhoid sepsis infection, increased use of dosage is proven to be effective. In infectious pathology, diarrhea and colitis are common among children. For treatment of those diseases, polyvalent bacteriophages are used with success. French scientist, F. d'Herelle, used the polyvalent intestine phage as a curative agent first.

The components of polyvalent intestine phages are: dysentery, typhoid sepsis, paratyphoid, salmonella, proteusis is staphylococcus, streptococcus, enterococcus, pseudomonas and intestinal bacillus, fighting bacteriophages.

Nowadays, the components of intestine phage depend on what kinds of micro- flora are circulating in a certain geographical zone. The activity of independent components in combined drugs is stable -their action is the same, as each of them in separate use.

The advantage of intestine phage, in comparison with other drugs in therapy of acute intestine diseases, lies in its polyvalent nature. It is effective to use this drug from the first days, before the beginning of investigation. Multiple content of phages gives us the chance to use this drug in the very first day of the treatment before any bacteriological research begins.

The majority of scientists, who used polyvalent intestine phages for treatments, concluded that bacteriophage and its biological qualities which are based on direct interaction with microbes, has to belong to the kind of drugs, that are effectively fighting intestinal infections. Use of intestine phage as preventative treatment gives us much better results than any other drug. This effect is based on its specific safety of its intake.

The research by Logoladze of intestine phage is quite interesting. According to this experiment, 452 children were treated by intestine phage, 110 with antibiotics and 29 with complex intestine phagetherapy. The recovery of its intake period among the children with intestine phage therapy was 9 days, with antibiotic therapy 29 days and with complex 15 days.

The length or recovery of its intake period in the group that used only intestine phage lasted for 9 days; recovery from antibiotic treatment for 29 days, and combined for 15 days. It has to be noted that use of intestine phage shows its effectiveness on the very next day. The recovery of the majority of children under phage therapy was starting at 4th, 5th, 6th day. Recovery period under antibiotic and combined treatment is much longer - starts at the third day and continues during 15 to 29 days.

There was interesting research done by scientists from Saratov Medical Institute's Children's Department of infectious Diseases (Stoliarova, German, Trifonov, and Milnikova). They used phagetherapy against dysentery caused by salmonella and proteusis.

The patents were treated with phages during 7-10 days. Also they were given symptomatic treatment. Some children took only antibiotics. This therapy lasted 17 days: 42.2% of the patients had unstable bowel movement, 26.7% of the same bowel movement symptom, 30.9% with secondary producer of cause.

The scientists decided to use extra therapy with polyvalent salmonella bacteriophage. After tile therapy, 66.2% of patients recovered completely, and 16.9 with better resistant conditions. The period of time spent in the clinic lasted 25.6 days. For ones that had not used phagetherapy, treatment stretched to30.5 days. 53.3% of patients recovered, 33.2% were in better condition, and 13.3% without any results. The patients were staying in the clinic 34.7 days. Scientists (Tsereteli, Kiknadze and others) studied the polyvalent -salmonellosis bacteriophage at the Tbilisi Institute of Vaccine.

Results of the 10 years of research proved that use of bacteriophages against intestinal infections has strong scientific background. It is very effective, safe and has no side effects. Use of bacteriophages for supuratavie inflammation, surgical, urological, gynecological and infectious allergies has to be noted. The reason for successful used of the bacteriophage in above-mentioned diseases, based on the existence of bacteriophages that are produced to fight against it, purulent infections and the possibility of a relationship between phage and its cause.

According to statistics of Saratov's Scientific Research Center, Staphylococcus and Streptococcus bacteriophages (made at the Tbilisi Institute of Vaccine) were used for infectious-allergic rinosinusopathy, infectious-allergic bronchial asthma, during neurodermatosis, caused by hyodermatosis. Among the 118 patients subjected to staphylococcus phage-therapy, 90.3% of the patients had positive results, streptococcus phage-therapy success rate was 94.1 %, combined phage- therapy of both gave 99% effectiveness.

Infections caused by diverse (phyogenic) microbes have mix infection character. In this condition, treatment is very difficult. There are very few active antibiotics with wide spectrum, and the quantities of resistant microbes are steadily increasing. Because of these conditions, demand to create new forms and possibilities occurred. Phyobacterophages are characterized to have positive results in cases of mixed infections, in short periods of time, an infected wound is healing faster and the patients' condition improves. Among newborn babies infected with pink eye, when etiologic factor is multifaceted and treatment with antibiotics is limited, staphylococcus and phyobacteriophages are used. A drug for treatment of infected eyes was given in quantities of two drops, 4-6 times a day. Results were positive in all cases. Purulence had disappeared on the second day. The complete sanitation and clinical effect were reached on the 3- 7th day without side effects.

During the process of phagetherapy and chronic phagetherapy, knowing the cause is important, along with etiological factors, which had gone through big changes since the 1940s. Different kinds of dysentery and other infections made it difficult to produce suitable bacteriophages, but research to increase the effectiveness of phage drugs, had positive results anyway. Nowadays, the medical institutions have produced effective and safe drugs with a wide spectrum.

In 1979, French scientist P. Nicole mentions in the bulletin of National Medical Academy that the growth of multiresistant bacteria against antibiotics in infectious pathology. He defined three ways of using bacteriophage in therapy: Treatment of post-surgical infections.

Liquidation of the process of infection, among children during epidemic of gram- negative microbes. The chronic infection of urethra.

The phagetherapy phage are not just a "primitive" drug, but a strong, effective agent:
Two groups of microbes: Salmonella and other bacteria, such as Klobsiella, Serratia, Proteus and Providencia, which spread epidemic and post-surgical non- epidemic infections, besides staphylococcus and acute purulent bacillus. Widespread different kinds of bacteria, which are resistant to all known antibiotics.

The different kind of bacteria, which are resistant against all known antibiotics. osteomylitis

The phages are curative agents for abscesses, boils, Septicemia and Pyelonephritis infection of the urethra, where gram-positive and gram-negative microbes cause diseases, afterburn purulent inflammation process, and skin infection. It has to be noted:
Post-surgical infections which cannot be controlled with antibiotic therapy --there are also staphylococcus, enterococcus, green purulent bacillus; Serratia, Klebsiella, Providencia, etc.
Epidermal infections among newborn babies --pathogenic intestinal bacillus, especially Enteritis and Salmonella. typhimurium,

The persistent urethra infection cause of Providencia, Serratia and Proteus microbes.

According to these infections, the medical institutions producing phages decided: to produce combined phages, with wide-spectrum of effectiveness, the Tbilisi Institute of Vaccine is the pioneer of producing phyo- and intestine phages –the best agents against purulent microbes and intestinal infections. Both drugs were tested in clinics successfully. The phyobacteriophage produced in the Tbilisi Institute of Vaccine was used as a therapy agent in Moscow Children's Medical Clinic for newborn babies with severe enterecolitis diseases. When it comes to heavy forms of the illness, combined bacteriophagetherapy is recommended. Within the middle ear, chronic purulent which was caused by staphylococcus and green purulent microbes, phagetherapy showed that the length of antibiotic therapy was 2.5 times longer than phagetherapy process.

The antibiotic therapy needs 8-10 doses, and phage therapy needs 2-4 doses (in the ear during 2-10 days they used 1-2 times antibiotics and bacteriophage eardrops.) It is known that all inflammation processes among newborn babies are followed by lack of intestinal bacteria, which is caused by Staphylococcus and fermentation qualities, changed by intestinal bacillus, Proteus and fungus.

The antibiotic therapy in many cases destroys the biosystem, which increases complications. The treatment of enterocolitis, caused by pathogenic microbes and method of decontamination (selective influence of antibiotics on microbes) antibiotics and other chemical therapy has some negative effects, which destroys intestinal normal micro-flora.

 In this case, as a clinical and bacteriological point of view, the polyvalent phage effects on conditional-pathogenic microbes without changing normal flora. The patients were given bacteriophage with Bifidobacteria and other patients were given antibiotics. The bacteriological researches confirmed that after 7-10 days of antibiotictherapy, the intestinal pathogenic flora was decreasing, without bifidobacteria flora, establishing dyspeptic syndrome on the background of lack of intestinal bacteria of II-III degree.

The phage therapy effect on conditional-pathogenic flora and on the increasing quantity of bifidoflora. The clinical recovery increase of patients weight were confirming and specification of phage. It is necessary to establish dosage of phages and of intake. It has to be established that the place of injection of drugs and to-select the quantity of patients according to bacterial researches. It is necessary to produce preventative-therapeutic bacteriophages, against conditional-pathogenic microbes like: Klebsiella, providencia, etc.

Air spray bacteriophages should be developed for local application. Half intetic phage drugs Should be as effective as an injection in the bone. The bacteriophage can be used not only like preventative therapy agent, but also diagnostic agent of infectious diseases, as identification and differentiation of cause. Today it is known the positive effect of bacteriophage against epidemics. Besides typhoid sepsis phages, there are Salmonella. paratyphi, Staphylococci. E. coli, Salmonella. typhimurium, Shigella. dysenteriae, Brucella and other phages.

The growth of titer of phage is a well-known method. It is necessary, represented by Timacov and Coldfavre.

The blueprint of the phage becomes more sophisticated and easy to get. The collection of typical phages is in reach with new specific phages like, cultivated enlarged and complete original phage blueprint toward Salmonella. typhimurium, pathogenic intestinal bacillus and salmonella paratyphoid microbes. It is a very interesting factor to use the phage to find out the level of pollution in the environment. Specific phage is produced to help to find out the source of infections and ways of spreading it.

It is known that a contact of patients with infected things and medical personnel cause the spread of infections. Also we have to know the mechanism of microbe pollution through air. According hou3e-contact infection phagetherapy the scientists received very impressive results -after phagetherapy, the level of dysentery decreased 3-6 times, hospital salmonella infections 3-14 times and in some cases it disappeared completely. After scientific experiments, it was found that phages not only are adsorbing on the microbe surface, but also self- reproducing in the environment.

In Kemerov's maternity house in Leningrad, combined drug-phyophage was used. Risk of supuratavie inflammation in the delivered baby's observation department was much higher than in the physiological department. That is why the physiological department was considered the control and the observation room the experimental area. Because of house-contact, the ways of supuratavie inflammation process, objects like tables, beds, sinks, floors were covered with phyophage. The comparative analysis of purulent infections in both departments was processing in two ways: the patients infected through the skin, mucopurulent infections belonged to the first groups. The acute intestinal infections to the second group. From the beginning of phagetherapy up to the end of the experiment, 2801 babies were delivered. 1641 babies were delivered in the physiological department and 1160 in the observation department.

The newborn babies' purulent infections were 2.3 times less in the experimental department than in the observation department. The coefficient of effectiveness was from 2-6. The maximum level of diseases in the physiological department was obvious after 2-3 weeks of delivery. 2-3 weeks after delivery, during the illnesses in the experimental group level of birth increased but in this group illness is 2.5 limes lower in every thousand child. It reaches from 4.3 to 6.0.

After phagetherapy decreased, the frequency of purulent infections increased by 2-3 times. In the department with phagetherapy, the acute intestinal infections decreased 2.4 times. The main sources of intestinal infections are Klebsiella, Citerobacter and unknown microorganisms. The intestinal infections among the newborn babies start within 1 week of birth, in 6.1 % of the patients. The infections are decreasing after 2-3 weeks, by half in the observational group.

The case is the opposite in the experimental group:


There is no incident of infection within 1 week of birth, which has certain epidemiological meaning. The level of the intestinal infections increases after 3- 4 weeks. To use phyophage in the environment decreases the purulent-sepsis infections 2-3 times and intestinal infections --2.4 times.

The bacteriophage can be used to study the mechanism of the purulent-sepsis epidemiological process. The patient, instruments and personnel are the sources of infections. As a rule, infecting by purulent infection happens in the procedural department. The Soviet scientists spent lots of time and energy to study theoretical and practical sides of bacteriophage. The Tbilisi Scientific Institute of Vaccine is one of the well-known and important centers producing bacteriophages.

Today there is ongoing research on development of bacteriophage and its use in medical practice for discovering new phages. The 60 years of experience of phagetherapy gives us the right not to agree with the American scientist, Stent's idea that the last paragraph in the history of phages is closed and there is not need to go back.




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

 Extracts from an Amazing Book on Bacteriophages

Viruses vs. Superbugs

A solution to the antibiotics crisis?
 Thomas Häusler

Translated by Karen Leube

Macmillan

London New York Melbourne Hong Kong



© Thomas Häusler 2006



First published 2006 by

Macmillan

Houndmills, Basingstoke, Hampshire RG21 6XS and

175 Fifth Avenue, New York, N.Y. 10010



ISBN-13: 978–1–4039–8764–8

ISBN-10: 1–4039–8764–5



contents

list of figures vi

foreword viii

preface xi

chapter 1 at the limits of medicine 1

chapter 2 invincible microbes 15

chapter 3 the wild pioneer era 48

chapter 4 the renaissance of phages during the war 105

chapter 5 a parallel universe 126

chapter 6 keepers of the grail in peril 175

chapter 7 resurrection 203

chapter 8 what’s the future of phage therapy? 248

appendix 1 a short list of bacteria 252

appendix 2 the advantages and

disadvantages of phage therapy 257

notes 261

figure sources 283

index 284



foreword


Just imagine life without antibiotics. It would be like it was 100 years ago, when pneumonia and tuberculosis were the most frequent causes of death, and the risk of infection turned a simple appendectomy into a dangerous operation.


Luckily we do have antibiotics. However, they are becoming increasingly ineffective. Doctors are more and more frequently confronted with infections they can’t do anything about because the bacteria have become resistant. This has dire consequences for patients. Many end up living with a chronic infection for years on end, some are forced to become amputees and yet others succumb to the infections. 


The crisis affects people in both industrialized and developing countries. In the US and the UK, the bug Staphylococcus aureus is wreaking havoc. Forty to fifty per cent of infections that people contract in hospitals are resistant to more than one antibiotic. The developing countries are groaning under the burden of tuberculosis, which claims the lives of 2 million victims throughout the world every year. The increase in multi-resistant TB is especially alarming. Treating it costs 100 times more than treating the regular form, making a cure unaffordable for many people in impoverished countries. And these are

only two examples.



Despite this, many pharmaceutical companies have stopped developing antibiotics. They see the financial risk as too big and potential profits too skimpy. This has led to very few new drugs for fighting bacterial infections being launched in recent years. A survey of 11 large pharmaceutical companies revealed that of 400 substances they were developing, only 5 were anti- bacterial drugs.


What can be done about the resistance crisis? One thing is needed for sure: new drugs. One of them could be bacteriophages, viruses that attack bacteria without harming people. So-called bacteriophage therapy had its heyday from 1920 to 1940, before it was pushed aside by penicillin. The former Soviet Union is the only place it continues to be used today. Most Western doctors do not even know that this method exists.

However, there are some scientists who have resumed research on bacteriophage therapy, and that’s a good thing. We need to pursue any and every approach that can contribute to solving the resistance crisis. Bacteriophage therapy may prove to be a particularly worthwhile area of  research. Its long history provides a large stock of knowledge that is freely accessible. Determined researchers now need to use this as a starting point and work out how to turn bacteriophages into drugs that meet today’s standards. This would be best carried out in cooperation with science departments at universities, along with private companies and non-profit foundations that support the projects. This is exactly the goal of the Foundation for Fatal Rare Diseases. The foundation supports the development of drugs for neglected infectious and pulmonary diseases and is especially committed to helping affected patients who have not been in the public eye, particularly those in Africa and India.



Thomas Häusler’s remarkable book plays a central part in this scheme, because it acquaints the public, researchers and decision makers with a therapy that has the potential to someday heal many patients who cannot be helped at the moment.


This is why the Foundation for Fatal Rare Diseases is supporting the realization of this English edition. The fact that the author writes about bacteriophage therapy in the form of such a

gripping story makes reading it all the more exciting.



Vaduz, October 2005





Vera Cavalli, Dorian Bevec and Fabio Cavalli

Founders of the Foundation for Fatal Rare Diseases


preface

 Why should anyone be interested in an old cure that hasn’t been used in the West for 50 years? It’s a method that many doctors aren’t even aware of today. The most telling answer to this question came when I received a call in my office from a man one Friday morning in January 2001. It was the day after my article on the Eliava Institute in Georgia had appeared in the German weekly newspaper Die Zeit. In the article I had described how this old remedy – phage therapy – had survived in the impoverished country.

Phages are viruses that attack and kill bacteria but not people. Since Stalin’s days, doctors in Russia and Georgia have been using phages to cure bacterial infections. In the West this method was also once popular but, in contrast to the Soviet Union, the triumph of penicillin pushed phage therapy aside here after 1940. The Eliava Institute in Tbilisi, Georgia is a place where phage therapy survived even after the collapse of the Soviet Union. It looks back on a glorious 80-year history.

However, because of Georgia’s economically and politically precarious situation, it is experiencing a gloomy present. From the point of view of today’s science, it is unclear how effective phages are in fighting infection. This is because the studies carried out by early pioneers and Soviet researchers do not meet today’s standards. All this was in my article in Die Zeit.

