Search Results for: neurofibromatosis type stem cell therapy

Systematic Review of Pediatric Brain Tumors in Neurofibromatosis Type 1: Status of Gene Therapy – Cureus

Posted: August 14, 2022 at 2:02 am

As oncology practice is rapidly shifting away from toxic chemotherapy, gene therapy provides a highly specific therapeutic approach for brain tumors. This treatment is rapidly evolvingto deliver specific therapeutic genes or oncolytic viruses to eliminate the tumor, which can lead to tumor cell death and increased immune responses to tumor antigens, and disruption of the tumor microenvironment (TME), including angiogenesis/neovascularization inhibition [1]. Oncolytic virotherapy (OV), suicide gene therapy, tumor suppressor gene delivery, immunomodulatory strategies, and gene target therapies are the various types of gene therapies. Gene therapy delivery methods include direct delivery of therapeutic genes into the tumor site, which include virus-mediated adenovirus, herpes simplex virus-1 (HSV-1), adeno-associated virus-2, nonviral vector-based nanoparticles, liposomes, and micelles. Neural stem cells and mesenchymal stem cells are tumor-tropic cell carriers that express therapeutic gene(s) in the tumor site. PH-sensitive drug release, pH-sensitive liposomal carriers, and stimuli-responsive particles are examples of intelligent carriers [2].

According to the National Brain Tumor Society (NBTS), approximately 700,000 Americans have been diagnosed with a primary brain tumor, with 63% being benign and 37% being malignant. Brain tumors were the 10th leading cause of death in 2020 [3]. The pediatric brain tumors associated with neurofibromatosis type 1 (NF1) are optic pathway gliomas (OPGs), brain stem gliomas, glioblastomas, and pilocytic astrocytoma [4]. Brain and central nervous system (CNS) tumors have been reported in approximately 20% of patients with NF1 and are typically discovered in childhood. Optic pathway gliomas (OPGs) account for approximately 70% of all CNS tumors in children with NF1, while brain stem glioma accounts for approximately 17% of all CNS tumors [5]. Despite recent advances in surgery, radiotherapy, and chemotherapy, brain tumor treatment regimens have only a limited impact on long-term disease control [6]. The price of the cure is frequently unacceptable, and it includes acute and chronic organ toxicity, resistance to therapy, and more concerning, an increased risk of secondary malignancy. As a result, new strategies are required to improve overall survival and reduce treatment-related morbidity [7]. To tackle this situation, a better understanding of the functions and control of genes was needed, which paved the way for the development of gene therapy in the last decades [6].

The current study aims to provide an advance in gene therapies for pediatric brain tumors with neurofibromatosis type 1. This includes different genomic alterations seen in brain tumors and gene delivery systems comprising viral and nonviral delivery platforms along with suicide/prodrug, oncolytic, cytokine, and tumor suppressor-mediated gene therapy approaches. Finally, we discuss the results of gene therapy-mediated human clinical trials and highlight the progress, prospects, and remaining challenges of gene therapies aiming at broadeningour understanding and highlighting the therapeutic approach for pediatric brain tumors.

This systematic review was performed in March 2022 usingthe Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 guidelines [8].

Eligibility Criteria

The inclusion criteria were cases of neurofibromatosis type 1 brain tumors in all age groups with the additional inclusion of English language, free full text, articles published within the last 20 years, randomized controlled trials (RCTs), observational studies, systematic reviews, and meta-analyses. Weexcluded case reports, case studies, and editorials.

Databases and Search Strategy

The search was conducted systematically using PubMed, Cochrane, Google Scholar, and ClinicalTrials.gov databases by the first and second authors separately. Table 1 summarizes the search strategy.

The search identified a total of 1,213 articles from the databases. EndNote is used to remove duplicated articles. The remaining articleswere screened manually by the first and second authors. A total of 145 articles from databases were sought for retrieval, and 25 articles from the databaseswere retrieved and sent for quality appraisal. The articles were assessed for quality by the first two authors separately using tools depending on the study type: Cochrane Collaboration Risk of Bias Tool (CCRBT) for randomized control trials [9], Scale for the Assessment of Narrative Review Articles 2 (SANRA 2) for narrative reviews [10], and Assessment of Multiple Systematic Reviews (AMSTAR) for systematic reviews and meta-analyses [11]. Nineteen studies included in the review were scored above 70% (Figure 1).

Table 2 shows the result of the summary of the quality assessment of narrative reviews by authors.

In the study by Immonen et al., compared to controls (n = 7 patients), there is a substantial rise in the mean number of tolerated O6-benzylguanine (O6BG)/temozolomide (TMZ) cycles (P = 0.05) with gene therapy. The median progression-free survival was nine months, and the overall survival was 20 months. The study revealed delayed tumor growth at lower cumulative TMZ doses in the study patients compared to those who received standardregimens, concluding that this supports the chemoprotective effect of gene therapy when used in combination with O6BG and TMZ [25]. In the study ofAdair et al., treatment of adenovirus-mediated herpes simplex virus thymidine kinase (AdvHSV-tk) resulted in a clinically and statistically significant increase in mean survival from 39.0 19.7 (standard deviation) to 70.6 52.9 weeks (P = 0.0095). From 37.7 to 62.4 weeks, the median survival time also increased, and treatment was well tolerated. The authors concluded that AdvHSV-tk gene therapy with ganciclovir (GCV) could be a promising new treatment[26].Table 3 summarizes the risk of bias in randomized controlled trials using the Cochrane Collaboration Risk of Bias Tool (CCRBT).

Table 4 summarizes the resultof critical appraisal for systematic reviews and meta-analyses by review authors.

Brain tumors account for 21% of childhood malignancies and are the primary cause of solid tumor cancer death in children.Children affected with neurofibromatosis type 1 (NF1) are prone to optic pathway gliomas, brain stem gliomas, glioblastomas, and pilocytic astrocytoma. Two-thirds of gliomas are found in the optic pathway, with the brain stem, cerebellum, cerebral hemispheres, and subcortical structures accounting for the remaining locations. Chemotherapy is used to treat clinical progression, but surgery and radiation are difficult to use in the case of NF1-associated optic pathway gliomas since surgical resection is usually unachievable due to the tumors position. Radiation is not suggested for children with NF1 because of the possibility of secondary tumors (glioma and malignant peripheral nerve sheath tumors) in the context of this tumor predisposition syndrome, as well as the risk of late neurocognitive sequelae in children. Vincristine and carboplatin are used in first-line optic pathway glioma treatment. Vinblastine, vinorelbine, and temozolomide are the second-line chemotherapy options [28].

Overall, five-year survival rates for children less than 15 years of age are currently around 75% and 77% for those aged 15-19. Despite these advancements in treatment, a considerable number of individuals continue to be resistant to typical treatments. Acute and chronic organ damage, as well as an increased risk of secondary malignancy, are all disadvantages. Successful glioma treatment is hampered by ineffective medication distribution across the blood-brain barrier (BBB), an immunosuppressive tumor microenvironment (TME), and the development of drug resistance. Because gliomas are caused by the accumulation of genetic changes over time, gene therapy, which allows for the altering of the genetic makeup of target cells, appears to be a viable way to overcome the challenges that existing therapeuticstrategies face [7].

Figure 2 explains the pathways involved in oncogenesis. By converting the active form of guanosine triphosphate (GTP)-bound Kirsten rat sarcoma virus (KRAS) to its inactive, guanosine diphosphate (GDP)-bound state, neurofibromin directly suppresses KRAS activation. Mitogen-activated protein kinases (MAPKs) and extracellular signal-regulated kinases 1 and 2 (ERK1 and ERK2) are activated by GTP-bound KRAS. The activation of rapidly accelerated fibrosarcoma gene (RAF)/MAPK causes transcription and cell proliferation to increase. Unchecked KRAS activation can also result in the cross-activation of the phosphoinositide 3 kinase (PI3K)-mammalian target of rapamycin (mTOR) pathway, which is critical for cell proliferation and survival. GTP-bound KRAS, for example, can bind and activate PI3K, resulting in survival and proliferation effects via AK strain transforming (AKT) and mTOR activity. As a result, neurofibromin deficiency can cause disease in a variety of ways. In gliomas, the KRAS, PI3K/phosphatase and tensin homolog (PTEN)/AKT pathways and neurogenic locus notch homolog protein (NOTCH) signaling are linked to cancer cell proliferation[29].

Glioblastoma Multiforme (GBM)

Complete resection of GBM is virtually impossible due to its intrusive nature and sensitive location. The current standard of care is a maximum surgical resection followed by radiation and temozolomide chemotherapy; however, the median survival time is still fewer than 15 months. This necessitates the creation of gene therapy, which delivers oncolytic viruses to the tumor in a precise manner to destroy it and lead to tumor cell death as well as increased immune responses to tumor antigens and disturbance of the tumor microenvironment, including angiogenesis/neovascularization inhibition [1].The common gene targets that are mutated or upregulated in glioblastoma are neurofibromin, epidermal growth factor receptor (EGFR), phosphate and tensin (PTEN) homolog, platelet-derived growth factor (PDGF) receptor-alpha, isocitrate dehydrogenase-1 (IDH1), and tumor suppressor p53. GBM is a suitable candidate for gene therapy for several reasons:tumors remain within the brain with only rare metastases to other tissues; most cells in the brain are postmitotic, which allows for precise targeting of dividing tumor cells; tumors are often accessible neurosurgically for vector administration; sophisticated imaging paradigms are available; and standard therapies are minimally successful.

Delivery Methods for Gene Therapy

Table 5 summarizes the advantages, limitations, and clinical trials of the viral vectors used for gene therapy.

Table 6 summarizes the advantages and limitationsof the nonviral vectors used for gene therapy.

Table 7 summarizes the advantages and limitations of tumor-tropic cell carriers expressing therapeutic gene(s) in the tumor site.

Oncolytic Virotherapy (OV)

OVs are intended to particularly infect cancer cells, self-replicate, induce oncolysis, and amplify therapeutic genes at tumor sites [27]. The advantages of OV include its high transduction efficiency and intra-tumoral viral spread, the capability of producing high viral titers, accessibility to genetic engineering, and adding additional therapeutic transgenes. Its limitations include host immune rejection/suppression of the virus, safety risks surrounding replication-competent viruses, and requirement of local administration during surgery[30]. Figure 3 explains the mechanism of action of oncolytic virotherapy.

Oncolytic herpes simplex virus (oHSV) are double-stranded deoxyribonucleic acid (DNA) viruses, a human pathogen with neurotropic properties. The challenge in designing oHSVs is to provide tumor selectivity while maintaining an acceptable safety profile [27]. Early clinical trial results showed that numerous oHSV vectors had high safety profiles with no signs of encephalitis but poor therapeutic effectiveness [31].

Conditionally replicating adenovirus (CRAd) are nonenveloped DNA viruses capable of infecting both the dividing and nondividing cells. An important advantage of CRAd viruses is that they are naturally non-neurotropic and have an enhanced safety profile over the oHSV vector. ONYX-015 and Ad5-Delta24 bare the most widely studied CRAd [14]. ONYX-015 contains a deletion in the viral protein early region 1B-55K (E1B-55K), which normally binds to and inactivates the host cell p53 protein. Therefore, it is assumed that cells with functional p53 cannot support viral replication in the absence of this protein, whereas tumor cells with a nonfunctional support viral replication.

Oncolytic measles, reovirus vectors, and recombinant nonpathogenic polio rhinovirus (PVS-RIPO) are reoviruses that only replicate in glioma cells because platelet-derived growth factor receptor (PDGFR) or EGFR stimulation of the KRASpathway suppresses ribonucleic acid (RNA)-activated protein kinase activation. Clinical trial demonstrates that they are safe and well-tolerated with no evidence of clinical encephalitis. Measles virus (MV) exhibits the mutated hemagglutinin envelope glycoprotein H, which targets the cluster of differentiation 46 (CD46) on glioma cells. The circulating carcinogenic embryonic antigen (CEA) was modified into MV, which can be used to measure virus replication and oncolytic function [27]. PVS-tumor RIPOs cell tropism is determined by the poliovirus receptor CD155, which is expressed on high-grade glioma cells. The clinical trialsfindings revealed satisfactory antitumor effectiveness but a low safety profile. Table 8 summarizes clinical trials and results on oncolytic virotherapy.

Suicide Gene Therapies

The suicide gene technique is based on virally delivering suicide genes to target cells, which produce enzymes that convert prodrugs to active compounds. The inert prodrug is given systematically and then activated by suicide enzymes at the tumor site, resulting in tumor cell apoptosis [27]. Its advantages include achieving a bystander effect, requiring short-term gene expression, selective tumor cell targeting, and enhancing sensitivity to conventional therapy. It is restricted by the limited spatial distribution of gene transfer vectors, poor gene transfer efficiency into tumor cells in vivo, inability to target dispersed tumor cells, and restricted intra-tumoral distribution.Figure 4 explains the mechanism of action of suicide gene therapy.

Herpes simplex virus thymidine kinase (HSV-tk) enzyme catalyzes ganciclovir/valacyclovir monophosphorylation, which occurs after the triphosphorylation and activation of intracellular kinases. The active medication inhibits DNA synthesis and tumor lysis by blocking the S phase and arresting the cell circle. Cytosine deaminase (CD) catalyzes the activation of the prodrug 5-fluorocytosine (5-FC). A replication-competent retrovirus called Toca 511 loads the CD and transinfects tumor cells. It stimulates the expression of CD, which activates the 5-FU, which blocks DNA synthesis irreversibly and causes cell death. Escherichia coli-derived purine nucleoside phosphorylase (PNP)transforms adenosine ribonucleosides, such as fludarabine, into the active adenine molecule, 2-fluoroadenine, which disrupts RNA replication and the cell cycle. Antibiotic therapy, which suppresses intestinal flora, may over-activate the PNP gene therapy, resulting in increased prodrug conversion [27]. Table 9 summarizes clinical trials and results on suicide gene therapies.

Tumor Suppressor Gene Therapies

High-grade gliomas frequently have deletions and mutations in tumor suppressor genes such as p53, p16, and phosphatase and tensin homologs (PTEN) [2]. Tumor suppressor gene techniques aim to restore normal function by transferring antitumoral functional genes to glioma cells. The advantages are safety in clinical trials, the potential to induce senescence within tumors, and the potential to sensitize tumor cells to other therapies. The limitations are as follows: multiple redundant pathways in tumors hinder efficacy, poor in vivo gene transfer, and limited distribution of therapy.Figure 5 explains the mechanism of action of tumor suppressor gene therapy.

P53 is involved in the inhibition of angiogenesis and DNA repair pathways. E1 gene is replaced by wild-type p53 in adenovirus and transmitted via a cytomegalovirus promoter (Ad5CMV-p53), which is the most widely used method. The E1 deletion prevents the virus from starting the infectious phase, while the cytomegalovirus promoter boosts the production of the p53 gene [27].

P16 prevents uncontrolled replication and oncogenesis by arresting the cell cycle during the G1-S transition [32]. Restoration of p16 function through an adenoviral vector has been found to decrease glioma growth and locoregional dissemination while also inhibiting matrix metalloprotease activity in the glioma microenvironment [33]. The adenovirus-mediated p16 gene was used to drive p16-null human glioma cell lines to enter phase G1 of the cell cycle. In HGG cells, data revealed that p16 expression is linked to tumor radiosensitivity through mechanisms of aberrant nucleation [34]. It is worth noting that the efficiency of the p16 gene approach is contingent on maintaining retinoblastoma protein (pRB) activity [35].

The PTEN gene has been shown to suppress glioma proliferation and induce oncolysis when delivered through an adenoviral vector [27]. Adenoviral vector transfer of the PTEN gene into glioma cells improved tumor sensitivity to temozolomide and radiation [36].Table 10 summarizes clinical trials and results on tumor suppressor gene therapies.

Immunomodulatory Gene Therapies

The objective of anti-glioma immunomodulatory gene therapy is to induce or augment the T-cell-mediated immune response against tumors using the delivery of genes for immunostimulatory cytokines and interferon beta/gamma (IFN-/) [27]. Its advantages include the following: this therapy can achieve passive or active tumor immunity, it has the possibility to eliminate tumor cells that remain post-surgery, and it regulates the tumor microenvironment. This therapy is limited by tumor-induced immunosuppression, lack of antigen-presenting dendritic cells within the brain, and overcoming the presence of immune-suppressive regulatory T-cells and cytokines.Figure 6 explains the mechanism of action of immunomodulatory gene therapy.

