Yearly Archives: 2015


CAR T-Cell Immunotherapy for ALL – National Cancer Institute

On This Page

For years, the cornerstones of cancer treatment have been surgery, chemotherapy, and radiation therapy. Over the last decade, targeted therapies like imatinib (Gleevec) and trastuzumab (Herceptin)drugs that target cancer cells by homing in on specific molecular changes seen primarily in those cellshave also emerged as standard treatments for a number of cancers.

Illustration of the components of second- and third-generation chimeric antigen receptor T cells. (Adapted by permission from the American Association for Cancer Research: Lee, DW et al. The Future Is Now: Chimeric Antigen Receptors as New Targeted Therapies for Childhood Cancer. Clin Cancer Res; 2012;18(10); 278090. doi:10.1158/1078-0432.CCR-11-1920)

And now, despite years of starts and stutter steps, excitement is growing for immunotherapytherapies that harness the power of a patients immune system to combat their disease, or what some in the research community are calling the fifth pillar of cancer treatment.

One approach to immunotherapy involves engineering patients own immune cells to recognize and attack their tumors. And although this approach, called adoptive cell transfer (ACT), has been restricted to small clinical trials so far, treatments using these engineered immune cells have generated some remarkable responses in patients with advanced cancer.

For example, in several early-stage trials testing ACT in patients with advanced acute lymphoblastic leukemia (ALL) who had few if any remaining treatment options, many patients cancers have disappeared entirely. Several of these patients have remained cancer free for extended periods.

Equally promising results have been reported in several small trials involving patients with lymphoma.

These are small clinical trials, their lead investigators cautioned, and much more research is needed.

But the results from the trials performed thus far are proof of principle that we can successfully alter patients T cells so that they attack their cancer cells, said one of the trial's leaders, Renier J. Brentjens, M.D., Ph.D., of Memorial Sloan Kettering Cancer Center (MSKCC) in New York.

Adoptive cell transfer is like giving patients a living drug, continued Dr. Brentjens.

Thats because ACTs building blocks are T cells, a type of immune cell collected from the patients own blood. After collection, the T cells are genetically engineered to produce special receptors on their surface called chimeric antigen receptors (CARs). CARs are proteins that allow the T cells to recognize a specific protein (antigen) on tumor cells. These engineered CAR T cells are then grown in the laboratory until they number in the billions.

The expanded population of CAR T cells is then infused into the patient. After the infusion, if all goes as planned, the T cells multiply in the patients body and, with guidance from their engineered receptor, recognize and kill cancer cells that harbor the antigen on their surfaces.

Although adoptive cell transfer has been restricted to small clinical trials so far, treatments using these engineered immune cells have generated some remarkable responses in patients with advanced cancer.

This process builds on a similar form of ACT pioneered by Steven Rosenberg, M.D., Ph.D., and his colleagues from NCIs Surgery Branch for patients with advanced melanoma.

The CAR T cells are much more potent than anything we can achieve with other immune-based treatments being studied, said Crystal Mackall, M.D., of NCIs Pediatric Oncology Branch (POB).

Even so, investigators working in this field caution that there is still much to learn about CAR T-cell therapy. But the early results from trials like these have generated considerable optimism.

CAR T-cell therapy eventually may become a standard therapy for some B-cell malignancies like ALL and chronic lymphocytic leukemia, Dr. Rosenberg wrote in a Nature Reviews Clinical Oncology article.

More than 80 percent of children who are diagnosed with ALL that arises in B cellsthe predominant type of pediatric ALLwill be cured by intensive chemotherapy.

For patients whose cancers return after intensive chemotherapy or a stem cell transplant, the remaining treatment options are close to none, said Stephan Grupp, M.D., Ph.D., of the Childrens Hospital of Philadelphia (CHOP) and the lead investigator of a trial testing CAR T cells primarily in children with ALL. This treatment may represent a much-needed new option for such patients, he said.

Trials of CAR T cells in adults and children with leukemia and lymphoma have used T cells engineered to target the CD19 antigen, which is present on the surface of nearly all B cells, both normal and cancerous.

In the CHOP trial, which is being conducted in collaboration with researchers from the University of Pennsylvania, all signs of cancer disappeared (a complete response) in 27 of the 30 patients treated in the study, according to findings published October 16 in the New England Journal of Medicine.

Nineteen of the 27 patients with complete responses have remained in remission, the study authors reported, with 15 of these patients receiving no further therapy and 4 patients withdrawing from the trial to receive other therapy.

According to the most recent data from a POB trial that included children with ALL, 14 of 20 patients had a complete response. And of the 12 patients who had no evidence of leukemic cells, called blasts, in their bone marrow after CAR T-cell treatment, 10 have gone on to receive a stem cell transplant and remain cancer free, reported the studys lead investigator, Daniel W. Lee, M.D., also of the POB.

Dr. Crystal Mackall

Our findings strongly suggest that CAR T-cell therapy is a useful bridge to bone marrow transplant for patients who are no longer responding to chemotherapy, Dr. Lee said.

Similar results have been seen in phase I trials of adult patients conducted at MSKCC and NCI.

In findings published in February 2014, 14 of the 16 participants in the MSKCC trial treated to that point had experienced complete responses, which in some cases occurred 2 weeks or sooner after treatment began. Of those patients who were eligible, 7 underwent a stem cell transplant and are still cancer free.

The NCI-led trial of CAR T cells included 15 adult patients, the majority of whom had advanced diffuse large B-cell lymphoma. Most patients in the trial had either complete or partial responses, reported James Kochenderfer, M.D., and his NCI colleagues.

Our data provide the first true glimpse of the potential of this approach in patients with aggressive lymphomas that, until this point, were virtually untreatable, Dr. Kochenderfer said. [NCI Surgery Branch researchers have also reported promising results from one of the first trials testing CAR T cells derived from donors, rather than the patients themselves, to treat leukemia and lymphoma.]

Other findings from the trials have been encouraging, as well. For example, the number of CAR T cells increased dramatically after infusion into patients, as much as 1,000-fold in some individuals. In addition, after infusion, CAR T cells were detected in the central nervous system, a so-called sanctuary site where solitary cancer cells that have evaded chemotherapy or radiation may hide. In two patients in the NCI pediatric trial, the CAR T-cell treatment eradicated cancer that had spread to the central nervous system.

If CAR T cells can persist at these sites, it could help fend off relapses, Dr. Mackall noted.

CAR T-cell therapy can cause several worrisome side effects, perhaps the most troublesome being cytokine-release syndrome.

