Stem Cell Basics | STEM Cell Information – National Institutes of Health

I. Introduction: What are stem cells, and why are they important?

Stem cells have the remarkable potential to renew themselves. They can develop into many different cell types in the body during early life and growth. Researchers study many different types of stem cells. There are several main categories: the pluripotent stem cells (embryonic stem cells and induced pluripotent stem cells) and nonembryonic or somatic stem cells (commonly called adult stem cells). Pluripotent stem cells have the ability to differentiate into all of the cells of the adult body. Adult stem cells are found in a tissue or organ and can differentiate to yield the specialized cell types of that tissue or organ.

Pluripotent stem cells

Early mammalian embryos at the blastocyst stage contain two types of cells cells of the inner cell mass, and cells of the trophectoderm. The trophectodermal cells contribute to the placenta. The inner cell mass will ultimately develop into the specialized cell types, tissues, and organs of the entire body of the organism. Previous work with mouse embryos led to the development of a method in 1998 to derive stem cells from the inner cell mass of preimplantation human embryos and to grow human embryonic stem cells (hESCs) in the laboratory. In 2006, researchers identified conditions that would allow some mature human adult cells to be reprogrammed into an embryonic stem cell-like state. Those reprogramed stem cells are called induced pluripotent stem cells (iPSCs).

Adult stem cells

Throughout the life of the organism, populations of adult stem cells serve as an internal repair system that generates replacements for cells that are lost through normal wear and tear, injury, or disease. Adult stem cells have been identified in many organs and tissues and are generally associated with specific anatomical locations. These stem cells may remain quiescent (non-dividing) for long periods of time until they are activated by a normal need for more cells to maintain and repair tissues.

Stem cells have unique abilities to self-renew and to recreate functional tissues.

Stem cells have the ability to self-renew.

Unlike muscle cells, blood cells, or nerve cellswhich do not normally replicate stem cells may replicate many times. When a stem cell divides, the resulting two daughter cells may be: 1) both stem cells, 2) a stem cell and a more differentiated cell, or 3) both more differentiated cells. What controls the balance between these types of divisions to maintain stem cells at an appropriate level within a given tissue is not yet well known.

Discovering the mechanism behind self-renewal may make it possible to understand how cell fate (stem vs. non-stem) is regulated during normal embryonic development and post-natally, or misregulated as during aging, or even in the development of cancer. Such information may also enable scientists to grow stem cells more efficiently in the laboratory. The specific factors and conditions that allow pluripotent stem cells to remain undifferentiated are of great interest to scientists. It has taken many years of trial and error to learn to derive and maintain pluripotent stem cells in the laboratory without the cells spontaneously differentiating into specific cell types.

Stem cells have the ability to recreate functional tissues.

Pluripotent stem cells are undifferentiated; they do not have any tissue-specific characteristics (such as morphology or gene expression pattern) that allow them to perform specialized functions. Yet they can give rise to all of the differentiated cells in the body, such as heart muscle cells, blood cells, and nerve cells. On the other hand, adult stem cells differentiate to yield the specialized cell types of the tissue or organ in which they reside, and may have defining morphological features and patterns of gene expression reflective of that tissue.

Different types of stems cells have varying degrees of potency; that is, the number of different cell types that they can form. While differentiating, the cell usually goes through several stages, becoming more specialized at each step. Scientists are beginning to understand the signals that trigger each step of the differentiation process. Signals for cell differentiation include factors secreted by other cells, physical contact with neighboring cells, and certain molecules in the microenvironment.

How are stem cells grown in the laboratory?

Growing cells in the laboratory is known as cell culture. Stem cells can proliferate in laboratory environments in a culture dish that contains a nutrient broth known as culture medium (which is optimized for growing different types of stem cells). Most stem cells attach, divide, and spread over the surface of the dish.

The culture dish becomes crowded as the cells divide, so they need to be re-plated in the process of subculturing, which is repeated periodically many times over many months. Each cycle of subculturing is referred to as a passage. The original cells can yield millions of stem cells. At any stage in the process, batches of cells can be frozen and shipped to other laboratories for further culture and experimentation.

How do you reprogram regular cells to make iPSCs?

Differentiated cells, such as skin cells, can be reprogrammed back into a pluripotent state. Reprogramming is achieved over several weeks by forced expression of genes that are known to be master regulators of pluripotency. At the end of this process, these master regulators will remodel the expression of an entire network of genes. Features of differentiated cells will be replaced by those associated with the pluripotent state, essentially reversing the developmental process.

How are stem cells stimulated to differentiate?

As long as the pluripotent stem cells are grown in culture under appropriate conditions, they can remain undifferentiated. To generate cultures of specific types of differentiated cells, scientists may change the chemical composition of the culture medium, alter the surface of the culture dish, or modify the cells by forcing the expression of specific genes. Through years of experimentation, scientists have established some basic protocols, or recipes, for the differentiation of pluripotent stem cells into some specific cell types (see Figure 1 below).

What laboratory tests are used to identify stem cells?

At various points during the process of generating stem cell lines, scientists test the cells to see whether they exhibit the fundamental properties that make them stem cells. These tests may include:

Given their unique regenerative abilities, there are many ways in which human stem cells are being used in biomedical research and therapeutics development.

Understanding the biology of disease and testing drugs

Scientists can use stem cells to learn about human biology and for the development of therapeutics. A better understanding of the genetic and molecular signals that regulate cell division, specialization, and differentiation in stem cells can yield information about how diseases arise and suggest new strategies for therapy. Scientists can use iPSCs made from a patient and differentiate those iPSCs to create organoids (small models of organs) or tissue chips for studying diseased cells and testing drugs, with personalized results.

Cell-based therapies

An important potential application is the generation of cells and tissues for cell-based therapies, also called tissue engineering. The current need for transplantable tissues and organs far outweighs the available supply. Stem cells offer the possibility of a renewable source. There is typically a very small number of adult stem cells in each tissue, and once removed from the body, their capacity to divide is limited, making generation of large quantities of adult stem cells for therapies difficult. In contrast, pluripotent stem cells are less limited by starting material and renewal potential.

To realize the promise of stem cell therapies in diseases, scientists must be able to manipulate stem cells so that they possess the necessary characteristics for successful differentiation, transplantation, and engraftment. Scientists must also develop procedures for the administration of stem cell populations, along with the induction of vascularization (supplying blood vessels), for the regeneration and repair of three-dimensional solid tissues.

To be useful for transplant purposes, stem cells must be reproducibly made to:

While stem cells offer exciting promise for future therapies, significant technical hurdles remain that will likely only be overcome through years of intensive research.

Note: Currently, the only stem cell-based products that are approved for use by the U.S. Food and Drug Administration (FDA) for use in the United States consist of blood-forming stem cells (hematopoietic progenitor cells) derived from cord blood. These products are approved for limited use in patients with disorders that affect the body system that is involved in the production of blood (called the hematopoietic system). TheseFDA-approved stem cell products are listed on the FDA website. Bone marrow also is used for these treatments but is generally not regulated by the FDA for this use. The FDA recommends that people considering stem cell treatments make sure that the treatment is either FDA-approved or being studied under an Investigational New Drug Application (IND), which is a clinical investigation plan submitted and allowed to proceed by the FDA.

NIH conducts and funds basic, translational, and clinical research with a range of different types of stem cells. NIH-supported research with human pluripotent stem cells is conducted under the terms of theNIH Guidelines for Human Stem Cell Research. NIH awards are listed in various categories of stem cell research through theNIH Estimates of Funding for Various Research, Condition, and Disease Categories (RCDC). NIH also supports a major adult stem cell and iPSC research initiative through theRegenerative Medicine Innovation Project.

Go here to read the rest:
Stem Cell Basics | STEM Cell Information - National Institutes of Health

Induced Pluripotent Stem Cell (iPSC) Global Market Report 2022: Development of iPSC-Derived Disease Models Driving Growth – ResearchAndMarkets.com -…

  1. Induced Pluripotent Stem Cell (iPSC) Global Market Report 2022: Development of iPSC-Derived Disease Models Driving Growth - ResearchAndMarkets.com  Business Wire
  2. Induced Pluripotent Stem Cells Production Market by Product Type (Hepatocytes, Fibroblasts, Keratinocytes, Amniotic Cells, Others), Application (Academic Research, Drug Development, Toxicity Screening, Regenerative Medicine), and Geography Global T  Skegness Siren
  3. Global Allogeneic Stem Cell Therapy Market is expected to grow at a CAGR of 9.85% by 2030  Digital Journal
  4. Stem Cell Therapy Global Market Size, Share, Trends Analysis, By Product, By Application, By Technology, By Th  openPR
  5. View Full Coverage on Google News

See original here:
Induced Pluripotent Stem Cell (iPSC) Global Market Report 2022: Development of iPSC-Derived Disease Models Driving Growth - ResearchAndMarkets.com -...

Discover Inspiring Animal Research Topics – Studyclerk.com

Why Choose Animal Research Topics For Writing Purposes?

Contrary to popular belief, animal research topics are not only used by veterinarians. They are also pursued by students majoring in Healthcare, Sound Engineering, and even subjects like Fashion Studies and Chemistry. Of course, it may require writing an excellent custom research paper because the trick here is to tailor things to what you need. The most challenging, however, is to choose your topic correctly and avoid being vague about what you must explore. Even if you would like to explore environmental issues, using animal research topics will be essential. You need to provide an explanation of your reasoning and the negative effects of human interaction with flora and fauna.

While there may be no universal topic that will reflect all sides of animal-related research, consider those subjects that you know well. It must inspire you and be an area where you feel comfortable. If you love marine life and can provide personal research examples, it would be good to choose something that will suit a reflection journal. Alternatively, consider animal topics for research papers that can be supported by reliable sources and statistical information.

Start with an outline or a list of arguments that you would like to explore. Once done, continue with the wording for your topic that introduces the problem and offers a solution. You may also pose a research question about a problem or make a claim that will be supported by what you include in your paper. Always refer to your grading rubric and choose your research paper type accordingly. For example, your nursing research paper may talk about the use of animals for rehabilitation purposes, while a legal student may talk about animal rights in various countries. It all should be approached through the lens of what you learn as a primary subject!

As you might already know, animal physiology studies anything related to the physical processes, changes in behaviors, breeding patterns, and more. As you think about choosing the animal physiology branch, always narrow things down if possible.

This aspect of animal research essay writing may not be everyones cup of tea, which is why it is necessary to explore the facts and provide information that represents both sides of the debate. Stay sensitive and avoid being too graphic unless it is necessary. Below are some ideas to consider:

The subject of animal rights is popular among students coming from all academic disciplines. Since you can approach it via the philosophical, legal, or medical lens, think about how to reflect your primary skills. It will make your research of animal right topics sound more confident.

In the majority of cases, you may refer to your veterinary branch first and proceed from there or take a look at the variety of veterinary research topics that we have presented below. Remember to quote every citation and idea that has been taken from other sources to avoid plagiarism.

Even though this subject seems to be discussed everywhere these days, finding good animals topics to write about that deal with animal testing is not easy. Think about what are the underlying reasons for testing and what forces scientists to use it as a method. It will help you come up with ideas and better exploration strategies.

Warning: writing about animal cruelty subject is not for everyone, which is why you must be aware that the facts and statistics you may find will be shocking. It should be explored only if you are ready to embrace this disturbing subject. At the same time, you can explore milder animal cruelty cases like using pets as influencers on social media or the use of donkeys at the beaches to entertain tourists. There is always something to think about!

When you would like to take a general approach to animals research, it is good to come up with a research question as a part of your thesis statement or main argumentation. See these animals research paper examples:

Without a doubt, it is easy to get stuck with a multitude of topics and ideas. If you are planning to write about animal rights but do not know how to include certain animal physiology principles, it is safer to consider timely help with research paper. Our skilled team of specialists in this field will provide you with relevant sources and will help you polish things to perfection when you need assistance or do not know how to continue.

The same relates to checking your existing draft and citations in terms of plagiarism and originality. Writing about animals is never easy, which is why we know how you feel and also realize what your college professors expect to see. Take a look at our research topics about animals, trust us with your concerns and we shall help you achieve success!

See more here:
Discover Inspiring Animal Research Topics - Studyclerk.com

Sex Hormones and Their Receptors Regulate Liver Energy Homeostasis

The liver is one of the most essential organs involved in the regulation of energy homeostasis. Hepatic steatosis, a major manifestation of metabolic syndrome, is associated with imbalance between lipid formation and breakdown, glucose production and catabolism, and cholesterol synthesis and secretion. Epidemiological studies show sex difference in the prevalence in fatty liver disease and suggest that sex hormones may play vital roles in regulating hepatic steatosis. In this review, we summarize current literature and discuss the role of estrogens and androgens and the mechanisms through which estrogen receptors and androgen receptors regulate lipid and glucose metabolism in the liver. In females, estradiol regulates liver metabolism via estrogen receptors by decreasing lipogenesis, gluconeogenesis, and fatty acid uptake, while enhancing lipolysis, cholesterol secretion, and glucose catabolism. In males, testosterone works via androgen receptors to increase insulin receptor expression and glycogen synthesis, decrease glucose uptake and lipogenesis, and promote cholesterol storage in the liver. These recent integrated concepts suggest that sex hormone receptors could be potential promising targets for the prevention of hepatic steatosis.

Obesity rapidly becomes a worldwide epidemic disease with increased risk of cardiovascular diseases, type 2 diabetes mellitus, and metabolic syndrome [1]. Metabolic syndrome is characterized by increased visceral adiposity, hyperlipidemia, insulin resistance, and hypertension [2]. The liver is the largest visceral organ for maintaining homeostasis in glucose, lipid, and protein. Hepatic steatosis is characterized by massive fat accumulation in the liver and thus is strongly related to several features of metabolic syndrome, including hyperlipidemia and insulin resistance [3]. Indeed, reduction or loss of insulin action in the liver leads to abnormally increased hepatic gluconeogenesis, glucose production, and lipogenesis, as well as decreased insulin clearance, hepatic glucose uptake, and lipolysis, consequently resulting in dyslipidemia [4].

Age and sex are physiologic factors that have strong association with the prevalence and features of metabolic syndrome. The state of estrogen deficiency as seen in postmenopausal women and the state of androgen deficiency as seen in aging men predispose older population to the metabolic syndrome and associated diabetes and cardiovascular diseases, indicating that sex hormones play important roles in regulating energy metabolism [5, 6]. Nonalcoholic fatty liver disease (NAFLD) disproportionally affects people with obesity, diabetes with insulin resistance, and dyslipidemia [79]. The prevalence of NAFLD varies among ethnicities, with the highest prevalence in Hispanics, correlated with the high prevalence of obesity and insulin resistance in this ethnic group, compared to whites and blacks [10]. Similar to the incidence of metabolic syndrome, the frequency of NAFLD varies between genders, with greater prevalence in men than in women among whites (42% in white men versus 24% in white women) but not in other ethnicities [10]. This is consistent with another epidemiology study showing that the rate of NAFLD is a little higher in men than in women with all ethnicities combined [7]. Interestingly, NAFLD is twice as common in postmenopausal women as in premenopausal women whose estrogen levels are higher than postmenopausal women [7, 11], which suggests the protective role of estrogens in NAFLD [12, 13].

In general, androgens are considered as hormones of the male sex due to their masculinizing effects and their roles in regulating male sexual behavior, whereas estrogens are considered as hormones of the female sex due to their roles in regulating female reproductive physiology and behaviors, although all sex hormones are present in both males and females, albeit at different levels between these two sexes. The most important biologically relevant forms of estrogens and androgens in humans are estradiol (E2) and testosterone, respectively. Understanding of how estrogens and androgens regulate energy metabolism via their receptors may shed light on potential pharmaceutical applications. In the present review, we discuss the roles of estrogens and androgens in regulating liver glucose and lipid homeostasis in rodents and humans. We also deliberate the distinct, important effects of estrogen receptors (ERs) and androgen receptors (ARs) on the regulation of liver metabolism.

In both males and females, E2 is derived from the aromatization of testosterone. In premenopausal women, E2 is mainly synthesized from cholesterol in the ovaries, with E2 concentration being approximately 5 times higher than that in men, while in postmenopausal women E2 is primarily converted from testosterone by aromatase in peripheral tissues, such as adipose tissue, adrenal glands, bones, vascular endothelium, and smooth muscle [14], with E2 concentration being similar compared with men (http://www.hemingways.org/GIDinfo/hrt_ref.htm).

Estrogens act on ERs, including classic nuclear receptors ER- and ER-, and membrane-bound receptors, including G protein-coupled ER (GPER, also known as GPR 30) and membrane-associated ER- and ER- variants [15]. All these nuclear and membrane ER subtypes are expressed in the livers of male and female humans and rodents, but at a lower level compared with reproductive organs such as uterus, prostate, testis, ovary, and breast [1618]. ER- is less abundant in liver cells than ER- [19, 20] and GPER (unpublished observation).

One study by Lax et al. determines levels of ERs in male and female rat livers and reports that the levels of nuclear ERs are not sex dependent but are age dependent, as levels of ERs are similar between male and female rats and vary with the course of life in a comparable manner in males and females [21]. Specifically, levels of ERs in the liver of both male and female rats are the highest during the perinatal period, decline till the onset of puberty, and increase to reach postpubertal peak. Additionally, levels of ERs are maintained as a stable level across the estrous cycles of female rats [21]. Consistently, Eisenfeld group has reported that ER concentration in the rat liver increases evidently at puberty [22]. Ovariectomy (OVX), a procedure that removes ovaries and thus majority of endogenous estrogens, is a suitable preclinical model to study postmenopausal diseases. Liver ER- expression does not change following OVX; however, it significantly increases by E2 treatment at a superphysiological level in rats with OVX, higher than sham-operated rats with intact ovaries and normal levels of endogenous estrogens [23]. These studies indicate that ER- expression in the liver is similar between gonad intact males and females and remains stable in postmenopausal females but could increase following hormone replacement therapy or during puberty. There is no available literature showing changes of expression of ER- and GPER during menstrual period or postmenopausal stage, and these questions remain unknown.

Males also express ERs in the liver, and aromatase metabolizes androgens to generate E2 and other estrogen metabolites locally in many target tissues. A growing body of evidence suggests that estrogens also have important metabolic functions in males. The aromatization of testosterone to E2 is beneficial for preventing intra-abdominal adiposity in men, demonstrated by a clinical study showing increased intra-abdominal fat in men by reduced estrogens due to aromatase inhibition [24]. The effects of estrogens on male and female reproductive organs have been extensively studied, but the beneficial effects of estrogens in nonclassical endocrine targets including the liver are less appreciated. We will discuss how hepatic estrogen signaling via ERs regulates metabolism in male and female animal and human models.

Upon estrogen binding, classic estrogen nuclear receptors ER- and ER- form homo- or heterodimers and bind to estrogen response element (ERE) in target gene promoters or to other transcription factors, such as activator protein-1 (AP-1) and stimulating protein-1 [25], to induce expression of target genes. The genomic action following E2-ER binding varies as the level of sex hormone changes. Specifically, the transcriptional activity of ER- alters during the 4-day estrous cycle, demonstrated by using ERE-luciferase reporter mice which have luciferase reporter controlled by activated ERs. The peak of the transcriptional activity of ER- in the liver occurs in proestrus [26], indicating dynamics of ER- transcriptional activity that is possibly modulated by different concentration of estrogens [27]. These findings suggest that liver ER- could recognize the changes in circulating E2 levels and response to reproductive cues during transition of different stages of the estrous cycles and select appropriate genetic programs to adapt the hepatic metabolism to the energy requirements of each stage. Thus, the hepatic ER- could serve as a peripheral coordinator of energy homeostasis. ER- also exists in the form of membrane-associated receptor. There are many lines of evidence showing that the full length ER- and truncated ER- may exert actions via nongenomic signaling which is faster than the classic genomic signaling. Such nongenomic signaling usually involves activation of intracellular second messenger systems, such as protein kinase A (PKA), protein kinase C, and mitogen-activated protein kinase (MAPK)/extracellular signal-regulated protein kinase (ERK) [2830]. GPER is structurally unrelated to ER- and ER- and is a seven-transmembrane domain G protein-coupled receptor located at the cell membrane and endoplasmic reticulum membrane. GPER is reported to rapidly activate different nongenomic estrogen signaling pathways, including PKA, MAPK/ERK, and phosphoinositide 3-kinase (PI3K) [31] (Figure 1).

Females, as compared with males, tend to store more energy in subcutaneous fat instead of in visceral fat. The liver is a key visceral organ for controlling energy storage, as the liver has high capacity for lipid transport, de novo lipogenesis, lipid oxidation, and lipolysis. Liver steatosis, as seen in the nonalcoholic fatty liver disease (NAFLD), is due to the excess of triglyceride (TG) accumulation within the hepatocytes. Incidence of hepatic steatosis is frequently associated with low levels of high density lipoprotein cholesterol (HDL-C) and high levels of low density lipoprotein cholesterol (LDL-C) in the circulation. Epidemiological studies have showed higher plasma level of LDL-C and lower plasma level of HDL-C in men and postmenopausal women compared with premenopausal women, suggesting that lower circulating estrogen levels may promote fat deposition in the liver [32]. Further evidence is supported by using OVX mouse model combined with pair-feeding between sham operation and OVX groups. Removal of the ovaries and thus the majority of endogenous estrogens in female mice results in increased fat proportion in the liver even when they are pair-fed with the same amount of calories as females with intact ovaries, which indicates the direct role of estrogens in inhibiting lipogenesis in the liver, rather than the secondary effects to OVX induced overfeeding [33]. In another E2-deficient aromatase knockout (ArKO) mouse model, spontaneous obesity and hepatic steatosis result from impaired fatty acid -oxidation and elevated fatty acid synthase (FAS) in the liver in both female and male mice [34]. These findings are further supported by previous studies demonstrating that E2 inhibits lipogenic gene expression and lipid uptake in the liver by decreasing lipoprotein lipase activity, as well as promoting lipolysis by increasing expression of hormone-sensitive lipase and adipose TG lipase in the liver [35, 36].

ER- is the predominant ER subtype presented in both male and female hepatocytes [19, 20]. Estrogen signaling is important in both males and females in the regulation of lipogenesis, demonstrated by using animal models and human studies. Specifically, estrogens regulate the activity and expression of lipogenic genes to directly inhibit lipogenesis in several animal species [37, 38]. Liver enzymes may also be regulated by circulating estrogen levels. One study of genome-wide analyses demonstrated that the subtle oscillations of estrogens occurring during the estrous cycle are sufficient to influence liver gene expression, and that ERs are involved in the pulsatile synthesis of fatty acids and cholesterol in the liver [27]. Thus this study demonstrated the importance of the maintenance of estrogen oscillation to limit fat deposition in the hepatic tissues in females [27]. Additionally, treatment of the specific ER- agonist PPT decreases weight, fat mass, and TG in the liver in both wild-type mice and obese ob/ob mice [39, 40]. Thus, the metabolically protective effect of estrogen may be attributed to estrogen signaling via ER- [41].

This is further demonstrated by investigation of estrogen and estrogen signaling using knockout or transgenic animal models. Male and female ER- knockout mice exhibit hepatic steatosis by increasing gene expression of lipogenic transcription factors such as sterol regulatory element binding protein 1c (SREBP-1c) and decreasing lipid transport genes [42, 43]. Mice with liver-specific ER- knockout [44, 45] or liver-specific GPER knockout [46] show increases in fat accumulation in the liver and develop disturbed insulin signaling under high-fat diet (HFD) feeding. Thus, hepatic steatosis has been observed in both of the above genetic models, one with liver-specific ER- knockout with functional GPER and the other with liver-specific GPER knockout with functional ER-. Thus, although it is widely recognized that estrogens regulate liver lipid metabolism and reduce triglyceride accumulation in the liver mainly via ER- [47, 48], both ER- and GPER are required to be present in the liver to maintain lipid homeostasis. Estrogen is produced in males by aromatization of testosterone. Male but not female mice in which the aromatase gene has been deleted (ArKO) develop hepatic steatosis that can be normalized by estrogen treatment [49]. Thus, E2 treatment reduces fatty acid synthesis and lipid accumulation and prevented NAFLD in castrated male rats [50].