The stimulation of natural killer cells and macrophages demonstrated potential antitumoral action [37]. INF- was also transferred using nanoparticles and liposomes. Clinical trial shows a reduction in volumetric glioma and mild toxicity [38]. Histological findings reported an elevated level of immune activation[39]. IFN- inhibits cancer cell proliferation and interactions with the extracellular matrix [40].

Interleukin-12 (IL-12) is one of the most important immunostimulant cytokines for strengthening the immune system and attracting cytotoxic cells in the tumor microenvironment. Nonreplicating adenoviruses and HSV were used in an earlier phase of research to deliver IL-12 to malignant glioma cells. Preclinical research revealed tumor cell death, active microglia cell infiltration, a favorable safety profile, and a significant local immune response [27].

Several clinical trials have shown that chemotherapy has a synergistic impact when combined with immunotherapy, challenging the conventional dogma that chemotherapy-induced immunosuppression prevents the formation of antitumor immune responses. In a limited phase I clinical trial, three pediatric patients with recurring brain tumors were given a combination of high-dose chemotherapy and adoptive immunotherapy [41]. Accumulating preclinical and clinical evidence suggests that combining tumor cell killing techniques with immunotherapy results in synergism between the two therapies, resulting in improved efficacy and lower toxicity. This collection of evidence refutes the conventional notion that tumor cell killing tactics hinder the immune systems ability to recognize and eradicate a brain tumor, and it supports the use of combined cytotoxic-immunotherapeutic strategies in the treatment of glioblastoma multiforme patients [42].Table 11 summarizes clinical trials and results on immunomodulatory gene therapy.

Gene Target Therapies

Gene target medicinesdirectly bind specific tumor antigens to block oncogenic pathways irreversibly.Figure 7 explains the target gene mechanism of action.

The epidermal growth factor receptor (EGFRvIII) variation, which is prevalent in 30% of high-grade gliomas, is involved in oncogenesis and tumor development processes. Antisense or short interfering RNA (siRNA) directed exclusively targeting the thymidine kinase domain of glioma EGFRvIII was delivered by viral vectors and nanoparticles [43]. The delivery of EGFRvIII siRNA using cyclodextrin-modified dendritic polyamine complexes (DexAMs) exhibited promising effects in malignant glioma cells, even when combined with erlotinib [44].

Direct intra-tumoral inoculation of polyethylenimine (PEI)/VEGF siRNA had a substantial antiangiogenic impact on xenografts [44]. In the Matrigel plug experiment, Ad-DeltaB7-shVEGF, an adenovirus construct, was developed, expressing a short hairpin RNA against VEGF; it showed excellent antiangiogenic action and better bioavailability than replication-incompetent adenoviruses [45]. In a human xenografted glioma model, Ad-DeltaB7-KOX,an oncolytic adenovirus, showed strong anticancer efficacy [46]. Another study looked at HGGs infected with adenovirus expressing vascular endothelial growth factor receptor (VEGFR) and the oncolytic virus dl922/947. This combination therapy was more successful than monotherapy [27].

Go here to see the original:
Systematic Review of Pediatric Brain Tumors in Neurofibromatosis Type 1: Status of Gene Therapy - Cureus

Posted in Gene therapy | Comments Off on Systematic Review of Pediatric Brain Tumors in Neurofibromatosis Type 1: Status of Gene Therapy – Cureus

Massachusetts Stem Cells | Stem Cell TV

Posted: September 10, 2019 at 7:43 pm

James F. Battey, Jr., MD, PhD; Laura K. Cole, PhD; and Charles A. Goldthwaite, Jr., PhD.

Stem cells are distinguished from other cells by two characteristics: (1) they can divide to produce copies of themselves (self-renewal) under appropriate conditions and (2) they are pluripotent, or able to differentiate into any of the three germ layers: the endoderm (which forms the lungs, gastrointestinal tract, and interior lining of the stomach), mesoderm (which forms the bones, muscles, blood, and urogenital tract), and ectoderm (which forms the epidermal tissues and nervous system). Pluripotent cells, which can differentiate into any mature cell type, are distinct from multipotent cells (such as hematopoietic, or blood-forming, cells) that can differ into a limited number of mature cell types. Because of their pluripotency and capacity for self-renewal, stem cells hold great potential to renew tissues that have been damaged by conditions such as type 1 diabetes, Parkinson's disease, heart attacks, and spinal cord injury. Although techniques to transplant multipotent or pluripotent cells are being developed for many specific applications, some procedures are sufficiently mature to be established options for care. For example, human hematopoietic cells from the umbilical cord and bone marrow are currently being used to treat patients with disorders that require replacement of cells made by the bone marrow, including Fanconi's anemia and chemotherapy-induced bone marrow failure after cancer treatment.

However, differentiation is influenced by numerous factors, and investigators are just beginning to understand the fundamental properties of human pluripotent cells. Researchers are gradually learning how to direct these cells to differentiate into specialized cell types and to use them for research, drug discovery, and transplantation therapy (see Figure 8.1). However, before stem cell derivatives are suitable for clinical application, scientists require a more complete understanding of the molecular mechanisms that drive pluripotent cells into differentiated cells. Scientists will need to pilot experimental transplantation therapies in animal model systems to assess the safety and long-term stable functioning of transplanted cells. In particular, they must be certain that any transplanted cells do not continue to self-renew in an unregulated fashion after transplantation, which may result in a teratoma, or stem cell tumor. In addition, scientists must ascertain that cells transplanted into a patient are not recognized as foreign by the patient's immune system and rejected.

Figure 8.1. The Scientific Challenge of Human Stem CellsThe state of the science currently lies in the development of fundamental knowledge of the properties of human pluripotent cells. The scientific capacity needs to be built, an understanding of the molecular mechanisms that drive cell specialization needs to be advanced, the nature and regulation of interaction between host and transplanted cells needs to be explored and understood, cell division needs to be understood and regulated, and the long-term stability of the function in transplanted cells needs to be established.

Stem cells derived from an early-stage human blastocyst (an embryo fertilized in vitro and grown approximately five days in culture) have the capacity to renew indefinitely, and can theoretically provide an unlimited supply of cells. It is also possible to derive stem cells from non-embryonic tissues, including amniotic fluid, placenta, umbilical cord, brain, gut, bone marrow, and liver. These stem cells are sometimes called "adult" stem cells, and they are typically rare in the tissue of origin. For example, blood-forming (hematopoietic) stem cell experts estimate that only 1 in 2000 to fewer than 1 in 10,000 cells found in the bone marrow is actually a stem cell.1 Because so-called "adult" stem cells include cells from the placenta and other early stages of development, they are more correctly termed "non-embryonic stem cells." Non-embryonic stem cells are more limited in their capacity to self renew in the laboratory, making it more difficult to generate a large number of stem cells for a specific experimental or therapeutic application. Under normal conditions, non-embryonic stem cells serve as a repair pool for the body, so they typically differentiate only into the cell types found in the organ of origin. Moreover, there is little compelling evidence for trans-differentiation, whereby a stem cell from one organ differentiates into a mature cell type of a different organ. New discoveries may overcome these limitations of stem cells derived from non-embryonic sources, and research directed toward this goal is currently underway in a number of laboratories.

Cultures of human pluripotent, self-renewing cells enable researchers to understand the molecular mechanisms that regulate differentiation (see Figure 8.2), including epigenetic changes (traits that may be inherited that do not arise from changes in the DNA sequence) in the chromatin structure, developmental changes in gene expression, exposure to growth factors, and interactions between adjacent cells. Understanding these basic mechanisms may enable future scientists to mobilize and differentiate endogenous populations of pluripotent cells to replace a cell type ravaged by injury or disease. Alternatively, scientists may some day be able to coax human pluripotent cells grown in the laboratory to become a specific type of specialized cell, which physicians could subsequently transplant into a patient to replace cells damaged by these same disease processes.

Scientists are gradually learning to direct the differentiation of pluripotent cell cultures into a specific type of cell, which can then be used as cellular models of human disease for drug discovery or toxicity studies. While it is not possible to predict the myriad ways that a basic understanding of stem cell differentiation may lead to new approaches for treating patients with cellular degenerative diseases, some avenues can be theorized. For example, in the case of Huntington's disease, a fatal neurodegenerative disorder, one could imagine that pluripotent cells derived from an embryo that carries Huntington's disease and differentiated into neurons in culture could be used to test drugs to delay or prevent degeneration.

Despite the incredible growth in knowledge that has occurred in stem cell research within the last couple of decades, investigators are just beginning to unravel the process of differentiation. Human pluripotent cell lines are an essential tool to understand this process and to facilitate the ultimate use of these cells in the clinic. To provide background on this fundamental topic, this article reviews the various potential sources and approaches that have been used to generate human pluripotent and multipotent cell lines, both of embryonic and non-embryonic origin.

Currently, at least six embryonic sources have been used to establish human pluripotent stem cell lines. All approaches involve isolation of viable cells during an early phase of development, followed by growth of these cells in appropriate culture medium. The various sources of these initial cell populations are discussed in brief below. It should be noted that the manipulation and use of embryonic tissues has raised a number of ethical issues.2,3 This article focuses on the scientific and technical issues associated with creating pluripotent cells, with the understanding that some of these techniques are currently subject to debates that extend beyond discussions of their scientific merits.

Figure 8.2. The Promise of Stem Cell ResearchStem cell research provides a useful tool for unraveling the molecular mechanisms that determine the differentiation fate of a pluripotent cell and for understanding the gene expression properties and epigenetic modifications essential to maintain the pluripotent state. In the future, this knowledge may be used to generate cells for transplantation therapies, whereby a specific cell population compromised by disease is replaced with new, functional cells. Differentiated derivatives of human pluripotent cells may also prove to be useful as models for understanding the biology of disease and developing new drugs, particularly when there is no animal model for the disease being studied. The greatest promise of stem cell research may lie in an area not yet imagined.

2008 Terese Winslow

Drawing upon twenty years of communal expertise with mouse ES cells,4 and on human inner cell mass culture conditions developed by Ariff Bongso and colleagues,5 James Thomson and colleagues at the University of Wisconsin generated the first hESC lines in 1998 using tissue from embryos fertilized in vitro.6 This method uses embryos generated for in vitro fertilization (IVF) that are no longer needed for reproductive purposes. During IVF, medical professionals usually produce more embryos than a couple attempting to start a family may need. Spare embryos are typically stored in a freezer to support possible future attempts for additional children if desired. It is estimated that there are approximately 400,000 such spare embryos worldwide.6 If these embryos are never used by the couple, they either remain in storage or are discarded as medical waste. Alternatively, these embryos can potentially be used to generate a hESC line.

To generate a hESC line, scientists begin with a donated blastocyst-stage embryo, at approximately five days after IVF (see Figure 8.3a). The blastocyst consists of approximately 150200 cells that form a hollow sphere of cells, the outer layer of which is called the trophectoderm. During normal development, the trophoblast becomes the placenta and umbilical cord. At one pole of this hollow sphere, 3050 cells form a cluster that is called the inner cell mass (ICM), which would give rise to the developing fetus. ICM cells are pluripotent, possessing the capacity to become any of the several hundred specialized cell types found in a developed human, with the exception of the placenta and umbilical cord.

Scientists remove the ICM from the donated blastocyst and place these cells into a specialized culture medium. In approximately one in five attempts, a hESC line begins to grow. Stem cells grown in such a manner can then be directed to differentiate into various lineages, including neural precursor cells,8 cardiomyocytes,9 and hematopoietic (blood forming) precursor cells.10

However, hESC lines are extremely difficult to grow in culture; the cells require highly specialized growth media that contain essential ingredients that are difficult to standardize. Yet the culture conditions are critical to maintain the cells' self-renewing and pluripotent properties. Culture requires the support of mouse or human cells, either directly as a "feeder" cell layer6,11,12 or indirectly as a source of conditioned medium in feeder-free culture systems.13 The feeder cells secrete important nutrients and otherwise support stem cell growth, but are treated so they cannot divide. Although the complete role of these feeder cells is not known, they promote stem cell growth, including detoxifying the culture medium and secreting proteins that participate in cell growth.14 hESC lines used to produce human cells for transplantation therapies may need to be propagated on a human feeder cell layer to reduce the risk of contamination by murine viruses or other proteins that may cause rejection. Thus, hESC lines often grow only under highly specific culture conditions, and the identification of ideal growth conditions presents a challenge regardless of the source of the hESCs.

Furthermore, human ES cell cultures must be expanded using an exacting protocol to avoid cell death and to control spontaneous differentiation. Since a limited number of laboratories in the United States are growing these cells, there is a shortage of people well-versed in the art and science of successful hESC culture. In the short term, challenges of working with these cells include developing robust culture conditions and protocols, understanding the molecular mechanisms that direct differentiation into specific cell types, and developing the infrastructure to advance this scientific opportunity. Once these challenges have been met, scientists will need to conduct transplantation studies in animal models (rodent and non-human primates) to demonstrate safety, effectiveness, and long-term benefit before stem cell therapies may enter clinical trials.

A second method for generating human pluripotent stem cell lines was published in 1998 by John Gearhart and coworkers at The Johns Hopkins Medical School.15

These researchers isolated specialized cells known as primordial germ cells (PGCs) from a 57-week-old embryo and placed these cells into culture (see Figure 8.3b). PGCs are destined to become either oocytes or sperm cells, depending on the sex of the developing embryo. The resulting cell lines are called embryonic germ cell lines, and they share many properties with ES cells. As with ES cells, however, PGCs present challenges with sustained growth in culture.16,17 Spontaneous differentiation, which hinders the isolation of pure clonal lines, is a particular issue. Therefore, the clinical application of these cells requires a more complete understanding of their derivation and maintenance in vitro.

Embryos that stop dividing after being fertilized in vitro are not preferentially selected for implantation in a woman undergoing fertility treatment. These embryos are typically either frozen for future use or discarded as medical waste. In 2006, scientists at the University of Newcastle, United Kingdom, generated hESC lines from IVF embryos that had stopped dividing.18 These scientists used similar methods as described under "Traditional hESC Line Generation" except that their source material was so-called "dead" IVF embryos (see Figure 8.3c). The human stem cells created using this technique behaved like pluripotent stem cells, including producing proteins critical for "stemness" and being able to produce cells from all three germ layers. It has been proposed that an IVF embryo can be considered dead when it ceases to divide.19 If one accepts this definition, such an embryo that "dies" from natural causes presumably cannot develop into a human being, thereby providing a source to derive human ES cells without destroying a living embryo.

Figure 8.3. Alternative Methods for Preparing Pluripotent Stem Cells

2008 Terese Winslow

Couples who have learned that they carry a genetic disorder sometimes use pre-implantation genetic diagnosis (PGD) and IVF to have a child that does not carry the disorder. PGD requires scientists to remove one cell from a very early IVF human embryo and test it for diseases known to be carried by the hopeful couple. Normally, embryos identified with genetic disorders are discarded as medical waste. However, Dr.Yuri Verlinsky and colleagues have capitalized on these embryos as a way to further our understanding of the diseases they carry (see Figure 8.3d) by deriving hESC lines from them.20 These stem cell lines can then be used to help scientists understand genetically-based disorders such as muscular dystrophy, Huntington's disease, thalessemia, Fanconi's anemia, Marfan syndrome, adrenoleukodystrophy, and neurofibromatosis.

In 2006, Dr. Robert Lanza and colleagues demonstrated that it is possible to remove a single cell from a pre-implantation mouse embryo and generate a mouse ES cell line.21 This work was based upon their experience with cleavage-stage mouse embryos. Later that same year, Dr. Lanza's laboratory reported that it had successfully established hESC lines (see Figure 8.3e) from single cells taken from pre-implantation human embryos.22 The human stem cells created using this technique behaved like pluripotent stem cells, including making proteins critical for "stemness" and producing cells from all three germ layers. Proponents of this technique suggest that since it requires only one embryonic cell, the remaining cells may yet be implanted in the womb and develop into a human being. Therefore, scientists could potentially derive human embryonic stem cells without having to destroy an embryo. However, ethical considerations make it uncertain whether scientists will ever test if the cells remaining after removal of a single cell can develop into a human being, at least in embryos that are not at risk for carrying a genetic disorder. Moreover, it is unclear whether the single cell used to generate a pluripotent stem cell line has the capacity to become a human being.