The infused T cells release cytokines, which are chemical messengers that help the T cells carry out their duties. With cytokine-release syndrome, there is a rapid and massive release of cytokines into the bloodstream, which can lead to dangerously high fevers and precipitous drops in blood pressure.

Cytokine-release syndrome is a common problem in patients treated with CAR T cells. In the POB and CHOP trials, patients with the most extensive disease prior to receiving the CAR T cells were more likely to experience severe cases of cytokine-release syndrome.

For most patients, trial investigators have reported, the side effects are mild enough that they can be managed with standard supportive therapies, including steroids.

The research team at CHOP noticed that patients experiencing severe reactions all had particularly high levels of IL-6, a cytokine that is secreted by T cells and macrophages in response to inflammation. So they turned to two drugs that are approved to treat inflammatory conditions like juvenile arthritis: etanercept (Enbrel) and tocilizumab (Actemra), the latter of which blocks IL-6 activity.

The patients had excellent responses to the treatment, Dr. Grupp said. We believe that [these drugs] will be a major part of toxicity management for these patients.

The other two teams subsequently used tocilizumab in several patients. Dr. Brentjens agreed that both drugs could become a useful way to help manage cytokine-release syndrome because, unlike steroids, they dont appear to affect the infused CAR T cells activity or proliferation.

Even with these encouraging preliminary findings, more research is needed before CAR T-cell therapy becomes a routine option for patients with ALL.

We need to treat more patients and have longer follow-up to really say what the impact of this therapy is [and] to understand its true performance characteristics, Dr. Grupp said.

We need to treat more patients and have longer follow-up to really say what the impact of this therapy is [and] to understand its true performance characteristics.

Dr. Stephan Grupp

Several other trials testing CAR T cells in children and adults are ongoing and, with greater interest and involvement from the pharmaceutical and biotechnology sector, more trials testing CAR T cells are being planned.

Researchers are also studying ways to improve on the positive results obtained to date, including refining the process by which the CAR T cells are produced.

Research groups like Dr. Brentjens are also working to make a superior CAR T cell, including developing a better receptor and identifying better targets.

For example, Dr. Lee and his colleagues at NCI have developed CAR T cells that target the CD22 antigen, which is also present on most B cells, although in smaller quantities than CD19. The CD22-targeted T cells, he believes, could be used in concert with CD19-targeted T cells as a one-two punch in ALL and other B-cell cancers. NCI researchers hope to begin the first clinical trial testing the CD22-targeted CAR T cells in November 2014.

Based on the success thus far, several research groups across the country are turning their attention to developing engineered T cells for other cancers, including solid tumorslike pancreatic and brain cancers.

The stage has now been set for greater progress, Dr. Lee believes.

NCI investigators, for example, now have a platform to plug and play better CARs into that system, without a lot of additional R&D time, he continued. Everything else should now come more rapidly.

Link:
CAR T-Cell Immunotherapy for ALL - National Cancer Institute

Adult vs. Embryonic Stem Cells – Brown University

Advantages of Adult Stem Cells

Both lines of stem cells have an enormous therapeutic potential. While embryonic stem cells offer the potential for wider therapeutic applications, adult stem cells avoid the ethical issues roused by embryonic stem cell research. Therefore, many stem cell therapies are currently being tested using adult stem cells. Additionally, adult stem cells offer the potential for autologous stem cell donation, which may help to avoid issues of immune rejection in certain situations.

It is also known that upon injection into mice with compromised immune systems, undifferentiated embryonic stem cells elicit the formation of a benign tumor called a teratoma. This tumor formation causes scientists to doubt the therapeutic applicability of embryonic stem cells. It is not yet known whether similar results are observed with adult stem cells [17].

Advantages of Embryonic Stem Cells

The advantages of embryonic stem cells is that they offer one cell source for multiple indications. They provide the potential for a wider variety of applications than do adult stem cells. Additionally, they theoretically have the possibility of being immuno-privileged, due to their highly undifferentiated state. A privileged immune status would remove one of the main barriers of stem cell therapies, as self rejection is one stem cell therapys main complications [17]. The idea that embryonic stem cells can be immune privilaged, must be viewed skeptically, however, as this theory has not yet been proven.

Another advantage of embryonic stem cells, is that they appear to be immortal in vitro, while adult and differentiated stem cells cannot be cultured indefinitely in the lab. Once differentiated, these stem cells seem to die off like typical tissue cells.

Click Here to go Back to: Stem Cell Types

Click Here to go to: Stem Cell Classification

Click Here to go to: Clinical Therapies

Click Here to go to: Stem Cells and Research

Click Here to go to: History of Stem Cells

Click Here to go to: Stem Cell Funding

Click Here to go to: Challenges

Read more:
Adult vs. Embryonic Stem Cells - Brown University

How are Adult Stem Cells Turned into Treatments? | Boston …

Currently, blood stem cells are the only type of adult stem cell used regularly for treatment; they have been used since the late 1960s in the procedure now commonly known as bone marrow transplant. Transplants of neural stem cells have been tried in small numbers of patients with brain disorders such as Parkinson disease, and the FDA recently approved a clinical trial of neural stem cells for spinal cord injury.

Preliminary research in animals has found that bone marrow stromal cells, injected into a damaged heart, can have beneficial effects. And researchers at Childrens Hospital Boston have shown in a mouse model that the same cells, injected into the blood, help protect against chronic lung disease in premature newborns.

In some cases, it may be possible to infuse the stem cells into the blood, as in a bone marrow transplant. The cells find their own way to the proper location and begin forming the cells and tissues needed. In other cases, the cells may need to be injected directly into the organ or tissue that needs them.

The ultimate goal is for the cells to take up residence in their proper places in the body, divide repeatedly and form functioning tissuesor repair diseased tissue. Its not always clear how this happens. In some cases, the transplanted cells may become part of the tissue or organ; in others, they may secrete growth factors that stimulate cells already residing there.

For adult stem cells to be successful treatments, they must:

1) Reproduce in large enough quantities to provide the amounts needed for treatment. Some adult stem cells have a very limited ability to divide, making it difficult to multiply them in large numbers. Scientists around the world are trying to find ways of encouraging them to multiply. The Stem Cell Program at Boston Childrens Hospital, for example, recently discovered that a drug called PGE2 can multiply numbers of blood stem cells. PGE2 is now being tested in patients with leukemia and lymphoma to see if it will help them rebuild their blood systems.

2) Create the needed cell types, either in the laboratory or after theyve been transplanted into the body.