Hepatic TG and diacylglyceride increase in the livers of ER- knockout male mice under HFD feeding, explained by dysregulation of insulin-stimulated ACC phosphorylation and DGAT1/2 protein levels [44]. Interestingly, a recent study using specific plasma membrane ER- knockout has demonstrated that it is the membrane-localized ER-, but not nuclear ER-, that is responsible for protection from hyperlipidemia by decreasing expressions of many hepatic genes involved in lipid synthesis, at least in female mice with OVX [51]. Although ER- is antilipogenic in the liver, the role of ER- in the liver is not consistent in the literature. ER- deficient mice have higher body weight but lower liver weight due to increased insulin sensitivity and decreased TG accumulation in the liver [52], indicating that ER- might be lipogenic and diabetogenic in the liver. Opposite finding has been reported where, different from treatment of E2 or ER- agonists that decrease hepatic PPAR expression, treatment of ER- agonist 8-VE2 comparably elevates PPAR expression to the same mRNA level as non-drug treated group in the liver of HFD-fed female rats with OVX [53]. Interestingly, all treatments of E2, ER- agonist, or ER- agonist are capable of reducing TG accumulation in the liver of HFD-fed rats with OVX [53]. Thus, the mechanism for reduced hepatic lipid accumulation in both suppressed ER- signaling as seen in ER- knockout mice [52] and activated ER- signaling as seen in ER- agonist-treated rats [53] is awaiting further elucidation. Hepatic steatosis is also found in GPER deficient female mice fed with HFD rather than male mice [46]. Although both 6-month-old female and male GPER KO mice display increased body weight, only female mice had glucose intolerance, while male mice developed glucose intolerance at the age of 18 months [54]. Furthermore, GPER agonist G-1 decreases fatty acid synthesis and TG accumulation in both human and rodent pancreatic cells [55], but the effect of G-1 treatment on lipid metabolism in the liver is not clear. Both liver GPER and membrane-associated ER- are critical for liver lipid metabolism. However, it is possible that GPER has greater impacts on male lipid regulation [54], whereas membrane-associated ER- variant [51] may have greater impacts on female lipid regulation, as female livers have markedly higher expression of all three membrane-associated ER- variants compared with male livers [56].

Hepatic glucose homeostasis is determined by glucose uptake and glucose production. The major glucose transporter (GLUT) in the liver is GLUT2 that bidirectionally transports glucose across liver cell plasma membrane, efflux of glucose formed from gluconeogenesis or glycogenolysis out of liver cells, and uptake of circulating glucose into liver cells. Hepatic GLUT2 is upregulated by glucose, FAS, and insulin [57]. Since estrogen treatment has been shown to increase insulin synthesis and release [58], estrogens might indirectly increase GLUT2 expression in the liver, which has not been demonstrated yet. A recent study demonstrates that it is estriol, instead of E2, that downregulates GLUT2 in pregnant women during late stages of pregnancy whose peak postprandial glucose levels are much lower than glucose levels of healthy nonpregnant women [59]. Estrogens are also important in hepatic insulin clearance. Several lines of evidence show that intravenous conjugated estrogen treatment or low dose of oral contraceptive does not significantly alter insulin sensitivity but slightly increases hepatic insulin clearance in postmenopausal women [60, 61]. Estrogens reduce gluconeogenesis and increase glycogen synthesis and storage in the liver, lowering circulating glucose level [43, 62]. Additional observations using rodents with OVX that lacks majority of endogenous estrogens support the notion that estrogens lower glucose levels [63, 64]. A recent study reports increased glucagon signaling due to increased amount of glucagon receptor that accounts for enhanced glucose production, accompanied with increased gluconeogenic enzymes in rats with OVX [65]. Interestingly, such changes cannot be prevented by E2 replacement, which indicates that disrupted liver glucose homeostasis following OVX is not merely caused by deficiency of endogenous E2 [65] but could be caused by deficiency of other ovarian hormones such as progesterone. Although classic nuclear progesterone receptor has not been found in the liver [22, 66], progestins can either bind to membrane-bound progesterone receptors [67] or bind to ARs [22] in human liver and carry metabolic effects. On the other hand, estrogens are also found to facilitate epinephrines action via 2-adrenergic receptor in regulating glycogenolysis and gluconeogenesis in the rat liver to increase circulating glucose level [68].

Estrogen signaling is important in both males and females in the regulation of glucose homeostasis, improving glucose tolerance and insulin sensitivity, demonstrated by using animal models and human studies [6971]. Additionally, although estrogens do not affect hepatic glucose metabolism in vivo, estrogens increase insulin receptor to enhance glucose metabolism in vitro [72, 73].

ER- deficient mice exhibit significantly impaired glucose tolerance and hepatic insulin resistance, while ER- deficient mice exhibit normal glucose tolerance, suggesting that ER- instead of ER- plays an important role in the regulation of hepatic glucose homeostasis [43]. The importance of ER- in the regulation of hepatic glucose tolerance is further supported by inadequate suppression of hepatic glucose production during hyperinsulinemic clamp study in ER- deficient mice [74]. Although impaired glucose tolerance is seen in GPER1 knockout mice, GLUT2 and glucokinase are not affected [1], and glucose production in liver has not been measured yet. Hepatic PPAR expression rises markedly following OVX in HFD-fed rats [53]. The rats treated with E2 or ER- agonist have reduced PPAR expression in the liver, whereas the rats treated with ER- agonist maintain a similarly high mRNA level of PPAR as non-drug treated HFD-fed rats with OVX. The sustained hepatic PPAR gene expression correlates with increased glucose uptake into the liver of rats with OVX [53].

Dyslipidemia is determined by decreased HDL but increased LDL and TG in the blood. The liver is the principal organ for cholesterol de novo biosynthesis, which is catalyzed by the rate limiting enzyme 3-hydroxy-3-methyl-glutaryl-CoA reductase (HMGR). The SREBP-1c is the master regulator of cholesterol by stimulating transcription of LDL and HMGR [75]. Postmenopausal women have elevated LDL and VLDL and lower HDL [76].

A previous in vitro study points out that HMGR promoter is induced by estrogen treatment in the breast cancer cell line MCF-7 but not in any hepatic cell line [77], indicating differential regulation of HMGR by estrogens among different tissues. Estrogen treatment does not increase cholesterol synthesis in liver cells in vitro. In an in vivo study using castrated male rats, DHT, but not E2, treatment increases phosphorylation of HMGR to decrease cholesterol synthesis in the liver [50]. Thus, at least in castrated male rats, androgen action is associated with downregulation of cholesterol biosynthesis in the liver.

Estrogens also decrease LDL level and increase HDL to promote cholesterol secretion into bile in postmenopausal women [78]. Total cholesterol and LDL are elevated in ArKO mice with E2 deficiency [79]. Increased hepatic HMGR activity and subsequently increased levels of cholesterol and LDL are seen in rats with OVX with reduced level of endogenous estrogens [65]. Estrogen replacement in both ArKO mice and rats with OVX normalizes the levels of LDL and cholesterol. The above mentioned cell, animal, and human studies collectively indicate important roles of estrogens in reducing LDL and increasing HDL.

ER- is able to protect the liver from hypercholesterolemia [47, 48]. To support this, lack of ER- (whole body) is associated with increased expression of genes involved in lipid biosynthesis and lipid metabolism [43]. A male patient without functional ER- has been reported with dyslipidemia [80], supporting the importance of ER- in regulating cholesterol homeostasis. Consistently, the expressions of ER- (and AR) and phosphorylated HMGR are significantly reduced in the human liver samples from male severe steatotic NAFLD patients compared with the liver samples from subnormal men [50].

Aromatase deficient mice without endogenous estrogen production exhibit obesity [79] and dyslipidemia [81] and mice with liver-specific ER- knockout accumulate liver triglycerides and diacylglycerides [42, 43]. In contrast, ER- agonist PPT increases the expression of genes involved in lipid oxidation and metabolism [82]. Additionally, ER- deficient mice and ER- and ER- double knockout mice display increased body fat and serum cholesterol level, but these changes are not found in ER- deficient mice [83].

In GPER KO mice, LDL levels increase approximately by 200%, but HDL levels do not show any significant differences from WT, which indicates that GPER mainly regulates the LDL metabolism instead of HDL [54]. A recent study shows that human individuals with hypofunctional P16L genetic variant of GPER have increased plasma LDL [84, 85]. In contrast, GPER activation upregulates LDL receptor expression in the liver via downregulation of proprotein convertase subtilisin kexin type 9 to enhance LDL metabolism [85]. Interestingly, animals with estrogen deficiency do not increase cholesterol synthesis; instead they decrease cholesterol catabolism by reducing activity of 7-hydroxylase, the enzyme that catalyzes the initial step in cholesterol catabolism and bile acid synthesis in calcium supplementation-induced hypercholesterolemia [86]. This study further demonstrates that estrogen treatment protects against increase in circulating level of cholesterol by activating of GPER [86].

The major circulating androgens include dehydroepiandrosterone, androstenedione, testosterone, and dihydrotestosterone (DHT), in descending order of circulating concentrations. Only testosterone and DHT bind to the AR whereas the rest are considered as proandrogens. Within target cells, testosterone can be converted to active androgen DHT via 5-reductase or converted to E2 by aromatase.

ARs are expressed in the liver of male and female humans and rodents, and AR expression in the liver is sex dependent. In adult rats, basal AR expression in the liver of male rats is about 20 times higher than that in the liver of female rats [87]. AR expression is also age dependent in the liver of either sex, which is very low, almost undetectable, before puberty, increases in postpubertal life, and gradually declines during aging, reaching an almost nondetectable level after about 2224 months of age in rats [88]. The sex- and age-dependent AR expression in the liver is programed by a regulatory element in the AR gene promoter [89].

There are isoforms of ARs which are AR-A with N-terminal truncated that resulted from proteolysis and AR-B with full length [90, 91]; among these two AR isoforms, the AR-B with full length is more potent than AR-A [92]. It is not clear, however, which isoform of AR is dominant in the liver. Androgens, like estrogens, work on both nuclear and nonnuclear receptors. The genomic effect of androgens is achieved through activation of nuclear receptor, followed by binding to specific DNA known as androgen response element (ARE) motifs in its target gene [93]. AR can recruit other transcription factors such as AP-1, nuclear factor-B, sex-determining region Y, and the E26 transformation-specific family of transcription factors and bind to DNA regions other than ARE, to participate in transcription activation of many other genes [94]. The nonnuclear receptor of androgens function is independent of DNA interaction and is more rapid by interacting with cytoplasmic signal transduction pathways, including PKA and MAPK/ERK [95] (Figure 1). The AR knockout animals are well developed, but the membrane-only AR knockout animals are not established yet, and that is why the exact role of membrane AR in liver metabolism is unclear.

Many studies have shown that androgens and androgen signaling suppress the development of hepatic steatosis [96, 97]. One population-based cross-sectional study has reported a close association between low serum testosterone level and hepatic steatosis in men [98]. Mice with 5-reductase knockout do not covert testosterone to DHT. These mice upregulate expression for the genes involved in lipid storage and downregulate genes for fatty acid oxidation and accumulate lipid in their livers when they are fed with HFD [99]. An inhibitor of 5-reductase induces liver steatosis in male obese Zucker rats [100]. Therefore, normal level of active androgen is critical to prevent liver steatosis.

Besides androgen level, ARs are also critical in maintaining lipid metabolism in the liver. Testicular feminized (Tfm) mice with nonfunctional AR and very low serum testosterone levels greatly increase HFD feeding-induced hepatic lipid deposition compared with control male mice with functional AR and normal circulating levels of testosterone. Replacement of testosterone reduces lipid deposition in the liver of Tfm mice to a similar level to control males [101]. Moreover, Kelly et al. [101] found that the expressions of key regulatory enzymes for fatty acid synthesis, including acetyl-CoA carboxylase (ACC) and FAS, are elevated in placebo-treated Tfm mice comparing with placebo-treated wild-type littermates and Tfm mice receiving testosterone treatment, indicating that the action of androgens on lipid deposition is independent of AR and at least partially via affecting key regulatory lipogenic enzymes to protect against hepatic steatosis [101]. Male but not female hepatic ArKO mice fed with a normal chow diet developed liver steatosis at 10 months with reduced fatty acid oxidation and increased de novo fatty acid synthesis [102]. Thus, males with either functional AR or normal circulating testosterone level would maintain normal level of fatty acid synthesis and avoid increased lipid deposition in the liver.

Although many studies have shown that androgens protect against NAFLD [50, 103], other studies have reported an opposite finding that androgens promote NAFLD development and progression [104, 105]. The inconsistencies might be due to different animal models employed and different treatments utilized in various studies. The findings reported by Mnzker et al. indicate that the testosterone/DHT ratio is more important for NAFLD development and progression than concentrations of testosterone and/or DHT [106]. In contrast, the role of AR in hepatic steatosis is less controversial. The total AR knockout mice develop liver steatosis and insulin resistance in both male and female mice [107]. Hepatic AR knockout mice with HFD feeding also show hepatic steatosis and insulin resistance, via upregulation of hepatic expression of SREBP-1c, ACC, and PPAR to increase lipid synthesis and downregulation of PPAR to decrease fatty acid oxidation; interestingly, such effects are evident in males but absent in females [102, 108]. Thus, hepatic AR plays more critical roles in maintaining liver lipid metabolism in males than in females.

Testosterone is either converted to E2 binding to ERs or converted to DHT binding to ARs. From the above studies, ARs are vital in regulating liver lipid homeostasis in both males and females [107], although hepatic ARs have greater impact in males than in females [102, 108]. In order to test the role of androgen-AR signaling in female metabolic process, Kanaya et al. replace DHT in female mice with OVX and find that those mice accumulate greater amount of fat in the liver and develop other symptoms and signs of metabolic dysfunction when these mice are fed with either a standard chow diet or HFD [109]. Therefore, androgen action has great impact on lipid metabolism in female livers.

Women have lower basal levels of androgens compared with males, and increased androgen level can affect metabolism in women. The role of androgens in females is not well established, but many lines of evidence indicate that hyperandrogenism in women with polycystic ovary syndrome (PCOS) increases risk of developing NAFLD. NAFLD is frequently present in PCOS women with excessive production of androgens by the ovaries and thus elevated circulating level of androgens, suggesting that abnormally high level of androgens in women may contribute to increased fat storage in the liver. It is noteworthy that the risk for NAFLD in women with PCOS is independent of obesity or insulin resistance but is triggered directly by the hepatotoxic, destructive effect in the liver, indicated by elevated level of alanine aminotransferase [110]. To summarize, normal level and signaling of androgens prevent hepatic lipid accumulation in males, while androgen deficiency in males is associated with fatty liver. Abnormally high level of androgens increases lipid deposition in the liver in females. Androgens therefore have differential effects in men and women.

Testosterone levels are lower in diabetic men than nondiabetic men [111]. Androgen deprivation therapy for prostate cancer patients lowers their circulating testosterone level and increases their risk of diabetes [112, 113] and not only increases circulating level of glucose but also diminishes pancreatic cell function [114]. Testosterone treatment markedly reduces circulating levels of glucose and TG in men [115].

GLUT2 directionally transports glucose across liver cell plasma membrane to maintain glucose homeostasis, as mentioned above in Section 2.3. Upregulation of GLUT2 plays a more critical role in regulating glucose export out of, rather than regulating glucose import into, the liver. It has been reported that blood glucose level, along with the mRNA and protein levels of GLUT2 in the liver, significantly increases following castration in male rats with deficiency of endogenous androgens [116]. Supplementation of testosterone or a combination of testosterone with E2 normalizes GLUT2 mRNA and protein levels in the livers of castrated rats, whereas treatment of E2 alone does not have any effect [116]. These findings suggest that testosterone maintains glucose homeostasis by regulating hepatic glucose output, and testosterone deprivation due to castration increases hepatic glucose output, induces hyperglycemia, and develops symptoms seen in type 2 diabetes and metabolic syndrome. Testosterone replacement restores GLUT2 mRNA and protein levels suggesting that testosterone may have a direct effect on GLUT2 transcription and translation of mRNA. Although the presence of ARE has not been identified in the promoter region of GLUT2, AR could function as a ligand-activated transcription factor by itself [117] or bind to some other coactivators [118, 119] to increase GLUT2 expression.

In contrast, estrogens have little effect on hepatic GLUT4 and insulin receptor in male rats, but estrogens increase level of insulin receptor in HepG2, a liver cancer cell line [72]. Interestingly, insulin receptor mRNA level as well as insulin sensitivity is increased in a human liver cell line when being treated with testosterone [73]. Similarly, replacement of testosterone in castrated male mice also increases insulin receptor mRNA and protein levels in the liver and normalizes castration-induced glucose metabolic impairment [120]. Treatment of testosterone induces glycogen synthesis in both intact and castrated male rats [108, 121].

High testosterone level is associated with a low risk of diabetes in men, whereas it is associated with a high risk of diabetes in women [111, 122124]. Excess androgen in women with PCOS impairs hepatic glucose metabolism by decreasing insulin-stimulated glucose uptake and glycogen synthesis and predisposes women with PCOS to insulin resistance [125, 126]. Metformin, the most commonly used first-line drug to treat diabetes, is found to be effective to treat NAFLD and also suppresses the serum androgen concentration in PCOS patients [127, 128]. Increased androgen activity in postmenopausal women correlates with impaired glucose tolerance [129, 130].

To summarize, testosterone in males favors hepatic glucose metabolism, whereas testosterone in females impairs it. Thus, androgens in males and females differentially regulate glucose homeostasis.

Old men have increased risks of developing dyslipidemia with increased serum cholesterol and LDL levels, and decreased HDL level, and testosterone replacement reverses such dyslipidemia [108]. Hepatic scavenger receptor class B member 1 (SR-1B) is important in regulating cholesterol uptake from circulating HDL. DHT treatment in castrated obese mice increases SR-1B compared with vehicle-treated castrated mice. At the same time, LDL secretion is decreased by DHT treatment. Cholesterol 7-hydroxylase, a key enzyme in bile formation and cholesterol removal, is also decreased after DHT treatment. All these above results provide a comprehensive explanation for how chronic androgen replacement can decrease serum levels of cholesterol and LDL via enhancing liver cholesterol uptake and via suppressing cholesterol removal, which in turn increases liver cholesterol accumulation [120]. A clinical study, however, shows that a single dose of testosterone treatment increases the serum cholesterol level after two days by increasing the expression of HMGR, the rate limiting enzyme for cholesterol de novo biosynthesis in the liver, but 15 days after the testosterone administration the cholesterol levels in the volunteers were back to baseline levels [131]. The mechanisms for the androgen induced upregulation of HMGCR transcription as well as the physiological consequences have not been investigated and need to be further elucidated.

The metabolic syndrome and its related diseases, such as obesity and diabetes, increase the health problems worldwide. The liver is the largest organ in the body that regulates lipid, glucose, and cholesterol homeostasis. Hepatic steatosis is one of the major manifestations of metabolic syndrome. Several lines of epidemiological data have suggested that sex hormones are associated in fatty different types of receptors. Estrogens seem to play protective roles against hepatic fat accumulation via suppressing lipogenesis and gluconeogenesis and promoting lipolysis and glycogen storage. Interestingly, estrogens increase both cholesterol synthesis and secretion. ER- and its membrane form are more important in regulating energy homeostasis than ER-. GPER and its roles in energy homeostasis are currently under intensive investigation; however, there is less evidence about the role of GPER in the liver compared with classic nuclear estrogen receptors. Since the GPER specific agonist and antagonist have been developed, further studies should apply these new chemicals to examine the role of GPER in liver energy homeostasis, yet the underlying molecular mechanisms are still unclear and longing for further investigation.

We review and discuss the roles played by estrogens, androgens, and their receptors in regulating liver energy homeostasis (Figure 2). The action mechanisms of estrogens are complicated in the body, as they work through multiple different subtypes of estrogen receptors. Estrogens promote liver glucose storage via increasing glucose transporters and glycogen synthesis and suppress liver glucose production via decreasing gluconeogenesis. Estrogens also actively participate in maintaining lipid and cholesterol balance and play protective roles against hepatic lipid accumulation, via suppressing lipogenesis, lipid uptake, and cholesterol synthesis and promoting lipolysis and cholesterol removal. Interestingly, estrogens increase both cholesterol synthesis and secretion. Classic nuclear ER- and its membrane form are more important in regulating energy homeostasis than ER-. GPER and its roles in energy homeostasis are currently under intensive investigation; however, there is less evidence about the roles of GPER in the liver compared with classic nuclear ERs. Since the GPER specific agonist and antagonist have been developed, further studies should apply these new chemical compounds to examine the role of GPER in liver energy homeostasis.

Androgens and nuclear AR have been shown to increase insulin receptor, decrease lipogenesis, and promote cholesterol storage in the liver. The membrane AR, however, is not well studied, which is also a potential research area to explore. It must be emphasized that the integration of nongenomic effects via membrane receptor signaling and genomic effects via nuclear receptor signaling of sex hormones is critical to produce the final sex hormone cellular outcomes.

Further investigation about differential androgen action in males and females is needed. Androgen deficiency, or excessive androgens as seen in women with PCOS, the most common endocrine disorder and cause of infertility among women of reproductive age, is closely associated with disturbed lipid and glucose metabolism in the liver.

The authors declare that there is no conflict of interests regarding the publication of this paper.

The authors thank funding from the Sigma Xi (G2012161930 and G20141015719335 to Minqian Shen), National Institutes of Health (R15 DK090823 to Haifei Shi), and Madalene and George Shetler Diabetes Research Award (to Haifei Shi). They also thank the Department of Biology at Miami University for providing Graduate Assistantship to Minqian Shen.

Go here to see the original:
Sex Hormones and Their Receptors Regulate Liver Energy Homeostasis

Gene therapy – Wikipedia

Medical field

Gene therapy is a medical field which focuses on the genetic modification of cells to produce a therapeutic effect[1] or the treatment of disease by repairing or reconstructing defective genetic material.[2] The first attempt at modifying human DNA was performed in 1980, by Martin Cline, but the first successful nuclear gene transfer in humans, approved by the National Institutes of Health, was performed in May 1989.[3] The first therapeutic use of gene transfer as well as the first direct insertion of human DNA into the nuclear genome was performed by French Anderson in a trial starting in September 1990. It is thought to be able to cure many genetic disorders or treat them over time.

Between 1989 and December 2018, over 2,900 clinical trials were conducted, with more than half of them in phase I.[4] As of 2017, Spark Therapeutics' Luxturna (RPE65 mutation-induced blindness) and Novartis' Kymriah (Chimeric antigen receptor T cell therapy) are the FDA's first approved gene therapies to enter the market. Since that time, drugs such as Novartis' Zolgensma and Alnylam's Patisiran have also received FDA approval, in addition to other companies' gene therapy drugs. Most of these approaches utilize adeno-associated viruses (AAVs) and lentiviruses for performing gene insertions, in vivo and ex vivo, respectively. AAVs are characterized by stabilizing the viral capsid, lower immunogenicity, ability to transduce both dividing and nondividing cells, the potential to integrate site specifically and to achieve long-term expression in the in-vivo treatment. (Gorell et al. 2014) ASO / siRNA approaches such as those conducted by Alnylam and Ionis Pharmaceuticals require non-viral delivery systems, and utilize alternative mechanisms for trafficking to liver cells by way of GalNAc transporters.

The concept of gene therapy is to fix a genetic problem at its source. If, for instance, a mutation in a certain gene causes the production of a dysfunctional protein resulting (usually recessively) in an inherited disease, gene therapy could be used to deliver a copy of this gene that does not contain the deleterious mutation and thereby produces a functional protein. This strategy is referred to as gene replacement therapy and is employed to treat inherited retinal diseases.[5][6]

While the concept of gene replacement therapy is mostly suitable for recessive diseases, novel strategies have been suggested that are capable of also treating conditions with a dominant pattern of inheritance.

Not all medical procedures that introduce alterations to a patient's genetic makeup can be considered gene therapy. Bone marrow transplantation and organ transplants in general have been found to introduce foreign DNA into patients.[13]

Gene therapy was conceptualized in 1972, by authors who urged caution before commencing human gene therapy studies.