Parthenogenesis is the creation of an embryo without fertilizing the egg with a sperm, thus omitting the sperm's genetic contributions. To achieve this feat, scientists "trick" the egg into believing it is fertilized, so that it will begin to divide and form a blastocyst (see Figure 8.3f). In 2007, Dr. E.S. Revazova and colleagues reported that they successfully used parthenogenesis to derive hESCs.23 These stem cell lines, derived and grown using a human feeder cell layer, retained the genetic information of the egg donor and demonstrated characteristics of pluripotency. This technique may lead to the ability to generate tissue-matched cells for transplantation to treat women who are willing to provide their own egg cells.24 It also offers an alternate method for deriving tissue-matched hESCs that does not require destruction of a fertilized embryo.

Amniotic fluid surrounding the developing fetus contains cells shed by the fetus and is regularly collected from pregnant women during amniocentesis. In 2003, researchers identified a subset of cells in amniotic fluid that express Oct-4, a marker for pluripotent human stem cells that is expressed in ES cells and embryonic germ cells.25 Since then, investigators have shown that amniotic fluid stem cells can differentiate into cells of all three embryonic germ layers and that these cells do not form tumors in vivo.26,27

For example, Anthony Atala and colleagues at the Wake Forest University have recently generated non-embryonic stem cell lines from cells found in human and rat amniotic fluid.27 They named these cells amniotic fluid-derived stem cells (AFS). Experiments demonstrate that AFS can produce cells that originate from each of the three embryonic germ layers, and the self-renewing cells maintained the normal number of chromosomes after a prolonged period in culture. However, undifferentiated AFS did not produce all of the proteins expected of pluripotent cells, and they were not capable of forming a teratoma. The scientists developed in vitro conditions that enabled AFS to produce nerve cells, liver cells, and bone-forming cells. AFS-derived human nerve cells could make proteins typical of specialized nerve cells and were able to integrate into a mouse brain and survive for at least two months. Cultured AFS-derived human liver cells secreted urea and made proteins characteristic of normal human liver cells. Cultured AFS-derived human bone cells made proteins expected of human bone cells and formed bone in mice when seeded onto scaffolds and implanted under the mouse's skin. Although scientists do not yet know how many different cell types AFS can generate, AFS may one day allow researchers to establish a bank of cells for transplantation into humans.

An alternative to searching for an existing population of stem cells is to create a new one from a population of non-pluripotent cells. This strategy, which may or may not involve the creation of an embryo, is known as "reprogramming." This section will summarize reprogramming approaches, including several recent breakthroughs in the field..

In SCNT (see Figure 8.3g), human oocytes (eggs) are collected from a volunteer donor who has taken drugs that stimulate the production of more than one oocyte during the menstrual cycle. Scientists then remove the nucleus from the donated oocyte and replace it with the nucleus from a somatic cell, a differentiated adult cell from elsewhere in the body. The oocyte with the newly-transferred nucleus is then stimulated to develop. The oocyte may develop only if the transplanted nucleus is returned to the pluripotent state by factors present in the oocyte cytoplasm. This alteration in the state of the mature nucleus is called nuclear reprogramming. When development progresses to the blastocyst stage, the ICM is removed and placed into culture in an attempt to establish a pluripotent stem cell line. To date, the technique has been successfully demonstrated in two primates: macaque monkeys28 and humans.29

However, successful SCNT creates an embryo-like entity, thereby raising the ethical issues that confront the use of spare IVF embryos. However, pluripotent cell lines created by embryos generated by SCNT offer several advantages over ES cells. First, the nuclear genes of such a pluripotent cell line will be identical to the genes in the donor nucleus. If the nucleus comes from a cell that carries a mutation underlying a human genetic disease such as Huntington's disease, then all cells derived from the pluripotent cell line will carry this mutation. In this case, the SCNT procedure would enable the development of cellular models of human genetic disease that can inform our understanding of the biology of disease and facilitate development of drugs to slow or halt disease progression. Alternatively, if the cell providing the donor nucleus comes from a specific patient, all cells derived from the resulting pluripotent cell line will be genetically matched to the patient with respect to the nuclear genome. If these cells were used in transplantation therapy, the likelihood that the patient's immune system would recognize the transplanted cells as foreign and initiate tissue rejection would be reduced. However, because mitochondria also contain DNA, the donor oocyte will be the source of the mitochondrial genome, which is likely to carry mitochondrial gene differences from the patient which may still lead to tissue rejection.

A technique reported in 2007 by Dr. Kevin Eggan and colleagues at Harvard University may expand scientists' options when trying to "reprogram" an adult cell's DNA30. Previously, successful SCNT relied upon the use of an unfertilized egg. Now, the Harvard scientists have demonstrated that by using a drug to stop cell division in a fertilized mouse egg (zygote) during mitosis, they can successfully reprogram an adult mouse skin cell by taking advantage of the "reprogramming factors" that are active in the zygote at mitosis. They removed the chromosomes from the single-celled zygote's nucleus and replaced them with the adult donor cell's chromosomes (see Figure 8.3h). The active reprogramming factors present in the zygote turned genes on and off in the adult donor chromosomes, to make them behave like the chromosomes of a normally fertilized zygote. After the zygote was stimulated to divide, the cloned mouse embryo developed to the blastocyst stage, and the scientists were able to harvest embryonic stem cells from the resulting blastocyst. When the scientists applied their new method to abnormal mouse zygotes, they succeeded at reprogramming adult mouse skin cells and harvesting stem cells. If this technique can be repeated with abnormal human zygotes created in excess after IVF procedures, scientists could use them for research instead of discarding them as medical waste.

Altered nuclear transfer is a variation on standard SCNT that proposes to create patient-specific stem cells without destroying an embryo. In ANT, scientists turn off a gene needed for implantation in the uterus (Cdx2) in the patient cell nucleus before it is transferred into the donor egg (see Figure 8.3i). In 2006, Dr. Rudolph Jaenisch and colleagues at MIT demonstrated that ANT can be carried out in mice.31 Mouse ANT entities whose Cdx2 gene is switched off are unable to implant in the uterus and do not survive to birth. Although ANT has been used to create viable stem cell lines capable of producing almost all cell types, the authors point out that this technique must still be tested with monkey and human embryos. Moreover, the manipulation needed to control Cdx2 expression introduces another logistical hurdle that may complicate the use of ANT to derive embryonic stem cells. Proponents of ANT, such as William Hurlbut of the Stanford University Medical Center, suggest that the entity created by ANT is not a true embryo because it cannot implant in the uterus.32, 33 However, the technique is highly controversial, and its ethical implications remain a source of current debate.4,32

In 2005, Kevin Eggan and colleagues at Harvard University reported that they had fused cultured adult human skin cells with hESCs (see Figure 8.3j).36 The resulting "hybrid" cells featured many characteristics of hESCs, including a similar manner of growth and division and the manufacture of proteins typically produced by hESCs. Some factor(s) within the hESCs enabled them to "reprogram" the adult skin cells to behave as hESCs. However, these cells raised a significant technical barrier to clinical use. Because fused cells are tetraploid (they contain four copies of the cellular DNA rather than the normal two copies), scientists would need to develop a method to remove the extra DNA without eliminating their hESC-like properties. The fusion method serves as a useful model system for studying how stem cells "reprogram" adult cells to have properties of pluripotent cells. However, if the reprogramming technique could be carried out without the fusion strategy, a powerful avenue for creating patient-specific stem cells without using human eggs could be developed.

In 2007, two independent research groups published manuscripts that described successful genetic reprogramming of human adult somatic cells into pluripotent human stem cells.34,35 Although some technical limitations remain, this strategy suggests a promising new avenue for generating pluripotent cell lines that can inform drug development, models of disease, and ultimately, transplantation medicine. These experiments, which are discussed below, were breakthroughs because they used adult somatic cells to create pluripotent stem cells that featured hallmarks of ES cells.

In 2006, Shinya Yamanaka and colleagues at Kyoto University reported that they could use a retroviral expression vector to introduce four important stem cell factors into adult mouse cells and reprogram them to behave like ES cells (see Figure 8.3k).37 They called the reprogrammed cells "iPSCs," for induced pluripotent stem cells. However, iPSCs produced using the original technique failed to produce sperm and egg cells when injected into an early mouse blastocyst and did not make certain critical DNA changes. These researchers then modified the technique to select for iPSCs that can produce sperm and eggs,38 results that have since been reproduced by Rudolph Jaenisch and colleagues at the Massachusetts Institute of Technology (MIT).39

In addition, the MIT scientists determined that iPSCs DNA is modified in a manner similar to ES cells, and important stem cell genes are expressed at similar levels. They also demonstrated that iPSCs injected into an early mouse blastocyst can produce all cell types within the developing embryo, and such embryos can complete gestation and are born alive.

Once these research advances were made in mice, they suggested that similar techniques might be used to reprogram adult human cells. In 2007, Yamanaka and coworkers reported that introducing the same four genetic factors that reprogrammed the mouse cells into adult human dermal fibroblasts reprogrammed the cells into human iPSCs.35 These iPSCs were similar to human ES cells in numerous ways, including morphology, proliferative capacity, expression of cell surface antigens, and gene expression. Moreover, the cells could differentiate into cell types from the three embryonic germ layers both in vitro and in teratoma assays. Concurrent with the Yamanaka report, James Thomson and coworkers at the University of Wisconsin published a separate manuscript that detailed the creation of human iPSCs through somatic cell reprogramming using four genetic factors (two of which were in common with the Yamanaka report).34 The cells generated by the Thomson group met all defining criteria for ES cells, with the exception that they were not derived from embryos.

These breakthroughs have spurred interest in the field of iPSCs research. In early 2008, investigators at the Massachusetts General Hospital40 and the University of California, Los Angeles41 reported generating reprogrammed cells. As scientists explore the mechanisms that govern reprogramming, it is anticipated that more reports will be forthcoming in this emerging area. Although these reprogramming methods require the use of a virus, non-viral strategies may also be possible in the future. In any case, these approaches have created powerful new tools to enable the "dedifferentation" of cells that scientists had previously believed to be terminally differentiated.42,43

Although further study is warranted to determine if iPS and ES cells differ in clinically significant ways, these breakthrough reports suggest that reprogramming is a promising strategy for future clinical applications. Induced pluripotent cells offer the obvious advantage that they are not derived from embryonic tissues, thereby circumventing the ethical issues that surround use of these materials. Successful reprogramming of adult somatic cells could also lead to the development of stem cell lines from patients who suffer from genetically-based diseases, such as Huntington's Disease, spinal muscular atrophy, muscular dystrophy, and thalessemia. These lines would be invaluable research tools to understand the mechanisms of these diseases and to test potential drug treatments. Additionally, reprogrammed cells could potentially be used to repair damaged tissues; patient-specific cell lines could greatly reduce the concerns of immune rejection that are prevalent with many transplantation strategies.

However, several technical hurdles must be overcome before iPSCs can be used in humans. For example, in preliminary experiments with mice, the virus used to introduce the stem cell factors sometimes caused cancers.37 The viral vectors used in these experiments will have to be selected carefully and tested fully to verify that they do not integrate into the genome, thereby harboring the potential to introduce genetic mutations at their site of insertion. This represents a significant concern that must be addressed before the technique can lead to useful treatments for humans. However, this strategy identifies a method for creating pluripotent stem cells that, together with studies of other types of pluripotent stem cells, will help researchers learn how to reprogram cells to repair damaged tissues in the human body.

Stem cell research is a rapidly evolving field, and researchers continue to isolate new pluripotent cells and create additional cell lines. This section briefly reviews other sources of pluripotent cells and the implications that their discovery may have on future research.

Epiblast Cells. While rodent and human ES cells are pluripotent, they maintain their respective pluripotencies through different molecular signaling pathways. It is not known why these differences exist. Recently, several research groups have reported the generation of stable, pluripotent cell lines from mouse and rat epiblast, a tissue of the post-implantation embryo that ultimately generates the embryo proper.44,45 These cells are distinct from mouse ES cells in terms of the signals that control their differentiation. However, the cells share patterns of gene expression and signaling responses with human ES cells. The establishment of epiblast cell lines can therefore provide insight into the distinctions between pluripotent cells from different species and illuminate ways that pluripotent cells pursue distinct fates during early development.

Existing Adult Stem Cells. As has been discussed in other chapters, numerous types of precursor cells have been isolated in adult tissues.46 Although these cells tend to be relatively rare and are dispersed throughout the tissues, they hold great potential for clinical application and tissue engineering. For example, tissues created using stem cells harvested from an adult patient could theoretically be used clinically in that patient without engendering an immune response. Moreover, the use of adult stem cells avoids the ethical concerns associated with the use of ES cells. In addition, adult-derived stem cells do not spontaneously differentiate as do ES cells, thus eliminating the formation of teratomas often seen with implantation of ES cells. The potential of adult stem cells for regenerative medicine is great; it is likely that these various cells will find clinical application in the upcoming decades.

Although the recent advances in reprogramming of adult somatic cells has generated a wave of interest in the scientific community, these cell lines will not likely replace hESC lines as tools for research and discovery. Rather, both categories of cells will find unique uses in the study of stem cell biology and the development and evaluation of therapeutic strategies. Pluripotent cells offer a number of potential clinical applications, especially for diseases with a genetic basis. However, researchers are just beginning to unlock the many factors that govern the cells' growth and differentiation. As scientists make strides toward understanding how these cells can be manipulated, additional applications, approaches, and techniques will likely emerge. As such, pluripotent cells will play a pivotal role in future research into the biology of development and the treatment of disease.

Chapter7|Table of Contents|Chapter9

Excerpt from:Alternate Methods for Preparing Pluripotent Stem Cells ...

See more here:
Massachusetts Stem Cells | Stem Cell TV

Posted in Massachusetts Stem Cells | Comments Off on Massachusetts Stem Cells | Stem Cell TV

Alternate Methods for Preparing Pluripotent Stem Cells …

Posted: September 25, 2017 at 3:48 pm

James F. Battey, Jr., MD, PhD; Laura K. Cole, PhD; and Charles A. Goldthwaite, Jr., PhD.

Stem cells are distinguished from other cells by two characteristics: (1) they can divide to produce copies of themselves (self-renewal) under appropriate conditions and (2) they are pluripotent, or able to differentiate into any of the three germ layers: the endoderm (which forms the lungs, gastrointestinal tract, and interior lining of the stomach), mesoderm (which forms the bones, muscles, blood, and urogenital tract), and ectoderm (which forms the epidermal tissues and nervous system). Pluripotent cells, which can differentiate into any mature cell type, are distinct from multipotent cells (such as hematopoietic, or blood-forming, cells) that can differ into a limited number of mature cell types. Because of their pluripotency and capacity for self-renewal, stem cells hold great potential to renew tissues that have been damaged by conditions such as type 1 diabetes, Parkinson's disease, heart attacks, and spinal cord injury. Although techniques to transplant multipotent or pluripotent cells are being developed for many specific applications, some procedures are sufficiently mature to be established options for care. For example, human hematopoietic cells from the umbilical cord and bone marrow are currently being used to treat patients with disorders that require replacement of cells made by the bone marrow, including Fanconi's anemia and chemotherapy-induced bone marrow failure after cancer treatment.

However, differentiation is influenced by numerous factors, and investigators are just beginning to understand the fundamental properties of human pluripotent cells. Researchers are gradually learning how to direct these cells to differentiate into specialized cell types and to use them for research, drug discovery, and transplantation therapy (see Figure 8.1). However, before stem cell derivatives are suitable for clinical application, scientists require a more complete understanding of the molecular mechanisms that drive pluripotent cells into differentiated cells. Scientists will need to pilot experimental transplantation therapies in animal model systems to assess the safety and long-term stable functioning of transplanted cells. In particular, they must be certain that any transplanted cells do not continue to self-renew in an unregulated fashion after transplantation, which may result in a teratoma, or stem cell tumor. In addition, scientists must ascertain that cells transplanted into a patient are not recognized as foreign by the patient's immune system and rejected.

Figure 8.1. The Scientific Challenge of Human Stem CellsThe state of the science currently lies in the development of fundamental knowledge of the properties of human pluripotent cells. The scientific capacity needs to be built, an understanding of the molecular mechanisms that drive cell specialization needs to be advanced, the nature and regulation of interaction between host and transplanted cells needs to be explored and understood, cell division needs to be understood and regulated, and the long-term stability of the function in transplanted cells needs to be established.