The caller explained that he had read the article. He was calling directly from the hospital and appeared to be under a great deal of pressure. Not mincing words, he explained that he had been suffering from an infection in his foot for two years. Doctors couldn’t get it under control because the bacteria were resistant to all antibiotics. He was scheduled to have his foot operated on a fourth time the next day. Could I put him in touch with someone in Georgia? He was afraid that before long he would lose his foot.

More than any research I have done, his call hit me between the eyes. Never before had I been so aware of the power that bacteria continue to wield over us. We have grown up with the certainty that every bacterial infection can be cured by antibiotics. Most of us have no idea of the destruction that bacteria are capable of rendering, because our doctors prescribe drugs at the slightest symptom.

One year after I received this call, I accompanied an expedition of botanists and fragrance researchers to a rain forest in Madagascar. One night, as I slept in my hammock, I woke up, and my right foot was hot, red and swollen. The next morning I could hardly walk. Bacteria must have entered places where my sandals had rubbed against my skin while we were hiking.

The doctor accompanying the expedition gave me some antibiotics that he found in his first-aid kit. The effect was hit or miss – more miss than hit, in fact. Four days later, I arrived home – with my foot still swollen. My GP prescribed some other antibiotics and luckily they worked. He cut to the chase: ‘That could have been the end of you.’

At that point, however, I no longer needed that kind of graphic demonstration of the power of bacteria, since I had already started doing research for this book. The 80-year-oldhistory of the tiny phages and their potential role in reining in the antibiotic resistance crisis were constantly on my mind.

The fascination produced by phage therapy is particularly striking as I write these lines. In Southeast Asia, veterinarians and doctors are combating bird flu. A pandemic is in the making. This was only just averted in the case of SARS, a new atypical kind of pneumonia. These health crises show viruses in their familiar role – as lethal villains. Phage therapy takes this image and turns it upside down, turning the bad guys into unexpected allies.

This book is not a health manual whose purpose is to testify to the efficacy of phages. First, it’s too early to reach a clear conclusion about their effectiveness. Researchers are still working on this. Second, I found the detective work on the origins of the captivating idea that bacteria can be fought with their natural enemies at least as interesting as the analysis of phages’ curative powers. I hope that this has led to a book that sheds some light on the sometimes winding paths of medical

research and in turn provides some insight into an area of our society that is becoming increasingly significant. Never has so much medical research been undertaken as at the present time,nor has so much money ever been spent to cure us of diseases.

This English edition came about some three years after the German edition was published. I have taken great pains to update the material in the book. As I did so, I saw that some companies had been confronted with scientific or financial obstacles, leading them to abandon their projects altogether. On the other hand, other companies and university researchers have joined the ranks of phage therapy research, contributing good ideas. What they require is support from public and private sponsors in order to produce drugs from phages. They are desperately needed.

I could not have written this book without the help of many researchers, doctors, patients, librarians and helpers. They provided me with information, books and photos, gave me accommodation, told me about their lives, interpreted or handed out advice. I extend my gratitude to all of them. I would specifically like to thank Elizabeth Kutter, Hans-Wolfgang Ackermann and Harald Brüssow for sharing their expertise. Zemphira Alavidze, Nino Chanishvili, Liana    Gache-chiladze, David Gamrekeli and Mzia Kutateladze not only provided me with exhaustive information, but made my research in Georgia possible in the first place. I will never forget their hospitality.

Reto Schneider, Elizabeth Kutter and my wife Susanne read the entire manuscript. I thank them for their countless suggestions for improvement in style and content. I also express my thanks to my translator Karen Leube, my editors Sara Abdulla (Macmillan) and Wolfgang Gartmann (Piper), and the team at Aardvark Editorial. Without the support of Tamedia AG, the publisher of Facts news magazine, this book would not have been possible. Facts, my employer, continued my salary while I worked on this book, and Tamedia’s media forum paid for the research expenses. I would like to thank my colleagues at Facts, Odette Frey, Beate Kittl and Rainer Klose, for their willingness to put up with the additional work and reorganization brought about by my absence.

The English translation was generously funded by the Foundation for Fatal Rare Diseases. Thomas Fritschi and Rich Weber drew the graphics for Figures 3.4 and 3.5. I thank Susanne and Julia for putting up with a husband and father who was more of a phantom for a year and, at times, an overworked, nervous one at that.

Thomas Häusler 


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

 Profile: Bacteria’s natural born killers

Winning the 2008 NSW BioFirst Commercialisation award has come at exactly the right time for Special Phage Holdings, which has treated a patient in the clinic and is currently looking at a second round of private fundraising to develop its portfolio of bacteriophages

02/10/2008 16:45:00
 

Australian Life Scientist

By Kate McDonald

Just before Christmas last year, a 60-year-old patient at Westmead Hospital got some good news. The woman was suffering from a chronic infection that nothing could shift.

Pseudomonas aeruginosa had taken up residence in her bladder and all of the available antibiotics proved useless. She was facing a decision: radical surgery or a new therapy that has been rarely tested in a western hospital.

She opted for the latter. Over five days just before Christmas 2007, she was infused with a cocktail of bacteriophages through a catheter. Bacteriophages are viruses that infect bacteria and represent the most prolific organism on the planet.

Phages are natural born bacteria killers and are nature’s solution to keeping bacteria in check. They are ancient, they are everywhere and there are an estimated 1031 of them in existence.

The patient began to feel better within a couple of days and was home for Christmas. Six months after her treatment, she remains infection-free.

Not only was this good news for the patient, but good news for the private Sydney company that is developing bacteriophage therapy, Special Phage Holdings(SPH), run by Dr Tony Smithyman.

Phage therapy is nothing new of course, having been used extensively in the countries of the former Soviet Union for close to a century. It is most definitely coming back into fashion in the west, however, as it represents a radically different – and safe - alternative to antibiotics.

Smithyman and his team at SPH have been investigating phage therapy for the last five years. They have managed to refine their technology for collecting, screening and growing phages, and now have a library of several hundred, all specific to particular bacteria and even specific to strains of bacteria.

“They are commonly found in soil, water, plants, sewage, mud – anywhere there is bacteria that they can go and eat,” Smithyman says.

“The question is, how do you get them? You’ve got to go looking for them in the most appropriate places, which means a lot of interesting times. For instance, we go looking in Manly lagoon and Hubert (SPH’s marketing manager, Hubert Mazure) has been on a sewage mission.

“However, you have to have the right systems to extract the phages. There is quite a lot of skill involved. It’s a bit like looking for monoclonal antibodies – you have a target and then you screen. And the longer you go the better you get.”

Bacteriophages come in two forms: lytic and lysogenic. Lytic phages have evolved purely to destroy bacteria, Smithyman says. “There is no messing around with them. They go straight in and destroy the bacteria, and because of that they have fairly small genomes.

“The lysogenic phages have evolved to live with the bacteria, so they have larger genomes and have a different lifecycle. They can transfer toxin genes to the bacteria, so one of the things we have to do in phage therapy is have a system for screening to make sure you don’t have the lysogenic ones.”

Smithyman recently returned from a phage conference in Edinburgh with tales of some of the fascinating research being done with phages throughout the world. Surprisingly, it was the business sector rather than academics that first took the idea of phage therapy and decided to run with it, but Smithyman says the universities are quickly catching up.

“They are looking at using phages for delivering vaccines, a lot of work on using phages for purification and more genetic work coming through to help understand the molecular biology. And then there is the clinical work. A lot of that is very interesting. It is an intrinsically fascinating topic.”

Classification and commercialisation

One of the issues phage therapy will face in the future is its categorisation for human health purposes. There are already phage therapy products on the market, but they are for plant health and to treat foodstuffs such as meat and cheese.

Those products were approved by the US FDA as Generally Regarded as Safe (GRAS), and this designation is expected for any future phage therapy product for humans.

“When it comes to treating patients, they aren’t a pharmaceutical so there is a lot of discussion around the world as to where they actually sit,” Smithyman says. “It may be that they will end up in their own category.

“They are extremely safe with no side-effects and no toxins, so there is no real need to go through animal studies, but the jury is still out on how they will be classified. It will probably be as a biological, not a chemical, but they might have to have their own category.”

Special Phage Holdings is one of a number of companies planning clinical trials for phages. The therapy has been used in parts of Europe, particularly in the world’s centre of excellence for phage therapy, Georgia, for most of the 20th century.

Smithyman says there are several American and UK companies in the Phase II trial stage, and there are university and clinical groups in France, Germany, the UK and Poland who are treating patients.

This is why both the successfully treated patient and the BioFirst Commercialisation award have come at exactly the right time for SPH. Smithyman says his company is at the stage of developing cocktails of phages for prototype products and was planning for the trial stage when the patient at Westmead came up.

SPH has strong research links with both Westmead and Royal North Shore Hospital, and has many international collaborators, and is planning a number of trials.

“We are planning trials now and working on the protocols,” he says. “We have three trials planned – one in Australia, again through Westmead and Royal North Shore; one overseas; and one a veterinary trial, also here in Australia.”

The company is also undertaking a second round of fundraising, approaching the group of original shareholders who provided the initial seed money, as well as sophisticated private investors and select corporate investors. The NSW Government award has come as a nice bit of publicity at exactly the right time, Smithyman says.

“We are going to be able to use the award to travel, there is some legal and patent advice and some accounting advice, as well as PR, so we’ll use them all very judiciously. It has come at exactly the right time.”

Broad spectrum delivery

Smithyman foresees a time when broad spectrum phages will be developed for immediate use once a patient presents with an infection. He also sees a time when patient-specific phages are produced for particular infections.

“If it is extremely urgent they will be hit with a broad spectrum, but before that you take a swab and isolate the type of infection, take that isolate and run it past a vast collection of phages, choose the right one and then grow it up quickly. In a couple of days the patient can be treated with their own specific phages.”

Delivery options are as versatile as the phages, he says. “The patient at Westmead was infused because we had to decide how to get the phages into the bladder, so a catheter was chosen. But normally they would be used in liquid form embedded in bandages, or as a tablet for straightforward oral delivery. We have also formulated a cream, and another way they will be used is through a nebulliser.”

Phages are extraordinarily powerful but obviously they won’t cure everything, particularly if there are co-morbidities. And they probably will meet the brick wall of bacterial resistance, just as antibiotics have done.

“That’s why we are developing cocktails – bacteria might become resistant to one type of phage, but not to many.”

And the market is potentially huge. As Smithyman points out, the large pharmaceuticals have been walking away from trying to develop new antibiotics over the last couple of years because there isn’t much profit in it, leaving it up to the smaller and generic pharma companies to look for biotechs to come up with novel ideas for antibiotics.

It is a $24 billion a year sector that is becoming a vacuum, he says, and perhaps phages are the right organism to fill it.

end



http://www.biotechnews.com.au/index.php/id;1656824057;pp;1;fp;4;fpid;3

                         .......................................................... 
Prologue

The Killers Within: The Deadly Rise of Drug-Resistant Bacteria 


By Michael Shnayerson, Mark J. Plotkin


Book Description
 
A battle is taking place on the frontiers of medicine between rapidly evolving bacteria that threaten our health and the doctors who are struggling to outwit them. These bacteria are everywhere: in and on our bodies, in homes, schools, hospitals, crowded airplanes, day-care centers. And, as this acclaimed book makes frighteningly clear, so far the bacteria are winning. The Killers Within is popular science writing at its best-a report from the front lines at once accessible and riveting.



Prologue

Dr. Glenn Morris was growing very worried. His patient was not supposed to die. 

The son of Southern Baptist missionaries, Morris had grown up in Bangkok, Thailand. There he had witnessed firsthand the magnitude of devastation that could be wrought by a bacterial plague. During the dreaded dry season after the monsoons had passed, waves of cholera would sweep through the canal-laced city, killing hundreds at a time. Morris would never forget the screams of ambulances racing through the streets. Cholera seemed to strike without warning: a man who'd sampled the food from a street vendor would be hideously sick just hours later, lying limp and helpless as his vital nutrients flowed out of him amid ceaseless diarrhea. Death would often follow. Was it the food? The water? Who knew? Morris had heard about the horrors of hell in church on Sunday. He didn't think it could be much worse than the dread permeating a city in the grip of a cholera epidemic. 

Morris's parents had assured the shaken child that by staying away from street food, drinking boiled water, and, most important, taking fluids and antibiotics at the slightest cholera-like symptom, he would be safe. But as Morris now looked down at his suffering patient, he knew that such soothing assurances had no relevance here.
A year before, Ed Burke* had taken his good health for granted. 

Forty years old, lean and fit, he was a recently divorced accountant living with his mother while he tried to put his life together again. But Burke had been feeling weak and tired when he went to the University of Maryland's Medical Center for a checkup. He told doctors he'd been having stomach pains and chronic colds. A routine blood test revealed that he was suffering from leukemia. Though Burke was shocked and frightened by the diagnosis of cancer, his physician explained that most forms of leukemia responded well to chemotherapy. In all likelihood, he'd be able to undergo the regimen and soon resume a normal life. 

That was the beginning of the end. 

Burke's physician initiated chemotherapy almost immediately. Though often effective against leukemia and other cancers, the drastic treatment-with its searing chemicals that course through the body like Drano-can have the undesired effect of suppressing the immune system as well, sometimes leading to bacterial infections that the weakened immune system cannot contain. Physicians use antibiotics to help eradicate these potentially life-threatening infections. Sometimes these bugs prove resistant to the initial antibiotic, in which case the physician simply switches to another one. For decades, there had always been plenty in reserve. But for this particular case, the reserve had been exhausted. 

Burke's infection was caused by the bacterium known as Enterococcus faecium. One expert calls E. faecium the cockroach of microbial pathogens: proliferating freely in the gastrointestinal tract, it usually causes no more trouble than roaches colonizing a dark cupboard. But when breakdowns in the immune system allow the bugs to escape, they begin to cause serious infections, anywhere from the heart down to the urinary tract. After proving resistant to the initial antibiotics used, Burke's E. faecium also showed resistance to vancomycin, an older but still powerful antibiotic that represented the last-chance treatment for resistant enterococci when all else failed. This time vancomycin failed as well: vancomycin-resistant E. faecium, better known as VRE, had appeared in Burke's bloodstream, a dangerous escalation. That was when Morris had been called in. 

With his stocky build and powerful arms and shoulders, the ruddy faced Morris looked more like an ex-linebacker than a man of medicine, though his soft-spoken manner made him, at first glance, seem shy and retiring. But when he was stationed behind a microphone at a scientific conference and given a rapt audience for his impassioned calls to action about disturbing bacterial advances, the Southern preacher inside him very clearly emerged. 

Awed by the bacterial devastation he had witnessed in Thailand, Morris had grown up determined to do what he could to prevent such suffering in the future. Through willpower, hard work, and a keen intellect, he had turned himself into a formidable microbe hunter: schooled in tropical medicine, public health, food safety, and genetics, he had a breadth of training possessed by few in his field. Now Head of Infectious Diseases at the University of Maryland's Baltimore Veterans Affairs Medical Center, Morris was one of the country's best-known experts in his field. And Maryland needed an expert. In the last few years, Morris had seen an explosive growth of VRE right in Baltimore. 

The full import of this trend was difficult for patients to absorb. Certain strains of E. faecium were resistant to nearly all of the more than one hundred antibiotics that modern science had produced. They were, quite simply, unstoppable. 

Even harder to explain to patients was that E. faecium was a hospital bug. Almost certainly, it had infected Burke after he was in the care of his oncologists. It had infected him right there in his hospital bed. And how had it gotten there? Probably by alighting from the unwashed hands of a busy doctor, nurse, or other healthcare worker who had just had contact with another patient carrying the bug. Oncology wards and intensive care units of nearly all hospitals were notoriously rife with resistant bugs, though few institutions would admit as much. All too often, these drug-resistant bugs took weeks to develop into infections, so that the doctors and nurses who had inadvertently passed them to a patient might not ever learn what they had done. 

For elderly patients with chronic illnesses and ravaged immune systems, VRE was proving lethal, the extra infection that nudged an already sick person over the edge. Younger patients were usually impervious to it -unless, that is, their own immune systems were compromised by chemotherapy, as Burke's was, or by drugs given to prevent rejection of a transplanted kidney, or by some life-threatening, out-of-the-blue calamity: a car accident, perhaps, or a third degree burn. Then they were as susceptible as patients twice their age. Given that well over one million Americans were diagnosed each year with some form of invasive cancer-15 million since 1990-the number of potential victims for VRE was surprisingly large. 

When Morris entered Burke's hospital room the first time, the accountant had looked up at him with desperate hope. Gently, Morris had had to explain that he had no magic bullet for Burke's VRE-no cure at all. He could only hope that with the end of chemotherapy, Burke's white blood cell count would bounce back up quickly enough for his immune system to handle the infection itself. 

As if in answer to the Burke family's prayers, that was what happened- at first. The leukemia disappeared-whether it was in remission or gone for good, the doctors could not yet tell-and chemotherapy was halted. Burke's immune system began to recover and started producing the white blood cells responsible for killing bacteria that invade the bloodstream. As the white blood cells attacked the infection, the patient's fever broke, and he felt stronger every day. Within a week, he was sent home. 

Just ten months later, Burke was back in the hospital with a relapse of leukemia. Reluctantly, his doctors gave him more chemotherapy. When they did, VRE reappeared in his bloodstream. It had been lurking in his intestinal tract, a killer within. Twice more the man recovered enough for his immune system to fight the VRE to a standoff. But the superbug was not yet beaten. 