3) Be safe. A host of clinics around the world offer supposed stem-cell therapies with claims of complete success, but these treatments must still be considered experimental and potentially risky until much more work is done to ensure their safety. For example, when adult stem cells are provided from a donor, precautions must be taken to avoid rejection by the patients immune system. Unless the patient is his or her own donor, or unless a donor is found with an identical tissue type, the patient will need to take powerful drugs to suppress the immune system so the transplanted cells or tissues wont be rejected. In addition, if adult stem cells are manipulated incorrectly, there is a risk of cancer.

4) Stay alive and remain functional for the rest of the patients life, continuing to maintain a healthy tissue or organ.

The rest is here:
How are Adult Stem Cells Turned into Treatments? | Boston ...

Woman Tells Doctors How Stem Cell Treatment Restored Her …

Posted Saturday, October 24th 2015 @ 1am

24 year old Sarah Hughes of Houston is a medical miracle, who is alive and active today thanks to major innovations in recent years in adult stem cell technology.

News Radio 1200 WOAI reports Sarah told her story this week to doctors in San Antonio who are part of the effort to develop stem cell technology.

Sarah has spent virtually her entire life in a hospital bed, suffering from a genetic disease called Systemic Juvenile Idiopathic Arthritis.

Also called Still's Disease, SJIA symptoms include enlargement of the liver and spleen, swollen lymph nodes, increased white blood cell count, and extreme fatigue.

But the major problem inside Sarah's body was her immune system. Just the opposite of reduced immunity diseases like HIV, Sarah's immune system was in overdrive, attacking her food, her organs, and her body.

So serious was Sarah's condition that others could not share a toilet seat with her because of the amount of radiation in her urine.

At the age of 22, Sarah made the life or death decision to try a therapy made possible by Houston-based Celltex Therapeutics to be injected with millions of her own stem cells.

"Before, I was bed ridden, so this is a huge improvement," a lively Sarah told me. "And I'm eating. Before I wasn't able to eat, I was artificially fed through a tube or intravenously."

In the eleven months since the therapy, Sarah says the results have been amazing, including increased appetite, a decrease in the number of medications needed, and an end to the chemotherapy, which has allowed her to gain weight and grow her hair.

Sarah says her greatest accomplishment is to be able to ride horses, an activity she loves, every day.

It is an amazing new life for Sarah, who says after spending her formative years in a hospital, she is just now learning, at the age of 24, to do basic things, like how to behave in social settings.

"I've never been normal," she joked. "I'm having to learn everything for the first time, things like what I like to eat. I've never been able to eat, and I have never enjoyed eating."

Celltex is the largest stem cell bank in America, dedicated to allowing patients like Sarah to receive treatment through their own mesenchymal stem cells through a process patented by Celltex.

Treatment with stem cells is a new but rapidly growing medical field. Cells are either harvested from fat in the patient's body, or, if the patient has the possibility of inheriting a known genetic disease, cord blood from the umbilical cord, which is rich in stem cells, is harvested.

Research is underway to use stem cells in treatment of cancer, aging, and Alzheimer's Disease, as well as other degenerative conditions.

Sarah says if her case is normal, it is a medical field which has endless potential and promise.

The rest is here:
Woman Tells Doctors How Stem Cell Treatment Restored Her ...

Cell Therapy Ltd

Founded in 2009 by Nobel prize winner Professor Sir Martin Evans and Ajan Reginald, former Global Head of Emerging Technologies at Roche, CTL develops life-saving and life altering regenerative medicines. CTLs team of scientists, physicians, and experienced management have discovered and developed a pipeline of world-class regenerative medicines.

Sir Martin Evans' unique expertise in discovering rare stem cells led to CTLs innovative drug discovery engine that can uniquely isolate very rare and potent tissue specific stem cells. This exceptional class of cells is then engineered into unique disease-specific cellular regenerative medicines. Each medicine is disease specific and forms part of CTLs world-class portfolio of four off the shelf blockbuster medicines all scheduled for launch before 2020.

The products in late stage clinical trials include Heartcel which regenerates the damaged heart of adults with coronary artery malformations and children with Kawasaki Disease and Bland White Garland Syndrome. In 2014, Heartcel reported unprecedented heart regeneration clinical trial results and is scheduled to launch in 2018 to treat ~400,000 patients worldwide. Myocardion is in Phase II/III trials and treats mild-moderate heart failure affecting 10 million patients worldwide. Tendoncel is the worlds first topical regenerative medicine, for early intervention of severe tendon injuries, and has completed Phase II trials. It is designed to treat the >1 million severe tendon injuries each year in the US and Europe. Skincel is for skin regeneration, and is due to complete Phase II trials in 2015. It is designed to address ulceration and wrinkles.

CTL combines world-class science and management expertise to bring life-saving regenerative medicines to market.

European Society of Gene and Cell Therapy Congress, 17-20 September 2015, Helsinki,Finland (ESGCT 2015)

4th International Conference and Exhibition on Cell & Gene Therapy, August 10-12, 2015, London (CGT 2015)

The International Society for Stem Cell Research Annual Meeting, 24th-27th June 2015, Stockholm, Sweden (ISSCR 2015)

British Society for Gene and Cell Therapy Annual Conference, 9th-11th June 2015, Strathclyde, Glasgow (BSGCT 2015)

Go here to read the rest:
Cell Therapy Ltd

Embryonic Stem Cell Maintenance & Differentiation (Human)

Reduce variation with the most complete, defined system for human embryonic stem cell (ES cell) and induced pluripotent stem cell (iPS cell) culture featuring mTeSR1 and the TeSR media family. From generation of iPS cells to maintenance, differentiation, characterization and cryopreservation of ES and iPS cells, see how you can "Maximize Your Pluripotential".

A Complete System for Supporting Your Human Pluripotent Stem Cell Research Human pluripotent stem cell (hPSC) research is an expanding field that has potential to change the way human diseases are studied and treated. The ability to differentiate ES cells and iPS cells to specific downstream cell types opens up new avenues for drug development and regenerative medicine.

STEMCELL Technologies offers an array of products designed to support the various steps of your ES and iPS cell culture workflow, from isolation, reprogramming and expansion to directed differentiation and characterization. For help with your hPSC workflow decision making, use ourinfographicsto find the right reagents for you.

Small Molecules for Reprogramming iPS cells have been traditionally generated through exogenous expression of pluripotency genes (via viral or episomal vectors). However, small molecules are increasingly being utilized and have been demonstrated to increase reprogramming efficiency:

Small Molecules for Maintenance Maintenance of stem cells in defined culture systems can reduce experimental variability. Small molecules have been used to stimulate the self-renewal capabilities of ES and iPS cells or increase viability of single cells.