Stem cells derived from an early-stage human blastocyst (an embryo fertilized in vitro and grown approximately five days in culture) have the capacity to renew indefinitely, and can theoretically provide an unlimited supply of cells. It is also possible to derive stem cells from non-embryonic tissues, including amniotic fluid, placenta, umbilical cord, brain, gut, bone marrow, and liver. These stem cells are sometimes called "adult" stem cells, and they are typically rare in the tissue of origin. For example, blood-forming (hematopoietic) stem cell experts estimate that only 1 in 2000 to fewer than 1 in 10,000 cells found in the bone marrow is actually a stem cell.1 Because so-called "adult" stem cells include cells from the placenta and other early stages of development, they are more correctly termed "non-embryonic stem cells." Non-embryonic stem cells are more limited in their capacity to self renew in the laboratory, making it more difficult to generate a large number of stem cells for a specific experimental or therapeutic application. Under normal conditions, non-embryonic stem cells serve as a repair pool for the body, so they typically differentiate only into the cell types found in the organ of origin. Moreover, there is little compelling evidence for trans-differentiation, whereby a stem cell from one organ differentiates into a mature cell type of a different organ. New discoveries may overcome these limitations of stem cells derived from non-embryonic sources, and research directed toward this goal is currently underway in a number of laboratories.

Cultures of human pluripotent, self-renewing cells enable researchers to understand the molecular mechanisms that regulate differentiation (see Figure 8.2), including epigenetic changes (traits that may be inherited that do not arise from changes in the DNA sequence) in the chromatin structure, developmental changes in gene expression, exposure to growth factors, and interactions between adjacent cells. Understanding these basic mechanisms may enable future scientists to mobilize and differentiate endogenous populations of pluripotent cells to replace a cell type ravaged by injury or disease. Alternatively, scientists may some day be able to coax human pluripotent cells grown in the laboratory to become a specific type of specialized cell, which physicians could subsequently transplant into a patient to replace cells damaged by these same disease processes.

Scientists are gradually learning to direct the differentiation of pluripotent cell cultures into a specific type of cell, which can then be used as cellular models of human disease for drug discovery or toxicity studies. While it is not possible to predict the myriad ways that a basic understanding of stem cell differentiation may lead to new approaches for treating patients with cellular degenerative diseases, some avenues can be theorized. For example, in the case of Huntington's disease, a fatal neurodegenerative disorder, one could imagine that pluripotent cells derived from an embryo that carries Huntington's disease and differentiated into neurons in culture could be used to test drugs to delay or prevent degeneration.

Despite the incredible growth in knowledge that has occurred in stem cell research within the last couple of decades, investigators are just beginning to unravel the process of differentiation. Human pluripotent cell lines are an essential tool to understand this process and to facilitate the ultimate use of these cells in the clinic. To provide background on this fundamental topic, this article reviews the various potential sources and approaches that have been used to generate human pluripotent and multipotent cell lines, both of embryonic and non-embryonic origin.

Currently, at least six embryonic sources have been used to establish human pluripotent stem cell lines. All approaches involve isolation of viable cells during an early phase of development, followed by growth of these cells in appropriate culture medium. The various sources of these initial cell populations are discussed in brief below. It should be noted that the manipulation and use of embryonic tissues has raised a number of ethical issues.2,3 This article focuses on the scientific and technical issues associated with creating pluripotent cells, with the understanding that some of these techniques are currently subject to debates that extend beyond discussions of their scientific merits.

Figure 8.2. The Promise of Stem Cell ResearchStem cell research provides a useful tool for unraveling the molecular mechanisms that determine the differentiation fate of a pluripotent cell and for understanding the gene expression properties and epigenetic modifications essential to maintain the pluripotent state. In the future, this knowledge may be used to generate cells for transplantation therapies, whereby a specific cell population compromised by disease is replaced with new, functional cells. Differentiated derivatives of human pluripotent cells may also prove to be useful as models for understanding the biology of disease and developing new drugs, particularly when there is no animal model for the disease being studied. The greatest promise of stem cell research may lie in an area not yet imagined.

2008 Terese Winslow

Drawing upon twenty years of communal expertise with mouse ES cells,4 and on human inner cell mass culture conditions developed by Ariff Bongso and colleagues,5 James Thomson and colleagues at the University of Wisconsin generated the first hESC lines in 1998 using tissue from embryos fertilized in vitro.6 This method uses embryos generated for in vitro fertilization (IVF) that are no longer needed for reproductive purposes. During IVF, medical professionals usually produce more embryos than a couple attempting to start a family may need. Spare embryos are typically stored in a freezer to support possible future attempts for additional children if desired. It is estimated that there are approximately 400,000 such spare embryos worldwide.6 If these embryos are never used by the couple, they either remain in storage or are discarded as medical waste. Alternatively, these embryos can potentially be used to generate a hESC line.

To generate a hESC line, scientists begin with a donated blastocyst-stage embryo, at approximately five days after IVF (see Figure 8.3a). The blastocyst consists of approximately 150200 cells that form a hollow sphere of cells, the outer layer of which is called the trophectoderm. During normal development, the trophoblast becomes the placenta and umbilical cord. At one pole of this hollow sphere, 3050 cells form a cluster that is called the inner cell mass (ICM), which would give rise to the developing fetus. ICM cells are pluripotent, possessing the capacity to become any of the several hundred specialized cell types found in a developed human, with the exception of the placenta and umbilical cord.

Scientists remove the ICM from the donated blastocyst and place these cells into a specialized culture medium. In approximately one in five attempts, a hESC line begins to grow. Stem cells grown in such a manner can then be directed to differentiate into various lineages, including neural precursor cells,8 cardiomyocytes,9 and hematopoietic (blood forming) precursor cells.10

However, hESC lines are extremely difficult to grow in culture; the cells require highly specialized growth media that contain essential ingredients that are difficult to standardize. Yet the culture conditions are critical to maintain the cells' self-renewing and pluripotent properties. Culture requires the support of mouse or human cells, either directly as a "feeder" cell layer6,11,12 or indirectly as a source of conditioned medium in feeder-free culture systems.13 The feeder cells secrete important nutrients and otherwise support stem cell growth, but are treated so they cannot divide. Although the complete role of these feeder cells is not known, they promote stem cell growth, including detoxifying the culture medium and secreting proteins that participate in cell growth.14 hESC lines used to produce human cells for transplantation therapies may need to be propagated on a human feeder cell layer to reduce the risk of contamination by murine viruses or other proteins that may cause rejection. Thus, hESC lines often grow only under highly specific culture conditions, and the identification of ideal growth conditions presents a challenge regardless of the source of the hESCs.

Furthermore, human ES cell cultures must be expanded using an exacting protocol to avoid cell death and to control spontaneous differentiation. Since a limited number of laboratories in the United States are growing these cells, there is a shortage of people well-versed in the art and science of successful hESC culture. In the short term, challenges of working with these cells include developing robust culture conditions and protocols, understanding the molecular mechanisms that direct differentiation into specific cell types, and developing the infrastructure to advance this scientific opportunity. Once these challenges have been met, scientists will need to conduct transplantation studies in animal models (rodent and non-human primates) to demonstrate safety, effectiveness, and long-term benefit before stem cell therapies may enter clinical trials.

A second method for generating human pluripotent stem cell lines was published in 1998 by John Gearhart and coworkers at The Johns Hopkins Medical School.15

These researchers isolated specialized cells known as primordial germ cells (PGCs) from a 57-week-old embryo and placed these cells into culture (see Figure 8.3b). PGCs are destined to become either oocytes or sperm cells, depending on the sex of the developing embryo. The resulting cell lines are called embryonic germ cell lines, and they share many properties with ES cells. As with ES cells, however, PGCs present challenges with sustained growth in culture.16,17 Spontaneous differentiation, which hinders the isolation of pure clonal lines, is a particular issue. Therefore, the clinical application of these cells requires a more complete understanding of their derivation and maintenance in vitro.

Embryos that stop dividing after being fertilized in vitro are not preferentially selected for implantation in a woman undergoing fertility treatment. These embryos are typically either frozen for future use or discarded as medical waste. In 2006, scientists at the University of Newcastle, United Kingdom, generated hESC lines from IVF embryos that had stopped dividing.18 These scientists used similar methods as described under "Traditional hESC Line Generation" except that their source material was so-called "dead" IVF embryos (see Figure 8.3c). The human stem cells created using this technique behaved like pluripotent stem cells, including producing proteins critical for "stemness" and being able to produce cells from all three germ layers. It has been proposed that an IVF embryo can be considered dead when it ceases to divide.19 If one accepts this definition, such an embryo that "dies" from natural causes presumably cannot develop into a human being, thereby providing a source to derive human ES cells without destroying a living embryo.

Figure 8.3. Alternative Methods for Preparing Pluripotent Stem Cells

2008 Terese Winslow

Couples who have learned that they carry a genetic disorder sometimes use pre-implantation genetic diagnosis (PGD) and IVF to have a child that does not carry the disorder. PGD requires scientists to remove one cell from a very early IVF human embryo and test it for diseases known to be carried by the hopeful couple. Normally, embryos identified with genetic disorders are discarded as medical waste. However, Dr.Yuri Verlinsky and colleagues have capitalized on these embryos as a way to further our understanding of the diseases they carry (see Figure 8.3d) by deriving hESC lines from them.20 These stem cell lines can then be used to help scientists understand genetically-based disorders such as muscular dystrophy, Huntington's disease, thalessemia, Fanconi's anemia, Marfan syndrome, adrenoleukodystrophy, and neurofibromatosis.

In 2006, Dr. Robert Lanza and colleagues demonstrated that it is possible to remove a single cell from a pre-implantation mouse embryo and generate a mouse ES cell line.21 This work was based upon their experience with cleavage-stage mouse embryos. Later that same year, Dr. Lanza's laboratory reported that it had successfully established hESC lines (see Figure 8.3e) from single cells taken from pre-implantation human embryos.22 The human stem cells created using this technique behaved like pluripotent stem cells, including making proteins critical for "stemness" and producing cells from all three germ layers. Proponents of this technique suggest that since it requires only one embryonic cell, the remaining cells may yet be implanted in the womb and develop into a human being. Therefore, scientists could potentially derive human embryonic stem cells without having to destroy an embryo. However, ethical considerations make it uncertain whether scientists will ever test if the cells remaining after removal of a single cell can develop into a human being, at least in embryos that are not at risk for carrying a genetic disorder. Moreover, it is unclear whether the single cell used to generate a pluripotent stem cell line has the capacity to become a human being.

Parthenogenesis is the creation of an embryo without fertilizing the egg with a sperm, thus omitting the sperm's genetic contributions. To achieve this feat, scientists "trick" the egg into believing it is fertilized, so that it will begin to divide and form a blastocyst (see Figure 8.3f). In 2007, Dr. E.S. Revazova and colleagues reported that they successfully used parthenogenesis to derive hESCs.23 These stem cell lines, derived and grown using a human feeder cell layer, retained the genetic information of the egg donor and demonstrated characteristics of pluripotency. This technique may lead to the ability to generate tissue-matched cells for transplantation to treat women who are willing to provide their own egg cells.24 It also offers an alternate method for deriving tissue-matched hESCs that does not require destruction of a fertilized embryo.

Amniotic fluid surrounding the developing fetus contains cells shed by the fetus and is regularly collected from pregnant women during amniocentesis. In 2003, researchers identified a subset of cells in amniotic fluid that express Oct-4, a marker for pluripotent human stem cells that is expressed in ES cells and embryonic germ cells.25 Since then, investigators have shown that amniotic fluid stem cells can differentiate into cells of all three embryonic germ layers and that these cells do not form tumors in vivo.26,27

For example, Anthony Atala and colleagues at the Wake Forest University have recently generated non-embryonic stem cell lines from cells found in human and rat amniotic fluid.27 They named these cells amniotic fluid-derived stem cells (AFS). Experiments demonstrate that AFS can produce cells that originate from each of the three embryonic germ layers, and the self-renewing cells maintained the normal number of chromosomes after a prolonged period in culture. However, undifferentiated AFS did not produce all of the proteins expected of pluripotent cells, and they were not capable of forming a teratoma. The scientists developed in vitro conditions that enabled AFS to produce nerve cells, liver cells, and bone-forming cells. AFS-derived human nerve cells could make proteins typical of specialized nerve cells and were able to integrate into a mouse brain and survive for at least two months. Cultured AFS-derived human liver cells secreted urea and made proteins characteristic of normal human liver cells. Cultured AFS-derived human bone cells made proteins expected of human bone cells and formed bone in mice when seeded onto scaffolds and implanted under the mouse's skin. Although scientists do not yet know how many different cell types AFS can generate, AFS may one day allow researchers to establish a bank of cells for transplantation into humans.

An alternative to searching for an existing population of stem cells is to create a new one from a population of non-pluripotent cells. This strategy, which may or may not involve the creation of an embryo, is known as "reprogramming." This section will summarize reprogramming approaches, including several recent breakthroughs in the field..

In SCNT (see Figure 8.3g), human oocytes (eggs) are collected from a volunteer donor who has taken drugs that stimulate the production of more than one oocyte during the menstrual cycle. Scientists then remove the nucleus from the donated oocyte and replace it with the nucleus from a somatic cell, a differentiated adult cell from elsewhere in the body. The oocyte with the newly-transferred nucleus is then stimulated to develop. The oocyte may develop only if the transplanted nucleus is returned to the pluripotent state by factors present in the oocyte cytoplasm. This alteration in the state of the mature nucleus is called nuclear reprogramming. When development progresses to the blastocyst stage, the ICM is removed and placed into culture in an attempt to establish a pluripotent stem cell line. To date, the technique has been successfully demonstrated in two primates: macaque monkeys28 and humans.29

However, successful SCNT creates an embryo-like entity, thereby raising the ethical issues that confront the use of spare IVF embryos. However, pluripotent cell lines created by embryos generated by SCNT offer several advantages over ES cells. First, the nuclear genes of such a pluripotent cell line will be identical to the genes in the donor nucleus. If the nucleus comes from a cell that carries a mutation underlying a human genetic disease such as Huntington's disease, then all cells derived from the pluripotent cell line will carry this mutation. In this case, the SCNT procedure would enable the development of cellular models of human genetic disease that can inform our understanding of the biology of disease and facilitate development of drugs to slow or halt disease progression. Alternatively, if the cell providing the donor nucleus comes from a specific patient, all cells derived from the resulting pluripotent cell line will be genetically matched to the patient with respect to the nuclear genome. If these cells were used in transplantation therapy, the likelihood that the patient's immune system would recognize the transplanted cells as foreign and initiate tissue rejection would be reduced. However, because mitochondria also contain DNA, the donor oocyte will be the source of the mitochondrial genome, which is likely to carry mitochondrial gene differences from the patient which may still lead to tissue rejection.

A technique reported in 2007 by Dr. Kevin Eggan and colleagues at Harvard University may expand scientists' options when trying to "reprogram" an adult cell's DNA30. Previously, successful SCNT relied upon the use of an unfertilized egg. Now, the Harvard scientists have demonstrated that by using a drug to stop cell division in a fertilized mouse egg (zygote) during mitosis, they can successfully reprogram an adult mouse skin cell by taking advantage of the "reprogramming factors" that are active in the zygote at mitosis. They removed the chromosomes from the single-celled zygote's nucleus and replaced them with the adult donor cell's chromosomes (see Figure 8.3h). The active reprogramming factors present in the zygote turned genes on and off in the adult donor chromosomes, to make them behave like the chromosomes of a normally fertilized zygote. After the zygote was stimulated to divide, the cloned mouse embryo developed to the blastocyst stage, and the scientists were able to harvest embryonic stem cells from the resulting blastocyst. When the scientists applied their new method to abnormal mouse zygotes, they succeeded at reprogramming adult mouse skin cells and harvesting stem cells. If this technique can be repeated with abnormal human zygotes created in excess after IVF procedures, scientists could use them for research instead of discarding them as medical waste.