A year later, another relapse of leukemia, and more chemotherapy, pushed Burke's white blood cell count down too far. The bug that had been his constant companion, as Morris grimly put it, once again infected his bloodstream. Back came the high fever, the chills, the irregular heartbeat, the shortage of breath. But this time there was no rebound, even when chemotherapy was halted. Burke began vomiting, and his blood pressure plunged. As the flow of blood to his brain slowed to a trickle, his vision dimmed and he became disoriented. At the same time, his ever weakening heartbeat pumped less and less blood to his other vital organs. One by one they began shutting down, like lights winking out during a power blackout. As the kidneys and liver ceased to operate and cleanse his body of waste materials, Burke essentially poisoned himself. Finally came full-blown septic shock. Burke went pale and delirious, cold and clammy to the touch. He suffered a series of small heart attacks, and began to suffocate as his lungs filled with fluid. Eight days after the VRE infected his bloodstream a final time, Burke succumbed.
That was in September 1995. 

It doesn't get much worse than this, Morris thought at the time: a forty-year-old man with an infection no antibiotic can stop. But he would be wrong. Over the next six years, a grim new era of multi-resistant bacteria would unfold in-and out of-hospitals around the globe, making Burke's case seem all too typical. Relentlessly, these newly hardy, invisible bugs would proliferate all around us, some festering on bed rails and seat cushions, telephones and thermometers, others passing through the air from one human host to the next. Silently, they would colonize even the healthiest of us, coating our skin, nestling in our noses, spreading in our throats, swimming through our stomachs and gastrointestinal tracts-until it could not be said that any of us was ever without at least a smattering of highly drug-resistant bugs, waiting for the chance to infect those among us who grew suddenly weak and sick. 

The bugs were everywhere, exponentially multiplying. And each year now, fewer drugs seemed able to stop them.

*Some names, including this one, have been changed to protect the privacy of patients and their families.
                     ...........................

The Silent War

The Killers Within: The Deadly Rise of Drug-Resistant Bacteria


By Michael Shnayerson, Mark J. Plotkin

Page 2 of 2)

1
Most mornings for Glenn Morris started with his daughters. Only after he loaded the three of them-aged fifteen, twelve, and nine-into his old Infiniti G20 and dropped them off at the carpool did he head in to the hospital. But on the mornings of July 2001, while the girls were on summer vacation, Morris bid his wife goodbye and drove off alone to the front lines of a war none of his neighbors could see or hear. 

As a doctor in his late forties who was both head of epidemiology at his hospital and chairman of the associated university department, Morris could have graduated from going on clinical rounds. Still, he made a point of doing it two months a year. You couldn't just teach and do research, he believed?you had to see what new infections patients were incurring. Also, going on rounds made him feel the same stomach-tightening anticipation of the unknown that he'd experienced as a medical resident more than two decades before. And so he headed in from his Tudor?style house in Roland Park?a leafy neighborhood of large, comfortable homes built a century ago as one of Baltimore's first suburbs?to spend his days treating half a hundred very sick patients, many of them indigent, in the general ward of Baltimore's Veterans Affairs Medical Center. 

On the fifteen-minute drive into the city, Morris liked to listen to country-and-western music, its trucks and trains and broken hearts weaving through his thoughts of doxycycline or ciprofloxacin for one patient, vancomycin or Synercid for another. After parking in the hospital's underground garage and ascending, white-jacketed, to the general ward on the third floor, he started by checking his charts. Six new patients, he saw, had been admitted to the ward by way of the emergency room. One's condition looked especially bad. 

Morris went from bed to bed, trailed by a note-taking team of medical students, interns, and residents. Because this was a VA hospital, most of the patients in the general ward were male, elderly, and afflicted with chronic conditions. Many also had symptoms that indicated bacterial infection. A decade ago, antibiotics would have knocked out all of these infections almost immediately. Now on average, about 20 percent of patients on Morris's clinical rounds had infections resistant to one, two, three, or more drugs. When he wrote for medical journals, Morris described this multi drug resistance in dry, clinical terms that expressed none of the emotions he felt when he witnessed the ravages of an almost unstoppable infection. What he felt was dismay, and alarm, and a little twitching of fear. 

When Morris pointed out antibiotic-resistant infections to his interns and residents, he didn't need to emphasize that these were bacterial infections. They'd had it drilled into them in medical school that most infections are either bacterial or viral, and that bacterial infections are the ones that respond to antibiotics. Viruses, they knew, were a whole other matter. A virus is a tiny squiggle of protein-covered DNA or RNA, so small it isn't even a living, cellular organism: its only function is to bore into the cells of other organisms and force those cells to produce more viruses. (AIDS is caused by a virus; so is the common cold.) Antibiotics are useless against viruses. Bacteria, on the other hand, are one-celled organisms: the smallest creatures on the planet. The cell has various parts that enable the bacterium to live and replicate. Those parts can be targets for antibiotics. Unless, that is, the bacteria figure out how to change or deflect the drugs and make themselves resistant. 

A decade ago, Morris liked to remind his entourage, doctors had only to reach for penicillin, or one of the third-generation cephalosporins, or the then new, brilliantly effective fluoroquinolones. Now for empiric therapy?immediate treatment of new patients, before a lab could determine exactly what bug they had?doctors often found themselves in the dark, guessing which antibiotic would work. Often there was time to correct the therapy once cultures provided a profile of which drugs still worked against a bug. Sometimes there wasn't. Whenever a newspaper obituary listed cause of death as “complications” following surgery, chances were that a doctor had guessed wrong in terms of antibiotics?or that a bug had proved resistant to all of them. This was code that all healthcare workers, hospital staff, and HMO providers understood but few outside the medical world knew.

Most at risk were the old and the infirm, their immune systems deteriorated, especially in hospitals: at the dawn of the twenty-first century, roughly a third of all people older than sixty-five were dying from infections. Nearly as vulnerable, however, were the very young. Their immune systems were immature, not ravaged, but the result was the same. Tough, sometimes unstoppable strains of the usual suspects especially Streptococcus pneumoniae caused terrible, recurrent ear infections, or meningitis, or systemic bloodstream infections that shut down a child's vital organs. Every year, 1.2 million children around the world were estimated to die of , the leading bacterial cause of pneumonia. In the United States alone, was said to cause 500,000 cases of pneumonia, many of them pediatric, as well as 7 million ear infections, most of them pediatric, too. Only a decade ago, nearly all strains of had been susceptible to penicillin, the drug of choice for these infections. Now 45 percent of all strains were penicillin resistant. Some skeptics observed that with , a doctor could increase the dose of antibiotics and still hope to prevail in many cases. But that was cold comfort to parents who saw their children's lives imperiled. Gary Doern, Director of Clinical Microbiology at the University of Iowa Hospital in Iowa City, tracked on a national, ongoing basis and was staggered by its fast-rising rates of resistance. “Do the math,” he said grimly. “Where will it be fifteen years from now?” claimed as many victims outside the hospital as it did because, unlike many bacterial pathogens, it was spread by droplets: coughing passed it from host to host. Enterococcus faecium and Staphylococcus aureus infected hospital patients for the most part. But with S. aureus, the most virulent of the three, there were signs that that was changing. 

In January 2001, Bryan Alexander, eighteen, was found guilty of assault and drunken driving and sentenced to a 180-day term at a correctional boot camp in Mansfield, Texas. On January 4, he filed a written request for medical attention. According to his father, he filed two more requests; all three requested treatment at the local hospital. The camp nurse chose to refuse them. On January 9, Alexander died of pneumonia caused by a infection: an otherwise healthy eighteen-year-old killed by microscopic organisms in just days. A few months later, talk show host Rosie O'Donnell very nearly died after cutting her finger with a fishing knife and incurring a multi drugresistant infection. “On Tuesday night, April 3 [2001], my hand started to hurt. A lot. It was an itchy-hot-burning-searing-what the- hell-is-happening pain,” she recalled. The pain became unbearable; by the next day, O'Donnell was in the hospital, her hand so swollen it looked “like a kid's bright-red baseball mitt.” Multiple surgeries were needed to debride her finger? to cut away the dead and infected tissue? and decontaminate the site. Neither good health nor celebrity had protected these victims. 

Strains of all three of these common bacterial infections?E. faecalis, , and were now multi drug-resistant and spreading into the community. Strains of other bacteria Acinetobacter baumannii, Pseudomonas aeruginosa, and E. faecium remained hospital bound but had become resistant to all antibiotics. So widely and quickly were bacteria of different species trading their resistance genes that the vast, invisible world of bacteria could be thought of as a single, miasmic, multi celled organism, its trillions of parts all working together for the common goal of survival against antibiotics. What this boded for humans, the bugs' primary source of food, was in no way good. 

At the bedside of the patient whose case history worried him the most, Morris offered greetings with a cheer he didn't feel. The patient, a man in his seventies, had come to the hospital some time ago for a routine knee replacement. Apparently, while his knee was cut open in surgery, he'd incurred a methicillin-resistant infection, or MRSA. Nearly all strains of were now resistant to penicillin; almost half the hospital strains were also resistant to methicillin, the drug once thought to be a permanent replacement for penicillin. The infection had manifested itself a month after the man was back home. In he came again to the hospital for surgery to decontaminate the joint, followed by a six-week course, also at the hospital, of vancomycin. 

Vancomycin was a last resort, but that didn't make it a great drug. It often failed to penetrate deep bone infections, and it had to be administered intravenously, which meant using catheters, which became conduits for other disease-causing, or pathogenic, bugs. In this case, when vancomycin failed to stem the infection, the man's doctors removed the artificial joint altogether and fused the joint that remained. Then they hit him with another six-week course of vancomycin. Now he was back again, this time with a fever that almost certainly signaled the return, yet again, of his resistant infection. He had bedsores, a urinary catheter, a fused knee that was essentially worthless, and deep infections that just wouldn't quit. He was almost pathologically depressed, as well. His wife had remained a constant presence at his hospital bedside, but she was on the verge of a breakdown herself, unsure whether the downward spiral of complication after complication could ever be reversed. 

Morris knew he had to prescribe vancomycin. He had no choice. But where to put the IV? The man had endured so many intravenous lines he was running out of veins. Reluctantly, Morris put him on vancomycin via a central line?a catheter introduced into one of his large veins?and wished him luck. Privately, Morris thought the man would be lucky to live out the year. 

This was a case, Morris thought, that should never have happened: a man who'd come into the hospital in basically good health and emerged with a dire strain of MRSA. Doctors had a phrase they used among themselves to refer to such patients?the ones with infections resistant to one or more drugs and who seemed too sick to respond to any antibiotics. 

Train wrecks, they called them. 

Not every doctor and microbiologist at the dawn of the twenty-first century felt, as Morris did, that the golden era of antibiotics might be coming to an end. Not all felt that bacterial resistance had become, in the words of one physician, one of the greatest threats to the survival of the human species. But many did. And all agreed that resistance had become an urgent global issue. Stuart Levy, M.D., a Tufts University professor whose Cassandra?like warnings on the subject two decades before had all come to pass, saw only worse things to come. “We are clearly in a public health crisis,” he said to anyone who would listen. “In fact, we're on the road to an impending public health disaster.” Joshua Lederberg, Ph.D., Nobel laureate and longtime leading expert in antibiotic resistance at New York's Rockefeller University, felt that by comparison, the Ebola virus was small potatoes. “The odds of Ebola breaking out are quite low, but the stakes are very high. With antibiotic resistance, the odds are certain and the stakes are just as high. It is happening right under our noses.” 

The principal cause was overuse?and misuse?of antibiotics. In 1954, 2 million pounds of antibiotics had been produced in the United States. By the end of the century, the annual figure had risen, by some estimates, to more than 50 million pounds. Yet researchers at the federal Centers for Disease Control and Prevention (CDC) in Atlanta, Georgia, judged that a full third of the 150 million outpatient prescriptions for antibiotics written each year in the United States were unnecessary: either the infection turned out to be viral or the wrong drug was prescribed. Doctors prescribed the drugs partly to placate demanding patients and partly to protect themselves legally if they failed to prescribe an antibiotic for an infection that turned out to be direly bacterial. The proliferation of antibiotics killed many bacteria but gave the hardiest few some more chances to learn how the drugs worked?and how to resist them. 

It was a phenomenon that biologists called selective pressure. Among the billions of bacteria in a drop of human blood, or on a pinpoint of skin, or in a minute isolate of phlegm in the throat or stomach acid, might be a few?just a few?with a chance mutation that enabled them to resist the antibiotic used against them. If the antibiotic was then removed because the patient felt better and stopped using it?or sometimes even if it wasn't?those few resistant bugs would have an ecological niche, or clear field, in which to run wild. Because bacteria replicated so quickly?some bugs created a whole new generation every twenty minutes?the mutants could soon fill the niche. The pressure of the antibiotic, rather than obliterating them, had selected them to survive. 

Only a small portion of blame could be pinned on doctors in the community. Lethally resistant bacteria now resided in every hospital and nursing home in the world. Every year in U.S. medical institutions, 2 million patients contracted infections?bacterial, viral, and otherwise?and 90,000 died. Of those 90,000, many had drug-resistant bacterial infections, mostly . The CDC estimated that 40,000 Americans died each year of those infections. That was more than half the number of servicemen who had died during the entire Vietnam War. These deaths occurred in ones and twos, in hospital beds spread across the country, not by the scores on a single battlefield, so they tended to be noticed only by the patients' family and friends; by the hospitals, which certainly did nothing to publicize deaths caused by organisms within their institutions; and by HMOs, which quietly raised their premiums to help cover the estimated $5 billion cost of treating drug-resistant infections each year in the United States. Doctors and researchers published academic papers on drug-resistant bacteria, and, every year, their concerns grew more urgent, their prognoses more bleak. But the public remained largely oblivious to the problem, and as it did, incomprehensibly large populations of bacteria grew more and more resistant to more and more drugs. 

Often these resistant bacteria, once established by selective pressure, were passed by contact, on the hands of doctors or nurses, from patient to patient. Many found easy access to their victims' bloodstream through surgical incisions or wounds or by lingering on catheters and prostheses. One study had found a high incidence of pathogenic bacteria on computer keyboards and faucet handles in intensive care units, or ICU s. Another had found the bugs in the cushions and fabric of chairs in hospital common rooms, and in the acoustical tiles of hospital ceilings?lingering there, sometimes, for years. A third had found them on rectal thermometers, a fourth on stethoscopes. 

These various reservoirs dramatized the other dimension of the problem. If misuse of antibiotics created drug-resistant bacteria in the first place, poor infection control in hospitals allowed the bugs to spread. Every time a doctor or nurse failed to wash his or her hands before entering a patient's room, millions of invisible pathogens potentially came along for the ride. Yet how feasible was it for emergency department doctors to wash their hands before and after treating each next desperate patient, at a rate of five or six patients an hour? Or for doctors seeing up to two dozen patients on clinical rounds to do the same? In fact, one recent study conducted at Duke University had determined that only 17 percent of doctors treating patients in an intensive care unit washed their hands thoroughly and consistently. But to the bugs, every patient in an ICU presented another irresistible meal, and, with lax infection control, the bugs got fed. 

The most prevalent pathogens were bacteria that people carried with them as part of their natural “flora.” In their stomach and intestinal tract milled billions of enterococci. In their throat resided billions more streptococci. In their nose, and on their skin, lived the most worrisome of the big three: staphylococci. Some of these bugs were essential to digestion; others promoted health by staking turf that might otherwise be colonized by more virulent bugs. But given access to a weakened host?often through a cut in the skin?certain strains of these three species could be very bad bugs indeed. Enterococci caused skin and bloodstream infections; under the right circumstances they infected heart valves, too. Streptococci caused all manner of infections, from sore throats and earaches to pneumonia to the horrific necrotizing fasciitis, better known as flesh-eating bacteria. , the most virulent of the staphylococci, was also alarmingly widespread: between 20-40 percent of people, both healthy and sick, carried , usually in their nose or on their skin. Once it managed to enter the bloodstream of an immunocompromised person, caused surgical infections, pneumonia, heart and brain infections, and systemic bloodstream infections that shut down vital organs one by one with an inexorable end result. 

In the last decade, the bugs had acquired intricate mechanisms of resistance more quickly, as if the bacterial world was mirroring humanity's own ever quickening pace of development. Some succeeded in making their cell walls impermeable to antibiotics. Others created tiny pumps that actually vomited them out of the cell. Many antibiotics targeted one enzyme or another of the cell wall itself, attaching to it just as the bacterium was making more cell wall enzymes in order to replicate; yet many bugs had figured out how to change or replace those enzymes so that the drug failed to attach. Still other bugs' enzymes attacked the drug itself, slicing its chemical rings. The broad-spectrum antibiotics that most doctors reached for first were the ones likeliest to provoke these mechanisms. They killed a wide range of bugs, as the term implied, but used frequently they also gave that wide range of bugs more chances to develop successful mutations or import resistance genes from other bacteria. As microbiologist Barry Kreiswirth of New York City's Public Health Research Institute put it, “The bugs are getting stronger?and they're getting stronger faster.” 