TeSR-E5 and TeSR-E6 are defined, serum- and xeno-free media that are based on the formulation of TeSR-E8, but do not contain transforming growth factor (TGF-), basic fibroblast growth factor (bFGF), or in the case of TeSR-E5, insulin. They may be used as basal media for differentiation of human ES or iPS cells or other applications where removal of the above cytokines and insulin is desirable. To learn more about the functions of the different cytokines in the TeSR media, click here.

Small Molecules for Differentiation Differentiation of pluripotent stem cells to specialized cell types requires selective activation or inhibition of specific signaling pathways. Small molecules have been used to identify pathways required for differentiation, and are often used in place of expensive growth factors to direct differentiation.

Here is the original post:
Embryonic Stem Cell Maintenance & Differentiation (Human)

Eli and Edythe Broad Center for Regenerative Medicine and …

Welcome

Welcome to the Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, located on the University of Southern Californias Health Sciences Campus.

Our investigators are exploring the normal mechanisms that build, maintain and repair our body systems, to develop knowledge-based approaches for regenerative medicine. Scientists are researching kidney, liver, neural, blood, cardiovascular, skeletal and skin disease models.

The center serves as a hub for USC Stem Cell, which connects researchers in stem cell biology and regenerative medicine across USC.

Oct 9, 2015

As a winner of the NIH Directors New Innovator Award, USC Stem Cell principal investigator Min Yu will strive to develop individualized medicine targeting rare and deadly breast cancer stem cells. The five-year, $2.475 million award is part of the High-Risk, High-Reward Research program supported by the NIH Common Fund.

Sep 22, 2015

How do you turn stem cells into nephrons, the functional unit of the kidney? Albert D. Kim, PhD, a postdoctoral fellow in the laboratory of Andy McMahon, PhD, is exploring this question with support from a Hearst Fellowship, an award recognizing an exceptional junior postdoctoral fellow pursuing stem cell research at USC.

Sep 21, 2015

Once the stuff of science fiction, genetic engineering is now offered on a fee-for-service basis at USC. On September 19, USC Stem Cell faculty and staff welcomed their supporters, the Chang and Choi families, and nearly 100 of their friends to celebrate the grand opening of the Chang Stem Cell Engineering Facility, located on the second floor of the Eli and Edythe Broad Center (BCC) for Regenerative Medicine and Stem Cell Research at USC on the Health Sciences Campus. Established with a generous gift from the Chang family, the stem cell engineering facility will serve researchers at USC as well as at other institutions.

See the rest here:
Eli and Edythe Broad Center for Regenerative Medicine and ...

My new ‘neighbor’ in Sacramento: a fat stem cell clinic …

For years Ive been writing about stem cell clinics that sell non-FDA approved stem cell treatments to vulnerable patients right here in America.

These clinics have been sprouting up like mushrooms across the US and their numbers may be above200 today overall. As a result perhaps it was inevitable that one would arrive in a locale near me.

Tomorrow, July 11, reportedly the Irvine Stem Cell Treatment Center will open a Sacramento, CA branch.The doctor there will apparently be Thomas A. Gionis (picture from press release). This private, for-profit clinic has no affiliation with UC Davis School of Medicine in Sacramento where Im located.

The stem cell clinic Sacramento branch will sell transplants of fat stem cells in the form of something called stromal vascular fraction or SVF, which I believe is almost certainly a drug. To my knowledge this clinic and the large chain that it belongs to called Cell Surgical Network (CSN), do not have FDA approval to use SVF.

Both publicly and to me on this blog, CSN continues to arguethat it doesnt need FDA approval (here,hereandhere), but recent FDA draft guidances sure suggest otherwise in my view. Of course if the FDA never takes action on the use of SVF then how are we all supposed to interpret that? WithoutFDA action or finalized guidelines, is it formally possible that the FDA could back down on SVF?

This clinic will reportedly sell SVF to treat a dizzying array of conditions having nothing to do with fat:

Emphysema, COPD, Asthma, Heart Failure, Heart Attack, Parkinsons Disease, Stroke, Traumatic Brain Injury, Lou Gehrigs Disease, Multiple Sclerosis, Lupus, Rheumatoid Arthritis, Crohns Disease, Muscular Dystrophy, Inflammatory Myopathies, and Degenerative Orthopedic Joint Conditions (Knee, Shoulder, Hip, Spine).

To me as a scientist the use of SVF to treat all these very different conditions does not make good common sense.

It would also seem arguably to be quite likely be considered non-homologous use by the FDA, a standing that would also automatically make this a drug requiring FDA pre-approval. Non-homologous use means using a biological product of a certain kind that is not homologous (not the same or similar in origin) to the tissue being treated. For example, fat is not the same as the brain or other central nervous system tissue that is involved in several of the conditions on the clinic menu. Same goes for cardiac muscle, airways, etc.

The use of a non-FDA approved product in a largely non-homologous manner increases risks for patients. Note that these stem cell transplants are also very expensive with little evidence in the way of published data of benefit.

The CSN stem cell clinic in Sacramento will be located at the New Body MD Surgical Center, just about 10 minutes from my office. I plan on paying them a visit at some point. Lets see how that goes. Will they let me in?

. Bookmark the

.

Follow this link:
My new 'neighbor' in Sacramento: a fat stem cell clinic ...

Stem-cell therapy – Wikipedia, the free encyclopedia

This article is about the medical therapy. For the cell type, see Stem cell.

Stem-cell therapy is the use of stem cells to treat or prevent a disease or condition.

Bone marrow transplant is the most widely used stem-cell therapy, but some therapies derived from umbilical cord blood are also in use. Research is underway to develop various sources for stem cells, and to apply stem-cell treatments for neurodegenerative diseases and conditions, diabetes, heart disease, and other conditions.

With the ability of scientists to isolate and culture embryonic stem cells, and with scientists' growing ability to create stem cells using somatic cell nuclear transfer and techniques to create induced pluripotent stem cells, controversy has crept in, both related to abortion politics and to human cloning. Additionally, efforts to market treatments based on transplant of stored umbilical cord blood have been controversial.