The first attempt, an unsuccessful one, at gene therapy (as well as the first case of medical transfer of foreign genes into humans not counting organ transplantation) was performed by Martin Cline on 10 July 1980.[14][15] Cline claimed that one of the genes in his patients was active six months later, though he never published this data or had it verified[16] and even if he is correct, it's unlikely it produced any significant beneficial effects treating beta thalassemia.[medical citation needed]

After extensive research on animals throughout the 1980s and a 1989 bacterial gene tagging trial on humans, the first gene therapy widely accepted as a success was demonstrated in a trial that started on 14 September 1990, when Ashanthi DeSilva was treated for ADA-SCID.[17]

The first somatic treatment that produced a permanent genetic change was initiated in 1993.[18] The goal was to cure malignant brain tumors by using recombinant DNA to transfer a gene making the tumor cells sensitive to a drug that in turn would cause the tumor cells to die.[19]

The polymers are either translated into proteins, interfere with target gene expression, or possibly correct genetic mutations. The most common form uses DNA that encodes a functional, therapeutic gene to replace a mutated gene. The polymer molecule is packaged within a "vector", which carries the molecule inside cells.[medical citation needed]

Early clinical failures led to dismissals of gene therapy. Clinical successes since 2006 regained researchers' attention, although as of 2014[update], it was still largely an experimental technique.[20] These include treatment of retinal diseases Leber's congenital amaurosis[5][21][22][23] and choroideremia,[24] X-linked SCID,[25] ADA-SCID,[26][27] adrenoleukodystrophy,[28] chronic lymphocytic leukemia (CLL),[29] acute lymphocytic leukemia (ALL),[30] multiple myeloma,[31] haemophilia,[27] and Parkinson's disease.[32] Between 2013 and April 2014, US companies invested over $600 million in the field.[33]

The first commercial gene therapy, Gendicine, was approved in China in 2003, for the treatment of certain cancers.[34] In 2011, Neovasculgen was registered in Russia as the first-in-class gene-therapy drug for treatment of peripheral artery disease, including critical limb ischemia.[35] In 2012, Glybera, a treatment for a rare inherited disorder, lipoprotein lipase deficiency, became the first treatment to be approved for clinical use in either Europe or the United States after its endorsement by the European Commission.[20][36]

Following early advances in genetic engineering of bacteria, cells, and small animals, scientists started considering how to apply it to medicine. Two main approaches were considered replacing or disrupting defective genes.[37] Scientists focused on diseases caused by single-gene defects, such as cystic fibrosis, haemophilia, muscular dystrophy, thalassemia, and sickle cell anemia. Glybera treats one such disease, caused by a defect in lipoprotein lipase.[36]

DNA must be administered, reach the damaged cells, enter the cell and either express or disrupt a protein.[38] Multiple delivery techniques have been explored. The initial approach incorporated DNA into an engineered virus to deliver the DNA into a chromosome.[39][40] Naked DNA approaches have also been explored, especially in the context of vaccine development.[41]

Generally, efforts focused on administering a gene that causes a needed protein to be expressed. More recently, increased understanding of nuclease function has led to more direct DNA editing, using techniques such as zinc finger nucleases and CRISPR. The vector incorporates genes into chromosomes. The expressed nucleases then knock out and replace genes in the chromosome. As of 2014[update] these approaches involve removing cells from patients, editing a chromosome and returning the transformed cells to patients.[42]

Gene editing is a potential approach to alter the human genome to treat genetic diseases,[7] viral diseases,[43] and cancer.[44][45] As of 2020[update] these approaches are being studied in clinical trials.[46][47]

Gene therapy may be classified into two types:

In somatic cell gene therapy (SCGT), the therapeutic genes are transferred into any cell other than a gamete, germ cell, gametocyte, or undifferentiated stem cell. Any such modifications affect the individual patient only, and are not inherited by offspring. Somatic gene therapy represents mainstream basic and clinical research, in which therapeutic DNA (either integrated in the genome or as an external episome or plasmid) is used to treat disease.[48]

Over 600 clinical trials utilizing SCGT are underway[when?] in the US. Most focus on severe genetic disorders, including immunodeficiencies, haemophilia, thalassaemia, and cystic fibrosis. Such single gene disorders are good candidates for somatic cell therapy. The complete correction of a genetic disorder or the replacement of multiple genes is not yet possible. Only a few of the trials are in the advanced stages.[49][needs update]

In germline gene therapy (GGT), germ cells (sperm or egg cells) are modified by the introduction of functional genes into their genomes. Modifying a germ cell causes all the organism's cells to contain the modified gene. The change is therefore heritable and passed on to later generations. Australia, Canada, Germany, Israel, Switzerland, and the Netherlands[50] prohibit GGT for application in human beings, for technical and ethical reasons, including insufficient knowledge about possible risks to future generations[50] and higher risks versus SCGT.[51] The US has no federal controls specifically addressing human genetic modification (beyond FDA regulations for therapies in general).[50][52][53][54]

The delivery of DNA into cells can be accomplished by multiple methods. The two major classes are recombinant viruses (sometimes called biological nanoparticles or viral vectors) and naked DNA or DNA complexes (non-viral methods).[55]

In order to replicate, viruses introduce their genetic material into the host cell, tricking the host's cellular machinery into using it as blueprints for viral proteins. Retroviruses go a stage further by having their genetic material copied into the genome of the host cell. Scientists exploit this by substituting a virus's genetic material with therapeutic DNA. (The term 'DNA' may be an oversimplification, as some viruses contain RNA, and gene therapy could take this form as well.) A number of viruses have been used for human gene therapy, including retroviruses, adenoviruses, herpes simplex, vaccinia, and adeno-associated virus.[4] Like the genetic material (DNA or RNA) in viruses, therapeutic DNA can be designed to simply serve as a temporary blueprint that is degraded naturally or (at least theoretically) to enter the host's genome, becoming a permanent part of the host's DNA in infected cells.

Non-viral vectors for gene therapy[56] present certain advantages over viral methods, such as large scale production and low host immunogenicity. However, non-viral methods initially produced lower levels of transfection and gene expression, and thus lower therapeutic efficacy. Newer technologies offer promise of solving these problems, with the advent of increased cell-specific targeting and subcellular trafficking control.

Methods for non-viral gene therapy include the injection of naked DNA, electroporation, the gene gun, sonoporation, magnetofection, the use of oligonucleotides, lipoplexes, dendrimers, and inorganic nanoparticles.

More recent approaches, such as those performed by companies such as Ligandal, offer the possibility of creating cell-specific targeting technologies for a variety of gene therapy modalities, including RNA, DNA and gene editing tools such as CRISPR. Other companies, such as Arbutus Biopharma and Arcturus Therapeutics, offer non-viral, non-cell-targeted approaches that mainly exhibit liver trophism. In more recent years, startups such as Sixfold Bio, GenEdit, and Spotlight Therapeutics have begun to solve the non-viral gene delivery problem. Non-viral techniques offer the possibility of repeat dosing and greater tailorability of genetic payloads, which in the future will be more likely to take over viral-based delivery systems.

Companies such as Editas Medicine, Intellia Therapeutics, CRISPR Therapeutics, Casebia, Cellectis, Precision Biosciences, bluebird bio, and Sangamo have developed non-viral gene editing techniques, however frequently still use viruses for delivering gene insertion material following genomic cleavage by guided nucleases. These companies focus on gene editing, and still face major delivery hurdles.

BioNTech, Moderna Therapeutics and CureVac focus on delivery of mRNA payloads, which are necessarily non-viral delivery problems.

Alnylam, Dicerna Pharmaceuticals, and Ionis Pharmaceuticals focus on delivery of siRNA (antisense oligonucleotides) for gene suppression, which also necessitate non-viral delivery systems.

In academic contexts, a number of laboratories are working on delivery of PEGylated particles, which form serum protein coronas and chiefly exhibit LDL receptor mediated uptake in cells in vivo.[57]

In in vivo gene therapy, a vector (typically, a virus) is introduced to the patient, which then achieves the desired biological effect by passing the genetic material (e.g. for a missing protein) into the patient's cells. In ex vivo gene therapies, such as CAR-T therapeutics, the patient's own cells (autologous) or healthy donor cells (allogeneic) are modified outside the body (hence, ex vivo) using a vector to express a particular protein, such as a chimeric antigen receptor.[58]

In vivo gene therapy is seen as simpler, since it does not require the harvesting of mitotic cells. However, ex vivo gene therapies are better tolerated and less associated with severe immune responses.[59] The death of Jesse Gelsinger in a trial of an adenovirus-vectored treatment for ornithine transcarbamylase deficiency due to a systemic inflammatory reaction led to a temporary halt on gene therapy trials across the United States.[60] As of 2021[update], in vivo and ex vivo therapeutics are both seen as safe.[61]

Athletes may adopt gene therapy technologies to improve their performance.[62] Gene doping is not known to occur, but multiple gene therapies may have such effects. Kayser et al. argue that gene doping could level the playing field if all athletes receive equal access. Critics claim that any therapeutic intervention for non-therapeutic/enhancement purposes compromises the ethical foundations of medicine and sports.[63]

Genetic engineering could be used to cure diseases, but also to change physical appearance, metabolism, and even improve physical capabilities and mental faculties such as memory and intelligence. Ethical claims about germline engineering include beliefs that every fetus has a right to remain genetically unmodified, that parents hold the right to genetically modify their offspring, and that every child has the right to be born free of preventable diseases.[64][65][66] For parents, genetic engineering could be seen as another child enhancement technique to add to diet, exercise, education, training, cosmetics, and plastic surgery.[67][68] Another theorist claims that moral concerns limit but do not prohibit germline engineering.[69]

A recent issue of the journal Bioethics was devoted to moral issues surrounding germline genetic engineering in people.[70]

Possible regulatory schemes include a complete ban, provision to everyone, or professional self-regulation. The American Medical Association's Council on Ethical and Judicial Affairs stated that "genetic interventions to enhance traits should be considered permissible only in severely restricted situations: (1) clear and meaningful benefits to the fetus or child; (2) no trade-off with other characteristics or traits; and (3) equal access to the genetic technology, irrespective of income or other socioeconomic characteristics."[71]

As early in the history of biotechnology as 1990, there have been scientists opposed to attempts to modify the human germline using these new tools,[72] and such concerns have continued as technology progressed.[73][74] With the advent of new techniques like CRISPR, in March 2015 a group of scientists urged a worldwide moratorium on clinical use of gene editing technologies to edit the human genome in a way that can be inherited.[75][76][77][78] In April 2015, researchers sparked controversy when they reported results of basic research to edit the DNA of non-viable human embryos using CRISPR.[79][80] A committee of the American National Academy of Sciences and National Academy of Medicine gave qualified support to human genome editing in 2017[81][82] once answers have been found to safety and efficiency problems "but only for serious conditions under stringent oversight."[83]

Gene therapy approaches to replace a faulty gene with a healthy gene have been proposed and are being studied for treating some genetic diseases. Diseases such as sickle cell disease that are caused by autosomal recessive disorders for which a person's normal phenotype or cell function may be restored in cells that have the disease by a normal copy of the gene that is mutated, may be a good candidate for gene therapy treatment.[84][85] The risks and benefits related to gene therapy for sickle cell disease are not known.[85]

Some genetic therapies have been approved by the U.S. Food and Drug Administration (FDA), the European Medicines Agency (EMA), and for use in Russia and China.

Some of the unsolved problems include:

Three patients' deaths have been reported in gene therapy trials, putting the field under close scrutiny. The first was that of Jesse Gelsinger, who died in 1999, because of immune rejection response.[109][110] One X-SCID patient died of leukemia in 2003.[17] In 2007, a rheumatoid arthritis patient died from an infection; the subsequent investigation concluded that the death was not related to gene therapy.[111]

Regulations covering genetic modification are part of general guidelines about human-involved biomedical research.[citation needed] There are no international treaties which are legally binding in this area, but there are recommendations for national laws from various bodies.[citation needed]

The Helsinki Declaration (Ethical Principles for Medical Research Involving Human Subjects) was amended by the World Medical Association's General Assembly in 2008. This document provides principles physicians and researchers must consider when involving humans as research subjects. The Statement on Gene Therapy Research initiated by the Human Genome Organization (HUGO) in 2001, provides a legal baseline for all countries. HUGO's document emphasizes human freedom and adherence to human rights, and offers recommendations for somatic gene therapy, including the importance of recognizing public concerns about such research.[112]

No federal legislation lays out protocols or restrictions about human genetic engineering. This subject is governed by overlapping regulations from local and federal agencies, including the Department of Health and Human Services, the FDA and NIH's Recombinant DNA Advisory Committee. Researchers seeking federal funds for an investigational new drug application, (commonly the case for somatic human genetic engineering,) must obey international and federal guidelines for the protection of human subjects.[113]

NIH serves as the main gene therapy regulator for federally funded research. Privately funded research is advised to follow these regulations. NIH provides funding for research that develops or enhances genetic engineering techniques and to evaluate the ethics and quality in current research. The NIH maintains a mandatory registry of human genetic engineering research protocols that includes all federally funded projects.[114]

An NIH advisory committee published a set of guidelines on gene manipulation.[115] The guidelines discuss lab safety as well as human test subjects and various experimental types that involve genetic changes. Several sections specifically pertain to human genetic engineering, including Section III-C-1. This section describes required review processes and other aspects when seeking approval to begin clinical research involving genetic transfer into a human patient.[116] The protocol for a gene therapy clinical trial must be approved by the NIH's Recombinant DNA Advisory Committee prior to any clinical trial beginning; this is different from any other kind of clinical trial.[115]

As with other kinds of drugs, the FDA regulates the quality and safety of gene therapy products and supervises how these products are used clinically. Therapeutic alteration of the human genome falls under the same regulatory requirements as any other medical treatment. Research involving human subjects, such as clinical trials, must be reviewed and approved by the FDA and an Institutional Review Board.[117][118]

In 1972, Friedmann and Roblin authored a paper in Science titled "Gene therapy for human genetic disease?".[119] Rogers (1970) was cited for proposing that exogenous good DNA be used to replace the defective DNA in those with genetic defects.[120]

In 1984, a retrovirus vector system was designed that could efficiently insert foreign genes into mammalian chromosomes.[121]

The first approved gene therapy clinical research in the US took place on 14 September 1990, at the National Institutes of Health (NIH), under the direction of William French Anderson.[122] Four-year-old Ashanti DeSilva received treatment for a genetic defect that left her with adenosine deaminase deficiency (ADA-SCID), a severe immune system deficiency. The defective gene of the patient's blood cells was replaced by the functional variant. Ashanti's immune system was partially restored by the therapy. Production of the missing enzyme was temporarily stimulated, but the new cells with functional genes were not generated. She led a normal life only with the regular injections performed every two months. The effects were successful, but temporary.[123]

Cancer gene therapy was introduced in 1992/93 (Trojan et al. 1993).[124] The treatment of glioblastoma multiforme, the malignant brain tumor whose outcome is always fatal, was done using a vector expressing antisense IGF-I RNA (clinical trial approved by NIH protocol no.1602 24 November 1993,[125] and by the FDA in 1994). This therapy also represents the beginning of cancer immunogene therapy, a treatment which proves to be effective due to the anti-tumor mechanism of IGF-I antisense, which is related to strong immune and apoptotic phenomena.

In 1992, Claudio Bordignon, working at the Vita-Salute San Raffaele University, performed the first gene therapy procedure using hematopoietic stem cells as vectors to deliver genes intended to correct hereditary diseases.[126] In 2002, this work led to the publication of the first successful gene therapy treatment for ADA-SCID. The success of a multi-center trial for treating children with SCID (severe combined immune deficiency or "bubble boy" disease) from 2000 and 2002, was questioned when two of the ten children treated at the trial's Paris center developed a leukemia-like condition. Clinical trials were halted temporarily in 2002, but resumed after regulatory review of the protocol in the US, the United Kingdom, France, Italy, and Germany.[127]

In 1993, Andrew Gobea was born with SCID following prenatal genetic screening. Blood was removed from his mother's placenta and umbilical cord immediately after birth, to acquire stem cells. The allele that codes for adenosine deaminase (ADA) was obtained and inserted into a retrovirus. Retroviruses and stem cells were mixed, after which the viruses inserted the gene into the stem cell chromosomes. Stem cells containing the working ADA gene were injected into Andrew's blood. Injections of the ADA enzyme were also given weekly. For four years T cells (white blood cells), produced by stem cells, made ADA enzymes using the ADA gene. After four years more treatment was needed.[128]

Jesse Gelsinger's death in 1999 impeded gene therapy research in the US.[129][130] As a result, the FDA suspended several clinical trials pending the reevaluation of ethical and procedural practices.[131]

The modified gene therapy strategy of antisense IGF-I RNA (NIH n 1602)[125] using antisense / triple helix anti-IGF-I approach was registered in 2002, by Wiley gene therapy clinical trial - n 635 and 636. The approach has shown promising results in the treatment of six different malignant tumors: glioblastoma, cancers of liver, colon, prostate, uterus, and ovary (Collaborative NATO Science Programme on Gene Therapy USA, France, Poland n LST 980517 conducted by J. Trojan) (Trojan et al., 2012). This anti-gene antisense/triple helix therapy has proven to be efficient, due to the mechanism stopping simultaneously IGF-I expression on translation and transcription levels, strengthening anti-tumor immune and apoptotic phenomena.

Sickle cell disease can be treated in mice.[132] The mice which have essentially the same defect that causes human cases used a viral vector to induce production of fetal hemoglobin (HbF), which normally ceases to be produced shortly after birth. In humans, the use of hydroxyurea to stimulate the production of HbF temporarily alleviates sickle cell symptoms. The researchers demonstrated this treatment to be a more permanent means to increase therapeutic HbF production.[133]

A new gene therapy approach repaired errors in messenger RNA derived from defective genes. This technique has the potential to treat thalassaemia, cystic fibrosis and some cancers.[134]

Researchers created liposomes 25 nanometers across that can carry therapeutic DNA through pores in the nuclear membrane.[135]

In 2003, a research team inserted genes into the brain for the first time. They used liposomes coated in a polymer called polyethylene glycol, which unlike viral vectors, are small enough to cross the bloodbrain barrier.[136]

Short pieces of double-stranded RNA (short, interfering RNAs or siRNAs) are used by cells to degrade RNA of a particular sequence. If a siRNA is designed to match the RNA copied from a faulty gene, then the abnormal protein product of that gene will not be produced.[137]

Gendicine is a cancer gene therapy that delivers the tumor suppressor gene p53 using an engineered adenovirus. In 2003, it was approved in China for the treatment of head and neck squamous cell carcinoma.[34]

In March, researchers announced the successful use of gene therapy to treat two adult patients for X-linked chronic granulomatous disease, a disease which affects myeloid cells and damages the immune system. The study is the first to show that gene therapy can treat the myeloid system.[138]

In May, a team reported a way to prevent the immune system from rejecting a newly delivered gene.[139] Similar to organ transplantation, gene therapy has been plagued by this problem. The immune system normally recognizes the new gene as foreign and rejects the cells carrying it. The research utilized a newly uncovered network of genes regulated by molecules known as microRNAs. This natural function selectively obscured their therapeutic gene in immune system cells and protected it from discovery. Mice infected with the gene containing an immune-cell microRNA target sequence did not reject the gene.

In August, scientists successfully treated metastatic melanoma in two patients using killer T cells genetically retargeted to attack the cancer cells.[140]

In November, researchers reported on the use of VRX496, a gene-based immunotherapy for the treatment of HIV that uses a lentiviral vector to deliver an antisense gene against the HIV envelope. In a phase I clinical trial, five subjects with chronic HIV infection who had failed to respond to at least two antiretroviral regimens were treated. A single intravenous infusion of autologous CD4 T cells genetically modified with VRX496 was well tolerated. All patients had stable or decreased viral load; four of the five patients had stable or increased CD4 T cell counts. All five patients had stable or increased immune response to HIV antigens and other pathogens. This was the first evaluation of a lentiviral vector administered in a US human clinical trial.[141][142]

In May, researchers announced the first gene therapy trial for inherited retinal disease. The first operation was carried out on a 23-year-old British male, Robert Johnson, in early 2007.[143]

Leber's congenital amaurosis is an inherited blinding disease caused by mutations in the RPE65 gene. The results of a small clinical trial in children were published in April.[5] Delivery of recombinant adeno-associated virus (AAV) carrying RPE65 yielded positive results. In May, two more groups reported positive results in independent clinical trials using gene therapy to treat the condition. In all three clinical trials, patients recovered functional vision without apparent side-effects.[5][21][22][23]

In September researchers were able to give trichromatic vision to squirrel monkeys.[144] In November 2009, researchers halted a fatal genetic disorder called adrenoleukodystrophy in two children using a lentivirus vector to deliver a functioning version of ABCD1, the gene that is mutated in the disorder.[145]

An April paper reported that gene therapy addressed achromatopsia (color blindness) in dogs by targeting cone photoreceptors. Cone function and day vision were restored for at least 33 months in two young specimens. The therapy was less efficient for older dogs.[146]

In September it was announced that an 18-year-old male patient in France with beta thalassemia major had been successfully treated.[147] Beta thalassemia major is an inherited blood disease in which beta haemoglobin is missing and patients are dependent on regular lifelong blood transfusions.[148] The technique used a lentiviral vector to transduce the human -globin gene into purified blood and marrow cells obtained from the patient in June 2007.[149] The patient's haemoglobin levels were stable at 9 to 10 g/dL. About a third of the hemoglobin contained the form introduced by the viral vector and blood transfusions were not needed.[149][150] Further clinical trials were planned.[151] Bone marrow transplants are the only cure for thalassemia, but 75% of patients do not find a matching donor.[150]

Cancer immunogene therapy using modified antigene, antisense/triple helix approach was introduced in South America in 2010/11 in La Sabana University, Bogota (Ethical Committee 14 December 2010, no P-004-10). Considering the ethical aspect of gene diagnostic and gene therapy targeting IGF-I, the IGF-I expressing tumors i.e. lung and epidermis cancers were treated (Trojan et al. 2016).[152][153]

In 2007 and 2008, a man (Timothy Ray Brown) was cured of HIV by repeated hematopoietic stem cell transplantation (see also allogeneic stem cell transplantation, allogeneic bone marrow transplantation, allotransplantation) with double-delta-32 mutation which disables the CCR5 receptor. This cure was accepted by the medical community in 2011.[154] It required complete ablation of existing bone marrow, which is very debilitating.[155]

In August two of three subjects of a pilot study were confirmed to have been cured from chronic lymphocytic leukemia (CLL). The therapy used genetically modified T cells to attack cells that expressed the CD19 protein to fight the disease.[29] In 2013, the researchers announced that 26 of 59 patients had achieved complete remission and the original patient had remained tumor-free.[156]

Human HGF plasmid DNA therapy of cardiomyocytes is being examined as a potential treatment for coronary artery disease as well as treatment for the damage that occurs to the heart after myocardial infarction.[157][158]

In 2011, Neovasculgen was registered in Russia as the first-in-class gene-therapy drug for treatment of peripheral artery disease, including critical limb ischemia; it delivers the gene encoding for VEGF.[35] Neovasculogen is a plasmid encoding the CMV promoter and the 165 amino acid form of VEGF.[159][160]

The FDA approved Phase I clinical trials on thalassemia major patients in the US for 10 participants in July.[161] The study was expected to continue until 2015.[151]

In July 2012, the European Medicines Agency recommended approval of a gene therapy treatment for the first time in either Europe or the United States. The treatment used Alipogene tiparvovec (Glybera) to compensate for lipoprotein lipase deficiency, which can cause severe pancreatitis.[162] The recommendation was endorsed by the European Commission in November 2012,[20][36][163][164] and commercial rollout began in late 2014.[165] Alipogene tiparvovec was expected to cost around $1.6 million per treatment in 2012,[166] revised to $1 million in 2015,[167] making it the most expensive medicine in the world at the time.[168] As of 2016[update], only the patients treated in clinical trials and a patient who paid the full price for treatment have received the drug.[169]

In December 2012, it was reported that 10 of 13 patients with multiple myeloma were in remission "or very close to it" three months after being injected with a treatment involving genetically engineered T cells to target proteins NY-ESO-1 and LAGE-1, which exist only on cancerous myeloma cells.[31]

In March researchers reported that three of five adult subjects who had acute lymphocytic leukemia (ALL) had been in remission for five months to two years after being treated with genetically modified T cells which attacked cells with CD19 genes on their surface, i.e. all B cells, cancerous or not. The researchers believed that the patients' immune systems would make normal T cells and B cells after a couple of months. They were also given bone marrow. One patient relapsed and died and one died of a blood clot unrelated to the disease.[30]

Following encouraging Phase I trials, in April, researchers announced they were starting Phase II clinical trials (called CUPID2 and SERCA-LVAD) on 250 patients[170] at several hospitals to combat heart disease. The therapy was designed to increase the levels of SERCA2, a protein in heart muscles, improving muscle function.[171] The U.S. Food and Drug Administration (FDA) granted this a breakthrough therapy designation to accelerate the trial and approval process.[172] In 2016, it was reported that no improvement was found from the CUPID 2 trial.[173]

In July researchers reported promising results for six children with two severe hereditary diseases had been treated with a partially deactivated lentivirus to replace a faulty gene and after 732 months. Three of the children had metachromatic leukodystrophy, which causes children to lose cognitive and motor skills.[174] The other children had WiskottAldrich syndrome, which leaves them to open to infection, autoimmune diseases, and cancer.[175] Follow up trials with gene therapy on another six children with WiskottAldrich syndrome were also reported as promising.[176][177]

In October researchers reported that two children born with adenosine deaminase severe combined immunodeficiency disease (ADA-SCID) had been treated with genetically engineered stem cells 18 months previously and that their immune systems were showing signs of full recovery. Another three children were making progress.[27] In 2014, a further 18 children with ADA-SCID were cured by gene therapy.[178] ADA-SCID children have no functioning immune system and are sometimes known as "bubble children".[27]

Also in October researchers reported that they had treated six people with haemophilia in early 2011 using an adeno-associated virus. Over two years later all six were producing clotting factor.[27][179]

In January researchers reported that six choroideremia patients had been treated with adeno-associated virus with a copy of REP1. Over a six-month to two-year period all had improved their sight.[6][180] By 2016, 32 patients had been treated with positive results and researchers were hopeful the treatment would be long-lasting.[24] Choroideremia is an inherited genetic eye disease with no approved treatment, leading to loss of sight.