Stuart Levy, a puckish fellow given to bow ties and elegant suits, often observed in his lectures, and in his classic book The Antibiotic Paradox, that the answer had caused the problem. Or rather, the answer to one problem had led to the next problem. Antibiotics had changed the world, eradicating the horror of pervasive infections that killed young and old alike. They had transformed surgery from a butchery in which most patients died of infections into a modern medical science. Yet the development of novel invasive therapies like organ transplants, prosthetic implants, dialysis machines for kidney failure, and chemotherapy for cancer had resulted in more and more immunosuppressed patients, which in turn provided additional fodder for the microbes. And the better that modern medicine enabled patients to overcome once-lethal conditions like faulty hearts or cancer, the longer it enabled them to live, the more likely they were to decline gently into the clutches of invisible microbial pathogens. “We can close the books on infectious diseases,” U.S. Surgeon General William Stewart had declared in 1969, suggesting, in a breathtaking show of hubris, that humans had beaten the bugs once and for all. But the bacteria were fighting back?and gaining on us. 

In the early 1990s, only doctors and nurses in hospitals had worried about drug-resistant bacteria. Now, like so many microscopic prisoners, the bacteria were breaking out. They caught their rides on the skin or in the intestinal tract of recovering patients in home care. They clung to aging patients shuttled back and forth between hospitals and long-term care facilities or nursing homes, especially in crowded cities like New York, which had become the epicenter in the United States of drug-resistant bacteria. They migrated to other places where people crowded together: prisons, military barracks, college dormitories, and, most frightening of all, daycare centers. Among the most likely carriers?or vectors, as the literature had it? were the doctors and nurses themselves. Once out, the bugs passed their resistance genes on to other bacteria, and resistance spread exponentially. 

What rule, after all, had ever restricted resistant bugs to hospitals? No rule they knew of. 

Resistance flowed from hospitals, it radiated out from antibiotic misuse by doctors in outpatient settings, and it welled up, too, from a third, ubiquitous source in the community: the agriculture industry. Of those 50 million pounds of antibiotics used in the United States each year, nearly half was consumed by animals. At vast commercial farming operations, tens of thousands of chickens were fed antibiotics in their drinking water if even a few appeared to be sick, a practice that all but assured the spread of resistance as the bacteria of healthy birds became familiar with, and then impervious to, the drugs. Nearly all livestock in America were also fed small, daily doses of antibiotics?“sub therapeutic doses,” they were called?as a time tested, if scientifically unproven, way to make the animals grow faster and fatter. If scientists had tried to devise a means of their own to foster resistance, they could not have come up with a better one than this. The tiny, sub-therapeutic doses, also called “growth promoters,” enabled bacteria in the animals to get familiar with the drugs but not be remotely threatened by them, and so blithely develop resistance to them. Often, resistance then passed from the livestock to the person who handled the livestock or ate under-cooked meat. The agriculture industry had denied this for years, its high-paid lobbyists sounding, as they called for ever more scientific proof, eerily like tobacco lobbyists denying that cigarettes caused cancer. 

The most common of the resistant food-borne infections were Salmonella and Campylobacter. Neither was as virulent as , the most worrisome bug of all. But both affected so many people that deaths did occur. Each year, Salmonella infected 1.4 million Americans and killed 500; Campylobacter infected 2.4 million Americans and killed 100. To epidemiologists like Morris looking at the big picture, the more alarming fact was that strains of Salmonella and Campylobacter were now resistant to as many as five drugs. A relatively new, synthetic class of antibiotics was very effective against both Salmonella and Campylobacter. Unfortunately, that class was the quinolones, which included drugs being used in livestock. Animal use of the quinolones was provoking resistance in the animals' own Salmonella and Campylobacter, which were then passing to people who ate that meat. The quinolones included ciprofloxacin, the drug that untold tens of thousands of Americans had persuaded their doctors to prescribe for them as an antidote to anthrax in the aftermath of September 11, 2001. The likelihood of any one of those people receiving an anthrax-laced envelope in the U.S. mail was very, very small. It was extremely likely, however, that many of those people would take Cipro at the first flu or cold symptom they feared might be anthrax, accelerating the spread of resistance. An entire class of drugs-the most important new class in four decades? might be compromised far sooner than anyone would have imagined five years before. 

The social fabric on which drug-resistant bacteria spread did not flutter to an end at the far edge of town or stop at the city limits. It passed from state to state, country to country, continent to continent. Chaos theory held, famously, that a butterfly flapping its wings in Africa might displace enough molecules around it to set off a series of reactions that resulted in a tornado over Kansas. With drug resistant bacteria, such a journey was fact: molecular biologists had traced the spread of earlier generations of methicillin-resistant from a single genetic mutation in Spain, or Australia, or Brazil, clear around the world. Americans and Europeans liked to imagine that they were safe from the myriad infectious diseases that plagued developing nations, and to some extent they were right. Their water was not contaminated by cholera; their air was not abuzz with malarial mosquitoes. But agricultural products carrying resistant Salmonella were sent routinely across international borders. For that matter, resistant bacteria could travel across the world in a day by plane, and often did. 

Throughout poor and developing countries, the list of other microbes on the march was abysmally long. Either no antibiotics for them were available or, ironically, too many were available, leading to rampant overuse. In China and Mexico, antibiotics were sold over the counter, no prescription needed, like cough drops. More and more, they were about as effective as cough drops, too. New, more powerful antibiotics were needed. But the new drugs-unlike penicillin and its many offspring?were very, very expensive. When two or more had to be combined, the cost rose, usually far beyond what the citizens of poor nations, or their governments, could pay. Treating a single case of multidrug-resistant tuberculosis with a whole coterie of drugs over an infection period as long as twenty-four months cost as much as $180,000 in the United States. In poor nations, the cost might be somewhat less?but so would the levels of sanitation and infection control. 

“We are seeing a global resurgence of infectious diseases,” U.S. Surgeon General David Satcher warned the U.S. Congress on the eve of the twenty-first century, a dramatic reversal of his office's stance a generation ago. Infectious diseases included viral killers?among them AIDS. But resistant bacterial pathogens were a growing subset, and each threat exacerbated the other. Roughly a third of the world's population, for example, was infected with tuberculosis, the result of early childhood exposure to the bug. Most of those carriers lived their whole lives without having the walled-off tubercles in their lungs break out and cause disease; most remained unaware they even had tuberculosis. But as AIDS spread, ravaging the immune systems of everyone it infected, many of its victims then developed active tuberculosis. 

The more widely TB spread, the more widely, and indiscriminately, a host of drugs were used against it. The more that happened, the more resistant TB became to those drugs. At the end of a particularly wrenching day on clinical rounds, even Reba McEntire did nothing to soothe Morris as he drove home from the hospital to his tree-lined neighborhood. He would glance at the handsome houses of Roland Park and think, They have no idea. Cosseted in their plush living rooms, most of his neighbors simply had no clue how many disease-causing bacteria were growing resistant to antibiotics, how in the silent, invisible war of bugs against drugs, the bugs were beginning to win. 

The first thing Morris did when he walked into his big house was go to the kitchen and wash his hands?once more, with soap, just to be sure. Then he went in to hug his wife, a physician herself, and his daughters. Sometimes he marveled at how his daughters took their perfect health-and everything else?for granted. Like most children, they took limited interest in the details of their father's day. Perhaps that was just as well. Morris didn't want to scare them with details of his latest cases. Nor did he want to say that he doubted their children would have antibiotics for every need. More and more infections, he felt sure, would be unstoppable killers, just as they had before the age of antibiotics began. 

Could it be only a decade ago that most doctors and drug company scientists had believed the antibiotics they had on hand would work forever? In that whisker of time, the entire medical establishment had been forced to swallow a very bitter pill. No antibiotic would work forever. Eventually, every bacterial pathogen would learn how to become resistant to every drug used against it. Given how quickly bacteria were adapting now, Glenn Morris was only echoing the fears of most colleagues when he predicted that many bacterial pathogens would likely be resistant to all existing antibiotics in another human generation or two. In a decade, after all, while some modest fraction of humanity reproduced itself, bacteria reproduced 50,000 times, trying each time, in some soulless but utterly determined, Darwinian way, to adapt in order to prevail. 

How, Morris wondered, could our species have made such a monumental blunder? Sixty years ago, scientists had discovered the first of the natural antibiotics and seen how brilliantly they worked against various bacteria: the biggest medical find of the century. In their excitement, they had failed to remember that bacteria had existed for billions of years, probably before any other life on the planet. Their ancestors, trillions of microbial generations ago, had seen the appearance of brontosauruses, tyrannosauruses, woolly mammoths, and saber-toothed tigers. And they had feasted on their carcasses. After surviving unimaginable extremes of fire and ice, were those bugs really going to let themselves be vanquished by a brand-new arrival in geologic time, using weapons they themselves had devised?

END OF EXTRACTS


About the Author
For much of the past 20 years, Dr. Mark J. Plotkin has worked with and learned from the ancient shamans of Central and South America. These years in the rainforests have provided Dr. Plotkin with incomparable knowledge of healing plants and traditions of the shamans


The Killers Within: The Deadly Rise of Drug-Resistant Bacteria
  A battle is taking place on the frontiers of medicine between rapidly evolving bacteria that threaten our health and the doctors who are struggling to outwit them. These bacteria are everywhere: in and on our bodies, in homes, schools, hospitals, crowded


http://www.enotalone.com/article/3995.html

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

Phage Therapy

Phage as Antibiotics

PHAGE THERAPY: BACTERIOPHAGES AS ANTIBIOTICS 


Elizabeth Kutter, Evergreen State College, Olympia, Wa.98505 -- Nov. 15, 1997  t4phage@evergreen.edu; 360 867-6099 or 867-6523  



INTRODUCTION 
 
Bacteria resistant to most or all available antibiotics are causing increasingly serious problems, raising widespread fears of returning to a pre-antibiotic era of untreatable infections and epidemics. Despite intensive work by drug companies, no new classes of antibiotics have been found in the last 30 years. There are hopes that the newfound ability to sequence entire microbial genomes and to determine the molecular bases of pathogenicity will open new avenues for treating infectious disease, but other approaches are also being sought with increasing fervor. One result is a renewed interest in the possibilities of bacteriophage therapy -- the harnessing of a specific kind of viruses that attack only bacteria to kill pathogenic microorganisms (cf. Levin and Bull, 1996; Lederberg, 1996; Radetsky, 1996; Barrow and Soothill, 1997).  

Phage therapy was first developed early in this century and showed much promise but also aroused much controversy. It has been little used in the West since the advent of antibiotics in the 1940s. However, extensive clinical research and implementation of phage therapy continued to be carried out in Eastern Europe over the last 50 years. The results of that work effectively complement the limited recent animal work in the West that is primarily cited in the recent articles, encouraging optimism that phage can indeed play an important role in dealing with infections involving increasingly drug-resistant microbes. We need to draw as much as possible on the largely-unknown body of knowledge that has accumulated in Poland, France and many parts of the former Soviet Union (FSU) as we again explore phage therapy, and to give credit where it is due for the many years of hard, careful work they have invested in the field. This paper is written primarily to put phage therapy in historical and ecological context and to explore some of the more interesting and extensive work in Eastern Europe, little of which has been accessible in English.  
 
THE NATURE OF BACTERIOPHAGES 


Viruses are like space ships that are able to carry genetic material between susceptible cells and then reproduce in those cells, just as the AIDS virus, HIV, specifically infects human T lymphocytes which carry a particular surface protein called CD4. Each virus consists of a piece of genetic information, determining all of the properties of the virus, which is carried around packaged in a protein coat. In the case of bacteriophages, the targets are specific kinds of bacterial cells; they cannot infect the cells of more complex organisms because of major differences in key intracellular machinery as well as in cell-surface proteins. Most phages have tails, the tips of which have the ability to bind to specific molecules on the surface of their target bacteria. The viral DNA is then injected through the tail into the host cell, where it directs the production of progeny phages -- often over a hundred in half an hour. Each strain of bacteria has characteristic protein, carbohydrate and lipopolysaccharide molecules present in large quantities on its surface. These molecules are involved in forming pores, in motility, in binding of the bacteria to particular surfaces; each such molecule can act as a receptor for particular phages. Development of resistance to a particular phage generally reflects mutational loss of its specific receptor; this loss often has negative effects on the bacterium and does not protect it against the many other phage which use different receptors.  

Each kind of bacteria has its own phages, which can be isolated wherever that particular bacterium grows -- from sewage, feces, soil, even ocean depths and hot springs. The process of isolation is easy. Just let the sample sit in an appropriate nutrient broth, separate off the liquid part, and pass it through a filter with pores so tiny that bacteria can't get through. Then mix it (at several different dilutions) with a culture of the bacteria in question. Spread a few drops on a block of appropriate nutrient medium which is made firm with agar taken from seaweed. The next day, one sees a dense covering or lawn of bacteria with round clear spots, called plaques. Each plaque contains many million phage particles, all progeny of one phage which was immobilized there on the agar. That phage infected a cell, multiplied inside it, and caused it to burst. This released many phages, which infected nearby cells and repeated the process. One can stick a toothpick into one of these plaques, transfer it to a fresh culture of the bacteria in liquid medium, and grow up a homogeneous stock of descendants of that particular phage, whose properties can then be studied.  


HISTORICAL CONTEXT 


Discovery 


A century ago, Hankin (1896) reported that the waters of the Ganges and Jumna rivers in India had marked antibacterial action which could pass through a very fine porcelain filter; this activity was destroyed by boiling. He particularly studied the effects on Vibrio cholerae and suggested that the substance responsible was what kept cholera epidemics from being spread by ingestion of the water of these rivers. However, he did not further explore the phenomenon. Edward Twort (1915) and Felix d'Herelle (1917) independently reported isolating filterable entities capable of destroying bacterial cultures and of producing small cleared areas on bacterial lawns, seemingly implying that discrete particles were involved. They are jointly given credit for the discovery. It was d'Herelle, a Canadian working at the Pasteur Institute in Paris, who gave them the name "bacteriophages"-- using the suffix phage not in its strict sense of to eat, but in that of developing at the expense of (d'Herelle, 1922, p. 21) - and who made them a major part of his life's work. D'Herelle, a largely self-trained microbiologist, had just spent 10 years in Guatemala, Mexico and Argentina. There, he dealt with epidemics of dysentery, yellow fever and a coffee-killing fungus, isolated a bacterium from dying locusts to use in controlling locust plagues, and explored several interesting fermentation challenges - all good preparation for his later work with phages, as discussed in an interesting fashion by Summers (1998). At the Pasteur Institute, he was carrying out a careful study of vaccine preparation techniques using a model system - "B. typhimurium" in its natural host, mice; he felt strongly that meaningful data on immunity and pathogenicity could only be obtained when natural hosts were used. In his spare time, he was also doing research with dysentary patients - a frequent problem in wartime France. From the feces of several of these patients, he isolated a filterable anti-Shiga "microbe" which multiplied through many serial passages on its host bacterium, and which could produce tiny clear circles on a plate of this "Shiga bacillus" (d'Herelle, 1917).  

 D'Herelle went on to carefully characterize bacteriophages as viruses which multiply in bacteria and worked out the details of infection by various phages of different bacterial hosts under a variety of environmental conditions, always working to combine natural phenomena with laboratory findings, to better understand immunity and natural healing from infectious disease (Summers, 1998). The Ninetieth Annual Meeting of the British Medical Association in Glasgow featured a very interesting discussion between d'Herelle, Twort and several other eminent scientists of the day on the nature and properties of bacteriophages (d'Herelle et al, 1922). The main issue at that time was whether the observed bacteriolytic principle was an enzyme produced by bacterial activity or a form of tiny virus with some sort of life of its own, as claimed by d'Herelle; this controversy continued for many years, splitting the rapidly-growing community of people working with phages. 

D'Herelle summarized the early phage work in a 300-page book "The Bacteriophage" (1922). He wrote classic descriptions of plaque formation and composition, infective centers, the lysis process, host specificity of adsorption and multiplication, the dependence of phage production on the precise state of the host, isolation of phages from sources of infectious bacteria and the factors controlling stability of the free phage. He quickly became fascinated with the apparent role of phages in the natural control of microbial infections. He noted for example the frequent specificities of the phages isolated from recuperating patients for their own disease organisms and the rather rapid variations over time in their phage populations. He thus worked throughout his life to develop the potential of using properly selected phages as therapeutic agents against the most devastating health problems of the day. However, he initially focussed on simply understanding phage biology. Thus, the first known report of successful phage therapy came not from d'Herelle but from Bruynoghe and Maisin (1921), who used phage to treat staphylococcal skin infections. 