For over 30 years, bone marrow has been used to treat cancer patients with conditions such as leukaemia and lymphoma; this is the only form of stem-cell therapy that is widely practiced.[1][2][3] During chemotherapy, most growing cells are killed by the cytotoxic agents. These agents, however, cannot discriminate between the leukaemia or neoplastic cells, and the hematopoietic stem cells within the bone marrow. It is this side effect of conventional chemotherapy strategies that the stem-cell transplant attempts to reverse; a donor's healthy bone marrow reintroduces functional stem cells to replace the cells lost in the host's body during treatment. The transplanted cells also generate an immune response that helps to kill off the cancer cells; this process can go too far, however, leading to graft vs host disease, the most serious side effect of this treatment.[4]

Another stem-cell therapy called Prochymal, was conditionally approved in Canada in 2012 for the management of acute graft-vs-host disease in children who are unresponsive to steroids.[5] It is an allogenic stem therapy based on mesenchymal stem cells (MSCs) derived from the bone marrow of adult donors. MSCs are purified from the marrow, cultured and packaged, with up to 10,000 doses derived from a single donor. The doses are stored frozen until needed.[6]

The FDA has approved five hematopoietic stem-cell products derived from umbilical cord blood, for the treatment of blood and immunological diseases.[7]

In 2014, the European Medicines Agency recommended approval of Holoclar, a treatment involving stem cells, for use in the European Union. Holoclar is used for people with severe limbal stem cell deficiency due to burns in the eye.[8]

Research has been conducted to learn whether stem cells may be used to treat brain degeneration, such as in Parkinson's, Amyotrophic lateral sclerosis, and Alzheimer's disease.[9][10][11]

Healthy adult brains contain neural stem cells which divide to maintain general stem-cell numbers, or become progenitor cells. In healthy adult animals, progenitor cells migrate within the brain and function primarily to maintain neuron populations for olfaction (the sense of smell). Pharmacological activation of endogenous neural stem cells has been reported to induce neuroprotection and behavioral recovery in adult rat models of neurological disorder.[12][13][14]

Stroke and traumatic brain injury lead to cell death, characterized by a loss of neurons and oligodendrocytes within the brain. A small clinical trial was underway in Scotland in 2013, in which stem cells were injected into the brains of stroke patients.[15]

Clinical and animal studies have been conducted into the use of stem cells in cases of spinal cord injury.[16][17][18]

The pioneering work[19] by Bodo-Eckehard Strauer has now been discredited by the identification of hundreds of factual contradictions.[20] Among several clinical trials that have reported that adult stem-cell therapy is safe and effective, powerful effects have been reported from only a few laboratories, but this has covered old[21] and recent[22] infarcts as well as heart failure not arising from myocardial infarction.[23] While initial animal studies demonstrated remarkable therapeutic effects,[24][25] later clinical trials achieved only modest, though statistically significant, improvements.[26][27] Possible reasons for this discrepancy are patient age,[28] timing of treatment[29] and the recent occurrence of a myocardial infarction.[30] It appears that these obstacles may be overcome by additional treatments which increase the effectiveness of the treatment[31] or by optimizing the methodology although these too can be controversial. Current studies vary greatly in cell-procuring techniques, cell types, cell-administration timing and procedures, and studied parameters, making it very difficult to make comparisons. Comparative studies are therefore currently needed.

Stem-cell therapy for treatment of myocardial infarction usually makes use of autologous bone-marrow stem cells (a specific type or all), however other types of adult stem cells may be used, such as adipose-derived stem cells.[32] Adult stem cell therapy for treating heart disease was commercially available in at least five continents as of 2007.[citation needed]

Possible mechanisms of recovery include:[9]

It may be possible to have adult bone-marrow cells differentiate into heart muscle cells.[9]

The first successful integration of human embryonic stem cell derived cardiomyocytes in guinea pigs (mouse hearts beat too fast) was reported in August 2012. The contraction strength was measured four weeks after the guinea pigs underwent simulated heart attacks and cell treatment. The cells contracted synchronously with the existing cells, but it is unknown if the positive results were produced mainly from paracrine as opposed to direct electromechanical effects from the human cells. Future work will focus on how to get the cells to engraft more strongly around the scar tissue. Whether treatments from embryonic or adult bone marrow stem cells will prove more effective remains to be seen.[33]

In 2013 the pioneering reports of powerful beneficial effects of autologous bone marrow stem cells on ventricular function were found to contain "hundreds" of discrepancies.[34] Critics report that of 48 reports there seemed to be just five underlying trials, and that in many cases whether they were randomized or merely observational accepter-versus-rejecter, was contradictory between reports of the same trial. One pair of reports of identical baseline characteristics and final results, was presented in two publications as, respectively, a 578 patient randomized trial and as a 391 patient observational study. Other reports required (impossible) negative standard deviations in subsets of patients, or contained fractional patients, negative NYHA classes. Overall there were many more patients published as having receiving stem cells in trials, than the number of stem cells processed in the hospital's laboratory during that time. A university investigation, closed in 2012 without reporting, was reopened in July 2013.[35]

One of the most promising benefits of stem cell therapy is the potential for cardiac tissue regeneration to reverse the tissue loss underlying the development of heart failure after cardiac injury.[36]

Initially, the observed improvements were attributed to a transdifferentiation of BM-MSCs into cardiomyocyte-like cells.[24] Given the apparent inadequacy of unmodified stem cells for heart tissue regeneration, a more promising modern technique involves treating these cells to create cardiac progenitor cells before implantation to the injured area.[37]

The specificity of the human immune-cell repertoire is what allows the human body to defend itself from rapidly adapting antigens. However, the immune system is vulnerable to degradation upon the pathogenesis of disease, and because of the critical role that it plays in overall defense, its degradation is often fatal to the organism as a whole. Diseases of hematopoietic cells are diagnosed and classified via a subspecialty of pathology known as hematopathology. The specificity of the immune cells is what allows recognition of foreign antigens, causing further challenges in the treatment of immune disease. Identical matches between donor and recipient must be made for successful transplantation treatments, but matches are uncommon, even between first-degree relatives. Research using both hematopoietic adult stem cells and embryonic stem cells has provided insight into the possible mechanisms and methods of treatment for many of these ailments.[citation needed]

Fully mature human red blood cells may be generated ex vivo by hematopoietic stem cells (HSCs), which are precursors of red blood cells. In this process, HSCs are grown together with stromal cells, creating an environment that mimics the conditions of bone marrow, the natural site of red-blood-cell growth. Erythropoietin, a growth factor, is added, coaxing the stem cells to complete terminal differentiation into red blood cells.[38] Further research into this technique should have potential benefits to gene therapy, blood transfusion, and topical medicine.

Hair follicles also contain stem cells, and some researchers predict these follicle stem cells may lead to successes in treating baldness through activation of progenitor stem cells. This treatment is expected to work by activating already existing stem cells on the scalp. Later treatments may be able to simply signal follicle stem cells to give off chemical signals to nearby follicle cells which have shrunk during the aging process, which in turn respond to these signals by regenerating and once again making healthy hair.