In March researchers reported that 12 HIV patients had been treated since 2009 in a trial with a genetically engineered virus with a rare mutation (CCR5 deficiency) known to protect against HIV with promising results.[181][182]

Clinical trials of gene therapy for sickle cell disease were started in 2014.[183][184]

In February LentiGlobin BB305, a gene therapy treatment undergoing clinical trials for treatment of beta thalassemia gained FDA "breakthrough" status after several patients were able to forgo the frequent blood transfusions usually required to treat the disease.[185]

In March researchers delivered a recombinant gene encoding a broadly neutralizing antibody into monkeys infected with simian HIV; the monkeys' cells produced the antibody, which cleared them of HIV. The technique is named immunoprophylaxis by gene transfer (IGT). Animal tests for antibodies to ebola, malaria, influenza, and hepatitis were underway.[186][187]

In March, scientists, including an inventor of CRISPR, Jennifer Doudna, urged a worldwide moratorium on germline gene therapy, writing "scientists should avoid even attempting, in lax jurisdictions, germline genome modification for clinical application in humans" until the full implications "are discussed among scientific and governmental organizations".[75][76][77][78]

In October, researchers announced that they had treated a baby girl, Layla Richards, with an experimental treatment using donor T cells genetically engineered using TALEN to attack cancer cells. One year after the treatment she was still free of her cancer (a highly aggressive form of acute lymphoblastic leukaemia [ALL]).[188] Children with highly aggressive ALL normally have a very poor prognosis and Layla's disease had been regarded as terminal before the treatment.[189]

In December, scientists of major world academies called for a moratorium on inheritable human genome edits, including those related to CRISPR-Cas9 technologies[190] but that basic research including embryo gene editing should continue.[191]

Researchers successfully treated a boy with epidermolysis bullosa using skin grafts grown from his own skin cells, genetically altered to repair the mutation that caused his disease.[192]

In April the Committee for Medicinal Products for Human Use of the European Medicines Agency endorsed a gene therapy treatment called Strimvelis[193][194] and the European Commission approved it in June.[195] This treats children born with adenosine deaminase deficiency and who have no functioning immune system. This was the second gene therapy treatment to be approved in Europe.[196]

In October, Chinese scientists reported they had started a trial to genetically modify T cells from 10 adult patients with lung cancer and reinject the modified T cells back into their bodies to attack the cancer cells. The T cells had the PD-1 protein (which stops or slows the immune response) removed using CRISPR-Cas9.[197][198]

A 2016 Cochrane systematic review looking at data from four trials on topical cystic fibrosis transmembrane conductance regulator (CFTR) gene therapy does not support its clinical use as a mist inhaled into the lungs to treat cystic fibrosis patients with lung infections. One of the four trials did find weak evidence that liposome-based CFTR gene transfer therapy may lead to a small respiratory improvement for people with CF. This weak evidence is not enough to make a clinical recommendation for routine CFTR gene therapy.[199]

In February Kite Pharma announced results from a clinical trial of CAR-T cells in around a hundred people with advanced non-Hodgkin lymphoma.[200]

In March, French scientists reported on clinical research of gene therapy to treat sickle cell disease.[201]

In August, the FDA approved tisagenlecleucel for acute lymphoblastic leukemia.[202] Tisagenlecleucel is an adoptive cell transfer therapy for B-cell acute lymphoblastic leukemia; T cells from a person with cancer are removed, genetically engineered to make a specific T-cell receptor (a chimeric T cell receptor, or "CAR-T") that reacts to the cancer, and are administered back to the person. The T cells are engineered to target a protein called CD19 that is common on B cells. This is the first form of gene therapy to be approved in the United States. In October, a similar therapy called axicabtagene ciloleucel was approved for non-Hodgkin lymphoma.[203]

In October, biophysicist and biohacker Josiah Zayner claimed to have performed the very first in-vivo human genome editing in the form of a self-administered therapy.[204][205]

On 13 November, medical scientists working with Sangamo Therapeutics, headquartered in Richmond, California, announced the first ever in-body human gene editing therapy.[206][207] The treatment, designed to permanently insert a healthy version of the flawed gene that causes Hunter syndrome, was given to 44-year-old Brian Madeux and is part of the world's first study to permanently edit DNA inside the human body.[208] The success of the gene insertion was later confirmed.[209][210] Clinical trials by Sangamo involving gene editing using zinc finger nuclease (ZFN) are ongoing.[211]

View post:
Gene therapy - Wikipedia

Somatic evolution in cancer – Wikipedia

Somatic evolution is the accumulation of mutations and epimutations in somatic cells (the cells of a body, as opposed to germ plasm and stem cells) during a lifetime, and the effects of those mutations and epimutations on the fitness of those cells. This evolutionary process has first been shown by the studies of Bert Vogelstein in colon cancer. Somatic evolution is important in the process of aging as well as the development of some diseases, including cancer.

Cells in pre-malignant and malignant neoplasms (tumors) evolve by natural selection.[1][2] This accounts for how cancer develops from normal tissue and why it has been difficult to cure. There are three necessary and sufficient conditions for natural selection, all of which are met in a neoplasm:

Cells in neoplasms compete for resources, such as oxygen and glucose, as well as space. Thus, a cell that acquires a mutation that increases its fitness will generate more daughter cells than competitor cells that lack that mutation. In this way, a population of mutant cells, called a clone, can expand in the neoplasm. Clonal expansion is the signature of natural selection in cancer.

Cancer therapies act as a form of artificial selection, killing sensitive cancer cells, but leaving behind resistant cells. Often the tumor will regrow from those resistant cells, the patient will relapse, and the therapy that had been previously used will no longer kill the cancer cells. This selection for resistance is similar to the repeatedly spraying crops with a pesticide and selecting for resistant pests until the pesticide is no longer effective.

Modern descriptions of biological evolution will typically elaborate on major contributing factors to evolution such as the formation of local micro-environments, mutational robustness, molecular degeneracy, and cryptic genetic variation.[4] Many of these contributing factors in evolution have been isolated and described for cancer.[5]

Cancer is a classic example of what evolutionary biologists call multilevel selection: at the level of the organism, cancer is usually fatal so there is selection for genes and the organization of tissues[6][7] that suppress cancer. At the level of the cell, there is selection for increased cell proliferation and survival, such that a mutant cell that acquires one of the hallmarks of cancer[3] (see below), will have a competitive advantage over cells that have not acquired the hallmark. Thus, at the level of the cell there is selection for cancer.

The earliest ideas about neoplastic evolution come from Boveri[8] who proposed that tumors originated in chromosomal abnormalities passed on to daughter cells. In the decades that followed, cancer was recognized as having a clonal origin associated with chromosomal aberrations.[9][10][11][12]

Early mathematical modeling of cancer, by Armitage and Doll, set the stage for the future development of the somatic evolutionary theory of cancer. Armitage and Doll explained the cancer incidence data, as a function of age, as a process of the sequential accumulation of somatic mutations (or other rate limiting steps).[13]

Advances in cytogenetics facilitated discovery of chromosome abnormalities in neoplasms, including the Philadelphia chromosome in chronic myelogenous leukemia[14] and translocations in acute myeloblastic leukemia.[15] Sequences of karyotypes replacing one another in a tumor were observed as it progressed.[16][17][18] Researchers hypothesized that cancer evolves in a sequence of chromosomal mutations and selection[6][17][19][20] and that therapy may further select clones.[12]

In 1971, Knudson published the 2-hit hypothesis for mutation and cancer based on statistical analysis of inherited and sporadic cases of retinoblastoma.[21] He postulated that retinoblastoma developed as a consequence of two mutations; one of which could be inherited or somatic followed by a second somatic mutation. Cytogenetic studies localized the region to the long arm of chromosome 13, and molecular genetic studies demonstrated that tumorigenesis was associated with chromosomal mechanisms, such as mitotic recombination or non-disjunction, that could lead to homozygosity of the mutation.[22] The retinoblastoma gene was the first tumor suppressor gene to be cloned in 1986.

Cairns hypothesized a different, but complementary, mechanism of tumor suppression in 1975 based on tissue architecture to protect against selection of variant somatic cells with increased fitness in proliferating epithelial populations, such as the intestine and other epithelial organs.[6] He postulated that this could be accomplished by restricting the number of stem cells for example at the base of intestinal crypts and restraining the opportunities for competition between cells by shedding differentiated intestinal cells into the gut. The essential predictions of this model have been confirmed although mutations in some tumor suppressor genes, including CDKN2A (p16), predispose to clonal expansions that encompass large numbers of crypts in some conditions such as Barrett's esophagus. He also postulated an immortal DNA strand that is discussed at Immortal DNA strand hypothesis.

Nowell synthesized the evolutionary view of cancer in 1976 as a process of genetic instability and natural selection.[1] Most of the alterations that occur are deleterious for the cell, and those clones will tend to go extinct, but occasional selectively advantageous mutations arise that lead to clonal expansions. This theory predicts a unique genetic composition in each neoplasm due to the random process of mutations, genetic polymorphisms in the human population, and differences in the selection pressures of the neoplasm's microenvironment. Interventions are predicted to have varying results in different patients. What is more important, the theory predicts the emergence of resistant clones under the selective pressures of therapy. Since 1976, researchers have identified clonal expansions[23][24][25][26][27][28] and genetic heterogeneity[29][30][31][32][33][34] within many different types of neoplasms.

There are multiple levels of genetic heterogeneity associated with cancer, including single nucleotide polymorphism (SNP),[35] sequence mutations,[30] Microsatellite shifts[29] and instability,[36] loss of heterozygosity (LOH),[34] Copy number variation (detected both by comparative genomic hybridization (CGH),[31] and array CGH,[37]) and karyotypic variations including chromosome structural aberrations and aneuploidy.[32][33][38][39][40] Studies of this issue have focused mainly at the gene mutation level, as copy number variation, LOH and specific chromosomal translocations are explained in the context of gene mutation. It is thus necessary to integrate multiple levels of genetic variation in the context of complex system and multilevel selection.

System instability is a major contributing factor for genetic heterogeneity.[41] For the majority of cancers, genome instability is reflected in a large frequency of mutations in the whole genome DNA sequence (not just the protein coding regions that are only 1.5% of the genome[42]). In whole genome sequencing of different types of cancers, large numbers of mutations were found in two breast cancers (about 20,000 point mutations[43]), 25 melanomas (9,000 to 333,000 point mutations[44]) and a lung cancer (50,000 point mutations and 54,000 small additions and deletions[45]). Genome instability is also referred to as an enabling characteristic for achieving endpoints of cancer evolution.[3]

Many of the somatic evolutionary studies have traditionally been focused on clonal expansion, as recurrent types of changes can be traced to illustrate the evolutionary path based on available methods. Recent studies from both direct DNA sequencing and karyotype analysis illustrate the importance of the high level of heterogeneity in somatic evolution. For the formation of solid tumors, there is an involvement of multiple cycles of clonal and non-clonal expansion.[39][46] Even at the typical clonal expansion phase, there are significant levels of heterogeneity within the cell population, however, most are under-detected when mixed populations of cells are used for molecular analysis. In solid tumors, a majority of gene mutations are not recurrent types,[47] and neither are the karyotypes.[39][41] These analyses offer an explanation for the findings that there are no common mutations shared by most cancers.[48]

The state of a cell may be changed epigenetically, in addition to genetic alterations. The best-understood epigenetic alterations in tumors are the silencing or expression of genes by changes in the methylation of CG pairs of nucleotides in the promoter regions of the genes. These methylation patterns are copied to the new chromosomes when cells replicate their genomes and so methylation alterations are heritable and subject to natural selection. Methylation changes are thought to occur more frequently than mutations in the DNA, and so may account for many of the changes during neoplastic progression (the process by which normal tissue becomes cancerous), in particular in the early stages. For instance, when loss of expression of the DNA repair protein MGMT occurs in a colon cancer, it is caused by a mutation only about 4% of the time, while in most cases the loss is due to methylation of its promoter region.[49] Similarly, when loss of expression of the DNA repair protein PMS2 occurs in colon cancer, it is caused by a mutation about 5% of the time, while in most cases loss of expression is due to methylation of the promoter of its pairing partner MLH1 (PMS2 is unstable in the absence of MLH1).[50] Epigenetic changes in progression interact with genetic changes. For example, epigenetic silencing of genes responsible for the repair of mispairs or damages in the DNA (e.g. MLH1 or MSH2) results in an increase of genetic mutations.

Deficiency of DNA repair proteins PMS2, MLH1, MSH2, MSH3, MSH6 or BRCA2 can cause up to 100-fold increases in mutation frequency[51][52][53] Epigenetic deficiencies in DNA repair gene protein expression have been found in many cancers, though not all deficiencies have been evaluated in all cancers. Epigeneticically deficient DNA repair proteins include BRCA1, WRN, MGMT, MLH1, MSH2, ERCC1, PMS2, XPF, P53, PCNA and OGG1, and these are found to be deficient at frequencies of 13% to 100% in different cancers.[citation needed] (Also see Frequencies of epimutations in DNA repair genes.)

In addition to well studied epigenetic promoter methylation, more recently there have been substantial findings of epigenetic alterations in cancer due to changes in histone and chromatin architecture and alterations in the expression of microRNAs (microRNAs either cause degradation of messenger RNAs or block their translation)[54] For instance, hypomethylation of the promoter for microRNA miR-155 increases expression of miR-155, and this increased miR-155 targets DNA repair genes MLH1, MSH2 and MSH6, causing each of them to have reduced expression.[55]

In cancers, loss of expression of genes occurs about 10 times more frequently by transcription silencing (caused by somatically heritable promoter hypermethylation of CpG islands) than by mutations. As Vogelstein et al. point out, in a colorectal cancer there are usually about 3 to 6 driver mutations and 33 to 66 hitchhiker or passenger mutations.[56] In contrast, in colon tumors compared to adjacent normal-appearing colonic mucosa, there are about 600 to 800 somatically heritable heavily methylated CpG islands in promoters of genes in the tumors while these CpG islands are not methylated in the adjacent mucosa.[57][58][59]

Methylation of the cytosine of CpG dinucleotides is a somatically heritable and conserved regulatory mark that is generally associated with transcriptional repression. CpG islands keep their overall un-methylated state (or methylated state) extremely stably through multiple cell generations.[60]

One common feature of neoplastic progression is the expansion of a clone with a genetic or epigenetic alteration. This may be a matter of chance, but is more likely due to the expanding clone having a competitive advantage (either a reproductive or survival advantage) over other cells in the tissue. Since clones often have many genetic and epigenetic alterations in their genomes, it is often not clear which of those alterations cause a reproductive or survival advantage and which other alterations are simply hitchhikers or passenger mutations (see Glossary below) on the clonal expansion.

Clonal expansions are most often associated with the loss of the p53 (TP53) or p16 (CDKN2A/INK4a) tumor suppressor genes. In lung cancer, a clone with a p53 mutation was observed to have spread over the surface of one entire lung and into the other lung.[27] In bladder cancer, clones with loss of p16 were observed to have spread over the entire surface of the bladder.[61][62] Likewise, large expansions of clones with loss of p16 have been observed in the oral cavity[24] and in Barrett's esophagus.[25] Clonal expansions associated with inactivation of p53 have also appear in skin,[23][63] Barrett's esophagus,[25] brain,[64] and kidney.[65] Further clonal expansions have been observed in the stomach,[66] bladder,[67] colon,[68] lung,[69] hematopoietic (blood) cells,[70] and prostate.[71]

These clonal expansions are important for at least two reasons. First, they generate a large target population of mutant cells and so increase the probability that the multiple mutations necessary to cause cancer will be acquired within that clone. Second, in at least one case, the size of the clone with loss of p53 has been associated with an increased risk of a pre-malignant tumor becoming cancerous.[72] It is thought that the process of developing cancer involves successive waves of clonal expansions within the tumor.[73]

The term "field cancerization" was first used in 1953 to describe an area or "field" of epithelium that has been preconditioned by (at that time) largely unknown processes so as to predispose it towards development of cancer.[74] Since then, the terms "field cancerization" and "field defect" have been used to describe pre-malignant tissue in which new cancers are likely to arise. Field defects, for example, have been identified in most of the major areas subject to tumorigenesis in the gastrointestinal (GI) tract.[75] Cancers of the GI tract that are shown to be due, to some extent, to field defects include head and neck squamous cell carcinoma (HNSCC), oropharyngeal/laryngeal cancer, esophageal adenocarcinoma and esophageal squamous-cell carcinoma, gastric cancer, bile duct cancer, pancreatic cancer, small intestine cancer and colon cancer.

In the colon, a field defect probably arises by natural selection of a mutant or epigenetically altered cell among the stem cells at the base of one of the intestinal crypts on the inside surface of the colon. A mutant or epigenetically altered stem cell, if it has a selective advantage, could replace the other nearby stem cells by natural selection. This can cause a patch of abnormal tissue, or field defect. The figure in this section includes a photo of a freshly resected and lengthwise-opened segment of the colon that may represent a large field defect in which there is a colon cancer and four polyps. The four polyps, in addition to the cancer, may represent sub-clones with proliferative advantages.

The sequence of events giving rise to this possible field defect are indicated below the photo. The schematic diagram shows a large area in yellow indicating a large patch of mutant or epigenetically altered cells that formed by clonal expansion of an initial cell based on a selective advantage. Within this first large patch, a second such mutation or epigenetic alteration may have occurred so that a given stem cell acquired an additional selective advantage compared to the other stem cells within the patch, and this altered stem cell expanded clonally forming a secondary patch, or sub-clone, within the original patch. This is indicated in the diagram by four smaller patches of different colors within the large yellow original area. Within these new patches (sub-clones), the process may have been repeated multiple times, indicated by the still smaller patches within the four secondary patches (with still different colors in the diagram) which clonally expanded, until a stem cell arose that generated either small polyps (which may be benign neoplasms) or else a malignant neoplasm (cancer). These neoplasms are also indicated, in the diagram below the photo, by 4 small tan circles (polyps) and a larger red area (cancer). The cancer in the photo occurred in the cecal area of the colon, where the colon joins the small intestine (labeled) and where the appendix occurs (labeled). The fat in the photo is external to the outer wall of the colon. In the segment of colon shown here, the colon was cut open lengthwise to expose the inner surface of the colon and to display the cancer and polyps occurring within the inner epithelial lining of the colon.

Phylogenetics may be applied to cells in tumors to reveal the evolutionary relationships between cells, just as it is used to reveal evolutionary relationships between organisms and species. Shibata, Tavare and colleagues have exploited this to estimate the time between the initiation of a tumor and its detection in the clinic.[29] Louhelainen et al. have used parsimony to reconstruct the relationships between biopsy samples based on loss of heterozygosity.[76] Phylogenetic trees should not be confused with oncogenetic trees,[77] which represent the common sequences of genetic events during neoplastic progression and do not represent the relationships of common ancestry that are essential to a phylogeny. For an up-to-date review in this field, see Bast 2012.[78]

An adaptive landscape is a hypothetical topological landscape upon which evolution is envisioned to take place. It is similar to Wright's fitness landscape[79][80] in which the location of each point represents the genotype of an organism and the altitude represents the fitness of that organism in the current environment. However, unlike Wright's rigid landscape, the adaptive landscape is pliable. It readily changes shape with changes in population densities and survival/reproductive strategies used within and among the various species.

Wright's shifting balance theory of evolution combines genetic drift (random sampling error in the transmission of genes) and natural selection to explain how multiple peaks on a fitness landscape could be occupied or how a population can achieve a higher peak on this landscape. This theory, based on the assumption of density-dependent selection as the principal forms of selection, results in a fitness landscape that is relatively rigid. A rigid landscape is one that does not change in response to even large changes in the position and composition of strategies along the landscape.

In contrast to the fitness landscape, the adaptive landscape is constructed assuming that both density and frequency-dependent selection is involved (selection is frequency-dependant when the fitness of a species depends not only on that species strategy but also on the strategy of all other species). As such, the shape of the adaptive landscape can change drastically in response to even small changes in strategies and densities.[81]

The flexibility of adaptive landscapes provide several ways for natural selection to cross valleys and occupy multiple peaks without having to make large changes in their strategies. Within the context of differential or difference equation models for population dynamics, an adaptive landscape may actually be constructed using a fitness generating function.[82] If a given species is able to evolve, it will, over time, "climb" the adaptive landscape toward a fitness peak through gradual changes in its mean phenotype according to a strategy dynamic that involves the slope of the adaptive landscape. Because the adaptive landscape is not rigid and can change shape during the evolutionary process, it is possible that a species may be driven to maximum, minimum, or saddle point on the adaptive landscape. A population at a global maximum on the adaptive landscape corresponds an evolutionarily stable strategy (ESS) and will become dominant, driving all others toward extinction. Populations at a minimum or saddle point are not resistant to invasion, so that the introduction of a slightly different mutant strain may continue the evolutionary process toward unoccupied local maxima.

The adaptive landscape provides a useful tool for studying somatic evolution as it can describe the process of how a mutant cell evolves from a small tumor to an invasive cancer. Understanding this process in terms of the adaptive landscape may lead to the control of cancer through external manipulation of the shape of the landscape.[83][84]

In their landmark paper, The Hallmarks of Cancer,[3] Hanahan and Weinberg suggest that cancer can be described by a small number of underlying principles, despite the complexities of the disease. The authors describe how tumor progression proceeds via a process analogous to Darwinian evolution, where each genetic change confers a growth advantage to the cell. These genetic changes can be grouped into six "hallmarks", which drive a population of normal cells to become a cancer. The six hallmarks are:

Genetic instability is defined as an "enabling characteristic" that facilitates the acquisition of other mutations due to defects in DNA repair.

The hallmark "self-sufficiency in growth signals" describes the observation that tumor cells produce many of their own growth signals and thereby no longer rely on proliferation signals from the micro-environment. Normal cells are maintained in a nondividing state by antigrowth signals, which cancer cells learn to evade through genetic changes producing "insensitivity to antigrowth signals". A normal cell initiates programmed cell death (apoptosis) in response to signals such as DNA damage, oncogene overexpression, and survival factor insufficiency, but a cancer cell learns to "evade apoptosis", leading to the accumulation of aberrant cells. Most mammalian cells can replicate a limited number of times due to progressive shortening of telomeres; virtually all malignant cancer cells gain an ability to maintain their telomeres, conferring "limitless replicative potential". As cells cannot survive at distances of more than 100 m from a blood supply, cancer cells must initiate the formation of new blood vessels to support their growth via the process of "sustained angiogenesis". During the development of most cancers, primary tumor cells acquire the ability to undergo "invasion and metastasis" whereby they migrate into the surrounding tissue and travel to distant sites in the body, forming secondary tumors.

The pathways that cells take toward becoming malignant cancers are variable, and the order in which the hallmarks are acquired can vary from tumor to tumor. The early genetic events in tumorigenesis are difficult to measure clinically, but can be simulated according to known biology.[85] Macroscopic tumors are now beginning to be described in terms of their underlying genetic changes, providing additional data to refine the framework described in The Hallmarks of Cancer.

The theory about the monoclonal origin of cancer states that, in general, neoplasms arise from a single cell of origin.[1] While it is possible that certain carcinogens may mutate more than one cell at once, the tumor mass usually represents progeny of a single cell, or very few cells.[1] A series of mutations is required in the process of carcinogenesis for a cell to transition from being normal to pre-malignant and then to a cancer cell.[86] The mutated genes usually belong to classes of caretaker, gatekeeper, landscaper or several other genes. Mutation ultimately leads to acquisition of the ten hallmarks of cancer.

The first malignant cell, that gives rise to the tumor, is often labeled a cancer stem cell.[87]

The cancer stem-cell hypothesis relies on the fact that a lot of tumors are heterogeneous the cells in the tumor vary by phenotype and functions.[87][88][89] Current research shows that in many cancers there is apparent hierarchy among cells.[87][88][89] in general, there is a small population of cells in the tumor about 0.2%1%[88] that exhibits stem cell-like properties. These cells have the ability to give rise to a variety of cells in tumor tissue, self-renew indefinitely, and upon transfer can form new tumors. According to the hypothesis, cancer stem cells are the only cells capable of tumorigenesis initiation of a new tumor.[87] Cancer stem cell hypothesis might explain such phenomena as metastasis and remission.