After a year at the Pasteur Institute of Saigon, d'Herelle returned to tight physical conditions, personal conflict and intellectual controversy at the Pasteur Institute in Paris. He soon accepted an offer to move to the Netherlands, where he was provided better conditions for his work with the recovery from infectious disease and the properties of bacteriophages, published his first book and a number of papers, and received an honorary MD degree. In 1925, he became a health officer for the League of Nations, based in Alexandria, Egypt, with special responsibility for controlling infectious disease on ships passing through the Suez Canal and during some of the major Muslim pilgrimages. Phage therapy and sanitation measures were the primary tools in his arsenal to deal with major outbreaks of infectious disease throughout the Middle East and India. Throughout this period, he continued publishing on his research and clinical trials and assisting others who were willing to do so with phages and consultations, often undertaking extended travel at his own expense. One of the most extensive trials of phage therapy he helped set up was the Bacteriophage Inquiry of 1927-1936 (Summers, 1993), which led to "what seems to be convincing results, endorsed by august committees" yet still left many skeptics of phage therapy; these studies deserve closer scrutiny
 
In 1928, d'Herelle was invited to Stanford to give the prestigious Lane Lectures; his discussion of "The Bacteriophage and its Clinical Applications" was published as a monograph (d'Herelle and Smith, 1930). He gave many lectures for medical schools and societies as he crisscrossed the country. He then went on to Yale to take up a regular faculty position, arranged with the support of George Smith, who had translated his first two books into English. He continued to spend summers in Paris working with the phage company he had established there, run by his son-in-law, in response to strong demands for phage preparations with careful quality control; this period is discussed particularly well by Summers (1998). He returned permanently to Europe in 1933, spending much time the following two years in Tiflis (Tbilisi), Georgia, helping to set up an international Bacteriophage Institute there, as discussed further below.  

From early on, one major practical use of phages was for bacterial identification through a process called phage typing -- the use of patterns of sensitivity to a specific battery of phages to precisely identify microbial strains. This technique takes advantage of the fine specificity of many phages for their hosts and is still in common use around the world. The sophisticated ability of phages to destroy their bacterial hosts can also have a very negative commercial impact; phage contaminants occasionally spread havoc and financial disaster for the various fermentation industries that depend on bacteria, such as cheese production and fermentative synthesis of chemicals (cf. Saunders, 1994)

 Phage therapy was tried extensively and many successes were reported for a variety of diseases, including dysentery, typhoid and paratyphoid fevers, cholera, and pyogenic (pus-producing) and urinary-tract infections. Phages were poured directly into lesions, given orally or applied as aerosols or enemas. They were also given as injections -- intradermal, intravascular, intramuscular, intraduodenal, intraperitoneal, even into the lung, carotid artery and pericardium. The early strong interest in phage therapy is reflected in the fact that some 800 papers were published on the topic between 1917 and 1956; the results have been discussed in some detail by Ackermann and Dubow (1987). The reported results were quite variable. Many physicians and entrepreneurs became very excited by the potential clinical implications and jumped into applications with very little understanding of phages, microbiology or basic scientific process. Thus many of the studies were anecdotal and/or poorly controlled, many of the failures were predictable and some of the reported successes did not make much scientific sense. Often, uncharacterized phages at unknown concentrations were given to patients without specific bacteriological diagnosis, and there is no mention of follow up, controls or placebos

Much of the understanding gained by d'Herelle was ignored in this early work, and inappropriate methods of preparation, "preservatives" and storage procedures were often used. On one occasion, d'Herelle reported testing 20 preparations from various companies and finding that not one of them contained active phages (Summers, 1998). On another occasion, a preparation was advertised as containing a number of different phages, but it turned out that the technician responsible had decided it was easier to grow them up in one large batch than in separate batches. Not too surprisingly, checking the product showed that one phage had outcompeted all the others and this was not, in fact, a polyvalent preparation. This was the origin of the phage T7, whose RNA polymerase now plays a major role in biotechnology (William Summers, personal communication). In general, there was no quality control except in a few research centers. Large clinical studies were rare and the results of those few were largely inaccessible outside of Eastern Europe.  

In 1931, an extensive review of bacteriophage therapy was commissioned by the Council on Pharmacy and Chemistry of the American Medical Association (Eaton and Bayne-Jones, 1931). Its purpose was "(a) to present summaries and discussions of (1) the experimentally determined facts relating to the bacteriophage phenomenon, (2) the laboratory and clinical evidence for and against the therapeutic usefulness of bacteriophage and (3) the relation of so-called antivirus to materials containing bacteriophage, and (b) to serve as a basis for a survey of the status of some of the commercial preparations." With 150 references, this report made a major effort to survey at least what they considered the most significant papers and reviews. In evaluating this report, it is important to realize how little was yet known then about bacteriophages. In fact, their first conclusion was "Experimental studies of the lytic agent called "bacteriophage" have not disclosed its nature. D'Herelle's theory that the material is a living virus parasite of bacteria has not been proved. On the contrary, the facts appear to indicate that the material is inanimate, possibly an enzyme." In retrospect, the proof that phages are viruses looks solid and it is hard to see how they could have come to this conclusion, which clearly impacted all of their other findings.

 These included: "2.) Since it has not been shown conclusively that bacteriophage is a living organism, it is unwarranted to attribute its effect on cultures of bacteria or its possible therapeutic action to a vital property of the substance. 3.) While bacteriophage dissolves sensitive bacteria in culture and causes numerous modifications of the organisms, its lytic action in the body is inhibited or greatly impeded by blood and other bodily fluids. 4.) The material called bacterophage is usually a filtrate of dissolved organisms, containing, in addition to the lytic principle, antigenic bacterial substances, products of bacterial growth and constituents of the culture medium. The effects of all these constituents must be taken into consideration whenever therapeutic action is tested. 5) A review of the literature on the use of bacteriophage in the treatment of infections reveals that the evidence for the therapeutic value of lytic filtrates is for the most part contradictory. Only in the treatment of local staphylococcic infections and perhaps cystitis has evidence at all convincing been presented."   

 This assessment clearly had a strong influence on the investment of the medical community in exploring phage therapy seriously, at least in the United States. Points are raised which still need to be considered, particularly in terms of the many trials described there in animals or humans which seemed to show little or no success and in terms of such potentially confounding explanations of the successes as the apparent strong stimulation of natural immune mechanisms by the bacterial debris in the lysates used. Then in the 1940's, the new "miracle" antibiotics such as penicillin became became widely available, and phage therapy was largely abandoned in the western world. 


 
SPECIFIC PROBLEMS OF EARLY PHAGE THERAPY WORK 

Today, many believe that phage therapy was proven not to work in the early part of this century. However, it appears that it simply was never given sufficient and appropriate trial, and reassessment is warranted. It is thus important to consider in some detail potential reasons for the early problems and the questions as to efficacy. These included: 
  1. Paucity of understanding of the heterogeneity and ecology of both the phages and the bacteria involved.
  2. Failure to select phages of high virulence against the target bacteria before using them in patients.
  3. Use of single phages in infections which involved mixtures of different bacteria
  4. Emergence of resistant bacterial strains. This can occur by selection of resistant mutants (a frequent occurrence if only one phage strain is used against a particular bacterium) or by lysogenization (if temperate phages are used, as discussed below).
  5. Failure to appropriately characterize or titer phage preparations, some of which were totally inactive.
  6. Failure to neutralize gastric pH prior to oral phage administration.
  7. Inactivation of phages by both specific and nonspecific factors in body fluids.
  8. Liberation of endotoxins as a consequence of widespread lysis of bacteria within the body (which physicians call the Herxheimer reaction). This can lead to toxic shock, and is a potential problem with chemical antibiotics as well.
  9. Lack of availability or reliability of bacterial laboratories for carefully identifying the pathogens involved, necessitated by the relative specificity of phage therapy.
In making the choice to again explore the possibilities of phage therapy, we should also consider their many potential advantages, discussed in more detail below: 
  1. They are both self-replicating and self-limiting, since they will multiply only as long as sensitive bacteria are present and then are gradually eliminated from the individual and the environment.
  2. They can be targeted far more specifically than can most antibiotics to the specific problem bacteria, causing much less damage to the normal microbial balance in the body. The bacterial imbalance or "dysbiosis" caused by treatment with many antibiotics can lead to serious secondary infections involving relatively resistant bacteria, often extending hospitalization time, expense and mortality. Particular resultant problems include infection by pseudomonads, which are especially difficult to treat, and Clostridium difficile, cause of serious diarrhea and membranous colitis (cf. Fékéty, 1995).
  3. Phages can often be targeted to receptors on the bacterial surface which are involved in pathogenesis, so that any resistant mutants are attenuated in virulence.
  4. Few side effects have been reported for phage therapy.
  5. Phage therapy would be particularly useful for people with allergies to antibiotics.
  6. Appropriately selected phages can easily be used prophylactically to help prevent bacterial disease in people or animals at times of exposure, or to sanitize hospitals and help protect against hospital-acquired (nosocomial) infections.
  7. Especially for external applications, phages can be prepared fairly inexpensively and locally, facilitating their potential applications to underserved populations.
  8. Phage can be used either independently or in conjunction with other antibiotics to help reduce the development of bacterial resistance.
PROPERTIES OF PHAGES 
One major source of confusion in the early phage work was the perception that all phages were fundamentally similar, though subject to adaptive change depending on the recent conditions of growth. One consequence of this was that often new phages were isolated for each series of studies, so that there was little continuity or basis for comparison. Phages specific for over 100 bacterial genera have now been isolated (Ackermann, 1996); they have been found virtually everywhere that they have been sought. However, only few have yet been well studied or classified (cf. Ackermann and DuBow, 1987)  
A second early source of confusion affecting therapeutic uses was the question of whether the lytic principle termed "bacteriophage" simply reflected an inherent property of the specific bacteria or required regular reinfection by an external agent. During the 1930s and 1940s, it became increasingly clear that in some senses both were true -- that there were in fact two quite fundamentally different groups of bacteriophages. Lytic phages always have to infect from outside, reprogram the host cell and release a burst of phage through breaking open, or lysing, the cell after a relatively fixed interval. Lysogenic phages, on the other hand, have another option. They can actually integrate their DNA into the host DNA, much as HIV can integrate the DNA copy of its RNA, leading to virtually permanent association as a prophage with a specific bacterium and all its progeny. The prophage directs the synthesis of a repressor, which blocks the reading of the rest of its own genes and also those of any closely-related lysogenic phages -- a major advantage for the bacterial cell. Many prophages further aid their host by helping protect against various unrelated, lytic phages. Occasionally, a prophage escapes from regulation by the repressor, cuts its DNA back out of the genome by a sort of site-specific recombination and goes ahead to make progeny phage and lyse open the cell. Sometimes the cutting-out process makes mistakes and a few bacterial genes get carried along with the phage DNA to its new host; this process, called transduction, plays a significant role in bacterial genetic exchange. Such lysogenic phages are very bad candidates for phage therapy, both due to their mode of inducing resistance and to the fact that they can potentially lead to transfer of genes involved in bacterial pathogenicity; this is discussed in more detail below. However, their specificity often makes them very useful for phage typing in distinguishing between bacterial strains. 
Key technical developments that helped clarify the general nature and properties of bacteriophages included: (1) the concentration and purification of some large phages by means of high-speed centrifugation and the demonstration that they contained equal amounts of DNA and protein (Schlesinger, 1933 a, b) and (2) visualization of phages by means of the electron microscopic (EM) (Ruska,1940; Pfankuch and Kausche, 1940). Soon after, Ruska (1943) reported the first attempts to use the EM for phage systematics; this has since become a key tool of the field (cf. Ackermann and DuBow, 1987). Each phage was found to have its own specific shape and size, from the "lunar lander"-style complexity of T4 and its relatives to the globular heads with long or short tails of lambda and T7 to the small filamentous phages that looked much like bacterial pili (See adjacent figure, from Ackermann, 1996
return to top
    
LYTIC PHAGES
 →
A much better understanding of the interactions between lytic phage and bacteria came from detailed one-step growth curve experiments expanding on the work of d'Herelle (1922) (Ellis and Delbrück, 1939, Doermann,1952). These demonstrated an eclipse period during which the DNA began replicating and there were no free phage in the cell, a period of accumulation of intracellular phage, and a lysis process which released the phage to go in search of new hosts. An example of this phage infection cycle is outlined in the adjacent figure
→. 
In the early 1940's, developments occurred which were to have a major impact on the orientation of phage research in the United States and much of western Europe, strongly shifting the emphasis from practical applications to basic science. Physicist-turned-phage-biologist Max Delbrück began working with key phage biologists Alfred Hershey and Salvador Luria and formed the "Phage Group", which eventually expanded dramatically with aid of the summer Phage Courses at Cold Spring Harbor, Long Island. These ran for many years starting in 1945 and regular phage meetings still continue there.  
 →
The influence of the Phage Group on the origins of molecular biology has been well documented (cf. Cairns et al, 1966; Fischer and Lipson, 1988; Summers, 1993b). Virulent phages had just the right balance of complexity and simplicity to tease out the key concepts of cell regulation at the molecular level. However, a major element of the rapid success of phage as model systems was that Delbrück convinced most phage biologists in the United States to focus on one bacterial host (E. coli B) and 7 of its lytic phages, building a very strong, tightly focussed community all working on the same set of problems, able to build effectively on each other's work and communicate easily. The 7 phages were arbitrarily chosen and named T(type)1-T7. As it turned out, T2, T4 and T6 were quite similar to each other, defining the "T-even" family of phages, discussed in more detail below. These phages were key in demonstrating that DNA is the genetic material, that viruses can encode enzymes, that gene expression is mediated through special copies in the form of "messenger RNA", that the genetic code is triplet in nature, and many other fundamental concepts. The negative side of this strong focus on a few phages growing under rich laboratory conditions, however, was that there was very little study or awareness of the ranges, roles and properties of bacteriophages in the natural environment, or of phages that infect other kinds of bacteria. 
return to top
    →
RATIONAL PHAGE THERAPY 
 →
The rapid, powerful developments in the understanding of phage biology had the potential to facilitate more rational thinking about the therapeutic process and the selection of therapeutic phages. However, there was generally little interaction between those who were so effectively using phage as tools to understand molecular biology and those still working on phage ecology and therapeutic applications. Many in the latter group were spurred on by concern about the increasing incidence of nosocomial (hospital-acquired) infections and of bacteria resistant against most or all known antibiotics. This is particularly true in Poland, France and the former Soviet Union where use of therapeutic phages never fully died out and where there has been some ongoing research and clinical experience. In France, Dr. Jean-François Vieu led the therapeutic phage efforts until his retirement some 10 years ago, he worked in the "Service des Entérobactéries" of the Pasteur Institute in Paris and, for example, prepared Pseudomonas phages on a case-by-case basis for patients. The experience there is discussed in Vieu (1975) and Vieu et al. (1979). Phage therapy was used extensively in many parts of Eastern Europe as a natural part of clinical practice, and there are now companies in Moscow and several other Russian cities making phage preparations for this purpose. However most of the research and much of the phage preparation came under the direction of key centers in Tbilisi, Georgia and in Wroclaw, Poland. I will thus focus on the work of these two groups. 