Altered nuclear transfer is a variation on standard SCNT that proposes to create patient-specific stem cells without destroying an embryo. In ANT, scientists turn off a gene needed for implantation in the uterus (Cdx2) in the patient cell nucleus before it is transferred into the donor egg (see Figure 8.3i). In 2006, Dr. Rudolph Jaenisch and colleagues at MIT demonstrated that ANT can be carried out in mice.31 Mouse ANT entities whose Cdx2 gene is switched off are unable to implant in the uterus and do not survive to birth. Although ANT has been used to create viable stem cell lines capable of producing almost all cell types, the authors point out that this technique must still be tested with monkey and human embryos. Moreover, the manipulation needed to control Cdx2 expression introduces another logistical hurdle that may complicate the use of ANT to derive embryonic stem cells. Proponents of ANT, such as William Hurlbut of the Stanford University Medical Center, suggest that the entity created by ANT is not a true embryo because it cannot implant in the uterus.32, 33 However, the technique is highly controversial, and its ethical implications remain a source of current debate.4,32

In 2005, Kevin Eggan and colleagues at Harvard University reported that they had fused cultured adult human skin cells with hESCs (see Figure 8.3j).36 The resulting "hybrid" cells featured many characteristics of hESCs, including a similar manner of growth and division and the manufacture of proteins typically produced by hESCs. Some factor(s) within the hESCs enabled them to "reprogram" the adult skin cells to behave as hESCs. However, these cells raised a significant technical barrier to clinical use. Because fused cells are tetraploid (they contain four copies of the cellular DNA rather than the normal two copies), scientists would need to develop a method to remove the extra DNA without eliminating their hESC-like properties. The fusion method serves as a useful model system for studying how stem cells "reprogram" adult cells to have properties of pluripotent cells. However, if the reprogramming technique could be carried out without the fusion strategy, a powerful avenue for creating patient-specific stem cells without using human eggs could be developed.

In 2007, two independent research groups published manuscripts that described successful genetic reprogramming of human adult somatic cells into pluripotent human stem cells.34,35 Although some technical limitations remain, this strategy suggests a promising new avenue for generating pluripotent cell lines that can inform drug development, models of disease, and ultimately, transplantation medicine. These experiments, which are discussed below, were breakthroughs because they used adult somatic cells to create pluripotent stem cells that featured hallmarks of ES cells.

In 2006, Shinya Yamanaka and colleagues at Kyoto University reported that they could use a retroviral expression vector to introduce four important stem cell factors into adult mouse cells and reprogram them to behave like ES cells (see Figure 8.3k).37 They called the reprogrammed cells "iPSCs," for induced pluripotent stem cells. However, iPSCs produced using the original technique failed to produce sperm and egg cells when injected into an early mouse blastocyst and did not make certain critical DNA changes. These researchers then modified the technique to select for iPSCs that can produce sperm and eggs,38 results that have since been reproduced by Rudolph Jaenisch and colleagues at the Massachusetts Institute of Technology (MIT).39

In addition, the MIT scientists determined that iPSCs DNA is modified in a manner similar to ES cells, and important stem cell genes are expressed at similar levels. They also demonstrated that iPSCs injected into an early mouse blastocyst can produce all cell types within the developing embryo, and such embryos can complete gestation and are born alive.

Once these research advances were made in mice, they suggested that similar techniques might be used to reprogram adult human cells. In 2007, Yamanaka and coworkers reported that introducing the same four genetic factors that reprogrammed the mouse cells into adult human dermal fibroblasts reprogrammed the cells into human iPSCs.35 These iPSCs were similar to human ES cells in numerous ways, including morphology, proliferative capacity, expression of cell surface antigens, and gene expression. Moreover, the cells could differentiate into cell types from the three embryonic germ layers both in vitro and in teratoma assays. Concurrent with the Yamanaka report, James Thomson and coworkers at the University of Wisconsin published a separate manuscript that detailed the creation of human iPSCs through somatic cell reprogramming using four genetic factors (two of which were in common with the Yamanaka report).34 The cells generated by the Thomson group met all defining criteria for ES cells, with the exception that they were not derived from embryos.

These breakthroughs have spurred interest in the field of iPSCs research. In early 2008, investigators at the Massachusetts General Hospital40 and the University of California, Los Angeles41 reported generating reprogrammed cells. As scientists explore the mechanisms that govern reprogramming, it is anticipated that more reports will be forthcoming in this emerging area. Although these reprogramming methods require the use of a virus, non-viral strategies may also be possible in the future. In any case, these approaches have created powerful new tools to enable the "dedifferentation" of cells that scientists had previously believed to be terminally differentiated.42,43

Although further study is warranted to determine if iPS and ES cells differ in clinically significant ways, these breakthrough reports suggest that reprogramming is a promising strategy for future clinical applications. Induced pluripotent cells offer the obvious advantage that they are not derived from embryonic tissues, thereby circumventing the ethical issues that surround use of these materials. Successful reprogramming of adult somatic cells could also lead to the development of stem cell lines from patients who suffer from genetically-based diseases, such as Huntington's Disease, spinal muscular atrophy, muscular dystrophy, and thalessemia. These lines would be invaluable research tools to understand the mechanisms of these diseases and to test potential drug treatments. Additionally, reprogrammed cells could potentially be used to repair damaged tissues; patient-specific cell lines could greatly reduce the concerns of immune rejection that are prevalent with many transplantation strategies.

However, several technical hurdles must be overcome before iPSCs can be used in humans. For example, in preliminary experiments with mice, the virus used to introduce the stem cell factors sometimes caused cancers.37 The viral vectors used in these experiments will have to be selected carefully and tested fully to verify that they do not integrate into the genome, thereby harboring the potential to introduce genetic mutations at their site of insertion. This represents a significant concern that must be addressed before the technique can lead to useful treatments for humans. However, this strategy identifies a method for creating pluripotent stem cells that, together with studies of other types of pluripotent stem cells, will help researchers learn how to reprogram cells to repair damaged tissues in the human body.

Stem cell research is a rapidly evolving field, and researchers continue to isolate new pluripotent cells and create additional cell lines. This section briefly reviews other sources of pluripotent cells and the implications that their discovery may have on future research.

Epiblast Cells. While rodent and human ES cells are pluripotent, they maintain their respective pluripotencies through different molecular signaling pathways. It is not known why these differences exist. Recently, several research groups have reported the generation of stable, pluripotent cell lines from mouse and rat epiblast, a tissue of the post-implantation embryo that ultimately generates the embryo proper.44,45 These cells are distinct from mouse ES cells in terms of the signals that control their differentiation. However, the cells share patterns of gene expression and signaling responses with human ES cells. The establishment of epiblast cell lines can therefore provide insight into the distinctions between pluripotent cells from different species and illuminate ways that pluripotent cells pursue distinct fates during early development.

Existing Adult Stem Cells. As has been discussed in other chapters, numerous types of precursor cells have been isolated in adult tissues.46 Although these cells tend to be relatively rare and are dispersed throughout the tissues, they hold great potential for clinical application and tissue engineering. For example, tissues created using stem cells harvested from an adult patient could theoretically be used clinically in that patient without engendering an immune response. Moreover, the use of adult stem cells avoids the ethical concerns associated with the use of ES cells. In addition, adult-derived stem cells do not spontaneously differentiate as do ES cells, thus eliminating the formation of teratomas often seen with implantation of ES cells. The potential of adult stem cells for regenerative medicine is great; it is likely that these various cells will find clinical application in the upcoming decades.

Although the recent advances in reprogramming of adult somatic cells has generated a wave of interest in the scientific community, these cell lines will not likely replace hESC lines as tools for research and discovery. Rather, both categories of cells will find unique uses in the study of stem cell biology and the development and evaluation of therapeutic strategies. Pluripotent cells offer a number of potential clinical applications, especially for diseases with a genetic basis. However, researchers are just beginning to unlock the many factors that govern the cells' growth and differentiation. As scientists make strides toward understanding how these cells can be manipulated, additional applications, approaches, and techniques will likely emerge. As such, pluripotent cells will play a pivotal role in future research into the biology of development and the treatment of disease.

Chapter7|Table of Contents|Chapter9

Excerpt from:
Alternate Methods for Preparing Pluripotent Stem Cells ...

Posted in Massachusetts Stem Cells | Comments Off on Alternate Methods for Preparing Pluripotent Stem Cells …

Pediatric brain tumors traced to brain stem cells

Posted: July 10, 2012 at 8:14 am

ScienceDaily (July 9, 2012) Scientists showed in mice that disabling a gene linked to a common pediatric tumor disorder, neurofibromatosis type 1 (NF1), made stem cells from one part of the brain proliferate rapidly. But the same genetic deficit had no effect on stem cells from another brain region.

The results can be explained by differences in the way stem cells from these regions of the brain respond to cancer-causing genetic changes.

NF1 is among the world's most common genetic disorders, occurring in about one of every 3,000 births. It causes a wide range of symptoms, including brain tumors, learning disabilities and attention deficits.

Brain tumors in children with NF1 typically arise in the optic nerve and do not necessarily require treatment. If optic gliomas keep growing, though, they can threaten the child's vision. By learning more about the many factors that contribute to NF1 tumor formation, scientists hope to develop more effective treatments.

"To improve therapy, we need to develop better ways to identify and group tumors based not just on the way they look under the microscope, but also on innate properties of their stem cell progenitors," says David H. Gutmann, MD, PhD, the Donald O. Schnuck Family Professor of Neurology.

The study appears July 9 in Cancer Cell. Gutmann also is the director of the Washington University Neurofibromatosis Center.

In the new study, researchers compared brain stem cells from two primary sources: the third ventricle, located in the midbrain, and the nearby lateral ventricles. Before birth and for a time afterward, both of these areas in the brain are lined with growing stem cells.

First author Da Yong Lee, PhD, a postdoctoral research associate, showed that the cells lining both ventricles are true stem cells capable of becoming nerve and support cells (glia) in the brain. Next, she conducted a detailed analysis of gene expression in both stem cell types.

"There are night-and-day differences between these two groups of stem cells," Gutmann says. "These results show that stem cells are not the same everywhere in the brain, which has real consequences for human neurologic disease."

The third ventricle is close to the optic chiasm, the point where the optic nerves cross and optic gliomas develop in NF1 patients. Lee and Gutmann postulated that stem cells from this ventricle might be the source of progenitor cells that can become gliomas in children with NF1.

Read more from the original source:
Pediatric brain tumors traced to brain stem cells

Posted in Stem Cell Videos | Comments Off on Pediatric brain tumors traced to brain stem cells

Pediatric brain tumors traced to brain stem cells

Posted: July 10, 2012 at 3:14 am

ScienceDaily (July 9, 2012) Scientists showed in mice that disabling a gene linked to a common pediatric tumor disorder, neurofibromatosis type 1 (NF1), made stem cells from one part of the brain proliferate rapidly. But the same genetic deficit had no effect on stem cells from another brain region.

The results can be explained by differences in the way stem cells from these regions of the brain respond to cancer-causing genetic changes.

NF1 is among the world's most common genetic disorders, occurring in about one of every 3,000 births. It causes a wide range of symptoms, including brain tumors, learning disabilities and attention deficits.

Brain tumors in children with NF1 typically arise in the optic nerve and do not necessarily require treatment. If optic gliomas keep growing, though, they can threaten the child's vision. By learning more about the many factors that contribute to NF1 tumor formation, scientists hope to develop more effective treatments.

"To improve therapy, we need to develop better ways to identify and group tumors based not just on the way they look under the microscope, but also on innate properties of their stem cell progenitors," says David H. Gutmann, MD, PhD, the Donald O. Schnuck Family Professor of Neurology.

The study appears July 9 in Cancer Cell. Gutmann also is the director of the Washington University Neurofibromatosis Center.

In the new study, researchers compared brain stem cells from two primary sources: the third ventricle, located in the midbrain, and the nearby lateral ventricles. Before birth and for a time afterward, both of these areas in the brain are lined with growing stem cells.

First author Da Yong Lee, PhD, a postdoctoral research associate, showed that the cells lining both ventricles are true stem cells capable of becoming nerve and support cells (glia) in the brain. Next, she conducted a detailed analysis of gene expression in both stem cell types.

"There are night-and-day differences between these two groups of stem cells," Gutmann says. "These results show that stem cells are not the same everywhere in the brain, which has real consequences for human neurologic disease."

The third ventricle is close to the optic chiasm, the point where the optic nerves cross and optic gliomas develop in NF1 patients. Lee and Gutmann postulated that stem cells from this ventricle might be the source of progenitor cells that can become gliomas in children with NF1.

View original post here:
Pediatric brain tumors traced to brain stem cells

Posted in Stem Cells | Comments Off on Pediatric brain tumors traced to brain stem cells

These 3 Stocks Are the Future of Biotechnology – The Motley Fool

Posted: September 8, 2022 at 1:54 am

The biotech industry is heavily regulated by the federal government and its science watchdog, the Food and Drug Administration (FDA). As a consequence, biotech investors often evaluate a possible investment by looking at the company's pipeline of future assets. Research and development is a key factor in this sector.

So what biotech companies have the most exciting future prospects? Three Fool.com healthcare contributors like Vertex Pharmaceuticals (VRTX 1.88%), Repligen (RGEN 4.51%), and Recursion Pharmaceuticals (RXRX 9.43%). Here's why they believe these three stocks are the future of biotechnology.

George Budwell (Vertex Pharmaceuticals): Vertex Pharmaceuticals has become a cash-printing machine recently thanks to its groundbreaking cystic fibrosis treatments like Trikafta. This single drug generated a whopping $5.7 billion in sales in 2021.

The company has been using the proceeds from its mega-blockbuster cystic fibrosis franchise to expand into cutting-edge treatment modalities such as stem cell therapy and gene editing. For example, Vertex dipped into its cash reserves in the recent past to buy the privately held biotechs Semma Therapeutics and ViaCyte. These complementary transactions have suddenly made Vertex a potential juggernaut in the world of diabetes care.

The 2019 Semma Therapeutics deal gave the biotech the allogeneic stem-cell product VX-880, aimed at curing type 1 diabetes. And earlier this year, Vertex bought ViaCyte for its cell replacement therapy platform, which is also initially aimed at curing type 1 diabetes.

This ViaCyte transaction also stands out because the biotech has an ongoing collaboration with Vertex's blood disease collaborator, CRISPR Therapeutics. This ViaCyte/CRISPR partnership centers around the development of genetically modified beta cells for the treatment of both type 1 and type 2 diabetes. CRISPR and ViaCyte are reportedly on track to release top-line data from an early-stage trial in type 1 diabetes next year. Human trials for the duo's type 2 therapy could kick off in either late 2023 or early 2024.

In sum, Vertex's cell- and gene-editing-based diabetes care therapies have the potential to bend the curve in terms of the standard of care for this worldwide epidemic. Wall Street, however, doesn't appear to have taken notice of this deep value proposition -- at least not yet. Vertex's stock, after all, seems to be being valued mainly for its cystic fibrosis franchise and pipeline assets in various blood disorders.

Patrick Bafuma (Repligen): When it comes to the future, Repligen is creating the tools that make it all possible. From cell and gene therapy to cutting-edge mRNA vaccines to monoclonal antibody treatment, Repligen is there. This biotech provides the equipment and associated consumables needed for various steps in the manufacturing of today's most complex treatments.

And in a year marred by supply chain and geopolitical issues, Repligen has seemingly been insulated. There was hardly a mention of either of these issues in the last few earnings conference calls. Not bad considering its sales are spread throughout the globe with Europe, North America, and Asia representing 35%, 45%, and 20% of second-quarter revenue, respectively. Despite sales slowing down for COVID-19 treatments and vaccines that Repligen helps manufacture, the company's base business in Q2 was up 41% year over year. The record quarterly revenue of $207.6 million -- up 32% year over year on a constant currency basis -- is a welcome sight for biotech investors in 2022's uncertain market.

If you believe cell and gene therapy are the future of healthcare, then Repligen deserves a second look. The bioprocessing company's cell and gene therapy segment is seeing accelerated growth. Revenue from this business line increased 40% in full year 2021, and growth in Q2 2022 was up almost 70% from the same period a year ago. This puts Repligen on track to smash its own 40% growth target for the division for 2022. There are other signs the future is bright, too. At the end of 2020, there were over 1,200 clinical trials underway worldwide in the cell and gene therapy segment, according to the company. Then there are the FDA estimates for annual revenue growth of over 25% for the entirety of said market through 2025. Put it all together, and Repligen is well positioned to thrive as the backbone of the industry.