The monoclonal model of cancer and the cancer stem-cell model are not mutually exclusive.[87] Cancer stem cell arises by clonal evolution as a result of selection for the cell with the highest fitness in the neoplasm. This way, the heterogeneous nature of neoplasm can be explained by two processes clonal evolution, or the hierarchical differentiation of cells, regulated by cancer stem cells.[87] All cancers arise as a result of somatic evolution, but only some of them fit the cancer stem cell hypothesis.[87] The evolutionary processes do not cease when a population of cancer stem cells arises in a tumor. Cancer treatment drugs pose a strong selective force on all types of cells in tumors, including cancer stem cells, which would be forced to evolve resistance to the treatment. Cancer stem cells do not always have to have the highest resistance among the cells in the tumor to survive chemotherapy and re-emerge afterwards. The surviving cells might be in a special microenvironment, which protects them from adverse effects of treatment.[87]

It is currently unclear as to whether cancer stem cells arise from adult stem cell transformation, a maturation arrest of progenitor cells, or as a result of dedifferentiation of mature cells.[88]

Therapeutic resistance has been observed in virtually every form of therapy, from the beginning of cancer therapy.[90] In most cases, therapies appear to select for mutations in the genes or pathways targeted by the drug.

Some of the first evidence for a genetic basis of acquired therapeutic resistance came from studies of methotrexate. Methotrexate inhibits the dihydrofolate reductase (DHFR) gene. However, methotrexate therapy appears to select for cells with extra copies (amplification) of DHFR, which are resistant to methotrexate. This was seen in both cell culture[91] and samples from tumors in patients that had been treated with methotrexate.[92][93][94][95]

A common cytotoxic chemotherapy used in a variety of cancers, 5-fluorouracil (5-FU), targets the TYMS pathway and resistance can evolve through the evolution of extra copies of TYMS, thereby diluting the drug's effect.[96]

In the case of Gleevec (Imatinib), which targets the BCR-ABL fusion gene in chronic myeloid leukemia, resistance often develops through a mutation that changes the shape of the binding site of the drug.[97][98] Sequential application of drugs can lead to the sequential evolution of resistance mutations to each drug in turn.[99]

Gleevec is not as selective as was originally thought. It turns out that it targets other tyrosine kinase genes and can be used to control gastrointestinal stromal tumors (GISTs) that are driven by mutations in c-KIT. However, patients with GIST sometimes relapse with additional mutations in c-KIT that make the cancer cells resistant to Gleevec.[100][101]

Gefitinib(Iressa) and Erlotinib (Tarceva) are epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors used for non-small cell lung cancer patients whose tumors have somatic mutations in EGFR. However, most patients' tumors eventually become resistant to these drugs. Two major mechanisms of acquired resistance have been discovered in patients who have developed clinical resistance to Gefitinib or Erlotinib:[102] point mutations in the EGFR gene targeted by the drugs,[103] and amplification of MET, another receptor tyrosine kinase, which can bypass EGFR to activate downstream signaling in the cell. In an initial study, 22% of tumors with acquired resistance to Gefitinib or Erlotinib had MET amplification.[104] To address these issues, clinical trials are currently assessing irreversible EGFR inhibitors (which inhibit growth even in cell lines with mutations in EGFR), the combination of EGFR and MET kinase inhibitors, and Hsp90 inhibitors (EGFR and MET both require Hsp90 proteins to fold properly). In addition, taking repeated tumor biopsies from patients as they develop resistance to these drugs would help to understand the tumor dynamics.

Selective estrogen receptor modulators (SERMs) are a commonly used adjuvant therapy in estrogen-receptor positive (ER+) breast cancer and a preventive treatment for women at high risk of the disease. There are several possible mechanisms of SERM resistance, though the relative clinical importance of each is debated. These include:[105][106]

Most prostate cancers derive from cells that are stimulated to proliferate by androgens. Most prostate cancer therapies are therefore based on removing or blocking androgens. Mutations in the androgen receptor (AR) have been observed in anti-androgen resistant prostate cancer that makes the AR hypersensitive to the low levels of androgens that remain after therapy.[111] Likewise, extra copies of the AR gene (amplification) have been observed in anti-androgen resistant prostate cancer.[112] These additional copies of the gene are thought to make the cell hypersensitive to low levels of androgens and so allow them to proliferate under anti-androgen therapy.

Resistance to radiotherapy is also commonly observed. However, to date, comparisons of malignant tissue before and after radiotherapy have not been done to identify genetic and epigenetic changes selected by exposure to radiation. In gliomas, a form of brain cancer, radiation therapy appears to select for stem cells,[113][114] though it is unclear if the tumor returns to the pre-therapy proportion of cancer stem cells after therapy or if radiotherapy selects for an alteration that keeps the glioma cells in the stem cell state.

Cancer drugs and therapies commonly used today are evolutionary inert and represent a strong selection force, which leads to drug resistance.[115] A possible way to avoid that is to use a treatment agent that would co-evolve alongside cancer cells.

Anoxic bacteria could be used as competitors or predators in hypoxic environments within tumors.[115] Scientists have been interested in the idea of using anoxic bacteria for over 150 years, but until recently there has been little progress in that field. According to Jain and Forbes, several requirements have to be met by the cells to qualify as efficient anticancer bacterium:[116]

In the process of the treatment, cancer cells are most likely to evolve some form of resistance to the bacterial treatment. However, being a living organism, bacteria would coevolve with tumor cells, potentially eliminating the possibility of resistance.[116]

Since bacteria prefer an anoxic environment, they are not efficient at eliminating cells on the periphery of the tumor, where oxygen supply is efficient. A combination of bacterial treatment with chemical drugs will increase chances of destroying the tumor.[116]

Oncolytic viruses are engineered to infect cancerous cells. Limitations of that method include immune response to the virus and the possibility of the virus evolving into a pathogen.[115]

By manipulating the tumor environment, it is possible to create favorable conditions for the cells with least resistance to chemotherapy drugs to become more fit and outcompete the rest of the population. The chemotherapy, administered directly after, should wipe out the predominant tumor cells.[115]

Mapping between common terms from cancer biology and evolutionary biology:

Read this article:
Somatic evolution in cancer - Wikipedia

Human Induced Pluripotent Stem Cells | ATCC

Induced pluripotent stem cells (iPSCs) provide a powerful starting material to model human disease in relevant cell types. iPSCs may be generated from patients of any genetic background and possess the capacity to differentiate into almost any desired terminal cell type.

Although additional investigation is needed, researchers are beginning to focus on the potential utility of iPSCs as a tool for drug development, modeling of disease, and transplantation medicine.

Using ATCCs complete feeder- and xeno-free culture systems, researchers can generate standardized, quality controlled, and highly characterized human iPSCs lines. ATCCs iPSCs are derived by episomal, retroviral, or Sendai viral reprogramming. After gaining pluripotent status, the iPSCs may then be induced to differentiate into many cell types. These cells are valuable materials in the study of differentiation, tissue repair, disease pathogenesis, and drug discovery and development.

ATCC is a licensee of iPS Academia Japans induced pluripotent stem (iPS) cell patent portfolio and is able to bring complete cell culturing solutions for iPSCs to the research community.

ATCC iPSCs are tested for pluripotency, karyotype, growth potential, and sample purity. These authenticated materials are backed by meticulous quality control procedures, making them ideal as reference standards for physiologically relevant in vitro research.

View post:
Human Induced Pluripotent Stem Cells | ATCC

Stem Cell Transplant for Acute Myeloid Leukemia (AML)

The doses of chemotherapy drugs that doctors can give to treat acute myeloid leukemia (AML) are limited by the serious side effects they can cause. Even though higher doses of these drugs might kill more cancer cells, they cant be given because they could severely damage the bone marrow, which is where new blood cells are formed. This could lead to life-threatening infections, bleeding, and other problems caused by low blood cell counts.

Doctors can sometimes use a stem cell transplant (SCT), also called a bone marrow transplant,to give higher doses of chemotherapy than could normally be given. (Sometimes radiation therapy is given as well.) After the treatment is finished, the patient gets an infusion of blood-forming stem cells to restore their bone marrow.

The blood-forming stem cells used for a transplant can come either from blood or from bone marrow. Sometimes stem cells from a babys umbilical cord blood are used.

Stem cell transplants differ based on whom the blood-forming stem cells come from.

This is the most common type of SCT used to treat AML. In an allogeneic SCT, the stem cells come from someone other than the patient usually a donor whose tissue type (also known as the HLA type) closely matches the patients. Tissue type is based on certain substances on the surface of cells in the body. Differences in HLA types between the stem cell donor and recipient can cause the body's immune system to react against the cells. Therefore, the closer a tissue match is between the donor and the recipient, the better the chance the transplanted cells will take and begin making new blood cells.

The best donor is often a close relative, such as a brother or sister, if they are a good match. If no close relatives match, stem cells might be available from a matched unrelated donor (MUD), an unrelated volunteer whose tissue type matches that of the patient. But the use of stem cells from a MUD is linked to more complications. Sometimes umbilical cord stem cells are used. These stem cells come from blood drained from the umbilical cord and placenta after a baby is born and the umbilical cord is cut.

For most patients with AML, especially those at higher risk of having the leukemia return after treatment, using an allogeneic SCT is preferred over an autologous SCT (see below). Leukemia is a disease of the blood and bone marrow, so giving the patient their own cells back after treatment may mean giving them back some leukemia cells as well. Donor cells are also helpful because of the graft-versus-leukemia effect. When the donor immune cells are infused into the body, they may recognize any remaining leukemia cells as being foreign to them and attack them. This effect doesnt happen with autologous stem cell transplants.

Allogeneic transplants can have serious risks and side effects, so patients typically need to be younger and relatively healthy to be good candidates. Another challenge is that it can sometimes be difficult to find a matched donor.

One of the most serious complications of allogeneic SCTs is known as graft-versus-host disease (GVHD). It happens when the patients immune system is taken over by that of the donor. When this happens, the donor immune system may see the patients own body tissues as foreign and attack them.

Symptoms can include severe skin rashes, itching, mouth sores (which can affect eating), nausea, and severe diarrhea. Liver damage can cause yellowing of the skin and eyes (jaundice). The lungs can also be damaged. The patient may also become easily fatigued and develop muscle aches. Sometimes GVHD can become disabling, and if it's severe enough, it can be life-threatening. Drugs that affect the immune system may be given to try to control it.

Non-myeloablative transplant (mini-transplant): Many older people cant tolerate a standard allogeneic transplant that uses high doses of chemo. Some may still be able to get a non-myeloablative transplant (also known as a mini-transplant or reduced-intensity transplant), where they get lower doses of chemo and radiation that dont completely destroy the cells in their bone marrow. They then get the allogeneic (donor) stem cells. These cells enter the body and establish a new immune system, which sees the leukemia cells as foreign and attacks them (a graft-versus-leukemia effect).

A non-myeloablative transplant can still sometimes work with much less toxicity. In fact, a patient can get the transplant as an outpatient. The major complication is graft-versus-host disease.

Many doctors still consider this an experimental procedure for AML, and it is being studied to determine how useful it may be.

In an autologous transplant, a patients own stem cells are removed from their bone marrow or blood. They are frozen and stored while the person gets treatment (high-dose chemotherapy and/or radiation). In the lab, a process called purging may be used to try to remove any leukemia cells in the samples. The stem cells are then put back (reinfused) into the patients blood after treatment.

Autologous transplants are sometimes used for people with AML who are in remission after initial treatment and who dont have a matched donor for an allogeneic transplant. Some doctors feel that it is better than standard consolidation chemotherapy (see Typical Treatment of Acute Myeloid Leukemia (AML)) for these people, but not all doctors agree with this.

Autologous transplants are generally easier for patients to tolerate than allogeneic transplants, because they are getting their own cells back, which lowers the risk of some complications. But the high-dose chemo can still cause major side effects. This type of transplant can be done in any otherwise healthy person, although patients who are very old or have other health problems might not be suitable.

One problem with autologous transplants is that its hard to separate normal stem cells from leukemia cells in the bone marrow or blood samples. Even after purging (treating the stem cells in the lab to try to kill or remove any remaining leukemia cells), there is the risk of returning some leukemia cells with the stem cell transplant.

Read more:
Stem Cell Transplant for Acute Myeloid Leukemia (AML)

Guidelines International Society for Stem Cell Research

Academy of Medical Sciences (2011). Animals containing human material. http://www.acmedsci.ac.uk/policy/policy-projects/animals-containing-human-material/

American College of Obstetricians and Gynecologists (2006). Using preimplantation embryos for research. ACOG Committee Opinion No. 347. Obstet. Gynecol. 108, 13051317.

Bacman, S., Williams, S., Pinto, M. et al. Specific elimination of mutant mitochondrial genomes in patient-derived cells by mitoTALENs. Nat Med 19, 11111113 (2013). https://doi.org/10.1038/nm.3261

Bacman SR, Kauppila JHK, Pereira CV, et al. MitoTALEN reduces mutant mtDNA load and restores tRNAAla levels in a mouse model of heteroplasmic mtDNA mutation [published correction appears in Nat Med. 2018 Oct 5;:]. Nat Med. 2018;24(11):16961700. doi:10.1038/s41591-018-0166-8

Benjaminy S, Kowal SP, MacDonald IM, Bubela T. Communicating the promise for ocular gene therapies: challenges and recommendations. Am J Ophthalmol. 2015 Sep;160(3):408-415.e2. doi: 10.1016/j.ajo.2015.05.026. Epub 2015 May 30. PMID: 26032192.

Boer, G. J. (1994). Ethical guidelines for the use of human embryonic or fetal tissue for experimental and clinical neurotransplantation and research. Journal of Neurology 242, 113.

Boutron, I., Dutton, S., Ravaud, P., and Altman, D.G. (2010). Reporting and interpretation of randomized controlled trials with statistically nonsignificant results for primary outcomes. JAMA 303, 2058-2064.

Bubela T, McCabe C, Archibald P, Atkins H, Bradshaw SE, Kefalas P, Mujoomdar M, Packer C, Piret J, Raxworthy M, Soares M, Viswanathan S. Bringing regenerative medicines to the clinic: the future for regulation and reimbursement. Regen Med. 2015;10(7):897-911. doi: 10.2217/rme.15.51. Epub 2015 Nov 13. PMID: 26565607.

Camacho, L.H., Bacik, J., Cheung, A., and Spriggs, D.R. (2005). Presentation and subsequent publication rates of phase I oncology clinical trials. Cancer 104, 1497-1504.

Costa-Borges, N., Nikitos, E., Spath, K. et al. (2020) First registered pilot trial to validate the safety and effectiveness of maternal spindle transfer to overcome infertility associated with poor oocyte quality. Fertility & Sterility (Abstract only) 114, issue 3, supplement , e71-e72 https://doi.org/10.1016/j.fertnstert.2020.08.220

Council for International Organizations of Medical Sciences (2016). International Ethical Guidelines for Health-related Research Involving Humans. https://cioms.ch/wp-content/uploads/2017/01/WEB-CIOMS-EthicalGuidelines.pdf

Department of Health, and Education and Welfare (1979). Report of the National Commission for the Protection of Human Subjects of Biomedical and Behavioral Research (The Belmont Report). 44 Fed. Reg. 23, 192.

ESHRE Taskforce on Ethics and Law (2001). The moral status of the pre-implantation embryo. Hum. Reprod. 17, 1409-1419.

Easson EC, Russell MH. Cure of Hodgkins disease. BMJ. 1963;5347:17047.

Ethics Committee of American Society for Reproductive Medicine (2013). Donating embryos for human embryonic stem cell (hESC) research: a committee opinion. Fertil. Steril. 100, 935-939.

European Parliament and Council of the European Union (2001). Directive 2001/20/EC of the European Parliament and of the Council of 4 April 2001 on the approximation of the laws, regulations and administrative provisions of the Member States relating to the implementation of good clinical practice in the conduct of clinical trials on medicinal products for human use. (Official Journal of the European Union).

European Science Foundation (2000). Good scientific practice in research and scholarship. Science Policy Briefing. http://www.esf.org/fileadmin/Public_documents/Publications/ESPB10.pdf

Fisher, M., Feuerstein, G., Howells, D.W., Hurn, P.D., Kent, T.A., Savitz, S.I., and Lo, E.H. (2009). Update of the stroke therapy academic industry roundtable preclinical recommendations. Stroke 40, 2244-2250.

Flory, J., and Emanuel, E. (2004). Interventions to improve research participants' understanding in informed consent for research: a systematic review. JAMA 292, 1593-1601.

Food and Drug Administration (2017). FDA Announces Comprehensive Regenerative Medicine Policy Framework. https://www.fda.gov/news-events/press-announcements/fda-announces-comprehensive-regenerative-medicine-policy-framework

Freeman, G.A., and Kimmelman, J. (2012). Publication and reporting conduct for pharmacodynamic analyses of tumor tissue in early-phase oncology trials. Clinical Cancer Research 18, 6478-6484.

Frei, E. 3rd & Gehan, E. A. Definition of cure for Hodgkins disease. Cancer Res. 31, 18281833 (1971).

Fung M, Yuan Y, Atkins H, Shi Q, Bubela T. Responsible Translation of Stem Cell Research: An Assessment of Clinical Trial Registration and Publications. Stem Cell Reports. 2017 May 9;8(5):1190-1201. doi: 10.1016/j.stemcr.2017.03.013. Epub 2017 Apr 13. PMID: 28416287; PMCID: PMC5425617.

Haimes, E., Skene, L., Ballantyne, A.J., Caulfield, T., Goldstein, L.S., Hyun, I., Kimmelman, J., Robert, J.S., Roxland, B.E., Scott, C.T., et al. (2013). Position statement on the provision and procurement of human eggs for stem cell research. Cell Stem Cell 12, 285-291.

Henderson, V.C., Kimmelman, J., Fergusson, D., Grimshaw, J.M., and Hackam, D.G. (2013). Threats to validity in the design and conduct of preclinical efficacy studies: a systematic review of guidelines for in vivo animal experiments. PLoS Medicine 10, e1001489.

Hudson, G., Takeda, Y. & Herbert, M. Reversion after replacement of mitochondrial DNA. Nature 574, E8E11 (2019). https://doi.org/10.1038/s41586-019-1623-3

Human Fertilization and Embryology Authority (2016). Scientific review of the safety and efficacy of methods to avoid mitochondrial disease through assisted conception: 2016 update. https://www.hfea.gov.uk/media/2611/fourth_scientific_review_mitochondria_2016.pdf

Human Fertilization and Embryology Authority (2019). Code of Practice, 9th Edition. https://portal.hfea.gov.uk/media/1527/2019-12-16-code-of-practice-9th-edition-december-2019.pdf

Hyslop LA, Blakeley P, Craven L, Richardson J, Fogarty NM, Fragouli E, Lamb M, Wamaitha SE, Prathalingam N, Zhang Q, OKeefe H. Towards clinical application of pronuclear transfer to prevent mitochondrial DNA disease. Nature. 2016 Jun 8 534: 383-386.

Hyun, I. (2013). Therapeutic hope, spiritual distress, and the problem of stem cell tourism. Cell Stem Cell12, 505-507.

Hyun, I., Taylor, P., Testa, G., Dickens, B., Jung, K. W., McNab, A., Roberston, J., Skene, L., and Zoloth, L. (2007). Ethical standards for human-to-animal chimera experiments in stem cell research. Cell Stem Cell 1, 159163.

International Society for Stem Cell Research (ISSCR). (2006). Guidelines for the conduct of human embryonic stem cell research. http://www.isscr.org/docs/default-source/hesc-guidelines/isscrhescguidelines2006.pdf.

International Society for Stem Cell Research (ISSCR). (2008). Guidelines for the clinical translation of stem cells. http://www.isscr.org/docs/default-source/clin-trans-guidelines/isscrglclinicaltrans.pdf.

International Society for Stem Cell Research (ISSCR). (2016). Guidelines for Stem Cell Research and Clinical Translation. https://www.isscr.org/docs/default-source/all-isscr-guidelines/guidelines-2016/isscr-guidelines-for-stem-cell-research-and-clinical-translationd67119731dff6ddbb37cff0000940c19.pdf?sfvrsn=4

Madan K. Natural human chimeras: A review. Eur J Med Genet. 2020 Sep;63(9):103971. doi: 10.1016/j.ejmg.2020.103971. Epub 2020 Jun 18. PMID: 32565253.

National Academy of Sciences, National Academy of Engineering, and Institute of Medicine. 2009. On Being a Scientist: A Guide to Responsible Conduct in Research: Third Edition. Washington, DC: The National Academies Press. https://doi.org/10.17226/12192.

National Academy of Medicine, National Academy of Sciences, and the Royal Society. 2020. Heritable Human Genome Editing. Washington, DC.: The National Academies Press. doi: 10.17226/25665.

Institute of Medicine (2015). Sharing Clinical Trial Data: Maximizing Benefits, Minimizing Risks (Washington, DC: National Academies Press). doi:10.17226/18998.

Nuffield Council on Bioethics. 2016. Genome Editing: An Ethical Review. https://www.nuffieldbioethics.org/wp-content/uploads/Genome-editing-an-ethical-review.pdf

Institute of Medicine and National Research Council. 2010. Final Report of the National Academies' Human Embryonic Stem Cell Research Advisory Committee and 2010 Amendments to the National Academies' Guidelines for Human Embryonic Stem Cell Research. Washington, DC: The National Academies Press. https://doi.org/10.17226/12923.

Kamenova, K., and Caulfield, T. (2015). Stem cell hype: media portrayal of therapy translation. Science Translational Medicine 7, 278ps274.

Kang, E., Koski, A., Amato, P. et al. Reply to: Reversion after replacement of mitochondrial DNA. Nature 574, E12E13 (2019). https://doi.org/10.1038/s41586-019-1624-2

Kang E, Wu J, Gutierrez NM, Koski A, Tippner-Hedges R, Agaronyan K, Platero-Luengo A, Martinez-Redondo P, Ma H, Lee Y, Hayama T, Van Dyken C, Wang X, Luo S, Ahmed R, Li Y, Ji D, Kayali R, Cinnioglu C, Olson S, Jensen J, Battaglia D, Lee D, Wu D, Huang T, Wolf DP, Temiakov D, Izpisua Belmonte JC, Amato P, Mitalipov S. Mitochondrial replacement in human oocytes carrying pathogenic mitochondrial DNA mutations. Nature 2016 DOI: 10.1038/nature20592

Kilkenny C, Browne WJ, Cuthill IC, Emerson M, and Altman DG (2010). Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. Plos Biol 8, e1000412.

Kimmelman, J., Mogil, J.S., Dirnagl, U. (2014). Distinguishing between Exploratory and Confirmatory Preclinical Research Will Improve Translation. PLoS Biol. 12, e1001863.

Kuriyan AE, Albini TA, Townsend JH, et al. Vision Loss after Intravitreal Injection of Autologous "Stem Cells" for AMD. N Engl J Med. 2017; 376(11):10471053. doi:10.1056/NEJMoa1609583

Landis, S.C., Amara, S.G., Asadullah, K., Austin, C.P., Blumenstein, R., Bradley, E.W., Crystal, R.G., Darnell, R.B., Ferrante, R.J., Fillit, H., et al. (2012). A call for transparent reporting to optimize the predictive value of preclinical research. Nature 490, 187-191.

Lau, D., Ogbogu, U., Taylor, B., Stafinski, T., Menon, D., and Caulfield, T. (2008). Stem cell clinics online: the direct-to-consumer portrayal of stem cell medicine. Cell Stem Cell 3, 591-594.

Mazur, P et al. 2019 P-221 Mitochondrial Replacement Therapy Gives No Benefits to Patients of Advanced Maternal Age. Paper presented to ASRM Scientific Conference and Expo, Philadelphia, Oct. 14-16. https://asrm.confex.com/asrm/2019/meetingapp.cgi/Paper/2347

Medical Professionalism Project (2002). Medical professionalism in the new millennium: a physician charter. Annals of Internal medicine 136, 243-246.

Munsie, M., and Hyun, I. (2014). A question of ethics: selling autologous stem cell therapies flaunts professional standards. Stem Cell Research 13, 647-653.

National Academies of Sciences, Engineering, and Medicine (2016). Mitochondrial Replacement Techniques: Ethical, Social, and Policy Considerations. (Washington, DC: The National Academies Press). doi:10.17226/21871.

National Institutes of Health (2014). Informed Consent Guidance for Human Gene Trials subject to the NIH Guidelines for Research Involving Recombinant or Synthetic Nucleic Acid Molecules (Office of Science Policy: Office of Biotechnology Activities). http://osp.od.nih.gov/sites/default/files/resources/IC2013.pdf

Nissanka N, Moraes CT. Mitochondrial DNA heteroplasmy in disease and targeted nuclease-based therapeutic approaches. EMBO Rep. 2020;21(3):e49612. doi:10.15252/embr.201949612

Nuremberg Code (1949). In Trials of War Criminals before the Nuremberg Military Tribunals under Control Council Law No 10, Vol 2 (Washington, DC: U.S. Government Printing Office), pp 181-182.