In 2004, scientists at King's College London discovered a way to cultivate a complete tooth in mice[39] and were able to grow bioengineered teeth stand-alone in the laboratory. Researchers are confident that the tooth regeneration technology can be used to grow live teeth in human patients.

In theory, stem cells taken from the patient could be coaxed in the lab turning into a tooth bud which, when implanted in the gums, will give rise to a new tooth, and would be expected to be grown in a time over three weeks.[40] It will fuse with the jawbone and release chemicals that encourage nerves and blood vessels to connect with it. The process is similar to what happens when humans grow their original adult teeth. Many challenges remain, however, before stem cells could be a choice for the replacement of missing teeth in the future.[41][42]

Research is ongoing in different fields, alligators which are polyphyodonts grow up to 50 times a successional tooth (a small replacement tooth) under each mature functional tooth for replacement once a year.[43]

Heller has reported success in re-growing cochlea hair cells with the use of embryonic stem cells.[44]

Since 2003, researchers have successfully transplanted corneal stem cells into damaged eyes to restore vision. "Sheets of retinal cells used by the team are harvested from aborted fetuses, which some people find objectionable." When these sheets are transplanted over the damaged cornea, the stem cells stimulate renewed repair, eventually restore vision.[45] The latest such development was in June 2005, when researchers at the Queen Victoria Hospital of Sussex, England were able to restore the sight of forty patients using the same technique. The group, led by Sheraz Daya, was able to successfully use adult stem cells obtained from the patient, a relative, or even a cadaver. Further rounds of trials are ongoing.[46]

In April 2005, doctors in the UK transplanted corneal stem cells from an organ donor to the cornea of Deborah Catlyn, a woman who was blinded in one eye when acid was thrown in her eye at a nightclub. The cornea, which is the transparent window of the eye, is a particularly suitable site for transplants. In fact, the first successful human transplant was a cornea transplant. The absence of blood vessels within the cornea makes this area a relatively easy target for transplantation. The majority of corneal transplants carried out today are due to a degenerative disease called keratoconus.

The University Hospital of New Jersey reports that the success rate for growth of new cells from transplanted stem cells varies from 25 percent to 70 percent.[47]

In 2014, researchers demonstrated that stem cells collected as biopsies from donor human corneas can prevent scar formation without provoking a rejection response in mice with corneal damage.[48]

In January 2012, The Lancet published a paper by Steven Schwartz, at UCLA's Jules Stein Eye Institute, reporting two women who had gone legally blind from macular degeneration had dramatic improvements in their vision after retinal injections of human embryonic stem cells.[49]

In June 2015, the Stem Cell Ophthalmology Treatment Study (SCOTS), the largest adult stem cell study in ophthalmology ( http://www.clinicaltrials.gov NCT # 01920867) published initial results on a patient with optic nerve disease who improved from 20/2000 to 20/40 following treatment with bone marrow derived stem cells.[50]

Diabetes patients lose the function of insulin-producing beta cells within the pancreas.[51] In recent experiments, scientists have been able to coax embryonic stem cell to turn into beta cells in the lab. In theory if the beta cell is transplanted successfully, they will be able to replace malfunctioning ones in a diabetic patient.[52]

Human embryonic stem cells may be grown in cell culture and stimulated to form insulin-producing cells that can be transplanted into the patient.

However, clinical success is highly dependent on the development of the following procedures:[9]

Clinical case reports in the treatment orthopaedic conditions have been reported. To date, the focus in the literature for musculoskeletal care appears to be on mesenchymal stem cells. Centeno et al. have published MRI evidence of increased cartilage and meniscus volume in individual human subjects.[53][54] The results of trials that include a large number of subjects, are yet to be published. However, a published safety study conducted in a group of 227 patients over a 3-4-year period shows adequate safety and minimal complications associated with mesenchymal cell transplantation.[55]

Wakitani has also published a small case series of nine defects in five knees involving surgical transplantation of mesenchymal stem cells with coverage of the treated chondral defects.[56]

Stem cells can also be used to stimulate the growth of human tissues. In an adult, wounded tissue is most often replaced by scar tissue, which is characterized in the skin by disorganized collagen structure, loss of hair follicles and irregular vascular structure. In the case of wounded fetal tissue, however, wounded tissue is replaced with normal tissue through the activity of stem cells.[57] A possible method for tissue regeneration in adults is to place adult stem cell "seeds" inside a tissue bed "soil" in a wound bed and allow the stem cells to stimulate differentiation in the tissue bed cells. This method elicits a regenerative response more similar to fetal wound-healing than adult scar tissue formation.[57] Researchers are still investigating different aspects of the "soil" tissue that are conducive to regeneration.[57]

Culture of human embryonic stem cells in mitotically inactivated porcine ovarian fibroblasts (POF) causes differentiation into germ cells (precursor cells of oocytes and spermatozoa), as evidenced by gene expression analysis.[58]

Human embryonic stem cells have been stimulated to form Spermatozoon-like cells, yet still slightly damaged or malformed.[59] It could potentially treat azoospermia.

In 2012, oogonial stem cells were isolated from adult mouse and human ovaries and demonstrated to be capable of forming mature oocytes.[60] These cells have the potential to treat infertility.

Destruction of the immune system by the HIV is driven by the loss of CD4+ T cells in the peripheral blood and lymphoid tissues. Viral entry into CD4+ cells is mediated by the interaction with a cellular chemokine receptor, the most common of which are CCR5 and CXCR4.1 Because subsequent viral replication requires cellular gene expression processes, activated CD4+ cells are the primary targets of productive HIV infection.[61] Recently scientists have been investigating an alternative approach to treating HIV-1/AIDS, based on the creation of a disease-resistant immune system through transplantation of autologous, gene-modified (HIV-1-resistant) hematopoietic stem and progenitor cells (GM-HSPC).[62]

On 23 January 2009, the US Food and Drug Administration gave clearance to Geron Corporation for the initiation of the first clinical trial of an embryonic stem-cell-based therapy on humans. The trial aimed evaluate the drug GRNOPC1, embryonic stem cell-derived oligodendrocyte progenitor cells, on patients with acute spinal cord injury. The trial was discontinued in November 2011 so that the company could focus on therapies in the "current environment of capital scarcity and uncertain economic conditions".[63] In 2013 biotechnology and regenerative medicine company BioTime (NYSEMKT:BTX) acquired Geron's stem cell assets in a stock transaction, with the aim of restarting the clinical trial.[64]

Scientists have reported that MSCs when transfused immediately within few hours post thawing may show reduced function or show decreased efficacy in treating diseases as compared to those MSCs which are in log phase of cell growth(fresh), so cryopreserved MSCs should be brought back into log phase of cell growth in invitro culture before these are administered for clinical trials or experimental therapies, re-culturing of MSCs will help in recovering from the shock the cells get during freezing and thawing. Various clinical trials on MSCs have failed which used cryopreserved product immediately post thaw as compared to those clinical trials which used fresh MSCs.[65]

There is widespread controversy over the use of human embryonic stem cells. This controversy primarily targets the techniques used to derive new embryonic stem cell lines, which often requires the destruction of the blastocyst. Opposition to the use of human embryonic stem cells in research is often based on philosophical, moral or religious objections.[104] There is other stem cell research that does not involve the destruction of a human embryo, and such research involves adult stem cells, amniotic stem cells and induced pluripotent stem cells.