Taylor Carmichael (Recursion Pharmaceuticals): Vertex and Repligen are great picks. Both companies are already highly profitable, so the future prospects are grounded in what these companies have already accomplished. Recursion is a different animal. The company doesn't have profits yet, and revenue is still small. But I've opened up a small position here because the upside is so high. If this company has any success, it will dramatically transform drug discovery.

Here's the current paradigm for finding a new drug candidate. You notice a problem in the world of healthcare. You read academic articles to see what has been tried and what has not been tried. You formulate your own hypothesis. And then you collect data by testing your hypothesis in animals, and then in people. That's the basic model for drug discovery -- and it has a spectacular failure rate of over 90%.

Recursion is using supercomputers and artificial intelligence (AI) to create maps of biology. The company's software has already generated 16 petabytes of biological and chemical data. One petabyte equals 1.5 million CD-ROM discs. It's estimated the human brain can store 2.5 petabytes of memory data. So Recursion's AI solution now has more data than six human brains.

Even more importantly, the company's AI solution is running experiments with this data all the time. Recursion's computers have now made 2.4 trillion predictions about biological and chemical relationships. Research scientists can search all this data to create a more informed hypothesis. This is a fundamentally superior model, in my opinion. First you collect and analyze the scientific data, and then you form the hypothesis.

The company has major partners inBayerand Roche. And Recursion has quickly found drug candidates. In the first quarter, the company initiated a phase 2 study for a drug to treat cerebral cavernous malformation. In Q2, the company started a phase 2 study for a molecule to treat neurofibromatosis type 2. This quarter the company will initiate yet another phase 2 study, in a drug treating familial adenomatous polyposis (FAP).

Of course improving the odds does not mean that these drugs are sure things. But I definitely think Recursion is on the right track, and the future of drug discovery will follow this model. So I've made a small investment here while it's early and the company is still small. (Its market cap is under $2 billion.) If I'm right, Recursion will be a foundational holding for biotech investors for many years to come.

Read this article:
These 3 Stocks Are the Future of Biotechnology - The Motley Fool

Posted in Biotechnology | Comments Off on These 3 Stocks Are the Future of Biotechnology – The Motley Fool

ONLINE: The Future of Medicine – Isthmus

Posted: September 29, 2020 at 6:56 am

Watch here: https://www.youtube.com/watch?feature=youtu.be&v=VVkQU91KbEs

press release: The UW has a long history of pioneering medical advancements that have transformed the world. From performing the first bone marrow transplant in the United States to cultivating the first laboratory-derived human embryonic stem cells. Now, where will UW medical research go next?

On the next Wisconsin Medicine Livestream, meet trailblazing doctors, researchers, and medical leaders who are charting a bold course to completely alter the health care landscape. During this insightful panel discussion, well explore how gene therapy and cell replacements could hold the keys to treating inherited and acquired blindness. Youll also discover the remarkable potential in xenotransplantation where nonhuman animal source organs are transplanted into human recipients. In addition, you will learn about UW Healths journey to build a multidisciplinary program to serve the community. These, and other, fascinating developments in treatment and care are happening right now at the UW and are the future of medicine. The presentation will be moderated by Robert Golden, the dean of the University of WisconsinMadisons School of Medicine and Public Health.

Our Guests:

David Gamm, professor, Department of Ophthalmology and Visual Sciences; Emmett A. Humble Distinguished Director, McPherson Eye Research Institute; Sandra Lemke Trout Chair in Eye Research

Dr. Gamms lab is at the forefront in developing cell-based therapies to combat retinal degenerative diseases (RDDs). As the director of the McPherson Eye Research Institute and a member of the Waisman Center Stem Cell Research Program, the UW Stem Cell and Regenerative Medicine Center, and the American Society for Clinical Investigation, his efforts are directed toward basic and translational retinal stem cell research. The Gamm Lab uses induced pluripotent stem cells to create retinal tissues composed of authentic human photoreceptor cells rods and cones that can detect light and initiate visual signals in a dish. The aims of his laboratory are to investigate the cellular and molecular events that occur during human retinal development and to generate cells for use in retinal disease modeling and cell replacement therapies. In collaboration with other researchers at UWMadison and around the world, the lab is developing methods to produce and transplant photoreceptors and/or retinal pigment epithelium (RPE) in preparation for future clinical trials. At the same time, the Gamm Lab uses lab-grown photoreceptor and RPE cells to test and advance a host of other experimental treatments, including gene therapies. In so doing, the lab seeks to delay or reverse the effects of blinding disorders, such as retinitis pigmentosa and age-related macular degeneration, and to develop or codevelop effective interventions for these RDDs at all stages of disease.

Dhanansayan Shanmuganayagam, assistant professor, Department of Surgery, School of Medicine and Public Health; Department of Animal and Dairy Sciences, UWMadison; director, Biomedical, and Genomic Research Group

Dr. Shanmuganayagams research focuses on the development and utilization of pigs as homologous models to close the translational gap in human disease research, taking advantage of the overwhelming similarities between pigs and humans in terms of genetics, anatomy, physiology, and immunology. He and his colleagues created the human-sized Wisconsin Miniature Swine breed that is unique to the university. The breed exhibits greater physiological similarity to humans, particularly in vascular biology and in modeling metabolic disorders and obesity. He currently leads genetic engineering of swine at the UW. His team has created more than 15 genetic porcine models including several of pediatric genetic cancer-predisposition disorders such as neurofibromatosis type 1 (NF1). In the context of NF1, his lab is studying the role of alternative splicing of the nf1 gene on the tissue-specific function of neurofibromin and whether gene therapy to modulate the regulation of this splicing can be used as a viable treatment strategy for children with the disorder.

Dr. Shanmuganayagam is also currently leading the efforts to establish the University of Wisconsin Center for Biomedical Swine Research and Innovation (CBSRI) that will leverage the translatability of research in pig models and UWMadisons unique swine and biomedical research infrastructure, resources, and expertise to conduct innovative basic and translational research on human diseases. The central mission of CBSRI is to innovate and accelerate the discovery and development of clinically relevant therapies and technologies. The center will also serve to innovate graduate and medical training. As the only center of its kind in the United States, CBSRI will make UWMadison a hub of translational research and industry-partnered biomedical innovation.

Petros Anagnostopoulos, surgeon in chief, American Family Childrens Hospital; chief, Section of Pediatric Cardiothoracic Surgery; professor, Department of Surgery, Division of Cardiothoracic Surgery

Dr. Anagnostopoulos is certified by the American Board of Thoracic Surgery and the American Board of Surgery. He completed two fellowships, one in cardiothoracic surgery at the University of Pittsburgh School of Medicine and a second in pediatric cardiac surgery at the University of California, San Francisco School of Medicine. He completed his general surgery residency at Henry Ford Hospital in Detroit. Dr. Anagnostopoulos received his MD from the University of Athens Medical School, Greece. His clinical interests include pediatric congenital heart surgery and minimally invasive heart surgery.

Dr. Anagnostopoulos specializes in complex neonatal and infant cardiac reconstructive surgery, pediatric heart surgery, adult congenital cardiac surgery, single ventricle palliation, extracorporeal life support, extracorporeal membrane oxygenation, ventricular assist devices, minimally invasive cardiac surgery, hybrid surgical-catheterization cardiac surgery, off-pump cardiac surgery, complex mitral and tricuspid valve repair, aortic root surgery, tetralogy of Fallot, coronary artery anomalies, Ross operations, obstructive cardiomyopathy, and heart transplantation.

When: Tuesday, Sept. 29, at 7 p.m. CDT

Where: Wisconsin Medicine Livestream: wiscmedicine.org/programs/ending-alzheimers

Excerpt from:
ONLINE: The Future of Medicine - Isthmus

Posted in Molecular Genetics | Comments Off on ONLINE: The Future of Medicine – Isthmus