Office for Human Research Protections (OHRP) (1993). Research on Transplantation of Fetal Tissue SEC. 498A, National Institutes of Health Revitalization Act of 1993. http://www.hhs.gov/ohrp/policy/publiclaw103-43.htm.html

Percie du Sert N, Hurst V, Ahluwalia A, Alam S, Avey MT, Baker M, et al. (2020) The ARRIVE guidelines 2.0: Updated guidelines for reporting animal research. PLoS Biol 18(7): e3000410. https://doi.org/10.1371/journal.pbio.3000410

Ravi, P., Kumar, S.K., Cerhan, J.R. et al. Defining cure in multiple myeloma: a comparative study of outcomes of young individuals with myeloma and curable hematologic malignancies. Blood Cancer Journal 8, 26 (2018). https://doi.org/10.1038/s41408-018-0065-8:

Reddy, Pradeep et al. Selective Elimination of Mitochondrial Mutations in the Germline by Genome Editing. Cell, Volume 161, Issue 3 (2015) pp. 459 469. https://doi.org/10.1016/j.cell.2015.03.051

Saini, P., Loke, Y.K., Gamble, C., Altman, D.G., Williamson, P.R., and Kirkham, J.J. (2014). Selective reporting bias of harm outcomes within studies: findings from a cohort of systematic reviews. BMJ 349, g6501.

SB-180 Gene therapy kits: advisory notice and labels.(2019-2020). California State Legislature.

S.D. Tardif, K. Coleman, T.R. Hobbs, C. Lutz, IACUC Review of Nonhuman Primate Research, ILAR Journal 53, no. 2, 2013: DOI: 10.1093/ilar/ilt040.

Sena, E.S., van der Worp, H.B., Bath, P.M., Howells, D.W., and Macleod, M.R. (2010). Publication bias in reports of animal stroke studies leads to major overstatement of efficacy. PLoS Biol. 8, e1000344.

Sugarman J, Siegel A. How to determine whether existing human embryonic stem cell lines can be used ethically. Cell Stem Cell 2008; 3: 238-9. PMID: 18786411.

Sugarman J, Siegel AW. When embryonic stem cell lines fail to meet consent standards. Science 2008; 322: 379. PMID: 18927375.

Tsilidis, K.K., Panagiotou, O.A., Sena, E.S., Aretouli, E., Evangelou, E., Howells, D.W., Al-Shahi Salman. R., Macleod, M.R., Ioannidis, J.P. (2013). Evaluation of excess significance bias in animal studies of neurological diseases. PLoS Biol. 11, e1001609.

U.K. Department of Health (2014). Mitochondrial donation. Government response to the consultation on draft regulations to permit the use of new treatment techniques to prevent the transmission of a serious mitochondrial disease from mother to child. https://www.gov.uk/government/uploads/system/uploads/attachment_data/file/332881/Consultation_response.pdf

Wang, Tian et al. Polar Body Genome Transfer for Preventing the Transmission of Inherited Mitochondrial Diseases. Cell, 157 (2014), pp. 1591-1604. https://doi.org/10.1016/j.cell.2014.04.042

World Medical Association (2018). Declaration of Helsinki: Ethical principles for medical research involving human subjects. https://www.wma.net/policies-post/wma-declaration-of-helsinki-ethical-principles-for-medical-research-involving-human-subjects/

Wu, K., Zhong, C., Chen, T. et al. Polar bodies are efficient donors for reconstruction of human embryos for potential mitochondrial replacement therapy. Cell Res 27, 10691072 (2017). https://doi.org/10.1038/cr.2017.67

Yamada M, Emmanuele V, Sanchez-Quintero MJ, Sun B, Lallos G, Paull D, Zimmer M, Pagett S, Prosser RW, Sauer MV, Hirano M. Genetic Drift Can Compromise Mitochondrial Replacement by Nuclear Transfer in Human Oocytes. Cell stem cell. 2016 Jun 2;18(6):749-54.

Zhang, John et al. Pregnancy derived from human zygote pronuclear transfer in a patient who had arrested embryos after IVF. Reproductive biomedicine online vol. 33,4 (2016): 529-533. doi:10.1016/j.rbmo.2016.07.008