Polish Academy of Sciences, Wroclaw

The most detailed publications documenting phage therapy have come from Stefan Slopek's group at the Institute of Immunology and Experimental Medicine of the Polish Academy of Sciences in Wroclaw. This group published a series of detailed papers in the Archivum Immunologiae et Therapie Experimentalis (cf. Slopek et al, 1983, 1985, 1987), describing the results of phage treatments carried out from 1981 to 1986 with 550 patients. This set of studies involved ten Polish medical centers, including the Wroclaw Medical Academy Institute of Surgery Cardiosurgery Clinic. Children's Surgery Clinic and Orthopedic Clinic; the Institute of Internal Diseases Nephrology Clinic and Clinic of Pulmonary Diseases. The patients ranged in age from 1 week to 86 years; in 518 of the cases, phage use followed unsuccessful treatment with all available antibiotics. The major categories of infections treated were long-persisting suppurative fistulas, septicemia, abscesses, respiratory tract suppurative infections and bronchopneumonia, purulent peritonitis and furunculosis. In a final summary paper (Slopek et al, 1987), the authors carefully analyzed the results with regard to such factors as nature and severity of the infection and monoinfection vs. infection with multiple bacteria. Rates of success ranged from 75 to 100 % (92% overall), as measured by marked general improvement of health, tendency to heal of local wounds and disappearance of titratable bacteria; 84% demonstrated full elimination of the suppurative process and healing of local wounds. Infants and children did particularly well; not surprisingly, the poorest results came with the elderly and those in the final stages of extended serious illness, with weakened immune systems and generally poor resistance.  
The bacteriophages used all came from the extensive collection of the Bacteriophage Laboratory of the Institute of Immunology and Experimental Therapy; in the later studies, some of the specific phages used were named. All were virulent, capable of completely lysing the bacteria being treated. In the first study alone, 259 different phages were tested (116 for Staphylococcus, 42 for Klebsiella, 11 for Proteus, 39 for Escherichia, 30 for Shigella, 20 for Pseudomonas, and one for Salmonella); 40% of them were selected to use directly for therapy. All of the treatment was in a research mode, with the phage prepared at the Institute by standard methods and tested for sterility. Treatment generally involved 10 ml of sterile phage lysate orally half an hour before each meal, with gastric juices neutralized by (basic) Vichy water, baking soda or gelatin. In addition, phage-soaked compresses were generally applied three times a day where dictated by localized infection. Treatment ran for 1.5-14 weeks, with an average of 5.3; for intestinal problems, short treatment sufficed, while it was very long for such problems as pneumonia with pleural fistula and pyogenic arthritis. Bacterial levels and phage sensitivity were continually monitored, and the phage(s) being used were changed if the bacteria lost their sensitivity; therapy was generally continued for two weeks beyond the last positive test for the bacteria.  
Few side effects were observed; those that were seen seemed directly associated with the therapeutic process. Pain in the liver area was often reported around day 3-5, lasting several hours; the authors suggested that this might be related to extensive liberation of endotoxins as the phage were destroying the bacteria most effectively. In severe cases with sepsis, patients often ran a fever for 24 hours about days 7-8 (Slopek et al, 1981a). Various other methods of administration were successfully used, including aerosols and infusion rectally or in surgical wounds. Intravenous administration was not recommended for fear of possible toxic shock from bacterial debris in the lysates (Slopek et al, 1981a). However, it was clear that the phages readily got into the body from the digestive tract and multiplied internally wherever appropriate bacteria were present, as measured by their presence in blood and urine as well as by therapeutic effects (Weber-Dabrowska et al, 1987). This interesting and rather unexpected finding has been replicated in other studies and systems (** add refs.).  
 →
Detailed notes were kept throughout on each patient. The final evaluating therapist also filled out a special inquiry form that was sent to the Polish Academy of Science research team along with the notes. The Computer Center at Wroclaw Technical University carried out the extensive analyses of the data. The authors used the categories established in the WHO (1977) International Classification of Diseases in assessing results. They also looked at the effects of age, severity of initial condition, type(s) of bacteria involved, length of treatment and other concomitant treatments. The papers include many specific details on individual patients which help give insight into the ways phage therapy was used, as well as an in-depth analysis of difficult cases.  
 →
Bacteriophage Institute, Tbilisi 
 →
The most extensive and least widely known work on phage therapy was carried out under the auspices of the Bacteriophage Institute at Tbilisi, d'Herelle's institute in the former Soviet republic of Georgia. The work there will thus be discussed in some detail.
Georgia is an ancient and beautiful country of 5 million people, lying tucked in a valley between the High Caucasus mountains at the south of Russia, the Samtsxe-Dgavaxete range bordering on Turkey and Armenia, and the Black Sea. Through all the centuries of political upheaval at this crossroads of the ancient world, it has managed to keep its own culture and unique language, which is related only (and remotely) to Basque. It has been Christian since the third century, but prides itself strongly on its openness to all religions and cultures, its synagogue, mosque, and various Christian churches all clustered in the heart of old Tbilisi. Strong emphasis is placed on culture, intellectual pursuits and hospitality; the literacy rate is 100%, according to the 1996 UN Human Development Report on Georgia, and it has a long tradition of excellence in fields from music to mathematics to wine-making and cooking. 
 →
According to various Georgian physicians with whom I have spoken, phage therapy is part of the general standard of care there, used especially extensively in pediatric, burn and surgical hospital settings. Phage preparation was carried out on an industrial scale, employing 1,200 people just before the break-up of the Soviet Union. Tons of tablets, liquid preparations and spray containers of carefully-selected mixtures of phages for therapy and prophylaxis were shipped throughout the former Soviet Union (FSU) each day. They generally were available both over the counter and through physicians. The largest use was in hospitals, to treat both primary and nosocomial infections, alone or in conjunction with chemical antibiotics. They played a particularly important role when antibiotic-resistant organisms were found. The military is still one of the strongest supporters of phage therapy research and development, because phages have proven so useful for wound and burn infections as well as for preventing debilitating gastrointestinal epidemics among the troops. 
The historical background of the institute is interesting, and reflects a relatively unknown period in d'Herelle's caeer. The following material comes from a number of people at the Institute, from a recent article by Shrayer (1996) on d'Herelle in Russia, from Summers (1998) and from d'Herelle's own work. 
In 1917, George Eliava, of the Georgian Institute of Microbiology, noticed that the water of the Koura (Mtkvary) river in Tbilisi (Tiflis) had a bactericidal action - an observation that could be explained by d'Herelle's bacteriophage discovery. Eliava spent several extended periods in Paris at the Pasteur Institute and was a very early and staunch collaborator of d'Herelle's; several papers of his are cited in d'Herelle's first book on phages (1922). The two developed the dream of founding an Institute of Bacteriophage Research in Tbilisi, to be a world center of phage therapy for infectious disease, including scientific and industrial facilities and supplied with its own experimental clinics. The dream quickly became a reality due to the support of Sergo Ordjonikidze, the People's Commissar of Heavy Industry, despite KGB opposition to this "foreign project" and personal conflicts between Eliava and Beria, then the local KGB head. A large campus on the river Mtkvary was allotted for the project in 1926. For many years, d'Herelle sent supplies, equipment and library materials, most of which he paid for himself. In 1934 -1935 he and his wife spent two 6-month periods working in Tbilisi, during which time he visited Kamenski, the People's Commisar of Health Care, in Moscow and turned down an invitation to move there. He also wrote a book on "The Bacteriophage and the Phenomenon of Recovery", which was translated into Russian by Eliava and dedicated to Stalin. D'Herelle intended to eventually move to Georgia; in fact, the cottage built for his use still stands on the Institute grounds. However, in 1937 Eliava was arrested as a "People's Enemy" by Beria, then head of the KGB in Georgia and soon to direct the Soviet KGB as Stalin's much-feared henchman. Eliava was summarily executed without a trial, sharing the tragic fate of many Georgian and Russian progressive intellectuals of the time, and d'Herelle, disillusioned, never returned to Georgia or the USSR. However, their Institute survived, and is still functioning at its original site on the Mtkvary (which it now shares with the more modern Institutes of Molecular Biology and Biophysics and of Animal Physiology).  
In 1938, the Bacteriophage Institute was merged with the Institute of Microbiology & Epidemiology, under direction of the People's Commissary of Health of Georgia. In 1951, it was formally transferred to the All-Union Ministry of Health set of Institutes of Vaccines and Sera, taking on the leadership role in providing bacteriophages for therapy and bacterial typing throughout the former Soviet Union. Under orders from the Ministry of Health, hundreds of thousands of samples of pathogenic bacteria were sent to the Institute from throughout the Soviet Union, to isolate more effective phage strains and better characterize their usefulness. In 1988, the Scientific Industrial Union "Bacteriophage" was formed, centered in Tbilisi with Russian production facilities in Ufa, Khabarowsk and Nijnyi Novgorod. The industrial part was always run on a self-supporting basis. The institute's government-supported scientific branch included the electron microscope facility, permanent strain collection, laboratories studying phages of the enterobacteria, staphylococci and pseudomonads and formulating new phage cocktails, and groups involved in immunology, vaccine production, work with Lactobacillus and other therapeutic approaches. It also carried out the very extensive studies needed for approval by the Ministry of Health in Moscow of each new phage strain, therapeutic cocktail and means of delivery.  
This careful study of the host range, lytic spectrum and cross-resistance properties of the phages being used were a major factor in the reported successes of the phage therapy work carried out through the Institute. All of the phages used for therapy are lytic, avoiding the problems engendered by lysogeny. The problems of bacterial resistance were largely solved by the use of well-chosen mixtures of phages with different receptor specificities against each type of bacterium as well as of phages against the various bacteria likely to be causing the problem in multiple infections. The situation was further improved whenever the clinicians typed the pathogenic bacteria and monitored their phage sensitivity; where necessary, new cocktails were then prepared to which the given bacteria were sensitive. Not infrequently, using phage in conjunction with other antibiotics was shown to give better results than either the phage or the antibiotic alone.  
The depth and extent of the work involved is very impressive. For example, 1n 1983-85 alone, the Institute's Laboratory of Morphology and Biology of Bacteriophages carried out studies of growth, biochemical features and phage sensitivity of 2038 strains of Staphylococcus, 1128 of Streptococcus, 328 of Proteus, 373 of Ps. aeruginosa and 622 of Clostridium, received from clinics and hospitals in towns across the former Soviet Union. New broader-acting phage strains were isolated using these and other Institute cultures and included in a reformulation of their extensively-used Piophage preparation; it now inhibited 71% of their Staphylococcus strains instead of 58%, 76% of Pseudomonas instead of 55%, 51% of E. coli instead of 11%, 30% of Proteus instead of 3%, 60% of Streptococcus instead of 38%, and 80% of Enterococcus instead of 3% (Zemphira Alavidze, personal communication.) In the years since, there have been continued improvements in the formulation based on further studies, and phages against Klebsiella and Acinetobacter have been isolated and developed into therapeutic preparations. One of the latest developments is their IntestiPhage preparation, which includes 23 different phages active against a range of enteric bacteria.  
A good deal of work has gone into developing and providing the documentation to get approval from the Ministry of Health for specialized new delivery systems, such as a spray for use in respiratory-tract infections, in treating the incision area before surgery, and in sanitation of hospital problem areas such as operating rooms. An enteric-coated pill was also developed, using phage strains that could survive the drying process, and accounted for the bulk of the shipments to other parts of the former Soviet Union.  
Much of the focus in the last 12 years has been on combating nosocomial infections, where multi-drug-resistant organisms have become a particularly lethal problem and where it is also easier to carry out proper long-term research. Clinical studies of the effectiveness of the phage treatment and appropriate protocols were carried out in collaboration with a number of hospitals, but little has been published in accessible form. Zemphira Alavidze and her colleagues who are currently doing most of the actual therapeutic development and clinical application have manuscripts in preparation which describe their work in institutions such as the Leningrad (St. Petersburg) Intensive Burn Therapy Center, the Academy of Military Medicine in Leningrad, the Kazan Trauma Center, the Kemerovo Maternity Hospital. Some of the most intensive studies were carried out in Tbilisi, at the Pediatric Hospital, the Burn Center, the Center for Sepsis and the Institute for Surgery. Special mixtures were developed for dealing with strains giving problems of nosocomial infections in various hospitals, and they were very effectively used in sanitizing operating rooms and equipment, water taps and other sources of spread of the infections (most of them involving predominantly Staphylococcus). (Table 1).  
The Industrial Branch on the grounds of the Bacteriophage Institute had large vats for growing the selected phage, using appropriate nonpathogenic bacteria and broth they prepared themselves from high-quality beef. The resulting phage lysates were sterile filtered using ceramic filters which could themselves be sterilized in very hot ovens. The various different phages for each particular formulation were then combined and automatically packaged and sealed into 10-ml ampoules or otherwise prepared and packaged for administration. Approximate titers were determined by checking the dilution that would produce lysis after coinnoculation with specific numbers of bacteria of standard test strains, and each batch was also tested for any surviving bacterial contaminants. In those rare cases where injection was planned, the phages were concentrated and resuspended in physiological saline solution; testing in guinea pigs was added to the rest of the analytical regime, to make sure there were no residual bacterial surface fragments (endotoxins) that might cause problems if injected. (As mentioned above, phages have generally been reported to appear in the bloodstream and other body fluids rather shortly after being ingested or poured into a wound and to still be effective against systemic infections, so injection is usually not necessary.)  
Injectable forms accounted for only about 5% of the phage production at its height at the Bacteriophage Institute. None are made there now due to such factors as the expense and complexities of keeping animals for the necessary toxicity controls in the difficult situation in Georgia since the dissolution of the Soviet Union. The extensive fighting in Abkhasia left 350,000 refugees in a country of 5 million people and cut off the major rail, road and power routes to Russia, leaving only one significant highway across the High Caucausus mountains. Power is still often available for only a few hours a day and heating is a serious problem in winter. The country has very little money available for science, but some research continues, despite virtually no funding for salaries or supplies. The conflict did provide an interesting opportunity for widespread phage use. Each soldier in the Georgian army carried a spray-on phage cocktail which they used to disinfect their wounds (Alavadze, Meipariani, Gvasalia, manuscript in preparation). The industrial plant was privatized a few years ago and put to other uses, so the phages currently used for therapy must be grown in large carboys, the appropriate mixtures made, and then transferred to vials and sealed by hand. However, the checks for sterility and efficacy on the designated bacteria are still just as careful. Unfortunately, until the electron microscope is repaired and electricity made more predictable, the phage preparations can no longer be checked to be sure that the phage present are of the appropriate mixtures of morphotypes, or physical shape and size. The Institute scientists still continue to do the best they can under the circumstance, and many in Tbilisi feel they clearly owe their lives to the group's efforts. Extensive therapeutic work still goes on in local surgical, burn, pediatric and infectious disease hospitals, and in local clinics for ambulatory patients, including one on the grounds of the Institute.  

return to top
 

RECENT WORK IN THE WEST RELATED TO PHAGE THERAPY

Levin and Bull (1996) and Barrow and Soothill (1997) have provided good reviews of much of the work applying phage therapy in animals which has been carried out in Britain and the United States since interest in the possibilities of phage therapy began to resurface there in the early 80's. The results in general are in very good agreement with the clinical work described above in terms of efficacy, safety and importance of appropriate attention to the biology of the host-phage interactions, reinforcing trust in the reported extensive eastern European results. 
In Britain, H. W. Smith and M. B. Huggins (1982, 1983) carried out a series of studies on use of phages in systemic E. coli infections in mice and then in diarrheal disease in young calves. For example, they found that injecting 106 colony-forming units of a particular pathogenic strain intramuscularly killed 10/10 of the mice, but none died if they simultaneously injected 104 plaque-forming units of a phage selected against the K1 capsule antigen of that bacterial strain.This phage treatment was more effective than using such antibiotics as tetracycline, streptomycin, ampicillin or trimethoprim/sulfafurazole. Furthermore, the resistant bacteria that emerged had lost their capsule and were far less virulent. In calves, they found very high levels of protection even though they did not succeed in isolating phages specific for the K88 or K99 adhesive fimbriae, which play key roles in attachment within the small intestine. Still, the phage were able to reduce the number of bacteria bound there by many orders of magnitude and to virtually stop the fluid loss. The results were particularly effective if the phage were present before or at the time of bacterial presentation, and if multiple phages with different attachment specificities were used. Furthermore, the phage could be transferred from animal to animal, supporting the possibility of prophylactic use in a herd. If phage were given only after the development of diarrhea, the severity of the infection was still substantially reduced and none of the animals died (Smith et al, 1987). Levin and Bull (1996) carried out a detailed analysis of the population dynamics and tissue phage distribution of the 1982 Smith and Huggins study which can be helpful in assessing the parameters involved in successful phage therapy and its apparent superiority to antibiotics. They have gone on to do very interesting animal studies of their own (Levin and Bull, manuscript in preparation) and conclude that phage therapy is at least well worth further study.  
Soothill (1994) carried out a series of very nice studies preparatory to using phages for infections of burn patients. Using guinea pigs, he showed that skin-graft rejection could be prevented by prior treatment with phage against Pseudomonas aeruginosa. He also saw excellent protection of mice against systemic infections with both Pseudomonas and Acinetobacter when appropriate phages were used (Soothill, 1992). In the latter case, as few as 100 phages protected against infection with 100 million bacteria -- 5 times the LD50!  
 →
Merrill and coworkers (1996) have carried out a series of experiments designed to better understand the interactions of phages with the human immune system, and have started a company called "Exponential Biotherapies, Inc." to explore the possibilities of phage therapy. Their published work has been with a lytic derivative of the lysogenic phage lambda. While this particular strain would be a poor choice for therapeutic use, as discussed above and below, they have gathered very interesting and important data about factors affecting interactions between phages and the immune system.  


BACTERIAL PATHOGENICITY

 →
Most bacteria are not pathogenic; in fact, they play crucial roles in the ecological balance in various parts of our bodies, including the digestive system and all body surfaces. They actually help protect us from pathogens; this is one reason why the use of broad-spectrum antibiotics leaves us so vulnerable, and why more narrowly-targeted bactericidal agents would be highly advantageous. Furthermore, most of the serious pathogens are close relatives of non-pathogenic strains -- so what are the differences that make particular strains so lethal? Studies clarifying the mechanisms of pathogenesis at the molecular level have progressed remarkably in recent years (cf. Falkow 1996). These have now been crowned by the determination of the complete DNA base sequence of (nonpathogenic) E. coli K12 and several other bacterial species and extensive cloning and sequencing of pathogenicity determinants. Generally, a number of genes are involved, and these are clustered in so-called "pathogenicity islands", or "Pais", which may be 50,000-200,000 base pairs long. They generally have some unique properties indicating that the bacterium itself probably acquired them as a sort of "infectious disease" at some time in the past, and then kept them because they helped the bacterium infect new ecological niches where there was less competition. Many of these Pais are carried on small extrachromosomal circles of DNA called plasmids, which also can be carriers of drug-resistance genes. Others reside in the chromosome; there, they often are found imbedded in defective lysogenic prophages which have lost some key genes in the process and cannot be induced to form phage particles. However, they sometimes can recombine with related infecting phages. Therefore, it makes sense to avoid using lysogenic phages or their lytic derivatives for phage therapy to avoid any chance of picking up and moving such pathogenicity islands.  