Coverage Policy – Arkansas Blue Cross and Blue Shield

Posted: September 7, 2016 at 4:53 am

Select a Title: Select Policy Title... Autologous or Allogeneic Stem &/or Progenitor Cell Support- POEMS Abatacept (Orencia) for Rheumatoid Arthritis Ablation Ther Atrial Fibrillation (Pulmo Venous Isolation, Radiofreq, Cryo, Ablation Therapy for Atrial Arrhythmias other than Atrial Fibrillation Ablation Therapy, Radiofrequency and Cryoablation of Pulmonary Tumors Acupuncture Adipose-Derived Stem Cells in Autologous Fat Grafting to the Breast Ado-Trastuzumab Emtansine (Trastuzumab-DM1) for Tx of HER-2+ Malignancies Adoptive Immunotherapy Alcohol Injections for Treatment of Peripheral Neuromas Alemtuzumab ( Lemtrada) Allergen Specific IgE In Vitro Testing Allergy Immunotherapy Allergy Testing, Metal Allergy Testing, Serial Endpoint Testing Allogeneic Donor Leukocyte Infusion Ambulatory Blood Pressure Monitoring Amevive (Alefacept) Amniocentesis/Chorionic Villus Samp-Detect Fetal Hereditary/Chromosomal Abn Amniotic Membrane and Amniotic Fluid Injections Angioplasty/Stent, Percutaneous, Carotid Artery Angioplasty/Stenting/Atherectomy of LE, Abd Aortic, & Visceral Arteries Annuloplasty: Percutaneous Intradiscal: IDET, PIRFT or Biacuplasty Anti-PD-1 (programmed death receptor-1)Therapy (Pembrolizumab)(Nivolumab) Antigen Leukocyte Antibody Test (ALCAT) Antineoplaston Cancer Therapy Antiprothrombin Antibody Antithrombin III Replacement Apheresis, Therapeutic (Plasma Exchange Transfusion) Aqueous Shunts and Devices for Glaucoma Arthroereisis for Pes Planus (Flat Feet) (Subtalar stabilization) Artificial Heart, Total Artificial Vertebral Disc, Cervical Spine Artificial Vertebral Disc, Lumbar Spine Atezolizumab (Tecentriq) Auditory Evoked Potential Autism Spectrum Disorder, Early Behavioral Intervention Autism Spectrum Disorder, Interventions NOT Early Behavioral Intervention Autologous Chondrocyte Implant for Focal Articular Cartilage Lesions Autologous Stem-cell Therapy to Treat Peripheral Arterial Disease Automated Whole Breast Ultrasound Balloon Sinuplasty Biochemical Marker's, Alzheimer's Disease Biofeedback as a Trtmnt of Chronic Pain Biofeedback as a Trtmnt of Fecal Incontinence or Constipation Biofeedback as a Trtmnt of Headache Biofeedback as a Trtmnt of Urinary Incontinence in Adults Biofeedback for Miscellaneous Indications Bioimpedance Devices for Detection of Lymphedema Biomarker Panel Testing for Systemic Lupus Erythematosus Biomarker Test (Vectra DA) Monitoring Disease Activity-Rheumatoid Arthriti Biomarker Test, reDx, Diabetes Risk Score Biomarker, Methotrexate Polyglutamates Predict Response Methotrexate -RA Biomarker, Serum Human Epididymis Protein 4 (HE4) Biomarkers for Liver Disease Biventricular Pacemakers for the Trtmnt of Congestive Heart Failure Blepharoplasty/Blepharoptosis Blinatumomab (Blincyto) Blood/Platelet-Derived Growth Factors for Wound Healing Bone Growth Stim, Elec, Adjunct to Spinal Fusion Bone Growth Stim, Elect, Appendicular Skeleton Bone Markers (Collagen Crosslinks as Biological Markers of Bone Turnover) Bone Mineral Density Study Bone Morphogenetic Protein Boron Neutron Capture Therapy Brachytherapy, Brain Tumors Brachytherapy, Breast Brachytherapy, Endobronchial Brachytherapy, Prostate, High-Dose Rate Temporary Brachytherapy, Prostate, Low-dose Rate Brachytherapy, Radioembolization Primary & Metastatic Tumors of the Liver Bronchial Thermoplasty Capsaicin (Qutenza) for the Trtmnt of Post-Herpetic Neuralgia Cardiac Event Recorder, External Loop or Continuous Recorder Cardiac Event Recorder, Insertable Loop Recorder Cardiac Event Recorder, Mobile Telemetry Cardiac Rehabilitation Cardiovascular Risk Panels Cardioverter Defibrillator; Implantable, SubQ, & Wearable Carotid Intima -Media Thickness, US Meas Assess Subclinic Atherosclerosis Certified Nurse Midwives Certified Nurse Practitioners Chelation Therapy Chemical Ecology (Environ Illness, Multi Chem Sensitiv, Environ Hypersensi Chemical Labyrinthectomy in the Trtmnt of Meniere's Syndrome Chemical Peels Chemodenerv BOTOX Chemodenerv BOTOX for the Trtmnt of Chronic Migraine Headache Chemosensitivity and Chemoresistance Assays, In-Vitro Chemotherapy for Malignancy Chromoendoscopy as an Adjunct to Colonoscopy Chronic Cerebrospinal Venous Insufficiency in MS, Diagnosis & Trtmnt Chronic Intermittent Intravenous Insulin Therapy (CIIIT) Circulating Tumor Cells in the Mgmt of Patients with Cancer, Detection of Clinical Nurse Specialist Closure Devices-Atrial/Ventricular Septal Defects (ASD, VSD) or PFO, Percut Cochlear Implant Cognitive Rehabilitation Cold Therapy Computed Tomography (CT) Cardiac and Coronary Artery Computed Tomography (CT) Perfusion Imaging Computed Tomography (CT) Scanning for Lung Cancer Screening Computer-Assisted Musculoskeletal Surgical Navigational Orthopedic Procedur Confocal Laser Endomicroscopy Continuous Passive Motion Device In the Home Setting Cord Blood as a Source of Stem Cells for Potential Disease Cord Blood as a Source of Stem Cells for Trtmnt of Identified Disease Corneal Collagen Cross-linking Corneal Topography Coronary Artery Calcium Scoring, Screening, to Predict Risk for Coronary Ar Corticosteroid Infusion of Middle Ear for Sudden Hearing Loss Cryosurgical Ablation of Breast Tumors, Benign and Malignant Cryosurgical Ablation of Pancreatic Cancer Cryosurgical Ablation of Primary or Metastatic Liver Tumors Cryosurgical Ablation of Prostate Cancer Cryosurgical Ablation of Renal Tumors Current Perception Threshold Test Cutting Guides and Custom Knee Implants Cytoreduction Surgery with Hyperthermic Intraperitoneal Chemotherapy Daclatasvir (Daklinza) Daratumumab (Darzalex) Digital Breast Tomosynthesis Digital Imaging Systems for the Detection of Diabetic Retinopathy Digital Motion X-ray (Cineradiography/Videoradiography) MusculoskeletalCond Digitization: Computer Enhanced X Ray Analysis for Spinal Evaluation Dopamine Transporter Imaging with Single Photon Emission CT (DAT-SPECT) Droxidopa (Northera) Dry Needling of Myofascial Trigger Points Eculizumab (Soliris) EKG, Signal Averaged Elec Stim, Auricular Stimulation and Cranial Electrotherapy Stimulation Elec Stim, Baroreflex Stim for the Trtmnt of Hypertension Elec Stim, Deep Brain (e.g. Parkinson, Dystonia, MS, Post Traumatic Dyskine Elec Stim, Occipital Nerve Stim for Trtmnt of Headaches Elec Stim, Percutaneous Electrical Nerve Stim(PENS) or PNT Elec Stim, Percutaneous Tibial Nerve Stim for the Trtmt Voiding Dysfunction Elec Stim, Transcutaneous Elec Nerve Stim Elec Stim, Vagus Nerve Stim for the Trtmnt of Depression Elec Stim, Vagus Nerve Stim for the Trtmnt of Essential Tremors Elec Stim, Vagus Nerve Stim for the Trtmnt of Fibromyalgia Elec Stim, Vagus Nerve Stim for the Trtmnt of Headaches Elec Stim, Vagus Nerve Stim for the Trtmnt of Heart Failure Elec Stim, Vagus Nerve Stim for the Trtmnt of Obesity Elec Stim, Vagus Nerve Stim for the Trtmnt of Seizures Elec/Electromagnetic Stim for the Trtmnt of Arthritis Electric Breast Pump (Hospital Grade) Electrical/Magnetic Stim, Pelvic Floor Muscles-Urinary & Fecal Incontinence Electrocardiogram, Computerized 2-Lead Analysis for the Diagnosis of CAD Electrocardiographic Body Surface Mapping Electromagnetic Navigation Bronchoscopy Electrophrenic Pacemaker (Diaphragmatic Pacemaker) ElectroStim and Electromagnetic Therapy for the Trtmnt of Wounds Endobronchial Valves Endothelial Function Testing, Noninvasive Endothelial Keratoplasty Endovascular Procedures, Intracranial Arterial Dz & Extracranial Vertebral Endovascular Stent Grafts for Thoracic Aortic Aneurysms or Dissections Epidural Adhesiolysis, Percutaneous Epiduroscopy Epiretinal Rad Ther for Age-Related Macular Degeneration ESWL for Plantar Fasciitis and Other Musculoskeletal Conditions ESWL in the Trtmnt of Peyronie's Disease Etanercept (Enbrel) External Enhanced Cardiac Counterpulsation (EECP) Extracorporeal Membrane Oxygenation for Adult Conditions Extracranial-Intracranial Bypass Surgery in Cerebrovascular Disease Eyelid Thermal Pulsation for the Treatment of Dry Eye Syndrome Fecal Microbiota Transplantation for the Treatment of Clostridium Difficile Femoroacetabular Impingement, Surgical Trtmnt of Fetal Fibronectin Enzyme Immunoassay Focal Treatments for Prostate Cancer Food and Chemical Sensitivity Testing Functional Anesthetic Discography Functional Intracellular Analysis Gait Analysis Galectin Measurement Gastric NeuroStim for Morbid Obesity Gastric NeuroStim for Trtmnt of Gastric and/or Small Bowel Paresi Gastroesophageal Reflux Disease (GERD), Magnetic Esophageal Ring Genetic Test: Analysis of MGMT Promoter Methylation in Malignant Gliomas Genetic Test: Aspirin Trtmnt, Lipoprotein(a) Variant(s) as a Decision Aid Genetic Test: Breast Ca Predict; Risk of Recurrence Need for Adjuvant Chem Genetic Test: Molecular Markers in Fine Needle Aspirates of the Thyroid Genetic Test: X-Linked Opitz G/BBB Syndrome, MID1 Mutation Testing Genetic Test: Acute Myeloid Leukemia, (FLT3 and NPM1) Genetic Test: Adolescent Idiopathic Scoliosis; Prediction of Disease Prog Genetic Test: Aid in the Mgmt of Psychiatric Meds & Conditions Genetic Test: Allopurinol Sensitivity (HLA-B*5801) Genetic Test: Alpha Thalessemia Genetic Test: Alpha-1 Antitrypsin Deficiency Genetic Test: Alzheimer's Disease Genetic Test: Amyotrophic Lateral Sclerosis Genetic Test: Asthma, HLA-DR and HLA-DQ Genotyping Genetic Test: Azothiaprine 6MP Sensitivity,Genotyping Phenotyping (TPMT) Genetic Test: BRCA1 or BRCA2 Mutations Genetic Test: Breast Ca Predict; Risk of Distant Metastasis Adjuvant Chem Genetic Test: Breast Ca Predict; Risk of Distant Metastasis Need Adjuvant C Genetic Test: Breast Ca; Risk Recurrence Need Adjuvamt Chemo (Oncotype Dx) Genetic Test: Canavan Disease Genetic Test: Cancer Susceptibility Panels Using Next Generation Sequencing Genetic Test: Carbamazepine HLA-B*1502 Genetic Test: Cardiac Ion Channelopathies Genetic Test: Celiac Disease, HLA Typing (HLA-DQ) Genetic Test: Cerebral Autosomal Dominant Arteriopathy (CADASIL) (NOTCH3) Genetic Test: CHARGE Syndrome Genetic Test: Chromosomal Microarray (CMA) & NGS Eval Pat Devel Delay/Autis Genetic Test: CMA Testing for the Evaluation of Pregnancy Loss Genetic Test: CML and ALL (BCR-ABL) Genetic Test: Colon Ca, KRAS, NRAS,BRAF Mutation-Tumor Sensitivity to Chemo Genetic Test: Colon Cancer, Gene Expression Profiling Genetic Test: Coronary Artery Disease, Testing to Predict Risk (Corus CAD) Genetic Test: Diagnosis & CA Risk Assessment for Prostate Cancer Genetic Test: Dilated Cardiomyopathy Genetic Test: Duchenne and Becker Muscular Dystrophy Genetic Test: Epilepsy Genetic Test: Facioscapulohumeral Muscular Dystrophy Genetic Test: Factor V Leiden Genetic Test: Fanconi Anemia Genetic Test: Fecal DNA to Detect Colorectal Cancer, Screening Genetic Test: Fetal RHD Genotyping Using Maternal Plasma Genetic Test: FMR 1 Mutations Including Fragile X Syndrome Genetic Test: Genotyping for 9p21 Single Nucleotide Polymorphisms-Cardio Genetic Test: Germline Mutations RET Protooncogene in Medullary Cx Thyroid Genetic Test: Head & Neck Cancer, EGFR Mutation Analysis Genetic Test: Heart Transplantation Rejection, AlloMap Testing Genetic Test: Hemochromatosis Genetic Test: HER2 Testing Genetic Test: Hereditary Pancreatitis Genetic Test: HERmark, HER2 Breast Cancer Assay Meas HER2 Tot Protein Ex Genetic Test: HLA-B*5701 Testing for Abacavir Hypersensitivity Reaction Genetic Test: Hypertrophic Cardiomyopathy, Predisposition Genetic Test: Inherited Peripheral Neuropathies (Charcot Marie Tooth, HNPP) Genetic Test: JAK2 and MPL Mutation Test for Myeloproliferative Disorders Genetic Test: KIF6 Predict Risk for CVD and/or Effectiveness of Statin Tx Genetic Test: KIT (c-KIT, CD117) Genetic Test: Lactase Insufficiency (-13910 C>T) Genetic Test: Li-Fraumeni Syndrome Genetic Test: Lynch Syndrome and Inherited Intestinal Polyposis Syndromes Genetic Test: Macular Degeneration Genetic Test: Melanoma, Hereditary Genetic Test: Melanoma, V600E Mutation Testing - Vemurafenib (Zelboraf) Genetic Test: Microarray-based Gene Exp Profile Analysis Prostate CA Mang Genetic Test: Microarray-Based Gene Expression Testing, CUP Genetic Test: Miscellaneous Genetic and Molecular Diagnostic Tests Genetic Test: Mitochondrial Disorders Genetic Test: Molecular Testing of Tumors for Genomic Profiling Genetic Test: Multiple Myeloma, Gene Expression Profiling Genetic Test: Mutation Testing for Limb-Girdle Muscular Dystrophies Genetic Test: Neurofibromatosis Genetic Test: PALB2 Mutations Genetic Test: PathFinderTG Molecular Testing Genetic Test: Pharmacogenetic Testing for Pain Management Genetic Test: Prenatal Analysis of Fetal DNA Detect Fetal Aneuploidy Genetic Test: Prothrombin Thrombophilia (G20210A) and MTHFR Mutations Genetic Test: PTEN Hamartoma Tumor Syndrome Genetic Test: Rett Syndrome Genetic Test: Statin-Induced Myopathy (SLCO1B1) Genetic Test: Tamoxifen Trtmnt (CYP2D6) Genetic Test: Testing for Use of 5-FU in Patients with Cancer Genetic Test: UGT1A1 to Predict Toxicity to Irinotecan Genetic Test: Universal Gene Test (Counsyl) Genetic Test: Use of Common Genetic Variants (SNPS), Nonfamilial Breast CA Genetic Test: Uveal Melanoma, Gene Expr Profile Predict Risk of Metastasis Genetic Test: Warfarin Dose/Response Genetic Test: Whole Exome Sequencing Genetic Testing: CHEK2 Mutations for Breast Cancer Glaucoma Evaluation, Ophthalmologic Techniques Glucose Monitoring, Continuous Golimumab (Simponi) Growth Hormone, Human Handheld Radio Spectroscopy for Intraoperative Assessment of Surg Margins.. HDC & Allo-Acute Lymphocytic Leukemia HDC & Allo-Acute Myelogenous Leukemia HDC & Allo-Auto-Chronic Lymphocytic Leukemia and Small Lymphocytic Lymphoma HDC & Allo-Auto-Chronic Malignant Astrocytomas & Gliomas HDC & Allo-Chronic Myelogenous Leukemia HDC & Allo-Ewing's Sarcoma HDC & Allo-Genetic Diseases & Acquired Anamias HDC & Allo-Hematophagocytic lymphohistiocytosis HDC & Allo-Hodgkin's Disease HDC & Allo-Myelodysplastic Disease HDC & Allo-Non-Hodgkin's Lymphoma HDC & Allo-Primitive Neuroectodermal Tumors (PNET) & Ependymoma HDC & Allo-Solid Tumors of Childhood HDC & Auto-Acute Lymphocytic Leukemia HDC & Auto-Acute Myelogenous Leukemia HDC & Auto-Chronic Myelogenous Leukemia HDC & Auto-Ewing's Sarcoma HDC & Auto-followed by Nonmyeloablative Allogeneic SCT for Multiple Myeloma HDC & Auto-Germ Cell Tumors HDC & Auto-Hodgkin's Disease HDC & Auto-Multiple Myeloma HDC & Auto-Myelodysplastic Syndrome HDC & Auto-Non Hodgkin's Lymphomas HDC & Auto-Primitive Neuroectodermal Tumors (PNET) & Ependymoma HDC & Auto-Solid Tumors of Childhood HDC & Auto-Waldenstrom's Macroglobulinemia HDC & Autologous or Allogeneic Stem Cell Sup-Epith Ovarian Cancer HDC & Hematopoietic Stem Cell Support for Breast Cancer HDC & Hematopoietic Stem Cell Support-Autoim Diseases Include Mult HDC & Hematopoietic Stem Cell Support-Misc Solid Tumors Adults HDC -AL Amyloidosis (Light Chain Amyloidosis) Heartsbreath Test for Heart Transplant Rejection Detection Hemodynamic Monitoring of Heart Failure, Mgmt in the Outpatient Setting Hepatic Tumors, Ablative Procedures(PEI, Acetic Acid Inj, ILP, LITT) Hereditary Angioedema, Prophylaxis and Acute Treatment Hip Resurfacing Hippotherapy HIV Tropism, Testing Home Apnea Monitors Home Uterine Activity Monitor Homocysteine Measurement Hormone Pellet Implantation for Hormone Replacement Therapy Human Papilloma Virus Testing of Cervical Pap Smears Hyperbaric Oxygen Pressurization (HBO) Hyperhidrosis Trtmnt Hysteroscopic Placement Micro-Inserts in Fallopian Tubes Form Immune Cell Function Assay Immune Globulin, Intravenous and Subcutaneous Implantable Bone Conduction Hearing Aids Implantable Infusion Pump Implantable Telescope for the Trtmnt of Age-Related Macular Degeneration IMRT IMRT, Anus, Anal Canal IMRT, Breast IMRT, Lung IMRT, Prostate Infertility Services Infliximab (Remicade) Ingestible pH and Pressure Capsule Injection, Clostridial Collagenase Fibroproliferative Disorders Insulin Infusion Pumps, External Interferon Gamma-1B Interspinous and Interlaminar Stabilization/Distraction Devices (Spacers) Interventions for Progressive Scoliosis Intradialytic Parenteral Nutrition Intraepidermal Nerve Fiber Density Intraoperative Neurophysiologic Monitoring Intraoperative Radiation Therapy Intrastromal Corneal Ring Segments, Implantation Intrauterine Systems, Progesterone or Progestogen-Releasing Intravenous Lidocaine or Ketamine Outpatient Management of Chronic Pain Intravitreal Implant, Dexamethasone (Ozurdex) Intravitreal