Stem-cell research and treatment was practiced in the People's Republic of China. The Ministry of Health of the People's Republic of China has permitted the use of stem-cell therapy for conditions beyond those approved of in Western countries. The Western World has scrutinized China for its failed attempts to meet international documentation standards of these trials and procedures.[105]

State-funded companies based in the Shenzhen Hi-Tech Industrial Zone treat the symptoms of numerous disorders with adult stem-cell therapy. Development companies are currently focused on the treatment of neurodegenerative and cardiovascular disorders. The most radical successes of Chinese adult stem cell therapy have been in treating the brain. These therapies administer stem cells directly to the brain of patients with cerebral palsy, Alzheimer's, and brain injuries.[citation needed]

Since 2008 many centres and doctors tried a diversity of methods; in Lebanon proliferative and non-proliferative, in-vivo and in-vitro techniques were used. The regenerative medicine also took place in Jordan and Egypt.[citation needed]

Stem-cell treatment is currently being practiced at a clinical level in Mexico. An International Health Department Permit (COFEPRIS) is required. Authorized centers are found in Tijuana, Guadalajara and Cancun. Currently undergoing the approval process is Los Cabos. This permit allows the use of stem cell.[citation needed]

In 2005, South Korean scientists claimed to have generated stem cells that were tailored to match the recipient. Each of the 11 new stem cell lines was developed using somatic cell nuclear transfer (SCNT) technology. The resultant cells were thought to match the genetic material of the recipient, thus suggesting minimal to no cell rejection.[106]

As of 2013, Thailand still considers Hematopoietic stem cell transplants as experimental. Kampon Sriwatanakul began with a clinical trial in October 2013 with 20 patients. 10 are going to receive stem-cell therapy for Type-2 diabetes and the other 10 will receive stem-cell therapy for emphysema. Chotinantakul's research is on Hematopoietic cells and their role for the hematopoietic system function in homeostasis and immune response.[107]

Today, Ukraine is permitted to perform clinical trials of stem-cell treatments (Order of the MH of Ukraine 630 "About carrying out clinical trials of stem cells", 2008) for the treatment of these pathologies: pancreatic necrosis, cirrhosis, hepatitis, burn disease, diabetes, multiple sclerosis, critical lower limb ischemia. The first medical institution granted the right to conduct clinical trials became the "Institute of Cell Therapy"(Kiev).

Other countries where doctors did stem cells research, trials, manipulation, storage, therapy: Brazil, Cyprus, Germany, Italy, Israel, Japan, Pakistan, Philippines, Russia, Switzerland, Turkey, United Kingdom, India and many others.

See the rest here:
Stem-cell therapy - Wikipedia, the free encyclopedia

Derivation of Ethnically Diverse Human Induced Pluripotent …

In vitro culture of primary human fibroblasts and lentivirus reprogramming

Human fibroblasts for iPSCs derivation were obtained from Coriell Institute (Camden, NJ) and reprogrammed using a single polycistronic vector using four-factor 2A (4F2A) doxycycline (DOX)-inducible lentivirus encoding mouse cDNAs for Oct4, Sox2, Klf4, and c-Myc separated by three different 2A peptides (P2A, T2A, and E2A, respectively). The lentiviral plasmids are p20321 (TetO-FUW-OSKM) and p20342 (FUW-M2rtTA) (Addgene, Cambridge, MA) originally developed by Carey et al.9. Lentiviral particles (4F2A and M2rtTA) were packaged in HEK 293T cells. The primary fibroblast cells were co-transfected using the lentivirus construct, psPAX and pCMV-VSVG vectors by calcium phosphate co-precipitation. Viral supernatants from cultures packaging each of the two viruses were pooled, filtered through a 0.45 m filter and concentrated by ultracentrifugation and stored at 80C.

The 5 human fibroblast lines were transduced by viral particles in xenofree human fibroblast culture medium10 in the presence of polybrene (8g/mL). Forty-eight hours after infection, less than 15% of fibroblasts tested immunopositive for viral-derived OCT4. The procedure was carried out in 1 well of a 6-well plate with cells at 70% confluence to allow for cell growth after viral infection an appearance of stem cell colonies. The medium was replaced two days after infection, and then daily, with xenofree hES medium plus doxycycline (1g/ml) formulated to maintain stem cell pluripotency10,11,12. After 35 days of culture, small cell clumps distinguishable from the fibroblast morphology appeared. Those that formed cell colonies with hESC-like morphology were mechanically isolated and passed on to mitotically inactivated xenofree human foreskin feeder cells (ATCC PCS-201010). Overall reprogramming efficiency by this method was calculated to be 0.002 ~ 0.004%. The iPSC colonies were expanded for several passages under xenofree conditions without doxycycline and evaluated for expression of markers of pluripotency by quantitative RT-PCR (qRT-PCR) and immunocytology.

Quantitative PCR analysis was done by isolation of total RNA from the hESC or iPSC lines and parental fibroblast lines and purification using the NucleoSpin RNA XS Total RNA isolation kit (Clontech). Reverse transcription (RT) was performed in a 20ul reaction volume using Superscript II (Invitrogen) and the cDNA reaction was diluted to a 300ul working stock volume. Primers for use in qPCR were first validated by maximally amplifying cDNA from a range of samples to confirm that a single PCR reaction product was produced and that the amplicon was of the predicted length. For validation, 10ul of cDNA from H9 hESCs (WA09, Wicell, Madison, WI), control fibroblasts (line A-2), and two of iPSC lines (A-2.2.1 & A-2.2.2) for each primer set was amplified for 36 cycles (95C 30s, 55C 30s, 72C 30s). For endogenous and transgene expression, 5ul of cDNA from each iPSC lines for each primer set was amplified for 32 cycles and resolved on a 3% nusieve agarose gel and visualized by ethidium bromide staining. Quantitative PCRs contained 10ng of cDNA, 400nM of each primer, and SYBR Green PCR Master Mix (AppliedBiosystems). Each sample was analyzed by triplicate by an ABI PRISM 7000 sequence detection system. Data was analyzed using the systems software. The expression of gene of interest was normalized to GAPDH in all cases and compared with hESCs.