Read more:
Guidelines International Society for Stem Cell Research

People | Faculty of Engineering and Physical Sciences – University of Leeds

Abas Prades Sonia S.AbasPrades@leeds.ac.uk Research Fellow Abbas Noorhan +44(0)113 343 0084 N.H.Abbas@leeds.ac.uk Research & Teaching Fellow Abiri Jahromi Amir A.AbiriJahromi@leeds.ac.uk Lecturer in Smart Energy Systems Aboelazayem Omar O.Aboelazayem@leeds.ac.uk Research Fellow in Advanced Biofuels Actis Paolo +44(0)113 343 4625 P.Actis@leeds.ac.uk Associate Professor of Bio-nanotechnology Adam-Day Bea B.Adam-Day@leeds.ac.uk Postgraduate Researcher Adams Peter +44(0)113 343 9718 P.G.Adams@leeds.ac.uk Associate Professor Adesanya Elijah E.D.Adesanya@leeds.ac.uk Visiting Research Fellow Adler Isolde +44(0)113 343 6819 I.M.Adler@leeds.ac.uk Associate Professor Adu-Amankwah Samuel +44(0)113 343 2304 S.Adu-Amankwah@leeds.ac.uk Lecturer in Civil Engineering Agboh Wisdom W.C.Agboh@leeds.ac.uk EPSRC Doctoral Prize Fellow Aivaliotis Georgios +44(0)113 343 5162 G.Aivaliotis@leeds.ac.uk Lecturer Al Arefi Salma S.Alarefi@leeds.ac.uk Teaching Fellow Al-Hajjar Mazen +44(0)113 343 9401 M.Al-Hajjar@leeds.ac.uk Visiting Research Fellow Al-Sayaghi Maram M.Al-Sayaghi@leeds.ac.uk Experimental Support Officer Alazmani Ali +44(0)113 343 2178 A.Alazmani@leeds.ac.uk University Academic Fellow Aldred Josephine +44(0)113 343 5837 J.Aldred@leeds.ac.uk Marketing Manager Aldridge Matthew M.Aldridge@leeds.ac.uk Lecturer Ali Mannan +44(0)113 343 3833 M.Ali2@leeds.ac.uk Experimental Officer Ali Sharib S.S.Ali@leeds.ac.uk Lecturer Alsalka Mhd Ammar +44(0)113 343 1142 M.A.Alsalka@leeds.ac.uk Lecturer in Computing Altahhan Abdulrahman +44(0)113 343 1310 A.Altahhan@leeds.ac.uk Senior Teaching Fellow in Artificial Intelligence Alves Thorne Karine +44(0)113 343 5533 K.I.AlvesThorne@leeds.ac.uk Trainee Technician Amsdon Timothy +44(0)113 343 4800 T.J.Amsdon@leeds.ac.uk Lecturer (Assistant Professor) Anamari Mohandas Reshma R.AnamariMohandas@leeds.ac.uk Research Fellow Andrews Gordon +44(0)113 343 2493 G.E.Andrews@leeds.ac.uk Professor Anquandah Jason J.S.Anquandah@leeds.ac.uk Tutorial Assistant Ansell Richard +44(0)113 343 6415 R.J.Ansell@leeds.ac.uk Senior Lecturer Antony Joseph +44(0)113 343 2409 S.J.Antony@leeds.ac.uk Associate Professor Arkless Leslie +44(0)113 343 2309 L.Arkless@leeds.ac.uk Analytical Technician in Microstructure Materials Characterisati Armstrong Chris +44(0)113 343 2046 C.J.Armstrong@leeds.ac.uk Student Education Service Officer Arrowsmith Rachel R.Arrowsmith@leeds.ac.uk Student Ed Service Officer (Admissions) Asachi Maryam M.Asachi@leeds.ac.uk Lecturer in Water and Environmental Engineering Asaf Mohammed +44(0)113 343 2945 M.Asaf@leeds.ac.uk Technician Aslam Zabeada Z.P.Aslam@leeds.ac.uk Research Officer Aslanyan Vahagn V.Aslanyan@leeds.ac.uk Leverhulme Trust Early Career Fellow Asprey Stuart +44(0)113 343 3296 S.Asprey@leeds.ac.uk CAD/CAM Manufacturing Technician Astoul Aurelie A.A.V.Astoul@leeds.ac.uk Leverhulme Early Career Fellow (postdoctoral researcher) Atai Farrokh F.Atai@leeds.ac.uk Marie Curie Research Fellow Atkins Hayley +44(0)113 343 2080 H.Atkins@leeds.ac.uk Personal Assistant Atkinson Clair +44(0)113 343 7072 C.Atkinson@leeds.ac.uk School Administrative Manager Atwell Eric +44(0)113 343 5761 E.S.Atwell@leeds.ac.uk Professor of Artificial Intelligence for Language Auer Stefan +44(0)113 343 8669 S.Auer@leeds.ac.uk Lecturer Awais Yasar Y.Awais@leeds.ac.uk Senior Teaching Fellow in Architecture Aykroyd Robert +44(0)113 343 5167 R.G.Aykroyd@leeds.ac.uk Senior Lecturer Azam Kamran K.Azam@leeds.ac.uk Lead Technician Azizi Sadegh S.Azizi@leeds.ac.uk Lecturer in Smart Energy Systems Azmi Siddeequah +44(0)113 343 7795 S.M.Azmi@leeds.ac.uk Business Engagement Officer (WRIPA) Babaie Meisam M.Babaie@leeds.ac.uk Lecturer Babatunde Akintunde +44(0)113 343 2743 A.Babatunde@leeds.ac.uk Associate Professor Bacchetti Alistair A.Bacchetti@leeds.ac.uk Visiting Research Assistant Baker Alastair +44(0)113 343 9068 A.R.Baker@leeds.ac.uk Research Associate Baker Daniel D.L.Baker@leeds.ac.uk Experimental Officer Bakr Mustafa M.S.A.Bakr@leeds.ac.uk Visiting Researcher Balazs Boglarka +44(0)113 343 4023 B.N.Balazs@leeds.ac.uk School Administration Officer Baldwin Debra +44(0)113 343 2154 D.A.Baldwin@leeds.ac.uk Administrative Support Officer - iMBE Bale Catherine +44(0)113 343 8250 C.S.E.Bale@leeds.ac.uk Associate Professor Balijepalli Chandra +44(0)113 343 5345 N.C.Balijepalli@leeds.ac.uk Lecturer in Transport Engineering Model Balme Jessica +44(0)113 343 6307 J.M.Balme@leeds.ac.uk Student Education Service Officer Banaie Heravan Fatemeh F.BanaieHeravan@leeds.ac.uk Research Fellow Barber Stuart +44(0)113 343 5146 S.barber@leeds.ac.uk Senior Lecturer in Statistics Barker Joseph +44(0)113 343 3841 J.Barker@leeds.ac.uk Royal Society University Research Fellow Barker Joseph J.Barker2@leeds.ac.uk Research Fellow Barker Adrian +44(0)113 343 5165 A.J.Barker@leeds.ac.uk Associate Professor in Applied Mathematics Barker Richard +44(0)113 343 2206 R.J.Barker@leeds.ac.uk Associate Professor in Corrosion Science and Engineering Barns David D.G.Barns@leeds.ac.uk Research Fellow Barrington Dani +44(0)113 343 6260 D.J.Barrington@leeds.ac.uk Visiting Lecturer in Water, Sanitation & Health Barton David +44(0)113 343 2137 D.C.Barton@leeds.ac.uk Professor Bartram James J.K.Bartram@leeds.ac.uk Professor of Public Health and Environment Basheer Muhammed +44(0)113 343 2272 P.A.M.Basheer@leeds.ac.uk Chair in Structural Engineering Baur Karin K.U.Baur@leeds.ac.uk Professor Bayana Mary +44(0)113 343 6900 M.E.Bayana@leeds.ac.uk Laboratory Manager Bayly Andrew +44(0)113 343 0167 A.E.Bayly@leeds.ac.uk Chair in Chemical Engineering Beadling Rob +44(0)113 343 2135 A.R.Beadling@leeds.ac.uk Teaching & Research Fellow Beales Paul +44(0)113 343 9101 P.A.Beales@leeds.ac.uk Associate Professor Beaume Cedric +44(0)113 343 9916 C.M.L.Beaume@leeds.ac.uk Associate Professor in Applied Mathematics Beddows Angela +44(0)113 343 3855 A.Beddows@leeds.ac.uk Technician Beige Almut +44(0)113 343 3899 A.Beige@leeds.ac.uk Associate Professor Bell Andrew +44(0)113 343 2370 A.J.Bell@leeds.ac.uk Professor Ben Rached Nadhir N.BenRached@leeds.ac.uk Lecturer Bennett Brandon +44(0)113 343 1070 B.Bennett@leeds.ac.uk Lecturer Bernal Lopez Susan S.A.BernalLopez@leeds.ac.uk Professor of Structural Materials Berretta Christian C.Berretta@leeds.ac.uk Lecturer Berry Helen +44(0)113 343 5617 H.E.Berry@leeds.ac.uk University Academic Fellow Besharat Mohsen M.Besharat@leeds.ac.uk Lecturer (Assistant Professor) Bettadapura Subramanyam Shashank S.BettadapuraSubramanyam@leeds.ac.uk Marie Curie Research Fellow Bhat Pallavi P.Bhat@leeds.ac.uk Research Fellow Bhogal Gurdev +44(0)113 343 2519 G.S.Bhogal@leeds.ac.uk Research Technician Bilbrough Andrew A.D.Bilbrough@leeds.ac.uk Teaching Support Technician Billeter Markus M.Billeter@leeds.ac.uk Assistant Professor Black Leon +44(0)113 343 2283 L.Black@leeds.ac.uk Professor of Infrastructure Materials Blacker John +44(0)113 343 8239 J.Blacker@leeds.ac.uk Professor Blakeley Ian +44(0)113 343 6472 I.J.Blakeley@leeds.ac.uk Building Manager/Safety Adviser Blitz Mark M.Blitz@leeds.ac.uk Research Fellow Bogachev Leonid +44(0)113 343 4972 L.V.Bogachev@leeds.ac.uk Reader Bokhove Onno +44(0)113 343 9751 O.Bokhove@leeds.ac.uk Chair in Geophysical Fluid Dynamics Bollada Peter +44(0)113 343 2567 P.C.Bollada@leeds.ac.uk Research Fellow Bonner Faith +44(0)113 343 3810 F.R.C.Bonner@leeds.ac.uk Research and Mgmt Suppt Officer Boocock Hardy +44(0)113 343 2134 H.Boocock@leeds.ac.uk Teaching Support Technician Booth Andrew A.Booth@leeds.ac.uk Research Fellow Boral Soumava S.Boral@leeds.ac.uk Research Fellow in Artificial Intelligence (Circular Economy) Borissova Dimitrova Antonia +44(0)113 343 2421 A.Borissova@leeds.ac.uk Teaching Fellow Borman Duncan +44(0)113 343 2354 D.J.Borman@leeds.ac.uk Associate Professor Bourne Richard +44(0)113 343 6547 R.A.Bourne@leeds.ac.uk Professor of Digital Chemical Engineering Boustead Graham G.A.Boustead@leeds.ac.uk Research Fellow Bower Denise +44(0)113 343 2271 D.A.Bower@leeds.ac.uk Professor Boyle Jordan +44(0)113 343 2140 J.H.Boyle@leeds.ac.uk Lectureship in Engineering Systems Bradley Derek +44(0)113 343 2115 D.Bradley@leeds.ac.uk Research Professor Brady David D.J.Brady@leeds.ac.uk Research Project Manager Bray Edward +44(0)113 343 2077 E.P.Bray@leeds.ac.uk Technician Bray Nicky +44(0)113 343 9934 N.J.Bray@leeds.ac.uk Team Challenge Administrator Brennan Jessica +44(0)113 343 0075 J.M.Brennan@leeds.ac.uk Project Manager Bressloff Neil N.Bressloff@leeds.ac.uk Professor of Biomedical Engineering & Design Brickwood Alan +44(0)113 343 2224 A.L.Brickwood@leeds.ac.uk Formula Car Support Technician Brindley John +44(0)113 343 5134 J.Brindley@leeds.ac.uk Research Professor Brine Alison +44(0)113 343 2262 A.E.Brine@leeds.ac.uk Education Service Officer Briscoe Adam A.B.Briscoe@leeds.ac.uk Visiting Research Fellow Britton Andrew A.Britton@leeds.ac.uk Research Officer Broadbent Matthew +44(0)113 343 2942 M.Broadbent@leeds.ac.uk Mechanical Workshop Technician Brockett Claire +44(0)113 343 7472 C.L.Brockett@leeds.ac.uk Associate Professor of Biomechanical Engineering Brooke-Taylor Andrew +44(0)113 343 7101 A.D.Brooke-Taylor@leeds.ac.uk Associate Professor Brookes Amy A.Brookes@leeds.ac.uk Research Development Support Officer Brooks Peter +44(0)113 343 2122 P.C.Brooks@leeds.ac.uk Associate Professor Brown Aaron A.E.Brown@leeds.ac.uk EPSRC Doctoral Prize Fellow Brown Graham +44(0)113 343 8264 G.Brown@leeds.ac.uk Mechanical Services Manager Brown Andy +44(0)113 343 2382 A.P.Brown@leeds.ac.uk Professor of Materials Characterisation Brunk Rebecca R.L.Brunk@leeds.ac.uk Research Fellow Bryan-Kinns Emily +44(0)113 343 6937 E.Bryan-Kinns@leeds.ac.uk CDT Administration Officer Bryant Michael +44(0)113 343 2161 M.G.Bryant@leeds.ac.uk Associate Professor Buckley Matthew +44(0)113 343 2134 M.W.Buckley@leeds.ac.uk Senior Electronics Technician Bukhari-Bibi Ruksana +44(0)113 343 6788 R.Bukhari-Bibi@leeds.ac.uk Education Service Assistant Bulman Christopher C.A.Bulman@leeds.ac.uk Analytical Technician Bulpitt Andy +44(0)113 343 6816 A.J.Bulpitt@leeds.ac.uk Professor of Computer Science Burdon Samuel +44(0)113 343 9133 S.G.Burdon@leeds.ac.uk Research Support Technician Burnell Gavin +44(0)113 343 3843 G.Burnell@leeds.ac.uk Associate Professor of Condensed Matter Physics Burnett Jordan J.Burnett1@leeds.ac.uk Technician Burnett Andrew +44(0)113 343 0997 A.D.Burnett@leeds.ac.uk Associate Professor Burns Zoe +44(0)113 343 6331 Z.S.Burns@leeds.ac.uk Education Service Officer / Receptionist Burton Isobel +44(0)113 343 3860 I.R.Burton@leeds.ac.uk Education Service Officer Bushby Richard +44(0)113 343 6509 R.J.Bushby@leeds.ac.uk Research Professor Byrne Michelle +44(0)113 343 2113 M.Byrne@leeds.ac.uk Administrative Support Officer - iDRO Califano Marco +44(0)113 343 2088 M.Califano@leeds.ac.uk Associate professor Callaghan Martin +44(0)113 343 0273 M.Callaghan@leeds.ac.uk Lecturer Campbell Anya A.Campbell1@leeds.ac.uk Education Service Officer (Admissions) Cardie Jane +44(0)113 343 2171 J.E.Cardie@leeds.ac.uk Technician Carney John +44(0)113 343 2022 j.a.carney@leeds.ac.uk Faculty Research Manager Carpenter Thomas T.Carpenter@leeds.ac.uk Visitor (Research) Carr Hamish +44(0)113 343 7042 H.Carr@leeds.ac.uk Professor of Computer Science Cassidy Tyler T.Cassidy1@leeds.ac.uk Lecturer in Mathematical Biology Caudrelier Vincent +44(0)113 343 9522 V.Caudrelier@leeds.ac.uk Lecturer in Mathematical Physics Cayre Olivier +44(0)113 343 4939 O.J.Cayre@leeds.ac.uk Associate Professor (Academic) Cespedes Oscar +44(0)113 343 1885 O.Cespedes@leeds.ac.uk Professor of Condensed Matter Physics Chakhlevitch Natasha Shakhlevich +44(0)113 343 5444 N.Shakhlevich@leeds.ac.uk Senior Lecturer Chalykh Oleg +44(0)113 343 5112 O.Chalykh@leeds.ac.uk Lecturer Chamberlain Thomas +44(0)113 343 6468 T.W.Chamberlain@leeds.ac.uk Associate Professor Chambers Anthony A.Chambers@leeds.ac.uk Student Education Service Assistant Chandler James J.H.Chandler@leeds.ac.uk Lecturer in Surgical Robotics Chapman David D.W.Chapman@leeds.ac.uk Technician Chau Hau Hing +44(0)113 343 2111 H.H.Chau@leeds.ac.uk Research Fellow Chau Chalmers C.C.Chau@leeds.ac.uk Research Fellow Chen Li +44(0)113 343 5215 L.Chen@leeds.ac.uk Experimental Officer Chen Xiaohui +44(0)113 343 0350 X.Chen@leeds.ac.uk Associate Professor in Geotechnical Engineering Chester Nick +44(0)113 343 0929 N.J.Chester@leeds.ac.uk Reporting Officer in Medical Technology Childs Tom +44(0)113 343 2207 T.H.C.Childs@leeds.ac.uk Emeritus Professor Chittenden Richard +44(0)113 343 2176 R.J.Chittenden@leeds.ac.uk Teaching Fellow Chong Benjamin +44(0)113 343 9091 B.Chong@leeds.ac.uk Lecturer Choudhry Omar O.Choudhry@leeds.ac.uk Teaching Assistant Choudhury Charisma +44(0)113 343 2659 C.F.Choudhury@leeds.ac.uk Chair in Behaviour Modelling Chowney Steven S.J.Chowney@leeds.ac.uk Stores Technician Christie Jessica +44(0)113 343 0794 J.R.Christie@leeds.ac.uk Student Education Service Functional Manager (Student Support) Chuang HsiaoHan H.G.Chuang@leeds.ac.uk Research associate Clark Kate +44(0)113 343 2118 K.L.Clark@leeds.ac.uk Flexible Learning Officer Clark Laura L.Clark@leeds.ac.uk Marie Curie Research Fellow Clark Amanda +44(0)113 343 3839 A.Clark1@leeds.ac.uk Student Education Service Officer (PGR) Clarke Roland +44(0)113 343 2094 R.G.Clarke@leeds.ac.uk Associate Professor Clarke Robert +44(0)113 343 2277 R.Clarke@leeds.ac.uk Technician Clarke Barry +44(0)113 343 9454 B.G.Clarke@leeds.ac.uk Professor Clarke Kirsty +44(0)113 343 5055 K.J.Clarke@leeds.ac.uk Student Education Service Officer (PGR) Clarke Deborah D.Clarke2@leeds.ac.uk Administrative Support Assistant Clarkson Mandy +44(0)113 343 6451 M.J.Clarkson@leeds.ac.uk School Research Support Officer Clayton Adam A.D.Clayton@leeds.ac.uk University Academic Fellow Cochrane Diane +44(0)113 343 2363 D.Cochrane@leeds.ac.uk Technician Cochrane Robert +44(0)113 343 2359 R.F.Cochrane@leeds.ac.uk Senior Lecturer Cockerill Timothy +44(0)113 343 7678 T.Cockerill@leeds.ac.uk Chair in Efficient Energy Utilisation Cohen Netta +44(0)113 343 6789 N.Cohen@leeds.ac.uk Professor Cohn Anthony +44(0)113 343 5482 A.G.Cohn@leeds.ac.uk Professor of Automated Reasoning Collins Sean S.M.Collins@leeds.ac.uk University Academic Fellow Colombo Marco M.Colombo@leeds.ac.uk Research Fellow Connell Simon +44(0)113 343 8241 S.D.A.Connell@leeds.ac.uk Associate Professor Connolly David +44(0)113 343 6304 D.Connolly@leeds.ac.uk Professor of Railway Engineering Cook Ed E.R.Cook@leeds.ac.uk Research Fellow in Circular Economy Systems for Waste Plastics Cooper Robert +44(0)113 343 9210 R.J.Cooper@leeds.ac.uk KTP Associate Copeland Helen +44(0)113 343 3210 H.O.W.Copeland@leeds.ac.uk Administrative Assistant Correia Benjamin +44(0)113 343 3765 B.Correia@leeds.ac.uk Estates and Fabric Technician Cottom Josh J.W.Cottom@leeds.ac.uk Research Fellow in Plastics Pollution Coupe Jennifer +44(0)113 343 3628 J.A.Coupe@leeds.ac.uk Deputy Employability and Placement Manager Cousens Terry T.W.Cousens@leeds.ac.uk Senior Lecturer Cowell David +44(0)113 343 4965 D.M.J.Cowell@leeds.ac.uk Teaching & Research Fellow Emb Syst/Ultr Cowie Raelene +44(0)113 343 9112 R.Cowie@leeds.ac.uk Research Fellow Critchley Kevin +44(0)113 343 3873 K.Critchley@leeds.ac.uk Associate Professor Critchley Kevin +44(0)113 343 3873 K.Critchley@leeds.ac.uk Associate Professor Culmer Peter +44(0)113 343 2141 P.R.Culmer@leeds.ac.uk Associate Professor Cunliffe Adrian +44(0)113 343 2470 A.M.Cunliffe@leeds.ac.uk Analytical Spport Technician Cunningham John +44(0)113 343 0618 J.E.Cunningham@leeds.ac.uk Professor Cutillo Luisa L.Cutillo@leeds.ac.uk Lecturer in Statistics Dai Sheng +44(0)113 343 0313 S.Dai1@leeds.ac.uk Professor of Chemical Engineering Dang Minh M.T.Dang@leeds.ac.uk Teaching Assistant Danso-Boateng Eric +44(0)113 343 7125 E.Danso-Boateng@leeds.ac.uk Teaching Fellow Dareiotis Konstantinos K.Dareiotis@leeds.ac.uk Lecturer Darwood Kim K.M.Darwood@leeds.ac.uk General Support Assistant and Receptionist Das Chinmay +44(0)113 343 2930 C.Das@leeds.ac.uk Research Fellow Davies Giles +44(0)113 343 7075 G.Davies@leeds.ac.uk Professor Dawson Peter +44(0)113 343 2565 mtlpgd@leeds.ac.uk Technician Dawson David +44(0)113 343 9548 D.A.Dawson@leeds.ac.uk Lecturer in Transport Management & Resilience Day Gavin +44(0)113 343 9402 G.Day1@leeds.ac.uk Research Fellow Day Stephanie +44(0)113 343 8461 S.J.Day@leeds.ac.uk Faculty Office Administrator De Angelis Tiziano +44(0)113 343 0392 T.DeAngelis@leeds.ac.uk Lecturer in Actuarial/Financial Maths de Boer Gregory +44(0)113 343 2607 G.N.deBoer@leeds.ac.uk Lecturer in Aeronautical and Aerospace Engineering de Jong Anna +44(0)113 343 2054 A.deJong@leeds.ac.uk Clerk de Kamps Marc +44(0)113 343 5322 M.deKamps@leeds.ac.uk Lecturer De Leeuw Nora N.H.deLeeuw@leeds.ac.uk Professor of Computational Chemistry De Pennington Alan +44(0)113 343 2207 A.dePennington@leeds.ac.uk Visiting Professor Dean Paul +44(0)113 343 2095 P.Dean@leeds.ac.uk University Research Fellow Dechant Pierre-Philippe P.P.Dechant@leeds.ac.uk Curriculum Redefined Lecturer Deger Aydin A.Deger@leeds.ac.uk Visiting Research Fellow Dehghani-Sanij Abbas +44(0)113 343 2906 A.A.Dehghani-Sanij@leeds.ac.uk Chair in Bio-Mech & Medical Robotics Derks Didi +44(0)113 343 4177 D.Derks@leeds.ac.uk Teaching Fellow in Physical Chemistry Desai Amisha +44(0)113 343 9238 A.Desai@leeds.ac.uk Research Fellow Dhandapani Yuvaraj Y.Dhandapani@leeds.ac.uk Research Fellow in Durability of Cement and Concrete Materials Dickinson Nicola +44(0)113 343 2787 N.H.Dickinson@leeds.ac.uk Finance Officer Dickinson Sara +44(0)113 343 7049 S.D.J.Dickinson@leeds.ac.uk Administrative Support Assistant Dickinson Jennie +44(0)113 343 6522 J.Dickinson@leeds.ac.uk Technician Dillon Nicholas +44(0)113 343 8452 N.Dillon@leeds.ac.uk Senior Research Officer (Post Award) Dimitrova Vania +44(0)113 343 1674 V.G.Dimitrova@leeds.ac.uk Professor of Human-Centred Artificial Intelligence Dixon-Hardy Darron +44(0)113 275 1265 D.W.Dixon-Hardy@leeds.ac.uk Senior Lecturer Djemame Karim +44(0)113 343 6590 K.Djemame@leeds.ac.uk Professor Dogar Mehmet +44(0)113 343 5777 M.R.Dogar@leeds.ac.uk Associate Professor Doidge Fleur +44(0)113 343 2508 F.Doidge@leeds.ac.uk School Administration Manager Doorsamy Wesley W.Doorsamy@leeds.ac.uk Lecturer Dorgham Abdel A.Dorgham@leeds.ac.uk Research Fellow Dougan Lorna +44(0)113 343 8958 L.Dougan@leeds.ac.uk Professor of Physics Douglas Ben +44(0)113 343 2378 B.Douglas@leeds.ac.uk Research Technician Douglas Kevin K.M.Douglas@leeds.ac.uk Research Fellow Drewniok Michal M.P.Drewniok@leeds.ac.uk Research Fellow in Transforming Foundation Industries Drummond-Brydson Rik +44(0)113 343 2369 R.M.Drummond-Brydson@leeds.ac.uk Professor Dubey Shival S.Dubey@leeds.ac.uk Research Fellow in Robotics Duff Gerard G.Duff@leeds.ac.uk Lecturer Duke David +44(0)113 343 6800 D.J.Duke@leeds.ac.uk Professor Emeritus of Computer Science Dupont Valerie +44(0)113 343 2503 V.Dupont@leeds.ac.uk Reader Duxbury Toni +44(0)113 343 2490 T.J.Duxbury@leeds.ac.uk Research Support Officer Dyer Martin +44(0)113 343 5442 M.E.Dyer@leeds.ac.uk Professor Dyer Keith +44(0)113 343 9133 K.Dyer@leeds.ac.uk Research Support Technician Edmonds David +44(0)113 343 2386 D.V.Edmonds@leeds.ac.uk Visiting Professor Edwards Jen +44(0)113 343 3085 J.H.Edwards@leeds.ac.uk University Academic Fellow Efford Nick +44(0)113 343 6809 N.D.Efford@leeds.ac.uk Senior Teaching Fellow Eisele Heribert +44(0)113 343 7074 H.Eisele@leeds.ac.uk Reader Eke Paul P.E.Eke@leeds.ac.uk Royal Academy of Engineering [RAEng] Visiting Professor Eleftheriou Pantelis P.Eleftheriou@leeds.ac.uk University Academic Fellow Elgorashi Taisir T.E.H.Elgorashi@leeds.ac.uk Lecturer in Optical Networks Elliott David +44(0)113 343 9453 D.J.S.Elliott@leeds.ac.uk Technician Ellis Martin Noemy +44(0)113 343 3641 N.EllisMartin@leeds.ac.uk Student Education Service Manager (Programme Support) Elmirghani Jaafar +44(0)113 343 2013 J.M.H.Elmirghani@leeds.ac.uk Head of Institute Elumalai Jayaprasath J.Elumalai@leeds.ac.uk Research Fellow Elwes Richard +44(0)113 343 5864 R.H.Elwes@leeds.ac.uk Senior Teaching Fellow Emery Paul +44(0)113 392 4884 P.Emery@leeds.ac.uk Professor (Clinical) Esat Faye +44(0)113 343 4142 F.Esat@leeds.ac.uk Experimental Officer X-ray Diffraction Espejo Conesa Cayetano +44(0)113 343 9962 C.EspejoConesa@leeds.ac.uk SWJTU Teaching Fellow Etchels Lee +44(0)113 343 6360 L.W.Etchels@leeds.ac.uk Research Fellow Eterovic Sebastian S.Eterovic@leeds.ac.uk Research Fellow Evans Stephen +44(0)113 343 3852 S.D.Evans@leeds.ac.uk Professor Evans Craig +44(0)113 343 4557 C.A.Evans@leeds.ac.uk Associate Professor Evans R M L +44(0)113 343 5861 R.M.L.Evans@leeds.ac.uk Associate Professor Evans Barbara +44(0)113 343 1990 B.E.Evans@leeds.ac.uk Professor of Public Health Engineering Eyley Jenny J.Eyley@leeds.ac.uk Lecturer Faber Eleonore +44(0)113 343 5185 E.M.Faber@leeds.ac.uk Associate Professor Fabiyi Samson S.D.Fabiyi@leeds.ac.uk Transnational Teaching Fellow Fairweather Michael M.Fairweather@leeds.ac.uk Professor Falle Samuel +44(0)113 343 5138 S.A.E.G.Falle@leeds.ac.uk Professor Fang Han H.Fang1@leeds.ac.uk Lecturer in Structural Engineering Fedele Francesca F.Fedele@leeds.ac.uk Research Fellow Fernandes Bruno B.Fernandes@leeds.ac.uk Research Fellow in Durability of Circular Concrete Feroleto Samuel S.Feroleto@leeds.ac.uk Summer Intern Field Sarah S.L.Field@leeds.ac.uk Knowledge Exchange Manager Figueredo Luis L.Figueredo@leeds.ac.uk Marie Curie Fellow Finn Robyn R.Finn@leeds.ac.uk Research Support Technician Finn Adrian A.Finn@leeds.ac.uk Visiting Associate Professor Fisher Quentin +44(0)113 343 1920 Q.J.Fisher@leeds.ac.uk Professor Fisher John +44(0)113 343 2128 J.Fisher@leeds.ac.uk Emeritus Professor Fishwick Colin +44(0)113 343 6510 C.W.G.Fishwick@leeds.ac.uk Professor Fitzgerald Steve +44(0)113 343 4331 S.P.Fitzgerald@leeds.ac.uk Associate Professor Fleming Lauren L.T.Fleming@leeds.ac.uk Research Fellow Fletcher Louise +44(0)113 343 2328 L.A.Fletcher@leeds.ac.uk Lecturer in Environmental Engineering Flint Samuel S.E.Flint@leeds.ac.uk Lead Technician (iTF) Fogarty David +44(0)113 343 6491 D.Fogarty@leeds.ac.uk Technician Fordy Allan +44(0)113 343 5115 A.P.Fordy@leeds.ac.uk Professor Forsyth Helen H.L.Forsyth@leeds.ac.uk CPD, Conference and Events Coordinator Forth John +44(0)113 343 2270 J.P.Forth@leeds.ac.uk Prof of Concrete Engineering & Structure Foster Richard +44(0)113 343 5759 R.Foster@leeds.ac.uk Associate Professor Fox Natalie N.Fox@leeds.ac.uk Senior Research Technician Frame Douglas +44(0)113 343 6587 D.S.Frame@leeds.ac.uk PC Service Leader Francis Charlotte +44(0)113 343 5821 C.Francis@leeds.ac.uk Assistant School Education Service Mger Frangi Alex +44(0)113 343 9640 A.Frangi@leeds.ac.uk Diamond Jubilee Chair in Computational Medicine | Royal Academy Freear Steven +44(0)113 343 2076 S.Freear@leeds.ac.uk Professor of Ultrasonics and Embedded Systems Freeman Helen H.M.Freeman@leeds.ac.uk Network Manager Freeman Joshua +44(0)113 343 8195 J.R.Freeman@leeds.ac.uk Associate Professor Frittaion Emanuele E.Frittaion@leeds.ac.uk Research Fellow in Pure Mathematics Gailani Ahmed A.Gailani@leeds.ac.uk Research Fellow in Industrial Decarbonisation Gale William +44(0)113 343 2796 W.F.Gale@leeds.ac.uk Professor Gallagher Justin J.F.Gallagher@leeds.ac.uk Teaching and Research Fellow Galloway Johanna J.M.Galloway@leeds.ac.uk Post-doctoral Research Associate Garcia-Taengua Emilio +44(0)113 343 0698 E.Garcia-Taengua@leeds.ac.uk Associate Professor in Structures Gardner Sarah +44(0)113 343 3881 S.M.Gardner@leeds.ac.uk Student Education Service Officer (Adm) Garrity Stephen +44(0)113 343 5388 S.W.Garrity@leeds.ac.uk Professor George Midhun M.George@leeds.ac.uk Research Fellow Ghadiri Mojtaba +44(0)113 343 2406 M.Ghadiri@leeds.ac.uk Professor Ghalaii Masoud M.Ghalaii@leeds.ac.uk Research Fellow in Quantum Communications Ghanbarzadeh Ali A.Ghanbarzadeh@leeds.ac.uk Lecturer in Functional Surfaces Gilkeson Carl +44(0)113 343 6915 C.A.Gilkeson@leeds.ac.uk Lecturer in Aerospace Engineering Gilkeson Natalie N.Gilkeson@leeds.ac.uk Teaching Fellow Gleeson Helen +44(0)113 343 3863 H.F.Gleeson@leeds.ac.uk Cavendish Professor of Physics Golshani Shokoufeh S.Golshani@leeds.ac.uk Marie Curie Researcher Goncalves Faria Martins Joao +44(0)113 343 4433 J.FariaMartins@leeds.ac.uk Lecturer in Algebra Gonzalez Montoya Francisco F.GonzalezMontoya@leeds.ac.uk Research Associate Gorman Stephen +44(0)113 343 6512 S.A.Gorman@leeds.ac.uk Laboratory Manager Gorrell Ian I.B.Gorrell@leeds.ac.uk Visiting Research Fellow Gouldson Andy +44(0)113 343 9753 A.Gouldson@leeds.ac.uk Dean: Interdisciplinary Res within CS Graham Emma +44(0)113 343 3804 E.J.Graham@leeds.ac.uk School Administration Manager Grant Rowan +44(0)113 343 0923 R.H.Grant@leeds.ac.uk Communications and Engagement Manager for Medical Technologies Grant-Muller Susan +44(0)113 343 6618 S.M.Grant-Muller@its.leeds.ac.uk Chair in Technologies & Informatics Graves Daniel D.Graves@leeds.ac.uk Teaching Fellow Gray Lucy +44(0)113 343 3086 L.V.Gray@leeds.ac.uk Administrative Support Assistant Green Michael +44(0)113 343 2456 fbsmgr@leeds.ac.uk Technician Greenbank Rachel +44(0)113 343 2302 R.J.Greenbank@leeds.ac.uk Education Service Officer Grieve Peter +44(0)113 343 7276 P.W.Grieve@leeds.ac.uk Technician Griffiths Stephen +44(0)113 343 5186 