Brain tumor radiation resistance defeated

One of the biggest challenges to the treatment of cancer is overcoming the ability of cancer cells to become resistant to drugs or radiation. The problem of resistance is particularly relevant in brain cancers called gliomas. Research published in the Journal Stem Cells reports one method of overcoming this problem.

Scientists at Duke University and the Cleveland Clinic claim to have identified molecules associated with the ability of certain cells within a glioma, called tumor stem cells, to resist radiation. Specifically, they have identified a protein called "Notch", which is normally involved in embryonic development, that seems to be selectively found in resistant cells. Using genetic engineering methods they have made cells that were previously sensitive to radiation to become resistant by inducing expression of Notch.

Dr. Jialiang Wang of Duke University, who is the lead author of the study. said the finding marked the first report that Notch signaling in tumor tissue is related to the failure of radiation treatments.

"This makes the Notch pathway an attractive drug target," Wang said. "The right drug may be able to stop the real bad guys, the glioma stem cells."
The authors have also demonstrated that by inhibiting activity of the Notch protein, through administration of chemicals known as gamma-secretase inhibitors, can result in making resistant cells sensitive to radiation.

These findings are interesting in light of another very recent publication (Zhen et al. Arsenic trioxide-mediated Notch pathway inhibition depletes the cancer stem-like cell population in gliomas. Cancer Lett. 2009 Dec 3) in which the anti-leukemic drug Arsenic Trioxide was also demonstrated to alter glioma stem cells through manipulation of the Notch pathway. Notch has been found in numerous other types of tumor stem cells such as breast, colon, lung, and prostate cancer. The expression of a protein in cancer cells that is supposed to be only expressed during embryonic development supports the theory that during formation of cancer, mature cells tend to revert to an embryonic-like phenotype. This is also exemplified by the fact that adding cancer genes (oncogenes) in combination with some specific proteins can take an adult skin cell and transform it into a cell that resembles the embryonic stem cell, called an "iPS" cell. This is described in the following video:

Click play button

Sickle-cell disease can be cured with stem-cell transplant procedure

Sickle cell anemia is a genetic disorder that affects approximately 80,000 Americans, primarily of African-American origin. It is characterized by defective production of red blood cells which makes them extremely fragile and not capable of fully transporting oxygen.

In the December 10th issue of the New England Journal of Medicine a report describing a clinical trial performed at the Clinical Center of the National Institutes of Health in Bethesda, was published that described successful treatment of 9 out of 10 adults suffering from this condition.

"This trial represents a major milestone in developing a therapy aimed at curing sickle cell disease," said NIDDK Director Griffin P. Rodgers M.D. "Our modified transplant regimen changes the equation for treating adult patients with severe disease in a safer, more effective way."

The use of stem cell transplantation for genetic conditions is now becoming increasingly commonplace. For example, cord blood stem cells have been used to treat conditions such as Krabbe Disease, which is a lethal metabolic abnormality. The reason that the current study is so important was that it used a type of "conditioning regimen" that was substantially less toxic than previously performed.

In an older study about 200 children with severe sickle cell disease were cured with bone marrow transplants after receiving high doses of chemotherapy. That protocol was too toxic for adults, whose organs are damaged from the prolonged exposure to abnormal red blood cells and their breakdown products. The current treatment method is applicable to adults.

Stem cell transplantation for conditions like leukemias require the recipient bone marrow to be destroyed prior to administration of the new stem cells. In conditions such as sickle cell, one does not need to completely destroy the recipient bone marrow but merely to replace it with enough healthy stem cells so as to produce sufficient quantities of healthy red blood cells. Although this has theoretically been discussed in sickle cell anemia, to date, the current study is the first one to actually demonstrate this. It is anticipated that future studies of this sort will be conducted to attempt treatment of other metabolic abnormalities.

Stem cells may hold the key to the fight against HIV

Substantial progress has been made in the area of stem cells. Despite the Bush Administration’s 8 1/2 year ban on federal funding for embryonic stem cell research, and President Obama’s recent reversal, adult stem cell therapies have been making progress in terms of clinical implementation. This may be related to the safety concerns of embryonic stem cells, which have included differentiation into undesired tissues, as well as cancer. In contrast, adult stem cells have been used for more than 4 decades in the area of bone marrow transplantation and for over a decade in other areas. Primarily, non-bone marrow transplant studies have been focused in the area of heart failure, however smaller studies have investigated the use of stem cells in liver and kidney failure.