For bacteria in the human gut, pathogenicity involves 2 main factors: (1) the production of toxin molecules, such as shiga toxin (from Shigella and some pathogenic E. coli) or cholera toxin. These toxins modify proteins in the target host cells and thereby cause the problems. (2) the acquisition of new cell-surface adhesins which allow the bacterium to bind to specific receptor sites in the small intestine, rather than just moving on through to the colon. They also all contain the components of so-called type-III secretion machinery, related to those involved in assembly of flagella (for motility) and of filamentous phages and instrumental in many plant pathogens. For all of the pathogenic enteric bacteria, the infection process triggers changes in the neighboring intestinal cells. These include degeneration of the microvilli, formation of individual "pedestals" cupping each bacterium and, in the case of Salmonella and Shigella, induction of cell-signaling molecules that trigger engulfment of the bacterium and its subsequent growth inside the cell.  
 →
Recently, E. coli O157 has been the subject of much concern, with contamination of such products as hamburgers and unpasteurized fruit juices leading to serious epidemics (cf. Grimm et al., 1995). Deaths have occurred, particularly in young children and the elderly, usually from hemorrhagic colitis (bloody diarrhea) or hemolytic-uremic syndrome, where the kidneys are affected. Antibiotic therapy has shown no benefit (cf. Greenwald and Brandt, 1997). We find that the version of O157 from the Seattle fast-food-chain epidemic, at least, is susceptible to several of our T4-related phages (Mark Mueller, Kutter et al., unpublished). It is interesting to consider their potential use in feedlots and meat-packing plants and in prophylaxis and therapy during outbreaks. 
return to top
 
 
THE T-EVEN FAMILY OF PHAGES 
 →
A substantial fraction of the phages in therapeutic mixes are relatives of bacteriophage T4, which has played such a key role in the development of molecular biology (cf. Karam, 1994). As discussed above, the name "T-even family of phages" is a historical accident reflecting the fact that T2, T4 and T6 out of the original collection of Delbrück's "Phage Group" all turned out to be related. Large sets of T4-like phages have been isolated for study from all over the world -- for example, from Long Island sewage treatment plants, animals in the Denver zoo, and dysentery patients in Eastern Europe (the latter often using Shigella as host). Members of the family are found infecting most enteric bacteria and their relatives (Ackermann and Krisch, Archives of Virology, in press). Most of the T-even phages studied to date use 5-hydroxymethylcytosine instead of cytosine in their DNA, which protects them against most of the restriction enzymes bacteria make to protect themselves against invading DNA and gives them a much more effective host range. The entire DNA base sequence of phage T4 is known (Kutter, Stidham et al., 1994) and we know a great deal about its infection process in standard laboratory conditions and about the methods it uses to so effectively target bacteria. We can potentially use some of that knowledge in developing more targeted approaches to phage therapy, particularly as more is learned about the similarities and differences in its extended family (cf. Monod et al., 1997; Kutter et al., 1996.) We know that different members of the T-even family use different outer membrane proteins and oligosaccharides as their receptors, and understand the tail-fiber structures involved well enough to potentially predict which phages will work on given bacteria and engineer phages with new specificities (cf. Henning and Hashemolhosseini, 1994; Krisch, personal communication.)  
 →
There have still been far too few studies of T4 ecology and its behavior under conditions more closely approaching the natural environment and the circumstances it will encounter in phage therapy, where the environment is often anaerobic and/or the bacteria experience frequent periods of starvation. The limited available information in that regard was summarized by Kutter, Kellenberger et al (1994). A variety of studies are shedding light on the ability of these highly virulent phages to coexist in balance with their hosts in nature. For example, they can reproduce in the absence of oxygen as long as their bacterial host had been growing anaerobically for several generations. We have found that they can also survive for a period of time in a sort of state of hibernation inside of starved cells and then allow their host to readapt enough when nutrients are again supplied to go on and produce a few phage. This is particularly interesting and important since bacteria undergo many drastic changes to survive periods of starvation which increase their resistance to a variety of environmental insults (cf. Kolter, 1992).  
The T-even bacteriophages share a unique ability that contributes significantly to their widespread occurrence in nature and to their competitive advantage. They are able to control the timing of lysis in response to the relative availability of bacterial hosts in their environment. When E. coli cells are singly infected with T4, they lyse after 25-30 minutes at body temperature in rich media, releasing about 100-200 phage per cell. However, when additional T-even phages attack the cell more than 4 minutes after the initial infection, the cell does not lyse at the normal time. Instead, it continues to make phage for as long as 6 hours, with the exact time of eventual lysis affected by the multiplicity of superinfecting phage (cf. Doermann, 1948; Abedon, 1994). This delay is termed "lysis inhibition".  
Thus, for many reasons the T4-related family of phages make excellent candidates for therapeutic use in enteric and other gram-negative bacteria, and studies of their ecology and distribution are being carried out with these goals in mind both in Tbilisi and at The Evergreen State College. Developing this same sort of understanding of other phage families potentially useful in phage therapy is equally important, taking advantage of the many powerful tools now available. Work useful to this end is progressing in a number of labs around the world but is still in its infancy, particularly as one moves beyond the enteric bacteria. 
return to top
    
CONCLUSIONS 
It is clearly time to look more carefully at the potential of phage therapy, both through strongly supporting new research and examining carefully what is already available. As Barrow and Soothill conclude, "Phage therapy can be very effective in certain conditions and has some unique advantages over antibiotics. With the increasing incidence of antibiotic resistant bacteria and a deficit in the development of new classes of antibiotics to counteract them, there is a need to investigate the use of phage in a range of infections." The stipulations of Ackermann (1987) are important here: "Blind treatment is clearly of no value; phages have to be tested just as antibiotics, and the indications have to be right, but this holds everywhere in medicine. However, phage therapy requires the creation of phage banks and a close collaboration between the clinician and the laboratory. Phages have at least one advantage....While the concentration of antibiotics decreases from the moment of application, phage numbers should increase. Another advantage is that phages are able to spread and thus prevent disease. Nonetheless, much research remains to be done ... on the stability of therapeutic preparations; clearance of phages from blood and tissues; their multiplication in the human body; inactivation by antibodies, serum or pus; and the release of bacterial endotoxins by lysis... In addition, therapeutic phages should be characterized at least by electron microscopy." While it seems premature to generally introduce injectible phage preparations in the West without further extensive research, their carefully-implemented use for a variety of agricultural purposes and in external applications could potentially help reduce the emergence of antibiotic-resistant strains. Furthermore, compassionate use of appropriate phages seems warranted in cases where bacteria resistant against all available antibiotics are causing life-threatening illness. They are especially useful in dealing with recalcitrant nosocomial infections, where large numbers of particularly vulnerable people are being exposed to the same strains of bacteria in a closed hospital setting. In this case, the environment as well as the patients can be effectively treated.  
In 1925, Sinclair Lewis's classic novel Arrowsmith, for which he won the Nobel prize in literature, played a significant role in raising popular interest in the possibilities of phage therapy and the potential scientific and ethical dilemmas involved (Summers, 1991). Today, the growing scientific, public and commercial interest in phage therapy is being reflected and fanned in a number of ways. For example, the BBC recently produced a Horizon documentary on phage therapy, The Virus that Cures, building on the ideas in Radetsky's Discover article on Return of the Good Virus. Several companies are beginning to explore work with phage therapy. In addition, a nonprofit "PhageBiotics" foundation has been formed to help support communication, education and research in the field. Hopefully all of this attention will lead to increased support of badly-needed research in the field and to rapid progress in developing appropriate applications, providing at least one alternative to the growing problem of multi-drug-resistant bacteria.  
Acknowledgments: Special thanks to Drs. Rezo Adamia, Zemphira Alavidze, Teimuraz and Nino Chanishvili, Taras Gabisonia, Liana Gachechiladze, Mzia Kutateladze, Amiran Meipariani and their colleagues at the Bacteriophage Institute, Tbilisi, for their hospitality and efforts to help me understand the extensive therapeutic work carried out there. Others who have been particularly helpful with information and communication include Dr. Marina Shubladze, pediatrician in Tbilisi for 10 years, now residing in Seattle; Nino Mzavia, Nino Trapaidze, Timur and Natasha Zurabishvili, who have worked in my laboratory on basic T4 biology; Hans-Wolfgang Ackermann (Laval University), Eduard Kellenberger (Basel), William Summers (Yale), Steve Abedon (Ohio State) and Bruce Levin (Emory); Mansour Samadpour, University of Washington; Kathy d'Acci, clinical lab director, St. Peter's Hospital, Olympia); physicians Jess Spielholz, MD, and Robin Moore, ND; and, especially, the many colleagues and students involved in our laboratory at Evergreen, particularly Barbara Anderson, Pia Lippincott, Mark Mueller, Stacy Smith, Elizabeth and Chelsea Thomas, Burt Guttman and Jim Neitzel.  

    REFERENCES 
 
Abedon, S. T. (1994). Lysis and the Interaction between Free Phages and Infected Cells, p. 397-405. In J. D. Karam (ed.), Molecular Biology of Bacteriophage T4. American Society for Microbiology, Washington, DC. 
Ackermann, Hans-Wolfgang and Michael DuBow (1987) Viruses of Prokaryotes I: General Properties of Bacteriophages, ch. 7. Practical Applications of Bacteriophages. CRC Press, Boca Raton, Florida 
Ackermann, H.-W. (1996). Frequency of morphological phage descriptions in 1995. Arch. Virol. 141:209-218. 
Ackermann, H.-W. and H. Krisch (1997) A catalogue of T4-type bacteriophages. Archives in Virology. 142, in press. 
Adamia, R. et al: (1990) The virulent bacteriophage IRA of Salmonella typhimurium: cloning of phage genes that are potentially lethal for the host cell. J. Basic Microbiol. 30: 707-716 
Begley, Sharon. (1994). The End of Antibiotics. Newsweek, March 28, pg. 47-51. 
Burnet, F. and M. McKie (1929). Observations on a permanently lysogenic strain of B. enteridis gaerther. Austral. J. Exptl. Biol. Med. Sci. 6: 277-284.] 
Bruynoghe, R. and J. Maisin (1921). Essais de therapeutique au moyen du bacteriophage du Staphylocoque. C. R. Soc. Biol. 85:1120-1121; 
Cairns, John, George Stent and James Watson (1966). Phage and the Origins of Molecular Biology. Cold Spring Harbor Laboratory Press, CSH, Long Island N. Y. 
D'Herelle, F. (1917). Sur un microbe invisible antagoniste des bac. dysentÈriques. Cr. r. Acad. Sci. Paris 165:373. 
D'Herelle, F. (translated to English by Dr. George H. Smith, PhD) (1922). The Bacteriophage: Its Role in Immunity. Williams and Wickens Co./Waverly Press, Baltimore, USA. 
D'Herelle, F., F. W. Twort, J. Bordet and Andre Gratia. Discussion on the Bacteriophage (Bacteriolysin). from the Ninetieth Annual Meeting of the British Medical Association, Glasgow, July, 1922. Published in the British Medical Journal 2:289-297, and reproduced in G. Stent, Papers on Bacterial Viruses, second edition, Little, Brown and Co., Boston, 1965 
Doermann, A. D. (1948). Lysis and lysis inhibition with Escherichia coli bacteriophage. J. Bacteriol. 55:257-275. 
Doermann, A. D. (1952). The Intracellular Growth of Bacteriophages. I. Liberation of intracellular bacteriophage T4 by premature lysis with another phage or with cyanide. J. Gen. Physiol. 35:645-656. 
Eaton, Monroe D. and Stanhope Bayne-Jones (1934). Bacteriophage Therapy. JAMA 103:1769-1776; 1847-1853; 1934-1939. 
Ellis, E. L. and M. Delbrück (1939). The Growth of Bacteriophage. J. Gen. Physiol. 22:365-384. 
Falkow, Stanley (1996). The Evolution of Pathogenicity in Eschericia, Shigella and Salmonella. p. 2723-2729 in E. coli American Society for Microbiology, Washington, DC. 
Fékéty, Robert (1995). Antibiotic-associated Diarrhea and Colitis. Current Opinion in Infectious Diseases 8:391-397. 
Fischer, Ernst and Carol Lipson. (1988). Thinking about Science: Max Delbrück and the Origins of Molecular Biology. W. W. Norton and Co. 
Gabisonia T.G. et al. (1995). Phagotherapy of nosocomial strains of P. aeruginosa, belonging to different o-groups. Georg. Med. News. N.15, pp.19-21. 
Hankin, E. H. (1896). L'action bactericide des Eaux de la Jumna et du Gange sur le vibrion du cholera. Ann. de l'Inst. Pasteur 10:511 
Jacob, F. and J. Monod (1961). Genetic regulatory mechanisms in the synthesis of proteins. J. Mol. Biol. 3: 318-56. 
Kolter, R. (1992). Life and death in stationary phase. ASM News 58:75-79. 
Krueger, A. P. and J. H. Northrop (1931). The Kinetics of the Bacterium-Bacteriophage Reaction. J. Gen. Physiol. 14:223-** 
Kutter, E., E. Kellenberger, K. Carlson, S. Eddy, J. Neitzel, L. Messinger, J. North, and B. Guttman. (1994). Effects of Bacterial Growth Conditions and Physiology on T4 Infection, p. 406-420. In J. D. Karam (ed.), Molecular Biology of Bacteriophage T4. American Society for Microbiology, Washington, DC. 
Kutter, E., T. Stidham, B. Guttman, E. Kutter, D. Batts, S. Peterson, T. Djavakhishvili, F. Arisaka, V. Mesyanzhinov, W. Ruger, and G. Mosig. (1994). Genomic Map of Bacteriophage T4, p. 491-519. In J. D. Karam (ed.), Molecular Biology of Bacteriophage T4. American Society for Microbiology, Washington, DC. 
Levin, Bruce and J. J. Bull (1996). Phage Therapy Revisited: The Population Biology of a Bacterial Infection and its Treatment with Bacteriophage and Antibiotics. The American Naturalist 147:881-898. 
Pfankuch, E., Kausche, G.A. Isolierung und Ðbermikroskopische Abbildung eines Bakteriophagen. Naturwissenschaften 28, 46. 1940. 
Ptashne, Mark (1967). Isolation of the Lambda Phage Repressor. Proc. Nat. Acad. Sci. 57:306-** 
Ruska, H. Ðber die Sichtbarmachung der bakteriophagen Lyse im Ðbermikroskop. Naturwissenschaften 28: 45-46, 1940. 
Ruska, H. Ergebnisse der Bakteriophagenforschung und ihre Deutung nach morphologischen Befunden. Ergeb. Hyg. Bakteriol. Immunforsch. Exp. Ther. 25: 437-498, 1943. 
Saunders, Mary Ellen (1994). Bacteriophages in Industrial Fermentations. p. 116-121 in Encyclopedia of Virology, R. Webster and A. Granoff, ed. Academic Press. 
Schlesinger, M. (1933). Reindarstellung eines Bakteriophagen in mit freiem Auge sichtbaren Mengen. Biochem. Zschr. 264: 6 
Shrayer, David (1996). Felix d'Herelle in Russia. Bull. Inst. Pasteur 94:91-96. 
Slopek, Stefan, Irina Durlakova, Beata Weber-Dabrowska, Alina Kucharewica-Krukowska, Marek Dabrowski and Regina Bisikiewicz (1981). Results of Bacteriophage Treatment of Suppurative Bacterial Infections I. General Evaluation of the Results. Arch. Immunol. Ther. Exp. 31:267-291. 
Slopek, Stefan, Irina Durlakova, Beata Weber-Dabrowska, Alina Kucharewica-Krukowska, Marek Dabrowski and Regina Bisikiewicz (1981). Results of Bacteriophage Treatment of Suppurative Bacterial Infections II. Detailed Evaluation of the Results. Arch. Immunol. Ther. Exp. 31:293-327. 
Slopek, Stefan, Irina Durlakova, Beata Weber-Dabrowska, Marek Dabrowski and Alina Kucharewica-Krukowska (1984). Results of Bacteriophage Treatment of Suppurative Bacterial Infections III. Detailed Evaluation of the Results Obtained in Further 150 Cases. Arch. Immunol. Ther. Exp. 32:317-335. 
Summers, William C. (1991). On the Origins of the Science in Arrowsmith: Paul de Kruif, Felix d'Herelle and Phage. Journal of the History of Medicine and Allied Sciences 46:315-332. 
Summers, William C. (1993a). Cholera and Plague in India: The Bacteriophage Inquiry of 1927-1936. Journal of the History of Medicine and Allied Sciences 48:275-301. 
Summers, William C. (1993b). How Bacteriophage came to be Used by the Phage Group. J. Hist. Biol. 26:255-267. 
Summers, William C. (1998). D'Herelle **. Yale University Press, in press. 
Twort, F. W. (1915) An investigation on the nature of ultramicroscopic viruses. Lancet 1915 II:1241. 
Vieu, J.-F. (1975). Les Bacteriophages. In Traite de Therapeutique, Vol. Serums et Vaccins. Fabre, J., ed. Flammarion, Paris, p. 337-40 
Vieu, J.-F., F. Guillermet, R. Minck and P. Nicolle (1979). Donneees actueles sur les applications therapeutiques des bacteriophages. Bull. Acad.Natl.Med 163:61. 
The Virus That Cures, a BBC Horizon Documentary produced by Judith Bunting. http://www.bbc.co.uk/horizon/beta/www/html/trcure.html
Weber-Dabrowska, Beata, Marek Dabrowski and Stefan Slopek (1987). Studies on Bacteriophage Penetration in Patients Subjected to Phage Therapy. 
return to top

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


Phage production goes large scale

Feed stuffs.  April 30, 2007 v79 i18 p12(1).
Full Text: COPYRIGHT 2007 Miller Publishing Company, Inc.



BIOPHAGE Pharma Inc. reported that it has signed a groundbreaking contract with Alberta Agriculture & Food to conduct a pilot study for the large-scale production and purification of bacteriophage preparations in a bio-fermentor. 

Securing this contract marks the beginning of a new business opportunity for Biophage in large-scale production of phage preparations. This is highly opportune as phages have been ruled to be safe for use as food additives. 