Implant, Fluocinolone Acetonide (Retisert, Iluvien) Ipilimumab (Yervoy) Iron Therapy, Parenteral Irreversible Electroporation, Nanoknife Keratomileusis Keratophakia Keratoplasties, Refractive Keratoprosthesis Kyphoplasty, Percutaneous and Mechanical Vertebral Augmentation Lab Test: Identification of Microorganisms Using Nucleic Acid Probes Laser Treatment of Onychomycosis Laser Trtmnt of Congenital Port Wine Stain Hemangiomas Left Atrial Appendage, Closure Device, Percutaneous Leuprolide (Lupron) LITT for Brain Tumors Low Level Laser Therapy (LLLT) Lung Volume Reduction Surgery (LVRS) Lyme Disease, Intravenous Antibiotic Therapy and Associated Diagnostic Test Lymphedema Pumps (Pneumatic Compression Devices) Trtmnt of Lymphedema Macular Translocation Magnetic Resonance Spectroscopy Magnetoencephalography/Magnetic Source Imaging Mammoplasty, Reduction Mastectomy, Male Gynecomastia Mastectomy, Prophylactic Maze Procedure Measurement of Serum Antibodies to Infliximab and Adalimumab Measuremt of Exhaled Nitric Oxide Dx and Mgmt of Asthma & Other Resp Disord Measuremt of Lipoprotein-Associated Phospholipase A2 (Lp-PLA2) -Cardiovasul Measuremt of Serum Intermediate Density Lipoproteins (Remnant-like Partic Meniscal Allograft Transplantation and Synthetic Meniscal Implants Mepolizumab (Nucala) Metabolite Testing, Monitor Antimetabolite Therapy for IBS, CVD, ALL Microprocessor-Controlled Prostheses for the Lower Limb Microwave Ablation of Tumors Microwave Thermotherapy for Breast Cancer Minimally Invasive Image-Guided Lumbar Decompression for Spinal Stenosis Minimally Invasive Lumbar Interbody Fusion Mohs' Micrographic Surgery MR Guided Ultrasound Ablation - Uterine Fibroids and Other Tumors MRI Targeted Biopsy of the Prostate MRI, Breast MRI, Cardiac Applications MRI, Functional Multiple Sleep Latency/Maintenance of Wakefulness Test Multispectral Digital Skin Lesion Analysis (MelaFind) (MSDSLA) Multitarget PCR Testing for Dx of Bacterial Vaginosis (SureSwab) (NuSwab) Mutation Molecular Analysis for Targeted Therapy in Patients With NSCLC Myocardial Damage, Auto Cell Therapy(progenitor, hematopoietic SC, Myoblast Myocardial Sympathetic Innervation Imaging in Patients with Heart Failure Myoelectric Prosthesis for the Upper Limb Natalizumab (Tysabri) Navigated Transcranial Magnetic Stimulation Needle Arthroscopy Nerve Conduction Studies (NCS), EMG and SEMG Nerve Graft For Patients Undergoing Radical Non-Nerve-Sparing Prostatectomy Nesiritide (Natrecor) for Use in the Outpatient Setting Neural Therapy Neurofeedback Neuromuscular Stim, Functional Non-Myeloablative Allo SCT Noninvasive Imaging Technologies to Detect Liver Fibrosis or Cirrhosis Novel Lipid Risk Factors in Risk Assessment & Management of CV Disease Nutritional Panel Testing (NutrEval, ONE FMV) Nutritional Supplements OATS and/or Mosaicplasty For Osteochondral Defects of the Knee Ocriplasmin (Jetrea) for Symptomatic Vitreomacular Adhesion Omalizumab (Xolair) Oophorectomy, Prophylactic Oprelvekin (Neumega) Optical Coherence Tomography Anterior Eye Segment Imaging Optical Coherence Tomography Imaging of Coronary Arteries Optical Diagnostic Devices, Evaluating Skin Lesions Suspected of Malignancy Orthopedic Applications of Stem Cell Therapy Orthoptic Training for the Trtmnt of Vision and Learning Disabilities Oscillatory Devices for Chest Physical Therapy Osteochondral Allograft and/or Mosaicplasty for Osteochondral Defects Knee Ovarian & Internal Iliac Vein Embolization Trtmnt of Pelvic Congest Syndrom Pain Management, Facet Joint Block Pain Management, Facet Nerve Denervation, other than Radiofreq Pain Management, Radiofreq Facet Joint Denervation Paliperidone Palmitate (Injectables Invega Sustenna & Invega Trinza) pasireotide Patient-actuated End Range Motion Stretching Devices PCSK9 INHIBITORS (Evolocumab) (Alirocumab) PDE-5 Inhibitors for Benign Prostatic Hypertrophy (Tadalafil) Percutaneous Transluminal Endovascular Graft for Abdominal Aortic Aneurysm Peripheral Subcutaneous Field Stimulation Periureteral Bulking Agents Trtmnt of Vesicoureteral Reflux (VUR) Periurethral Bulking Agents Trtmnt of urinary and Fecal Incontinence Peroral Endoscopic Myotomy (POEM) for Treatment of Esophageal Achalasia Pertuzumab PET or PET/CT for Anal Carcinoma PET or PET/CT for Brain Imaging, Non-malignant Dis PET or PET/CT for Breast Cancer PET or PET/CT for Carcinoma of Unknown Primary (CUP) PET or PET/CT for Cervical Cancer PET or PET/CT for Colorectal Cancer PET or PET/CT for Esophageal or Esophagogastric Junction (EGJ) Cancer PET or PET/CT for Gastric Cancer PET or PET/CT for Head and Neck Malignant Disease PET or PET/CT for Hodgkin's Lymphoma PET or PET/CT for Lymphadenopathy of Unknown Cause PET or PET/CT for Malignant Brain Tumors PET or PET/CT for Melanoma PET or PET/CT for Mesothelioma PET or PET/CT for Neuroendocrine Tumors PET or PET/CT for Non-Hodgkin's Lymphoma PET or PET/CT for Non-Small Cell Lung Cancer PET or PET/CT for Ovarian Cancer PET or PET/CT for Pancreatic Cancer PET or PET/CT for Pulmonary Langerhans Cell Histiocytosis PET or PET/CT for Renal Cell Carcinoma PET or PET/CT for Small Cell Lung Cancer PET or PET/CT for Soft Tissue Sarcoma, including GIST PET or PET/CT for Testicular Germ Cell Cancer PET or PET/CT for Thymoma/Thymic Carcinoma PET or PET/CT for Thyroid Cancer PET or PET/CT for Vulvar Carcinoma PET or PET/CT, Choline C 11 or FDG, for Prostate Cancer PET Scan for Alzheimer's-Beta Amyloid Imaging PET Scan for Alzheimer's-FDG PET Scan for Cardiac Applications PET Scan for Multiple Myeloma, Plasmacytoma Photochemotherapy (PUVA) Photochemotherapy, Extracorporeal Trtmnt of Cutaneous T-Cell Lymphoma Photochemotherapy, Extracorporeal Trtmnt of Graft-versus-Host Disease Photochemotherapy, Extracorporeal Trtmnt to Prevent Reject after Transplant Photodynamic Therapy for Dermatologic Conditions Photodynamic Therapy for Malignancy Photodynamic Therapy for Ophthalmology Phototherapy for Psoriasis Phototherapy for Vitiligo Physician Assistants Platelet-Rich Plasma (Autologous Growth Factors), Orthopedic Applications Plugs for Anal Fistula Repair Pneumatic Compression Device, Intermittent, after Hip and Knee Arthroplasty Polysomnography for Non-Respiratory Sleep Disorders Positional Magnetic Resonance Imaging Positional Plagiocephaly & Craniosynostoses: Adjustable Cranial Orthoses Positron Emission Mammography (PEM) Posturography, Dynamic/Static Powered Exoskeleton for Ambulation in Patients with Lower Limb Disabilities PPACA FOR NGP: ABDOMINAL AORTIC ANEURYSM SCREENING PPACA FOR NGP: ALCOHOL MISUSE COUNSELING AND/OR SCREENING PPACA FOR NGP: ANEMIA, SCREENING, INFANTS, CHILDREN & ADOLESCENTS PPACA FOR NGP: ASPIRIN TO PREVENT CARDIOVASCULAR DISEASE IN ADULTS PPACA FOR NGP: ASPIRIN, TO PREVENT MORBIDITY & MORTALITY FROM PREECLAMPSIA PPACA FOR NGP: AUTISM SCREENING PPACA FOR NGP: BACTERIURIA SCREENING IN PREGNANT WOMEN PPACA FOR NGP: BICYCLE HELMET USE, CHILDREN & ADOLESCENTS PPACA FOR NGP: BLOOD PRESSURE, SCREENING, INFANTS, CHILDREN & ADOLESCENTS PPACA FOR NGP: BRCA TESTING; GENETIC COUNSELING AND EVALUATION PPACA FOR NGP: BREAST CANCER PREVENTIVE MEDICATION PPACA FOR NGP: BREAST CANCER SCREENING (MAMMOGRAPHY) PPACA FOR NGP: BREASTFEEDING COUNSELING PPACA FOR NGP: CARDIOMETABOLIC RISKS OF OBESITY, CHILDREN & ADOLESCENTS PPACA FOR NGP: CERVICAL CANCER SCREENING PPACA FOR NGP: CHLAMYDIAL INFECTION SCREENING IN WOMEN & ADOLESCENTS PPACA FOR NGP: COLORECTAL CANCER SCREENING PPACA FOR NGP: CONTRACEPTIVE USE AND COUNSELING PPACA FOR NGP: DENTAL CARIES PREVENTION IN PRESCHOOL CHILDREN PPACA FOR NGP: DEPRESSION SCREENING IN ADOLESCENTS PPACA FOR NGP: DEPRESSION SCREENING, ADULTS PPACA FOR NGP: DEVELOPMENTAL SCREENING PPACA FOR NGP: FOLIC ACID FOR PREVENTION OF NEURAL TUBE DEFECTS PPACA FOR NGP: GESTATIONAL DIABETES SCREENING PPACA FOR NGP: GONORRHEA PROPHYLAXIS, NEWBORN OPHTHALMIC PPACA FOR NGP: GONORRHEA SCREENING, WOMEN & ADOLESCENTS PPACA FOR NGP: HCV SCREENING PPACA FOR NGP: HEARING LOSS SCREENING IN NEWBORNS UP TO AGE 21 PPACA FOR NGP: HEP B VIRUS INFECTION, SCREENING, PREGNANCY, ADOL & ADULTS PPACA FOR NGP: HIGH BLOOD PRESSURE SCREENING IN ADULTS PPACA FOR NGP: HPV SCREENING FOR SEXUALLY ACTIVE WOMEN PPACA FOR NGP: HUMAN IMMUNODEFICIENCY VIRUS (HIV) COUNSELING & SCREENING PPACA FOR NGP: HYPOTHYROIDISM SCREENING IN NEWBORNS PPACA FOR NGP: IBC, TO PROMOTE HEALTHY DIET & EXERCISE, ADULTS PPACA FOR NGP: INTIMATE PARTNER VIOLENCE; SCREENING IN WOMEN PPACA FOR NGP: IRON DEFICIENCY ANEMIA SCREENING IN PREGNANT WOMEN PPACA FOR NGP: IRON SUPPLEMENTATION FOR CHILDREN PPACA FOR NGP: LEAD SCREENING, INFANTS & CHILDREN PPACA FOR NGP: LUNG CANCER SCREENING PPACA FOR NGP: MEDIA USE, SCREENING & COUNSELING PPACA FOR NGP: METABOLIC/HEMOGLOBIN SCREENING, NEWBORNS PPACA FOR NGP: OBESITY IN CHILDREN; SCREENING AND COUNSELING PPACA FOR NGP: OBESITY SCREENING IN ADULTS PPACA FOR NGP: OSTEOPOROSIS SCREENING IN WOMEN PPACA FOR NGP: OVERVIEW PPACA FOR NGP: PHENYLKETONURIA SCREENING IN NEWBORNS PPACA FOR NGP: PREGNANCY SCREENING, SEXUALLY ACTIVE FEMALES PPACA FOR NGP: PREVENTION OF FALLS IN COMMUNITY-DWELLING OLDER ADULTS PPACA FOR NGP: RH INCOMPATABILITY SCREENING PPACA FOR NGP: SERUM LIPIDS SCREENING PPACA FOR NGP: SEXUALLY TRANSMITTED INFECTIONS, BEHAVIORAL COUNSELING-PREVE PPACA FOR NGP: SICKLE CELL DISEASE, NEWBORN SCREENING PPACA FOR NGP: SKIN CANCER, COUNSELING PPACA FOR NGP: SYPHILIS SCREENING PPACA FOR NGP: TB SCREENING, INFANTS, CHILDREN & ADOLESCENTS PPACA FOR NGP: TOBACCO USE, SCREENING, COUNSELING AND INTERVENTIONS PPACA FOR NGP: TYPE 2 DIABETES MELLITUS SCREENING FOR ADULTS PPACA FOR NGP: VISUAL IMPAIRMENT SCREENING IN CHILDREN PPACA FOR NGP: WELL CHILD VISITS PPACA FOR NGP: WELL-WOMAN VISITS PPACA rule,NGP_Clinical Trials Preimplantation Genetic Diagnosis, Testing or Trtmnt Procalcitonin Prolotherapy (Sclerotherapy) Prostate Cancer Predicting Risk of Recurrence, Systems Pathology Prostate, Saturation Biopsy Prostatic Stent, Temporary Prostatic Urethral Lift (UroLift System) Proteomics Predict Response to Chemotherapy (VeriStrat) Proteomics, Evaluation of Ovarian (Adnexal) Masses (e.g., OVA1, ROMA) Proteomics, Screening and Detection of Cancer (e.g., OvaCheck) Pulmonary Arterial Hypertension, Pharm Trtmnt w Prostacyclin Analogues, ETC Pulmonary Rehabilitation Pulsed Pressure, Trtmnt for Meniere's Disease Quantitative Electroencephalography as a Diagnostic Aid for ADHD Rad Ther, Image Guidance for Prostate Cancer Rad Ther, Proton Beam for Trtmnt of Prostate Cancer Rad Ther, Proton Beam or Helium Ion Irradiation, Other than Prostate Radioactive Seed Localization of Nonpalpable Breast Lesions Radiofreq Ablation & Other Laparoscopic & Perc Techniques, Uterine Fibroids Radiofreq Ablation, Barrett's Esophagus Radiofreq Ablation, Bony Metastases Radiofreq Ablation, Breast Tumors Radiofreq Ablation, Osteoid Osteoma Radiofreq Ablation, Renal Tumors Radiofreq Trtmnt of Fecal Incontinence Radiofreq Trtmnt, Chronic Back Pain (Nucleoplasty) Radiofrequency Ablation of Peripheral Nerves to Treat Pain Radiofrequency Ablation of Primary or Metastatic Liver Tumors Radiofrequency Ablation of Renal Sympathetic Nerves, Tx for Resistant HTN Radioimmunoscintigraphy Imaging - In-111 Capromab Pendetide (ProstaScint) Radioimmunotherapy in the Trtmnt of Non-Hodgkin Lymphoma Radium Ra 223 dichloride for Symptomatic Osseous Metastatic Prostate Cancer Renal Artery, Angioplasty/Stenting, Percutaneous Repair Durable Medical Equipment (DME) and External Prosthetic Devices Repository Corticotropin Injection Respiratory Syncytial Virus Immune Prophylaxis with Palivizumab (Synagis) Responsive Neurostimulation for the Tx of Refractory Partial Epilepsy Rilonacept (Arcalyst) Rituximab (Rituxan), Off-label Use Sacral Nerve Stim Trtmnt of Fecal Incontinence Sacral Nerve Stim Trtmnt of Neurogenic Bladder Second to Spinal Cord Injury Sacral Nerve Stim Trtmnt of Urge Urinary Incontinence Sacroiliac Joint Fusion, Minimally Invasive Sacroplasty Scintimammography and Gamma Imaging of the Breast and Axilla Scleral Contact Lens, Gas Permeable Screening for Vertebral Fracture with Dual X-ray Absorptiornetry Semi-Implantable and Fully Implantable Middle Ear Hearing Aid Sensory Integration Therapy and Auditory Integration Therapy Serum Antibodies for Diagnosis of Inflammatory Bowel Disease Short Tandem Repeat Analysis for Specimen Provenance Test (know error) Sinus Spacers and Stents, Implantable, following Endoscopic Sinus Surgery Sipuleucel-T (Provenge) for the Trtmnt of Prostate Cancer Skin and Soft Tissue Substitutes, Bio-Engineered Products Sleep Apnea & Pulmonary Disease, Ventilation Support & Resp Assist Devices Sleep Apnea, Minimally Invasive Surgical Treatment Sleep Apnea, Testing Sofosbuvir (Sovaldi) Soft Tissue Substitutes, Orthobiologic Implant Spinal Cord NeuroStim for Trtmnt of Intractable Pain Spinal Decompression Ther (Internal Disc Decompression Ther, Spinal Distrac Spinal Manipulation Under General Anesthesia Spinal Unloading Devices Trtmnt Low Back Pain (Orthotrac Pneumatic Vest) ST2 Assay for Chronic Heart Failure Stem Cell Growth Factor, Romiplostim (Nplate) Stem Cell Growth Factors, ESAs, Darbepoetin, Epoetin, Peginesatide Stereotactic Radiosurgery Gamma Knife Surg ,Linear Accelerator, Cyberknife, Subconjunctival Retinal Prosthesis Surg Deactivation of Headache Trigger Sites Surg Interruption Pelvic Nerve Pathways for Prim & Second Dysmenorrhea Surgery for Morbid Obesity Teduglutide for SBS Telemedicine Temporomandibular Joint Dysfunction Testing For Drugs of Abuse including Controlled Substances Testosterone Replacement Therapy Thermography and Infrared Dermal Thermometry Tocolysis, Acute and Maintenance Therapy Total Facet Arthroplasty Transanal Endoscopic Microsurgery (TEMS/TAMIS) Transcatheter Aortic Valve Implantation Transcatheter Arterial Chemoembolization (TACE) Treat Prim or Metas Liver Transcatheter Mitral Valve Repair Transcatheter Pulmonary Valve Implantation Transcranial Magnetic Stim Trtmnt of Depression & Other Psychiatric Disorde Transesophageal Ther GERD, Endoscopic Radiofreq Transesophageal Ther GERD, Endoscopic Suturing, Transoral Incisionless Fund Transesophageal Therapy for GERD, Endoscopic Polymer Inj., Bulking Agents Transmyocardial Laser Revascularization Transplant, Allogeneic Islet Cell or Pancreas for Diabetes Mellitus Transplant, Autologous Islet Cell for Chronic Pancreatitis Transplant, Composite Tissue Allotransplantation, Hand and/or Face Transplant, Heart Transplant, Heart/Lung Transplant, Kidney Transplant, Liver Transplant, Lung and Lobar Lung Transplant, Small Bowel Transplant, Small Bowel/Liver and Multivisceral Transpupillary Thermotherapy for Trtmnt of Choroidal Neovascularization Trastuzumab Treatment of Varicose Veins/Venous Insufficiency Tumor Antigen, CA 125 (Carcinoembryonic Antigen 125) Tumor Antigen, Prostate Specific Antigen (PSA) Tumor Markers, Urinary Bladder Cancer Tumor Vaccines Tumor-Treating Fields Therapy for Glioblastoma (NovoTTF) Ultrafiltration in Decompensated Heart Failure Under Heavy Sedation or General Anesthesia as a Tech of Opioid Detoxificati Unicondylar Interpositional Spacer Trtmnt Unicompartmental Arthritis Knee Urinary Metabolite Tests for Adherence to DAA Medications for HEP C US Accelerated Fracture Healing Device US Maternity Care US Maternity Care, 1st Trim Detect Down Syndrome-Fetal US Assess Nuchal Tra US Trtmnt for Wounds, Non-Contact Uterine Artery Embolization Trtmnt of Leiomyomas or Abnormal Uterine Bleedi Vacuum Assisted Closure Device Vagal Nerve Blocking Therapy for the Treatment of Obesity Vedolizumab (Entyvio) for Inflammatory Bowel Disease Ventricular Assist Devices Ventricular Restoration/Remodeling, Surgical Vertebroplasty, Percutaneous Vertical Expandable Prosthetic Titanium Rib Viekira Pak Virtual Colonoscopy/CT Colonography Viscocanalostomy and Canaloplasty Viscosupplementation for Trtmnt of Osteoarthritis of the Knee Whole Body Computed Tomography Scan as a Screening Test Wireless Capsule Endoscopy, Small Bowel Study, Esophagus & Colon Wireless Pressure Sensors in Endovascular Aneurysm Repair

When you select a policy, you will see its title, category and effective date at the top of the page. A description of the treatment and the actual policy, which explains what is covered, follow. At the bottom of the page, you will see related CPT codes and references.

Originally posted here:
Coverage Policy - Arkansas Blue Cross and Blue Shield

Posted in Arkansas Stem Cells | Comments Off on Coverage Policy – Arkansas Blue Cross and Blue Shield