The field of stem cell therapies has recently been expanded. In a study published December 7in the medical journalPloS ONE, scientists from the University of California Los Angeles reported that human blood cells derived from adult stem cells can be engineered into cells that can target and kill HIV-infected cells – a process that could potentially be used against a range of chronic viral diseases.

The leader of the study, Dr. Scott G. Kitchen, Assistant Professor of Medicine in the Division of Hematology and Oncology at the David Geffen School of Medicine at UCLA and a Member of the UCLA AIDS Institute stated "We have demonstrated in this proof-of-principle study that this type of approach can be used to engineer the human immune system, particularly the T-cell response, to specifically target HIV-infected cells," Additionally, he commented on the possibility of future studies. "These studies lay the foundation for further therapeutic development that involves restoring damaged or defective immune responses toward a variety of viruses that cause chronic disease, or even different types of tumors."

Possible methods of manipulating blood cells to make them resistant to HIV infection includes genetically altering proteins called receptors. T cells have a specific receptor called CXCR5 which when mutated cannot be infected with HIV. Certain subsets of the human population who are resistant to HIV have this mutation in CXCR5, but also have normal T cell activities. One of the possible genetic alterations that can be performed in patients with HIV is to induce a similar CXCR5 mutation to endow resistance. Stem cell types that could be used include bone marrow, cord blood, or expanded peripheral blood stem cells.

Stem Cell Derived Neurons for Research Relevant to Alzheimer’s and Niemann-Pick Type C Diseases

One of the major hurdles to curing diseases is finding ways in which to test potential cures (or treatments) without having to use people, or even animals. One way of trying to cure a disease is to associate it with a gene or series of genes that are either mutated or abnormally acting. When the cause of the disease is known, then scientists use computers to generate molecules that theoretically would inhibit the disease. These are then tested in the test-tube, and subsequently in animals having the disease. If it works on animals and is relatively non-toxic, then Phase I human trials are conducted to assess safety and what dosage can be tolerated.

Subsequently Phase II trials are performed to assess whether there is an effect of the drug on the disease. Finally Phase III trials are conducted, which assess the efficacy of the drug but in a manner that is double blind and placebo controlled. If the drug is successful, then the FDA or EMEA (in Europe) grants approval. The other way to approach diseases is to randomly screen compounds. The issue with random screening is that one needs to have a replica of the disease in a test tube that can be rapidly assessed whether there is or is not an effect.

The contributive role of stem cells in human medicine has to some extent been underestimated. For example, while it is well-known that embryonic stem cells have not been used in humans to date, embryonic stem cells have contributed tremendously to human medicine. Mouse embryonic stem cells are the key to development of genetically engineered animals in which a gene of interest to humans is either made to be artificially highly expressed in the animal (called transgenic animals), or in which the animal is selectively depleted of the gene of interest (called knockout animals). The development of genetically engineered animals for human testing was the basis of identifying numerous "Achilles Heal’s" of diseases. For example, using knockout mice it was demonstrated that the molecule TNF-alpha is essential for animals to get rheumatoid arthritis. The development of antibodies to TNF-alpha has heralded a revolution in the therapeutic of not only rheumatoid arthritis but also several other inflammatory diseases such as Crohn’s Disease and Psoriasis.

Today the group of Lawrence B. Goldstein, Ph.D., of the University of California, San Diego, School of Medicine and Howard Hughes Medical Institute (HHMI) presented data at the American Society for Cell Biology (ASCB) 49th Annual Meeting, in San Diego describing a new "model" of disease that they developed. The scientists wanted to develop means of testing drugs against the neurological disorders Alzheimer’s disease (AD) and the rarer but always fatal disease, Niemann-Pick Type C (NPC).

In order to do this, the investigators needed to obtain the cells that develop the disease, specific types of neurons, from individuals with the disease. The problem with this approach is that it is in general very difficult to extract neurons, and it is even more difficult to grow them from patients with AD or NPC. To overcome this, stem cells were created from the skin of patients with these diseases and then the stem cells were made into disease-specific neurons by treatment with growth factors. Previous to this, researchers had to perform experiments in neurons from fruitflies which obviously have many differences as compared to humans.