Biophage has seen a renewed interest in phage therapy since the first groundbreaking announcement by the Food & Drug Administration (August 2006) regarding the safe use of a phage preparation made of six phages against Listeria monocytogenes on ready-to-eat meat and poultry products. 

Dr. Rosemonde Mandeville, Biophage president and chief executive officer, said, "We are very pleased to announce Biophage's first contract for the large-scale production of phages in a bio-fermentor. This new business opportunity is expected to generate a continuous stream of revenues in the coming years. 

"Since FDA's approval of applying phages as food additives, we received numerous inquiries from producers who understand the commercial potential of this innovative, safe and environment-friendly solution for the control of bacterial contamination," she continued. 

Mandeville added, "Biophage is continuously seeking strategic partnerships for the co-development, out-licensing and commercialization of its phage-based proprietary approach for the detection, prevention and control of deadly microorganisms."


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

Phages channel their resources.
(Antibiotic resistance)

 

Cath O'Driscoll. Chemistry and IndustryJan 29, 2007 i2 p10(1).
Full Text: COPYRIGHT 2007 Society of Chemical Industry



Harnessing the channel-forming power of certain bacteriophages, viruses that target bacteria, could be useful for fighting resistant disease. Scientists have discovered that some types of bacteriophages can boost antibiotic effectiveness, lowering the doses required against some pseudomonas bacteria--involved in diseases from pneumonia to blood infections--by up to 50 times. 

Microbiologist Steven Hagens, previously at the University of Vienna," used unusual phages that parasitise the host by creating channels in the outer membrane rather than using enzymes to kill them. It is this channel-forming ability that allows them to bolster antibiotic effectiveness. 

'Pseudomonas bacteria are particularly multi-resistant to antibiotics because they have efflux pump mechanisms that enable them to throw out antibiotics. A pore in the cell wall would obviously cancel the efflux effect,' according to Hagens. 'The ["channelling"] method is useful in principle for all Gram-negative organisms, which is roughly half of all pathogens, including salmonella, E. coli and campylobacter,' he said. 

Dose-lowering effects were also observed with the antibiotic tetracycline. And doses of gramicidin were lower with E. coli bacteria in the presence of phages, although in this case the effect was not measurable because this particular, nonresistant, E. coli strain was already highly sensitive to the antibiotic. 

Hagens and coworkers recently reported that 75% of mice infected with a lethal dose of pseudomonas survived if the antibiotic gentamicin was administered in the presence of bacteriophages. None survived without the bacteriophages (Microb. Drug Resist. 2006, 12 (3), 164). 

'Given the current lack of antibacterials with good anti-Gramnegative activity, the prospect of using such treatments to prolong the life of existing agents and delay the onset of widespread resistance is very much to be welcomed,' commented Jim Spencer, a lecturer in microbial pathogenesis at the University of Bristol. 'Of particular interest is the authors' proposal that the sensitisation occurs through expression of phage proteins that form membrane-bound pores required for the extrusion of viral progeny, as much of the resistance of Gram-negative pathogens such as P. aeruginosa arises from their impermeability.' 

The new approach could be particularly useful for treating food poisoning, because the lower doses of antibiotic needed would be unlikely to disrupt the friendly bacteria in the gut--a problem with conventional antibiotic treatments. 

Phage therapy has been used since the 1920s. But the big drawback is that they must be matched perfectly to the exact strain of bacteria. '[Our method] wouldn't necessarily be any better, but if you don't find any useful phages, you could use these [channel-forming types] instead,' said Hagens. It may also prove possible to replicate the channel-forming ability of the phages by other means. 

With the increases in bacterial resistance, phages can only get more popular. Although bacteria can become resistant to both antibiotics and phages, phages are themselves capable of adapting and so new varieties should be available to outwit the bacteria. 

 
* Hagens currently works for EBI Food Safety in the Netherlands.



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

FDA approves viral defense against Listeria in RTE foods

Zachary Richardson. Food Chemical NewsAugust 28, 2006 v48 i29 p15.
Full Text: COPYRIGHT 2006 Agra Informa, Inc.



FDA has approved a food additive petition that will allow for the use of a mixture containing bacteriophages (bacteria-eating viruses) in combating Listeria monocytogenes, marking the first time such an approval has been issued in the United States--but, according to the additive's manufacturer, not the last.

Bacteriophages are viruses that target and infect bacteria with their own genetic material. They "hijack" the bacteria's DNA, forcing it to produce more bacteriophages, which then lyse (destroy) the bacteria by bursting it from within.

The approved mixture, manufactured by Intralytix, Inc., contains six varieties of bacteriophages, each specific against different strains of Listeria, including strains responsible for foodborne illness.

"They all target Listeria monocytogenes, but we collected almost 300 different samples of Listeria and then we took those samples and tested our phage library against them," Intralytix president and CEO John Vazzana told Food Chemical News. "Based on that testing, we selected the six phages that we believed would give us the best protection," he said. 

According to FDA, "The phage preparation will be used as an antimicrobial agent to control L. monocytogenes in the production of ready-to-eat (RTE) meat and poultry products. The phage preparation is directly sprayed on the surface of the RTE food articles at a level of approximately 1 milliliter of the preparation per 500 square centimeters of food surface area just prior to packaging."

"The target market has always been RTE foods," Vazzana continued. "During the four years of the approval process--which is a very long time for an approval--we've seen the problems with Listeria in RTE foods growing. If you back to the records that the CDC maintains, 99% of the Listeria monocytogenes cases have been in RTE foods, so that's always been what we've targeted," he said.

The preparation must comply with the Federal Meat Inspection Act and the Poultry Products Inspection Act, FDA noted, both of which are regulated by USDA, and must be listed on the label as an ingredient. FSIS has been convinced of the treatment's safety, FDA said.

Oddly enough, phage technology is both time-tested and novel, Vazzana said. "Phage technology has been around for 100 years; it was used in the U.S. for human health prior to the advent of antibiotics, and it's been used in Europe and the former Soviet Union since the early Twenties," he said.

FDA noted that while it was the first time the agency has allowed the use of a phage preparation in foods, phages are utilized on crops and in pesticide applications.
The technology could also represent another line of defense in light of increasing rates of antibiotic resistance, Vazzana suggested. "Phages are the most ubiquitous organism on the planet today--they're nature's way of controlling bacteria. One of the things that we think is an advantage ... is that they are very specific, those same phages will not lyse or kill any other strain of bacteria, unlike antibiotics, which are very broad-spectrum. Several bacteria have become resistant to antibiotics, and that's a problem that's growing every day," he said.

E. coli and Salmonella in the crosshairs

Intralytix is currently conducting research on similar treatments for other foodborne pathogens, notably E. coli 0157:H7 and Salmonella, Vazzana said. "Preparations for both have been developed, and we're going through some efficacy testing and screening at this point. We would hope to file a petition with FDA in the next four to six months for the E. coli product," with the Salmonella preparation to follow.

Those products would make the leap from RTE foods to those that require cooking, he added. "We're testing putting [the E. coli preparation] on beef before it would be ground up; the belief is and so far the tests indicate that we can eliminate the presence of E. coli by grinding the phages into the meat. We believe that we could significantly reduce the E. coli contamination and then maybe my wife will be able to make rare burgers again," Vazzana said.

Moreover, he added, the approval process for bacteriophage products may be a little less grueling in the future. "Four years is an extraordinarily long time to get a food additive approved, but this is a new technology, and frankly we were kind of neophytes in regard to what FDA expected from us," he explained. "We now feel like we know what FDA wants and FDA knows what we can provide."

Zachary Richardson
zachary.richardson@informa.com

 

            .....................................
  Bacteriophage therapy a revitalized therapy against bacterial infectious diseases.


Shigenobu Matsuzaki, Mohammad Rashel, Jumpei Uchiyama, Shingo Sakurai, Takako Ujihara, Masayuki Kuroda, Masahiko Ikeuchi, Toshikazu Tani, Mikiya Fujieda, Hiroshi Wakiguchi, Shosuke Imai. Journal of Infection and Chemotherapy.  Oct 2005 v11 i5 p211.

Author's Abstract:
 
Byline: Shigenobu Matsuzaki (1), Mohammad Rashel (1), Jumpei Uchiyama (1), Shingo Sakurai (1), Takako Ujihara (1), Masayuki Kuroda (1), Masahiko Ikeuchi (2), Toshikazu Tani (2), Mikiya Fujieda (3), Hiroshi Wakiguchi (3), Shosuke Imai (1)
Keywords: 

Phage therapy; Multidrug-resistant bacteria; Genetic modification; Lysin; Protein antibiotics
Abstract: 

Bacteriophage (phage) therapy involves using phages or their products as bioagents for the treatment or prophylaxis of bacterial infectious diseases. Much evidence in support of the effectiveness of phage therapy against bacterial infectious diseases has accumulated since 1980 from animal model studies conducted in Western countries. Reports indicate that appropriate administration of living phages can be used to treat lethal infectious diseases caused by gram-negative bacteria, such as Escherichia coli, Pseudomonas aeruginosa, Acinetobacter baumannii, Klebsiella pneumoniae, Vibrio vulnificus, and Salmonella spp., and gram-positive bacteria, such as Enterococcus faecium and Staphylococcus aureus. The phage display system and genetically modified nonreplicating phages are also effective for treatment of Helicobacter pylori and P. aeruginosa, respectively. In addition to phage particles per se, purified phage-encoded peptidoglycan hydrolase (lysin) is also reported to be effective for the treatment of bacterial infectious diseases caused by gram-positive bacteria such as Streptococcus pyogenes, S. pneumoniae, Bacillus anthracis, and group B streptococci. All phage lysins that have been studied to date exhibit immediate and strong bacteriolytic activity when applied exogenously. Furthermore, phage-coded inhibitors of peptidoglycan synthesis (protein antibiotics), search methods for novel antibacterial agents using phage genome informatics, and vaccines utilizing phages or their products are being developed. Phage therapy will compensate for unavoidable complications of chemotherapy such as the appearance of multidrug resistance or substituted microbism. 

Author Affiliation:
(1) Department of Molecular Microbiology and Infections, Kochi Medical School, Kohasu, Oko-cho, Nankoku, Kochi, 783-8505, Japan
(2) Department of Orthopaedics, Kochi Medical School, Kochi, Japan
(3) Department of Pediatrics, Kochi Medical School, Kochi, Japan
Article History:
Registration Date: 01/01/2005
Received Date: 08/07/2005

http://galenet.galegroup.com/servlet/HWRC/hits?r=d&origSearch=false&bucket=per&o=&rlt=2&n=10&l=d&items=0&c=24&tcit=0_1_0_0_0_0&docNum=A154218409&sgPhrase=false&locID=cityadelaide&secondary=false&t=RK&s=1&SU=Phage+Therapy

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

  Georgian biomedical preparations to be used by a US owned company; bacteriophage-based antibacterial products from world-renowned Eliava Institute to be used by US owned Phage Therapy Center in Mexico.

Asia Africa Intelligence Wire.  May 18, 2005 pNA.
Full Text: COPYRIGHT 2005 Financial Times Ltd.



(From Chemical Business NewsBase - Press Release) 

The George Eliava Institute of Bacteriophage, Microbiology and Virology (Eliava), Republic of Georgia, and Phage International Inc, Los Altos, CA, have signed an agreement whereby Eliava will become the supplier of bacteriophages for Phage International's new Phage Therapy Center (PTC) in Tijuana Mexico. This is the first commercial use of phages in a medical clinic in the western world and marks a starting point in the promising collaboration between a scientific research centre in the Republic of Georgia and a US owned company. The Institute will test bacterial cultures from PTC patients for PIO-phage susceptibility, produce the appropriate phage and deliver it to PTC to eradicate the bacteria in suffering patients. Bacteriophages represent a viable and effective alternative for fighting drug-resistant infections. Phage International Inc, a primary enabler of phage therapy in the western world, is in the process of opening a number of clinics.



                    ,,,,,,,,,,,,,,,,,,,,,,,,,

Activity of E. coli phages have implications for phage therapy.

Obesity, Fitness & Wellness WeekSept 11, 2004 p633.
Full Text: COPYRIGHT 2004 NewsRX



2004 SEP 11 - (NewsRx.com & NewsRx.net) -- The in vitro and in vivo bacteriolytic activities of Escherichia coli phages have implications for phage therapy

"Four T4-like coliphages with broad host ranges for diarrhea-associated Escherichia coli serotypes were isolated from stool specimens from pediatric diarrhea patients and from environmental water samples. All four phages showed a highly efficient gastrointestinal passage in adult mice when added to drinking water. Viable phages were recovered from the feces in a dose-dependent way. The minimal oral dose for consistent fecal recovery was as low as 10[superscript]3 PFU of phage per mL of drinking water," investigators in Switzerland report. 

"In conventional mice, the orally applied phage remained restricted to the gut lumen, and as expected for a noninvasive phage, no histopathological changes of the gut mucosa were detected in the phage-exposed animals," said Sandra Chibani-Chennoufi at Nestec Ltd. in Switzerland and collaborators in Switzerland, the U.S., and Bangladesh. "E. coli strains recently introduced into the intestines of conventional mice and traced as ampicillin-resistant colonies were efficiently lysed in vivo by phage added to the drinking water. Likewise, an in vitro phage-susceptible E. coli strain freshly inoculated into axenic mice was lysed in vivo by an orally applied phage, while an in vitro-resistant E. coli strain was not lysed." 

"In contrast, the normal E. coli gut flora of conventional mice was only minimally affected by oral phage application despite the fact that in vitro the majority of the murine intestinal E. coli colonies were susceptible to the given phage cocktail," reported the researchers. "Apparently, the resident E. coli gut flora is physically or physiologically protected against phage infection."
Chibani-Chennoufi and her coauthors published their study in Antimicrobial Agents and Chemotherapy (In vitro and in vivo bacteriolytic activities of Escherichia coli phages: Implications for phage therapy. Antimicrob Agents Chemother, 2004;48(7):2558-2569). 

For additional information, contact Harald Brussow, Nestle Research Center, Nestec Ltd., Vers-chez-les-Blanc, CH-1000 Lausanne 26, Switzerland. E-mail: harald.bruessow@rdls.nestle.com. 

The publisher of the journal Antimicrobial Agents and Chemotherapy can be contacted at: American Society for Microbiology, 1752 N Street NW, Washington, DC 20036-2904, USA. 

The information in this article comes under the major subject areas of Gastroenterology Vaccine, Vaccine Development, Immunology, Immunotherapy, and Gastroenterology. 

This article was prepared by Obesity, Fitness & Wellness Week editors from staff and other reports. Copyright 2004, Obesity, Fitness & Wellness Week via NewsRx.com & N

http://galenet.galegroup.com/servlet/HWRC/hits?r=d&origSearch=false&bucket=per&o=&rlt=2&n=10&l=d&items=0&c=28&tcit=0_1_0_0_0_0&docNum=A121589640&sgPhrase=false&locID=cityadelaide&secondary=false&t=RK&s=1&SU=Phage+TherapyewsRx.net

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

Phage therapy could remove E. coli O157:H7 from livestock.

TB & Outbreaks WeekJune 10, 2003 p15.
Full Text: COPYRIGHT 2003 NewsRX



2003 JUN 10 - (NewsRx.com & NewsRx.net) -- A bacteria-killing virus found in the feces of some sheep could help remove the dangerous food-borne bacteria Escherichia coli O157:H7 from livestock. 

Researchers from Evergreen State College in Olympia, Washington, discussed their findings at the 103rd General Meeting of the American Society for Microbiology on May 19, 2003. 

"Here we report a promising new natural way of reducing pathogen concentrations in livestock. This takes advantage of bacteriophages - bacteria-killing viruses, harmless to humans and other animals, which have been used extensively as antibiotics in Eastern Europe and the former Soviet Union for over 50 years," says Michael Dyen, one of the study researchers. 

Dyen and his colleagues reported on a new bacteriophage (CEV1) that they isolated from the feces of sheep naturally resistant to gut colonization by E. coli O157:H7. Preliminary trials of CEV1 in the lab have shown that it can be produced easily and can efficiently infect and kill the bacteria under proper conditions. In model systems reflecting the cow/sheep gut, CEV1 completely eliminated the bacteria in 11 days. 

"CEV1 and other carefully-selected phages against E. coli O157:H7 could be used to develop an effective management strategy to eradicate this pathogen from livestock," says Dyen. 

Outbreaks of E. coli O157:H7 have been linked to the consumption of hamburger meat, alfalfa sprouts, unpasteurized fruit juice, and even drinking water; more than 75% of the cases can be directly traced to contamination from carrier ruminants. The most recent data suggest that about 28% of the cattle presented for slaughter in the U.S. harbor O157:H7, and similar numbers have been reported in Canada and Europe. Infected livestock show no signs of illness and the levels are generally low, making contaminated animals hard to identify. Current prevention methodologies have centered on reducing meat contamination in the slaughterhouse and testing all products for human consumption as they leave. 

"At present, there are few therapeutic treatments for victims of this potentially deadly infectious agent except supportive therapy to manage the complications of cellular damage," says Dyen. "Our work focuses on removing O157:H7 from the food chain." 

This article was prepared by TB & Outbreaks Week editors from staff and other reports.



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