We used the MycoAlertTM PLUS Assay mycoplasma detection kit (Lonza, Allendale, NJ) essentially as manufacturers instructions. Briefly, after centrifugation (1500rpm, 5min) of cell supernatant during passage of suspension iPSC cultures, the supernatants were transferred into luminescence compatible tubes (Corning Inc., Corning, NJ). The viable mycoplasma was lysed to allow enzymes to react with MycoAlertTM PLUS substrate, catalyzing the conversion of ADP to ATP. The level of ATP in the sample both before (reading A; ATP background) and after (reading B) the addition of MycoAlertTM PLUS substrate was assessed using a luminometer (Victor3, Perkin-Elmer, Waltham, Massachusetts, USA), so that a ratio B/A was obtained. Reading B assesses the conversion of ADP to ATP and is a monitor of contaminated samples. If the ratio of B/A is greater than 1 the cell culture was considered to be contaminated by mycoplasma. For control samples, the MycoAlert TM assay positive and negative control set was used.

Ethnically diverse-induced pluripotent stem cell (ED-iPSC) lines maintained on human foreskin fibroblast feeders were transferred to feeder-free conditions in non-tissue culture treated dishes coated with xenofree vitronectin (StemCell Technologies, Vancouver, Canada) or 1:100 Matrigel (10mg/ml; BD Biosciences, San Jose, CA) diluted into Hanks Buffered Saline Solution (Gibco HBSS; Life Technologies, Grand Island, NY). Cells were maintained in mTeSR2 complete media (StemCell Technologies, Vancouver, Canada) and mechanically passaged between days 5 and 7. Media was replaced on day 1 after the first passage of the series and cells grown overnight. On day 2, slow release Stem Beads FGF2 (20 microliters of PLGA beads loaded with hFGF2; StemBeads; Stem Culture Inc., Rensselaer, NY) were added with fresh mTeSR2 media. Media changes were done every 3 days with Stem Beads FGF2 and mTeSR2. Preparation of uniform sized EBs from iPSCs colonies was done in custom lithography template microarrays (LTA) generated in-house. Chemical dissociation of the stem cell colonies into single cell suspension was done before and loading of the cells into LTA- polydimethylsiloxane (PDMS) grids in mTeSR2 media in the presence of 10M Rock inhibitor (Sigma-Aldrich, St. Louis, MO) at day 0. Stem cells were maintained in grids for five days with media changes every two days. For directed multi-lineage early differentiation we used the Human Pluripotent Stem Cell Functional Identification Kit (R&D Systems, Minneapolis, MN).

For immunocytology of biomarkers in iPSC colonies, cells were prepared by two methods. Cells were fixed using 4% paraformaldehyde in PBS for 15min at room temperature and blocked by incubating cells for 90min in a solution containing 3% normal donkey serum and permeabilized by 0.1% Triton-X 100 for 10min before antibody addition. Incubations with the primary antibodies of anti-Nanog (Santa Cruz Biotechnology, Dallas, TX) and anti-SSEA4 (Santa Cruz Biotechnology, Dallas, TX) were done at 4C overnight, followed by incubation with a secondary antibody conjugated with Alexa 647 or Alexa 488 (Abcam, Cambridge, MA). After rinsing with phosphate buffered saline (PBS), the DNA was stained with bisBenzimide H 33258 (Sigma-Aldrich, St. Louis, MO) and cells imaged using a digital camera connected to a Nikon TE-2000 inverted microscope.

Phase imaging for in vitro differentiated samples was done on a Nikon 80i epifluorescence microscope using a PLAN 100.30 NA DL objective and images captured with a cooled QICam CCD camera. Fluorescent images were obtained on a Leica SP5 Laser Scanning Confocal Microscope using either HC PL FLUOTAR 100.30 NA or HCX PL APO CS 20X .70 NA objectives and also on a Zeiss AxioObserver Z1 Inverted Microscope with Colibri LED illumination, using a 100X oil 1.45 NA PlanFLUAR or 63X Plan-Apochromat 1.4 NA oil DIC objectives. Images were captured with a Hamamatsu ORCA ER CCD camera and Zeiss Axiovision Rel 4.8 acquisition software. Figures were compiled using Adobe Photoshop (Adobe Systems Inc., San Jose, CA) and Microsoft PowerPoint (Microsoft Corp., Redmond, WA) software.

The immunocytology of 2D cell cultures or three dimensional EBs was done by first fixing cells for 10 minutes at room temperature in 4% paraformaldehyde and stored overnight in PBS+0.1% Tween20 at 4C. Immediately before incubation with antibodies, the cells were permeabilized with PBS+0.5% Triton X-100 for 1 hour at 4C. Nonspecific binding was blocked by 20 minute incubation in 1% BSA in HBSS and followed by a single HBSS wash. Antibodies used for gauging pluripotency recognized Oct4A C-10 (Santa Cruz Biotechnology, Dallas, TX) and anti-SSEA4 (Millipore, Billerica, MA) (1:1000 each). Analysis of lineage commitment to differentiation was done using antibodies to OTX2 (ectoderm), SOX17 (endoderm), and Brachyury (mesoderm; 1:100 each) provided in the Human Pluripotent Stem Cell Functional Identification Kit (R&D Systems, Minneapolis, MN). Secondary antibodies were either AlexaFluor 488 or AlexaFluor 594 (A-11001, A-11037, Invitrogen, Carlsbad, CA). Nuclei were stained with bisBenzimide H 33258 (Sigma-Aldrich, St. Louis, MO) at 4C overnight and followed by washing one hour in HBSS at 4C. Samples were mounted in ProLong Gold antifade reagent (Life Technologies, Grand Island, NY) at 20C overnight in the dark before imaging immediately or storing at 4C.

Approximately 2 million ED-iPSCs were injected subcutaneously in the flank region of NOD scid gamma (NSG) mice (The Jackson Lab, Bar harbor, ME). After 1224 weeks, teratomas were formed from 10 iPSC lines, and tumors were excised & fixed in 10% normal buffered formalin (NBF) overnight. The samples were processed for histology by the Division of Human Pathology at MSU. Hematoxylin- and eosin (H&E)-stained sections were examined under a microscope.

Follow this link:
Derivation of Ethnically Diverse Human Induced Pluripotent ...