According to Dr. Goldstein, who is a professor in the Department of Cellular & Molecular Medicine, an HHMI investigator and director of UC San Diego’s Stem Cell Program "Such research may yield an understanding of what components of sporadic disease are defined by genetic characteristics"

Studies are currently being performed using these "in vitro" models of disease to assess random chemical compounds, a process called "screening" in order to identify potential drugs that may be useful in these conditions.

Human Umbilical Stem Cells Cleared Mice’s Cloudy Eyes

The cornea is front part of the eye that is transparent and allows light to enter. The cornea is does not have a blood supply and receives its nutrients from diffusion and oxygen directly from the air. Corneal scarring is a major cause of vision loss and blindness. Today researchers at the University of Cincinnati reported a new method of reducing corneal scarring using stem cells in mice.

Mice that suffer from corneal scarring are used for assessment of possible new treatments for this condition. The researchers conducting the study, lead by Winston Whei-Yang Kao, PhD, professor of ophthalmology, at the University of Cincinnati, used a mouse that was genetically engineered to lack a protein called lumican, which is involved in maintaining a clear cornea.

At the 49th Annual Meeting of the American Society of Cell Biology in San Diego today, the researchers presented data indicating that treatment of the lumican deficient mice with stem cells derived from human umbilical cords leads to preservation of vision. The specific type of stem cells used were called mesenchymal stem cells. These cells have been previously demonstrated to be capable of becoming a variety of other tissues when exposed to specific chemicals.

Although corneal transplantation is a relatively established procedure that has saved the vision of many, researchers believe that the use of umbilical cord blood stem cells in this area still has significant potential. "Corneal transplantation is currently the only true cure for restoration of eyesight that may have been lost due to corneal scarring caused by infection, mechanical and chemical wounds and congenital defects of genetic mutations," Kao says. "However, the number of donated corneas suitable for transplantation is decreasing as the number of individuals receiving refractive surgeries, like LASIK, increases."

Dr. Kao also commented on the potential treatment applications possible with the umbilical cord stem cells. "Our results suggest a potential treatment regimen for congenital and/or acquired corneal diseases," he says, adding that the availability of human umbilical stem cells is almost unlimited. These stem cells are easy to isolate and can be recovered quickly from storage when treating patients. "These findings have the potential to create new and better treatments — and an improved quality of life — for patients with vision loss due to corneal injury."

Mesenchymal stem cells have already been demonstrated safe in clinical trials, however, to date efficacy studies are still underway for a variety of conditions. The data presented today suggests how many new possible uses of stem cells exist, and the almost limitless possibilities in the area of regenerative medicine.

Cleveland Clinic receives $2.75M grant to study stem cell use in treating MS

The use of stem cells for multiple sclerosis can be categorized into two main approaches. The first involves transplantation of blood making stem cells, called hematopoietic stem cells, after the immune response of the patient is destroyed. This is performed because multiple sclerosis is an immunological disease in which the T cells are attacking the "insulator" of the nerves, a protein called myelin basic protein. By destroying the immune system and subsequently adding stem cells that will make a new immune system, this approach "resets the clock" and has yielded success in early clinical studies. Unfortunately, the problem with destroying the patient immune system is that they undergo a period of immune compromise during which they are susceptible to bacterial, fungal, and viral infections. The second method of using stem cells in multiple sclerosis is to administer a type of stem cell called mesenchymal stem cells, which actually reprogram the pathogenic T cells so that they slow down their immune attack. Mesenchymal stem cells also possess two other important properties: a) they induce the generation of T regulatory cells, which block pathologic T cells from attacking myeling&; and b) they help to regenerate the injured neurons through producing growth factors, as well as becoming new neurons.

For the study of this second approach, the Cleveland Clinic has received a $2.75 million federal grant from the Department of Defense. This is a 4-year grant that will fund a 24-patient study which will be conducted by the Center for Stem Cell and Regenerative Medicine. The study will investigate patients with relapse-remitting MS that are still able to walk but have moderate to severe disability. Collaborators in the study will include the stem cell company Athersys Inc., Case Western Reserve University, the Clinic, Ohio State University and University Hospitals Case Medical Center.

"Mesenchymal stem cells are primitive cells in the bone marrow that have a wide range of effects that decrease the activity of immune cells which are over-active in MS," said Dr. Jeffrey Cohen of the Clinic’s Mellen Center for Multiple Sclerosis Treatment and Research. "In addition, in numerous laboratory studies, MSC’s were able to migrate from the blood in to areas of inflammation or injury in the nervous system and reduce damage by developing into cells resembling neurons (nerve cells) and glia (support cells) and, probably more importantly, by creating a tissue environment that encourages intrinsic repair mechanisms," he said.

The proposed study is similar to work performed by the Cellmedicine (www.cellmedicine.com ) stem cell treatment clinic which has published on 3 patients with MS undergoing a recovery after treatment with their own fat derived stem cells, without immune suppression. This was published with collaborators at the company Medistem Inc, the University of California San Diego, Indiana University, the company Vet-Stem and the University of Utah. The publication is freely available at this link www.translational-medicine.com/content/7/1/29.

The use of fat as a source of mesenchymal stem cells for treatment of MS is appealing for several reasons. Firstly, the high content of these stem cells in the fat makes expansion of the cells unnecessary for certain uses. The process of cell expansion is technically complex and can only be performed at specialized institutions with experience in cell processing. Secondly, fat contains high concentrations of T regulatory cells, therefore in addition to administering mesenchymal stem cells, the presence of these T cells is theoretically beneficial since they are known to inhibit pathological immune responses. An explanation of the importance/relevance of T regulatory cells in fat is provided in this video:

Click play button

Other cells found in fat include endothelial progenitor cells (EPC), these are useful for healing injured tissue by creating new blood vessels, a critical part of the healing process.

Researchers Launch Phase II Trial of Stem Cells and Acute Heart Attack

Doctors at the University of Texas Medical School at Houston have announced initiation of an efficacy-finding study in the area of heart failure using a "universal donor" stem cell product called "Prochymal". This cell therapy drug is under development by the company Osiris Therapeutics and is the subject of substantial scientific interest internationally. Prochymal has made it to Phase III trials in the area of Graft Versus Host Disease, a side effect of bone marrow transplantation, however, data was not sufficiently strong to warrant FDA approval. Prochymal is made from the bone marrow mesenchymal stem cells of healthy human volunteers. It is a unique stem cell product in that it does not require matching with the recipient.

Data from Phase I clinical trial of Prochymal have been published in the Journal of the American College of Cardiology. The researchers involved in the Phase I trial reported that patients who received Prochymal intravenously after a heart attacked did not have adverse effects associated with the stem cell infusion. Therapeutic benefits were observed in the treated but not control patients, including reduction in number of arrhythmias, improved heart and lung function, and improvement in overall condition.

"We are able to use a stem cell product that is on the shelf without prior preparation of anything from the patient, and this product appears to be able to help the heart muscle recover after a heart attack," said Ali E. Denktas, M.D., the trial’s Houston site principal investigator and assistant professor of cardiology at the UT Medical School at Houston. "This means patients have the potential to recover quicker with less risk of an immediate secondary attack."

The first patient for the Phase II study at the Houston site was recruited today. The heart attack victim Melvin Dyess, 49, received an intravenous infusion of either the stem cells or placebo as part of the protocol of the double-blind study. The procedure took place at the Memorial Hermann Heart & Vascular Institute-Texas Medical Center. Denktas said UT Medical School researchers will continue to enroll willing patients into the Phase II study who are admitted to Memorial Hermann-Texas Medical Center. Neither patients nor their physicians know whether they received the stem cell drug.

Stem cells from umbilical cord used for cerebral palsy

Cerebral palsy is a major health problem, affecting approximately 1 in 500 newborns. It is caused by damage to the brain by lack of oxygen before birth. The scientific rationale for the use of stem cells for this condition has been discussed previously in the video Stem Cell Therapy for Cerebral Palsy.

In a recent news announcement, a case of a child in Singapore with cerebral palsy that was treated with their own cord blood stem cells was discussed.
"It is quite a safe procedure. It is like a standard blood transfusion, except that you are using the cord blood cells that were stored. So there is no risk of a reaction, apart from perhaps minor hypersensitivity reactions, as in all blood transfusions," said Dr Keith Goh, neurosurgeon, Mount Elizabeth Hospital.

After the administration, the patient, 2-year-old Georgia Conn is reportedly calmer, with a decrease in constant crying an seizures. The parents, Michael and Louise Conn, previously stored Georgia’s umbilical cord blood cells. "Within two days, Georgia was noticeably happier. Just instantly more smiley, chatty and more energetic. That was the first real indication that something was going on," said Louise Conn. "And since then we all feel, and all her therapists feel, that her muscle tone has reduced, which is enabling her to achieve a lot more within her therapy sessions," she added.

Theoretically the risks of using a patient’s own cord blood stem cells are minimal since they are not manipulated, and are of the same genetic make up as the patient. However there are certain considerations, for example, "are there enough cells" to actually cause a meaningful effect? Additionally, what if the patient needs the cord blood cells later in life?

Other approaches to cord blood stem cell therapy include using cells from non-related cords, as well as expansion of the cord blood stem cells before using. The rationale for the non-related use of cord blood has been previously published (Riordan et al. Cord blood in regenerative medicine: do we need immune suppression? J Transl Med. 2007 Jan 30;5:8). Expansion of cord blood stem cells has previously been attempted by the companies Viacell and Aastrom. Although the technology is still a work in progress, some clinical trials have been performed with expanded cord blood cells in the area of hematological malignancies such as leukemias.

French scientists create skin fast from stem cells

After severe burn wounds patients are susceptible to infections, which unless properly managed can result in sepsis and dead. The current standard of care involves taking skin cells from the patient in an area that has not been injured, expanding the cells in tissue culture, and subsequently placing the cells on the area that has been burned. The drawback with this approach is that it takes about three weeks. In the interim many patients develop infections. Therefore new technologies are needed to create cells that can be ready to use in an expedient manner. A previous approach has been to use skin cells from the foreskin of infants after circumcision and expand these cells. However immune rejection occurs and these cells are not as protective as using the patient’s own cells.

A French team of scientists created human skin from stem cells and demonstrated that the skin was functional by placing it on the back of a mouse that was lacking an immune system (so to avoid rejection). The skin engrafted on the mouse and remained alive for the evaluation period of 3 months. Most strikingly, the skin was able to allow for healing of the mouse’s own epidermis. Since the skin cells were derived from stem cells, the scientists believe that they represent cells that are less visible to the immune system.

Dr. Marc Peschanski, research director at the French Research Institute I-Stem, stated" What our findings can provide is a way to cover the burns during those three weeks with skin epidermis … produced in that factory and sent to the physician at the moment they receive a severely burned patient," He continued "They call the factory and then, immediately, they will get a square meter of epidermis which will be a temporary way to cover the burns."

Stem cell therapies for burn wounds have been relatively underexplored. Previous studies have demonstrated that bone marrow stem cells can accelerate wound healing. While this approach is promising, it is difficult to perform bone marrow extraction in patients with severe burn wounds. Additionally, few hospitals have the facilities to process bone marrow cells in order to be administered on the skin.

The advantage of the work described in the publication (Guenou et al. Lancet. 2009 Nov 21;374(9703):1745-53) is that the cells can be used in a "universal donor" fashion. That is, they can be stockpiled and ready for application when the need arises.

Killing Breast Cancer Stem Cells

Cancer stem cells are a very new and scary concept. Traditionally researchers tested new drugs on cancer cells in test tubes. When a drug was found that killed the cells in the test tube, it was then experimented with on animals. If there was an effect on animals, then it would be tested in humans. The concept of the cancer stem cell threatens to overturn this traditional "way of doing things" that the cancer drug development industry has been based upon practically for the last century.

Specifically, the cancer stem cell concept is that the tumor is not one population of cells that all have the same properties. The cancer stem cell concept teaches that within a tumor there is a small population of "seed cells" or "stem cells" that perpetually make copies of themselves as well as differentiate or become the bulk of the tumor. In the same way that if you cut a tree but the trunk is left, the tree will re-sprout, according to the cancer stem cell concept, if you treat the bulk of the tumor but not address the cancer stem cell, the cancer will come back. But you may ask, if this is the case, then why is it that in animal studies some drugs actually produce complete cures? The answer may be in that animal tumors are very different than naturally occurring tumors. Specifically, when one gives cancer to an animal there are two approaches that are used. The first involves giving an animal cancer "cell line" to an animal that has an immune system. The second approach is giving a human "cell line" to an animal that does not have an immune system. Both of these conventional approaches involve "cell lines", which are cancer cells that have been growing in tissue culture for a long time. When tumors grow in tissue culture, certain cells of the tumor will grow faster than others. According to the old hypothesis that tumor cells are the same throughout the tumor, the use of cell lines is acceptable because it would mean that the cell line represents the tumor. According to the tumor stem cell hypothesis, the use of cell lines is unacceptable because when the tumor cells were growing in the test tube, certain cell populations may have overgrown the cell populations that "really" represented the tumor.

Evidence of the existence of tumor "subpopulations" came originally from studies in leukemia. Work from Dr. John Dick’s group at the University of Toronto, in Canada, demonstrated that of the leukemic cells in the blood of patients, only a small percentage (<0.0001&) were capable of causing leukemias in mice when freshly isolated from patients. The difference between the cells capable of causing leukemia in mice and those not having this ability, resided in whether the cells expressed markers of stem cells. Specifically, the cells that contained the stem cell associated protein CD34 were capable of causing leukemia formation, but the cells that lacked expression of this molecule could not (Lapidot, T., et al., A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature, 1994. 367(6464): p. 645-8)

Subsequent studies by other groups demonstrated similar cancer stem cells existed in other tumors, including in breast cancer (Al-Hajj et al. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A. 2003 Apr 1;100(7):3983-8) and colon cancer (O’Brien et al. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature. 2007 Jan 4;445(7123):106-10).

One of the common characteristics of the "tumor stem cell" is that they all express high levels of molecular pumps that spit out chemotherapeutic drugs. This is a mechanism by which cancer stem cells protect themselves. The question is asked, "how would the cancer stem cells specifically know to express such proteins that specifically spit out the drugs we use against them?" The answer is actually associated with the fact that the cancer stem cells in many ways resemble normal stem cells. Normal stem cells also express high quantities of these "drug pumps", the reason for this is because the normal stem cell has to protect itself from DNA damage. By pumping out things that could damage the DNA (such as chemotherapeutic drugs), the stem cell protects itself.

So if cancer stem cells express pumps that make chemotherapy ineffective, how can one develop therapies against them?

Researchers at the Broad Institute and Whitehead Institute have an idea. In a recently published paper (Gupta et al. Identification of selective inhibitors of cancer stem cells by high-throughput screening. Cell. 2009 Aug 21;138(4):645-59) a novel method of keeping breast cancer stem cells alive and growing in tissue culture was reported. The scientists proved that they were able to expand cancer stem cells based on cell characteristics and ability of the cells to induce tumor growth when administered to animals. Using this system, the scientists screened thousands of compounds randomly to see which ones may inhibit the cancer stem cell.

"Evidence is accumulating rapidly that cancer stem cells are responsible for the aggressive powers of many tumors," says Robert Weinberg, a Member of Whitehead Institute for Biomedical Research and one of the authors of the study. "The ability to generate such cells in the laboratory, together with the powerful techniques available at the Broad Institute, made it possible to identify this chemical. There surely will be dozens of others with similar properties found over the next several years."
"Many therapies kill the bulk of a tumor only to see it regrow," says Eric Lander, Director of the Broad Institute of MIT and Harvard, and an author of the Cell paper.

"This raises the prospect of new kinds of anti-cancer therapies."

The scientists identified one compound as particularly promising: salinomycin. This compound was 100-fold more potent than standard chemotherapeutic drugs such as paclitaxel at inhibiting tumor stem cell proliferation. Additionally, salinomycin was capable of inhibiting human tumors grown in immune deficient mice. In the studies performed salinomycin appeared to have minimal toxicity. Interestingly the mechanism of action appeared to be through induction of tumor stem cell differentiation. That is, instructing the tumor cell to become a type of cell that is still alive but has lower or absent potential for continued growth and metastasis.

Salinomycin is an ionophoric coccidiostat agent that is used as a supplement in chicken feed to control infection with coccidia and Clostridium perfringens (Bolder et al. The effect of flavophospholipol (flavocin) and salinomycin sodium (sacox) on the excretion of Clostridium perfringens, Salmonella enteritidis, and Campylobacter jejuni in broilers after experimental infection. Poult Sci. 1999;78:1681-1689). Salinomycin is commercially available for veterinary use.