S.D.Griffiths@leeds.ac.uk Lecturer in applied mathematics Grigorova Miryana M.R.Grigorova@leeds.ac.uk Lecturer in Financial and Actuarial Mathematics Gronqvist Marcus +44(0)113 343 0543 M.N.Gronqvist@leeds.ac.uk Senior Teaching Fellow Guarino Maria Vittoria M.Guarino@leeds.ac.uk Research Fellow Guest Robert +44(0)113 343 2184 R.Guest@leeds.ac.uk Technician Guseva Anna A.Guseva@leeds.ac.uk Marie Curie Research Fellow Gusnanto Arief +44(0)113 343 5135 A.Gusnanto@leeds.ac.uk Associate Professor Hainsworth Tim +44(0)113 343 5163 T.J.Hainsworth@leeds.ac.uk Computer Officer Halcrow Malcolm +44(0)113 343 6506 M.A.Halcrow@leeds.ac.uk Professor Hall Richard M Hall +44(0)113 343 2132 R.M.Hall@leeds.ac.uk Professor Hammersley Camille +44(0)113 343 2190 C.Hammersley@leeds.ac.uk Technician Hammond Robert +44(0)113 343 2428 R.B.Hammond@leeds.ac.uk Lecturer Hanson Bruce +44(0)113 343 0475 B.C.Hanson@leeds.ac.uk Professor Harbottle David +44(0)113 343 4154 D.Harbottle@leeds.ac.uk Associate Professor Hardcastle Thomas T.Hardcastle@leeds.ac.uk KTP Associate Hardie Michaele +44(0)113 343 6458 M.J.Hardie@leeds.ac.uk Professor Harding Dawn +44(0)113 343 2229 D.E.Harding@leeds.ac.uk Education Service Officer Harland Derek +44(0)113 343 5152 D.G.Harland@leeds.ac.uk Associate Professor in Geometry Harlen Oliver +44(0)113 343 5189 O.G.Harlen@leeds.ac.uk Reader Harrington John +44(0)113 343 2559 J.P.Harrington@leeds.ac.uk Facility Manager/Senior Experimental Officer Harris Sarah +44(0)113 343 3816 S.A.Harris@leeds.ac.uk Associate Professor Harris Allen +44(0)113 343 3820 A.Harris@leeds.ac.uk Technician Harris Russell +44(0)113 343 2155 R.Harris@leeds.ac.uk Professor Harris Robert R.I.Harris@leeds.ac.uk Technician Harrison Darren +44(0)113 343 3296 D.R.Harrison@leeds.ac.uk Robotics Manufacturing Technician Hartquist Thomas +44(0)113 343 3885 T.W.Hartquist@leeds.ac.uk Professor Hassanpour Ali +44(0)113 343 2405 A.Hassanpour@leeds.ac.uk Associate Professor Hawksworth Louise +44(0)113 343 6465 L.Hawksworth@leeds.ac.uk School Administrator Hayler Anne +44(0)113 343 2228 A.Hayler@leeds.ac.uk School Education Service Manager Haynes David +44(0)113 343 7907 D.I.Haynes@leeds.ac.uk Senior Administration Assistant Hazlehurst Thomas T.Hazlehurst@leeds.ac.uk Research Fellow Head David +44(0)113 343 4693 D.Head@leeds.ac.uk Lecturer Heard Dwayne +44(0)113 343 6471 D.E.Heard@leeds.ac.uk Professor of Atmospheric Chemistry Heath George G.R.Heath@leeds.ac.uk University Academic Fellow Heggs Peter +44(0)113 343 2386 P.J.Heggs@leeds.ac.uk Visiting Professor Heitor Ana A.Heitor@leeds.ac.uk Lecturer in Geotechnical Engineering Herbert Anthony +44(0)113 343 7371 A.Herbert@leeds.ac.uk Lecturer in Medical and Biological Engineering Herbert Megan M.C.Herbert@leeds.ac.uk Project Manager Heyam Alex A.Heyam@leeds.ac.uk Experimental Officer for NMR Hickey B J +44(0)113 343 3836 B.J.Hickey@leeds.ac.uk Professor Higgins Luke L.J.R.Higgins@leeds.ac.uk EPSRC Doctoral Prize Fellow Hilditch Beth +44(0)113 343 5465 B.A.Hilditch@leeds.ac.uk Education Service Officer Hill Peter +44(0)113 343 8983 P.R.Hill@leeds.ac.uk Pre+Post Award Administrator Hine Peter +44(0)113 343 3827 P.J.Hine@leeds.ac.uk Associate Professor Hiwar Waseem W.F.M.Hiwar@leeds.ac.uk Researcher Hoare Melvin +44(0)113 343 3864 M.G.Hoare@leeds.ac.uk Professor Hobson Susan +44(0)113 343 2070 S.Hobson@leeds.ac.uk Secretary to Institute & PA to Pro Dean Hodges Christopher C.S.Hodges@leeds.ac.uk Research Fellow Hodgson Daniel D.R.E.Hodgson@leeds.ac.uk Demonstrator/Module Assistant Hogg David +44(0)113 343 5765 D.C.Hogg@leeds.ac.uk Professor of Artificial Intelligence Holdsworth Nick +44(0)113 343 1940 N.J.Holdsworth@leeds.ac.uk Faculty Accountant Holdsworth Alex A.Holdsworth1@leeds.ac.uk Apprentice Laboratory Technician Holland Andrew A.D.Holland@leeds.ac.uk Research Fellow in Architectural Heritage and VR Modelling Hollerbach Rainer +44(0)113 343 5134 R.Hollerbach@leeds.ac.uk Professor Hollins Andrew +44(0)113 343 2308 A.T.Hollins@leeds.ac.uk School Education Service Manager Holmes Kimberley +44(0)113 343 6553 K.Holmes@leeds.ac.uk Technician Holmes Stephen +44(0)113 343 8460 S.Holmes@leeds.ac.uk Technician Holroyd Tom +44(0)113 343 2330 T.Holroyd@leeds.ac.uk Education Service Officer Holroyd Sadie S.Holroyd@leeds.ac.uk School Administrative Assistant Holt Raymond +44(0)113 343 7936 R.J.Holt@leeds.ac.uk Lecturer Hondow Nicole +44(0)113 343 2056 N.Hondow@leeds.ac.uk Associate Professor Honore Teresa +44(0)113 343 5222 M.Honore@leeds.ac.uk School Office and Projects Manager Houston Kevin +44(0)113 343 5136 K.Houston@leeds.ac.uk Senior Lecturer Houwing-Duistermaat Jeanne +44(0)113 343 9821 J.Duistermaat@leeds.ac.uk Chair in Data Analytics and Statistics Howling Graeme +44(0)113 343 0908 G.Howling@leeds.ac.uk Technology Manager Hoyle Brian +44(0)113 343 2386 B.S.Hoyle@leeds.ac.uk Visiting Professor Hoz de Vila Eduardo Kattia K.HozdeVila@leeds.ac.uk Research Software Engineer Huang Wengui W.Huang@leeds.ac.uk Research Fellow Huang Yanlong +44(0)113 343 3505 Y.L.Huang@leeds.ac.uk University Academic Fellow Huerta Omar O.I.HuertaCardoso@leeds.ac.uk Lecturer Huggan Michael +44(0)113 343 2196 M.Huggan@leeds.ac.uk Technician Hughes David +44(0)113 343 5105 D.W.Hughes@leeds.ac.uk Professor Hunter Ian +44(0)113 343 2055 I.C.Hunter@leeds.ac.uk Emeritus Professor Hunter Timothy +44(0)113 343 2790 T.N.Hunter@leeds.ac.uk Associate Professor (Academic) Hutchings Paul P.Hutchings@leeds.ac.uk Lecturer in Water, Sanitation & Health Iglesias Vallejo Daniela D.P.Iglesias@leeds.ac.uk Research Fellow Ihara Larissa L.M.Ihara@leeds.ac.uk Marie Curie ESR Researcher Ikonic Zoran +44(0)113 343 7320 Z.Ikonic@leeds.ac.uk Reader Ilee John J.D.Ilee@leeds.ac.uk STFC Ernest Rutherford Fellow & University Academic Fellow Indjin Dragan +44(0)113 343 2082 D.Indjin@leeds.ac.uk Reader Ingham Eileen +44(0)113 343 5691 E.Ingham@leeds.ac.uk Chair of Medical Immunology Ingham Trevor T.Ingham@leeds.ac.uk Senior Research Fellow Ingham Nancy N.Ingham@leeds.ac.uk Centre Manager Iqbal Nasser +44(0)113 343 3388 M.N.Iqbal@leeds.ac.uk Management Accountant Irabor Kenneth K.Irabor@leeds.ac.uk Experimental Officer Isaac Dec D.T.Isaac@leeds.ac.uk Apprentice Technician Isaac Graham G.H.Isaac@leeds.ac.uk Professor Issoglio Elena +44(0)113 343 4660 E.Issoglio@leeds.ac.uk Lecturer (Academic) Iuorio Ornella +44(0)113 343 2294 O.Iuorio@leeds.ac.uk Professor of Architecture & Structures Jackson Andrew +44(0)113 343 6480 A.E.Jackson@leeds.ac.uk Research/Teaching Fellow James Alexander (Sandy) A.James1@leeds.ac.uk Research Fellow Jaramillo Cevallos Pablo P.Jaramillo@leeds.ac.uk Research Fellow Javed Mohammed +44(0)113 343 2396 M.Javed@leeds.ac.uk Technician Jennings Louise +44(0)113 343 2100 L.M.Jennings@leeds.ac.uk Professor of Medical Engineering Jennings David D.Jennings@leeds.ac.uk Lecturer and University Academic Fellow Jha Animesh +44(0)113 343 2342 A.Jha@leeds.ac.uk Professor Ji Lanpeng +44(0)113 343 5891 L.Ji@leeds.ac.uk Lecturer in Actuarial/Financial Maths Jia Xiaodong +44(0)113 343 2801 X.Jia@leeds.ac.uk Lecturer (Academic) Jimack Peter +44(0)113 343 2002 P.K.Jimack@leeds.ac.uk Professor of Scientific Computing Jimenez-Cruz David D.Jimenez-Cruz@leeds.ac.uk Research Fellow Jin Zhongmin Z.Jin@leeds.ac.uk Visiting Professor Jinks Michael M.Jinks@leeds.ac.uk Postdoctoral Research Fellow Johnson Owen +44(0)113 343 5459 O.A.Johnson@leeds.ac.uk Senior Teaching Fellow Johnson Jenny +44(0)113 343 2127 J.E.Johnson@leeds.ac.uk Education Service Officer Johnson Peter +44(0)113 343 6515 P.Johnson@leeds.ac.uk Emertius Professor Johnson Benjamin +44(0)113 343 7127 B.R.G.Johnson@leeds.ac.uk Experimental Officer Johnston Katharine +44(0)113 343 8279 K.G.Johnston@leeds.ac.uk Research Fellow Jones Cliff +44(0)113 343 7311 J.C.Jones@leeds.ac.uk Professor Jones Alison +44(0)113 343 2099 A.C.Jones@leeds.ac.uk Associate Professor of Computational Biomedical Engineering Jones Margaret +44(0)113 343 5101 medsjon@leeds.ac.uk School Administrator Jones Jenny +44(0)113 343 2477 J.M.Jones@leeds.ac.uk Professor Jones Christopher +44(0)113 343 5107 C.A.Jones@maths.leeds.ac.uk Professor Jose Gin +44(0)113 343 2536 G.Jose@leeds.ac.uk Chair in Functional Materials Kaddouh Bilal +44(0)113 343 2201 B.Kaddouh@leeds.ac.uk Lecturer in Aerial Robotics (Assistant Professor) Kailas Lekshmi L.Kailas@leeds.ac.uk Experimental Officer in AFM Kale Girish +44(0)113 343 2805 G.M.Kale@leeds.ac.uk Reader Kamarol Zaman Faizal M.F.KamarolZaman@leeds.ac.uk Marie Curie Early Stage Researcher Kamde Deepak D.K.Kamde@leeds.ac.uk UKRI Research Fellow Kanuganti Sandeep S.R.Kanuganti@leeds.ac.uk Research Fellow Kapur Nik +44(0)113 343 2152 N.Kapur@leeds.ac.uk Professor of Applied Fluid Mechanics Karagila Asaf A.Karagila@leeds.ac.uk University Academic Fellow Karim Mshell +44(0)113 343 2269 M.Karim@leeds.ac.uk Education Service Officer/Reception Kay Robert +44(0)113 343 2139 R.W.Kay@leeds.ac.uk Associate Professor in Advanced Manufacturing Kazlauciunas Algy +44(0)113 343 2939 A.Kazlauciunas@leeds.ac.uk Laboratory Manager Kechidi Smail S.Kechidi@leeds.ac.uk KTP Associate - Cold-Formed Steel Specialist Kee Terence +44(0)113 343 6421 T.P.Kee@leeds.ac.uk Reader Keller Philipp P.Keller1@leeds.ac.uk Marie Curie (ESR) Researcher Kelmanson Mark +44(0)113 343 5150 M.Kelmanson@leeds.ac.uk Professor Kelsall Robert +44(0)113 343 2068 R.W.Kelsall@leeds.ac.uk Professor Kemp Andrew +44(0)113 343 2078 A.H.Kemp@leeds.ac.uk Professor of Communications Kennedy Joanne +44(0)113 343 2514 J.E.Kennedy@leeds.ac.uk CPD Marketing & Development Officer Kent John +44(0)113 343 5103 J.T.Kent@leeds.ac.uk Professor Kersale Evy +44(0)113 343 5149 E.Kersale@leeds.ac.uk Lecturer Khaliq Kishwer K.A.Khaliq@leeds.ac.uk Research Assistant Khan Yasir +44(0)113 343 3854 Y.Khan@leeds.ac.uk Laboratory Technician Khan Amirul +44(0)113 343 2286 A.Khan@leeds.ac.uk Lecturer Khatir Zinedine +44(0)113 343 2220 Z.Khatir@leeds.ac.uk Visiting Research Fellow Khodaparast Sepideh S.Khodaparast@leeds.ac.uk University Academic Fellow Kidd Matthew +44(0)113 343 6542 M.Kidd@leeds.ac.uk Temporary Clerk Kilburn Paul P.Kilburn@leeds.ac.uk IFS ResearchTechnician Kim Yi-Yeoun +44(0)113 343 9407 Y.Y.Kim@leeds.ac.uk Senior Research and Teaching Fellow Kim Jongrae +44(0)113 343 2159 menjkim@leeds.ac.uk Associate Professor King Marco-Felipe +44(0)113 343 1957 M.F.King@leeds.ac.uk Research Fellow King Jenna +44(0)113 343 5746 J.King1@leeds.ac.uk CPD, Conference & Events Coordinator King Sarah S.King@leeds.ac.uk Project Manager King Stuart S.King3@leeds.ac.uk Analytical Technician Kirk Daniel +44(0)113 343 3807 D.R.Kirk@leeds.ac.uk Tutorial Assistant Kisil Vladimir V. +44(0)113 343 5173 V.Kisil@leeds.ac.uk Reader in Applied Analysis Kitayama Shoma S.Kitayama@leeds.ac.uk Research Fellow Knaggs Eve +44(0)113 343 0034 E.Knaggs@leeds.ac.uk Senior Administration Assistant Kokarev Gerasim +44(0)113 343 0599 G.Kokarev@leeds.ac.uk Lecturer Komissarov Serguei +44(0)113 343 5127 S.S.Komissarov@leeds.ac.uk Professor Kortantamer Dicle D.Kortantamer@leeds.ac.uk Lecturer in Project Management Kosarieh Shahriar +44(0)113 343 9741 S.Kosarieh@leeds.ac.uk Lecturer In Mechanical Engineering Krishnan Sreejith S.Krishnan1@leeds.ac.uk Research Fellow in Cementitious Materials Chemistry Kubiak Krzysztof +44(0)113 343 8333 K.Kubiak@leeds.ac.uk Associate Professor Kuffner Dos Anjos Rafael +44(0)113 343 3625 R.KuffnerdosAnjos@leeds.ac.uk Lecturer in Computer Graphics Kulak Alexander A.Kulak@leeds.ac.uk Materials Characterisn Istrmn Speclst Kulikowski Anoushka +44(0)113 343 0554 A.kulikowski@leeds.ac.uk Centre Manager Kumar Satish +44(0)113 343 3722 S.Kumar3@leeds.ac.uk Research Fellow Kumari Pallavi P.Kumari1@leeds.ac.uk Research Fellow in Plant Cell Biophysics Kumi Barimah Eric +44(0)113 343 2540 E.Kumi-Barimah@leeds.ac.uk Experimental Officer Kundu Iman I.Kundu@leeds.ac.uk Research Fellow Kwan Raymond +44(0)113 343 5760 R.S.Kwan@leeds.ac.uk Professor of Scheduling Lai Chun sing C.S.Lai@leeds.ac.uk Visiting Research Fellow Lai Xiaojun +44(0)113 343 2439 X.Lai@leeds.ac.uk Lecturer Lambert Benjamin B.S.Lambert@leeds.ac.uk Lecturer in Pure Mathematics Langfeld Kurt +44(0)113 343 5414 K.Langfeld@leeds.ac.uk Head of School of Mathematics, Professor in Theoretical Physics Lassila Toni +44(0)113 343 3724 T.Lassila@leeds.ac.uk Lecturer Lau Hui +44(0)113 343 3748 H.K.Lau@leeds.ac.uk Teaching Fellow Laughton Tom +44(0)113 343 4480 T.Laughton@leeds.ac.uk Functional Education Service Manager Lawey Ahmed A.Q.Lawey@leeds.ac.uk Lecturer in Communication Networks Lawlor Rob R.S.Lawlor@leeds.ac.uk Lecturer Lawrie Ian I.D.Lawrie@leeds.ac.uk Emeritus Professor Laycock Campbell Jeremy +44(0)113 343 7190 J.Laycock@leeds.ac.uk Resarch & Innovation Development Manager Le Khoa K.Le@leeds.ac.uk Lecturer Lebrero Alejandro A.Lebrero@leeds.ac.uk Research Software Engineer Lecheval Valentin V.Lecheval@leeds.ac.uk Research Fellow Lee Andrew +44(0)113 343 9712 A.Lee@leeds.ac.uk Centre Manager, Bragg Centre for Materials Research Lee Jaemin J.Lee2@leeds.ac.uk University Academic Fellow Leng Joanna +44(0)113 343 3809 J.Leng@leeds.ac.uk Senior Research Software Engineering Fellow Leonardo Diaz Roberto R.LeonardoDiaz@leeds.ac.uk Research Fellow Leonetti Matteo +44(0)113 343 5792 M.Leonetti@leeds.ac.uk Lecturer Lesnic Daniel +44(0)113 343 5181 D.Lesnic@leeds.ac.uk Professor in Applied Mathematics Levesley Martin +44(0)113 343 2110 M.C.Levesley@leeds.ac.uk Professor Lewis Kelly +44(0)113 343 0095 K.Lewis@adm.leeds.ac.uk Human Resources Officer Li Kang K.Li1@leeds.ac.uk Professor of Smart Energy Systems Li Hu +44(0)113 343 7754 H.Li3@leeds.ac.uk Associate Professor Li Zhenhong Z.H.Li@leeds.ac.uk Research Fellow Li Lianhe +44(0)113 343 6887 L.H.Li@leeds.ac.uk Senior Research Fellow/Experimental Officer Li Xiang X.Li11@leeds.ac.uk Research Fellow Lifshitz Ron R.Lifshitz@leeds.ac.uk Visiting Cheney Professor Lima Dos Santos Pedro P.L.LimaDosSantos@leeds.ac.uk Marie Curie Early Stage Researcher Linfield Edmund +44(0)113 343 2015 E.H.Linfield@leeds.ac.uk Professor Linyard Andy +44(0)113 343 5440 A.J.Linyard@leeds.ac.uk Education Service Officer (Admissions) Liu Jian J.Liu9@leeds.ac.uk Lecturer Liu Jason J.H.W.Liu@leeds.ac.uk Research Fellow Liu Ronghui +44(0)113 343 5338 R.Liu@its.leeds.ac.uk Professor Liu Haiyan +44(0)113 343 2930 H.Liu1@leeds.ac.uk University Academic Fellow in Statistical and Machine Learning Liwski Lukasz L.Liwski@leeds.ac.uk Student Education Service Assistant Lloyd Simon +44(0)113 343 2681 S.R.Lloyd@leeds.ac.uk Technician Lockwood Alexander A.P.G.Lockwood@leeds.ac.uk Visiting Researcher: Sludge Centre of Expertise Loganathan Sarathkumar S.Loganathan@leeds.ac.uk Postdoctoral Research Fellow Lopez Garcia Martin +44(0)113 343 8951 M.LopezGarcia@leeds.ac.uk Associate Professor Loveridge Fleur +44(0)113 343 2248 F.A.Loveridge@leeds.ac.uk Associate Professor of Geostructures Lumsden Stuart +44(0)113 343 6691 S.L.Lumsden@leeds.ac.uk Associate Professor Lunn Justin S +44(0)113 343 2320 J.S.Lunn@leeds.ac.uk Associate Professor Lynch Tom +44(0)113 343 1397 T.O.Lynch@leeds.ac.uk Project Manager Lythe Grant +44(0)113 343 5132 G.D.Lythe@leeds.ac.uk Professor of Applied Mathematics M Roufechaei Kamand K.MRoufechaei@leeds.ac.uk Teaching and Thesis supervisor Ma CaiYun +44(0)113 343 7809 C.Y.Ma@leeds.ac.uk Senior Research Fellow Macdonald Geraldine +44(0)113 343 8914 G.Macdonald@leeds.ac.uk Administrative Assistant Macente Alice A.Macente@leeds.ac.uk SEM - XCT Experimental Officer MacIntyre Jay +44(0)113 343 2096 J.M.MacIntyre@leeds.ac.uk Deputy Faculty Finance Manager Macpherson H Dugald +44(0)113 343 5166 H.D.MacPherson@leeds.ac.uk Professor Magee Derek +44(0)113 343 6819 D.R.Magee@leeds.ac.uk Lecturer Mahdi Faiz +44(0)113 343 9965 F.M.Mahdi@leeds.ac.uk Research Fellow Mahmud Tariq +44(0)113 343 2431 T.Mahmud@leeds.ac.uk Associate Professor Mandle Richard R.Mandle@leeds.ac.uk UKRI Future Leaders Fellow and University Academic Fellow Manga Mohamed M.S.Manga@leeds.ac.uk Post-Doctoral Research Fellow Mangan Thomas T.P.Mangan@leeds.ac.uk Research Fellow Mann Richard +44(0)113 343 8988 R.P.Mann@leeds.ac.uk Associate Professor Mansfield Jane +44(0)113 343 8324 J.Mansfield@leeds.ac.uk Student Ed Service Officer (Admissions) Mantova Vincenzo +44(0)113 343 8126 V.L.Mantova@leeds.ac.uk Lecturer Mao Xiaoan +44(0)113 343 4807 X.Mao@leeds.ac.uk Lecturer (Academic) Mardia Kanti +44(0)113 343 5100 K.V.Mardia@leeds.ac.uk Senior Research Professor Marrows Christopher +44(0)113 343 3780 C.H.Marrows@leeds.ac.uk Professor of Condensed Matter Physics Marsden Steve +44(0)113 343 6425 S.P.Marsden@leeds.ac.uk Professor of Organic Chemistry Marsh Daniel +44(0)113 343 9296 D.Marsh@leeds.ac.uk Priestley Chair in Comparative Planetary Atmospheres Marsh Bethany +44(0)113 343 5164 B.R.Marsh@leeds.ac.uk Professor Marsh Alastair A.Marsh@leeds.ac.uk Research Fellow in Alkali-Activated Materials Martin Adrian A.P.Martin@leeds.ac.uk Tutorial Assistant Martin Elaine +44(0)113 343 0889 E.Martin@leeds.ac.uk Professor of Chemical and Process Engineering Martin Paul +44(0)113 343 7787 P.P.Martin@leeds.ac.uk Professor Martin Sue +44(0)113 343 2000 S.Martin@leeds.ac.uk Education Service Officer / Receptionist Mason Lee +44(0)113 343 3735 L.A.Mason@leeds.ac.uk Research & Innovation Development Manager Matamoros Veloza Adriana A.MatamorosVeloza@leeds.ac.uk Research Fellow Mathai Basil B.Mathai@leeds.ac.uk UKRI Research Fellow Mattar Suhaila +44(0)113 343 0800 S.Mattar@leeds.ac.uk Lecturer Matthews Kimberley K.Matthews@leeds.ac.uk CDT Manager (based in Sheffield) Mattsson Johan +44(0)113 343 3815 K.J.L.Mattsson@leeds.ac.uk Associate Professor Matzkin Victor V.F.Matzkin@leeds.ac.uk PhD Student Mayambala Francisca F.J.N.Mayambala@leeds.ac.uk Internship: Equality and Inclusion Project Assistant Mayne Kelly Kaeyo +44(0)113 343 0907 K.Kelly@leeds.ac.uk Communication and Engagement Assistant, Grow MedTech McCaffrey Bill +44(0)113 343 6625 W.D.McCaffrey@leeds.ac.uk Professor McCall Blake B.McCall@leeds.ac.uk Research Fellow McCall Sam +44(0)113 343 8813 S.I.McCall@leeds.ac.uk Senior Marketing Executive McCann Sarah +44(0)113 343 1960 S.McCann@leeds.ac.uk Student Education Service Officer McConnell Claire +44(0)113 343 2380 C.L.McConnell@leeds.ac.uk Senior Education Service Officer McCormack Paul +44(0)113 343 2322 P.J.McCormack@leeds.ac.uk Research Finance Assistant McGonagle Dennis +44(0)113 392 4747 D.G.McGonagle@leeds.ac.uk Professor (Clinical) McGowan Patrick +44(0)113 343 6404 P.C.McGowan@leeds.ac.uk Professor McIntosh Andy Emeritus Professor McKay Alison +44(0)113 343 8175 A.McKay@leeds.ac.uk Professor McKay James +44(0)113 343 2556 J.McKay@leeds.ac.uk CDT Manager Mclaughlan James +44(0)113 343 0956 J.R.McLaughlan@leeds.ac.uk Associate Professor McLean Christopher +44(0)113 343 8573 C.J.McLean@leeds.ac.uk Linux Systems Manager McLernon Des +44(0)113 343 2050 D.C.McLernon@leeds.ac.uk Reader McNeill Erin +44(0)113 343 4065 E.McNeill@leeds.ac.uk Physics Outreach Officer McPhillie Martin +44(0)113 343 6513 M.J.McPhillie@leeds.ac.uk Lecturer in Organic Chemistry Meggs Andrew +44(0)113 343 8845 A.W.S.Meggs@leeds.ac.uk Research Support Officer (Pre Award) Megone Christopher +44(0)113 343 7888 C.B.Megone@leeds.ac.uk Professor Meldrum Fiona +44(0)113 343 6414 F.Meldrum@leeds.ac.uk Professor Meng Qingen Q.Meng@leeds.ac.uk Teaching Fellow Mengoni Marlne +44(0)113 343 5011 M.Mengoni@leeds.ac.uk Associate Professor in Computational Medical Engineering Menzel Robert +44(0)113 343 6407 R.Menzel@leeds.ac.uk Associate Professor Messmer Margit +44(0)113 343 5104 M.Messmer@leeds.ac.uk Principal Teaching Fellow Mhamdi Lotfi +44(0)113 343 6919 L.Mhamdi@leeds.ac.uk Lecturer Micklethwaite Stuart +44(0)113 343 2559 S.L.Micklethwaite@leeds.ac.uk Electron Microscopy Support Technician Mikaitis Mantas M.Mikaitis@leeds.ac.uk Lecturer Mikhailov Alexandre +44(0)113 343 5176 A.V.Mikhailov@leeds.ac.uk Professorial Research Fellow Miles Danielle +44(0)113 343 0921 D.E.Miles@leeds.ac.uk Technology Innovation Manager Milne Steven +44(0)113 343 2539 S.J.Milne@leeds.ac.uk Professor of Materials Chemistry Minton-Taylor Jasper +44(0)113 343 5746 J.Minton-Taylor@leeds.ac.uk CPD, Conference and Events Coordinator Mistry Devesh D.A.Mistry@leeds.ac.uk Leverhulme Trust Early Career Fellow and UAF Mistry Nimesh +44(0)113 343 7459 N.Mistry@leeds.ac.uk Senior Teaching Fellow Mitseas Ioannis +44(0)113 343 5784 I.Mitseas@leeds.ac.uk Lecturer in Structural Engineering Mobilia Mauro +44(0)113 343 1591 M.Mobilia@leeds.ac.uk Professor of Applied Mathematics Mohammed Abdulah A.Mohammed@leeds.ac.uk Chem stores technician Molina-Paris Carmen +44(0)113 343 5151 C.MolinaParis@leeds.ac.uk Professor Molzahn Ben +44(0)113 343 5104 B.Molzahn@leeds.ac.uk Senior Student Education Service Officer Montoya Pachongo Carolina C.MontoyaPachongo@leeds.ac.uk Research Fellow Moodley Kris +44(0)113 343 2329 K.Moodley@leeds.ac.uk Senior Lecturer (Teaching + Scholarship) Moore Thomas +44(0)113 343 3896 T.A.Moore@leeds.ac.uk Assoc Prof in Condensed Matter Physics Moore Rhys +44(0)113 343 8493 R.A.Moore@leeds.ac.uk Senior Technician - Additive Manufacturing / 3D Printing Moorsom Timothy T.Moorsom@leeds.ac.uk Royal Academy of Engineering Fellow Morina Ardian +44(0)113 343 8965 A.Morina@leeds.ac.uk Professor Morris Lindsay +44(0)113 343 2694 L.Morris@leeds.ac.uk Education Service Officer Morris Lisa-Dionne +44(0)113 343 6665 L.D.Morris@leeds.ac.uk Associate Professor of Human Activity & Product Design Developme Morris Kevin +44(0)113 343 9366 K.Morris1@leeds.ac.uk Professor of Radio Frequency Engineering Morrison Ciaran C.M.Morrison@leeds.ac.uk Aviation Simulator Manager Morsy Mohamed M.Morsy@leeds.ac.uk Research Fellow Mortimer Sally +44(0)113 343 2246 S.Mortimer@leeds.ac.uk Senior Education Service Officer Moseley Katy-anne K.A.Moseley@leeds.ac.uk Impact Fellow Motamen Salehi Farnaz F.MotamenSalehi@leeds.ac.uk Teaching Fellow Muhit Imrose I.Muhit@leeds.ac.uk Research Fellow in Masonry Arch Bridges Muller Frans +44(0)113 343 2933 F.L.Muller@leeds.ac.uk Chair in Chemical Process Engineering Muller Haiko +44(0)113 343 5445 H.Muller@leeds.ac.uk Senior Lecturer Mullis Andrew +44(0)113 343 2568 A.M.Mullis@leeds.ac.uk Professor Murphy William +44(0)113 343 5232 W.Murphy@leeds.ac.uk Senior Lecturer Murphy Julie +44(0)113 343 2157 J.F.Murphy@leeds.ac.uk Finance Officer Murphy Graham +44(0)113 343 5187 G.J.Murphy@leeds.ac.uk Senior Teaching Fellow Murray Rachael +44(0)113 343 0893 R.Murray@leeds.ac.uk Marketing and Development Officer Nadin Timothy T.J.Nadin@leeds.ac.uk School Manager Nagaraj Mamatha +44(0)113 343 8475 M.Nagaraj@leeds.ac.uk Lecturer Nahil Mohamad M.A.Nahil@leeds.ac.uk Research/Teaching Fellow Nelson Adam +44(0)113 343 6502 A.S.Nelson@leeds.ac.uk Professor Nelson Andrew +44(0)113 343 6409 A.L.Nelson@leeds.ac.uk Professor Nelson Andrew +44(0)113 343 6409 A.L.Nelson@leeds.ac.uk Professor Neville Anne +44(0)113 343 6812 A.Neville@leeds.ac.uk Professor Newisar May M.Newisar@leeds.ac.uk Research Fellow Nezami Zeinab Z.Nezami@leeds.ac.uk Research Fellow Nguyen Bao +44(0)113 343 0109 B.Nguyen@leeds.ac.uk Associate Professor Ngwana Adama A.Ngwana@leeds.ac.uk Administrative Assistant Nie Luzhen L.Nie@leeds.ac.uk Research Fellow Niesen Jitse +44(0)113 343 5870 J.Niesen@leeds.ac.uk Lecturer Nijhoff Frank +44(0)113 343 5120 F.W.Nijhoff@leeds.ac.uk Professor of Mathematical Physics Nikitas Nikolaos +44(0)113 343 0901 N.Nikitas@leeds.ac.uk Associate Professor in Structural Dynamics and Engineering Nix Michael M.G.Nix@leeds.ac.uk Visiting Senior Research Fellow Nlebedim Valentine V.U.Nlebedim@leeds.ac.uk Teaching Fellow Noakes Catherine +44(0)113 343 2306 C.J.Noakes@leeds.ac.uk Professor of Environmental Engineering for Buildings Noble Lydia L.R.Noble@leeds.ac.uk Student Education Service Assistant Norbertczak Halina +44(0)113 343 5607 H.T.Norbertczak@leeds.ac.uk PDRA Norman Alistair +44(0)113 343 7818 A.W.T.Norman@lubs.leeds.ac.uk Lecturer (Teaching and Scholarship) Normington Chris C.Normington1@leeds.ac.uk Student Education Services Manager O'Reilly Gerard G.A.OReilly@leeds.ac.uk Tutorial Assistant Okoro Shekwaga Cynthia C.K.OkoroShekwaga@leeds.ac.uk BBSRC Discovery Fellow Oliver Richard +44(0)113 343 3832 R.G.Oliver@leeds.ac.uk Technician Onel Lavinia L.Onel@leeds.ac.uk Research Fellow Ong Zhan +44(0)113 343 0051 Z.Y.Ong@leeds.ac.uk Associate Professor Ordyniak Sebastian S.Ordyniak@leeds.ac.uk Lecturer (Algorithms and Complexity) Orlova Ekaterina E.E.Orlova@leeds.ac.uk Research Fellow Oudmaijer Rene +44(0)113 343 3886 R.D.Oudmaijer@leeds.ac.uk Professor Owen Joshua J.J.Owen@leeds.ac.uk Lecturer Ozdemir Servet S.Ozdemir@leeds.ac.uk Post Doctoral Research Associate Pachos Jiannis +44(0)113 343 3817 J.K.Pachos@leeds.ac.uk Professor of Theoretical Physics Paesani Giacomo G.Paesani@leeds.ac.uk Research Fellow (Algorithms and Complexity) Palczewski Jan +44(0)113 343 5180 J.Palczewski@leeds.ac.uk Associate Professor Pallipurath Anuradha A.R.Pallipurath@leeds.ac.uk Royal Society Olga Kennard Fellow Panic Olja O.Panic@leeds.ac.uk Dorothy Hodgkin Fellow Papallas Rafael R.Papallas@leeds.ac.uk Research Fellow Papic Zlatko +44(0)113 343 3882 Z.Papic@leeds.ac.uk Associate Professor in Theoretical Physics Parker Douglas +44(0)113 343 6739 D.J.Parker@leeds.ac.uk Professor of Meteorology (School of Mathematics and School of Ea Parker Shelly +44(0)113 343 2934 M.Parker@leeds.ac.uk School Support Officer Parker Alison +44(0)113 343 5126 A.E.Parker@leeds.ac.uk Associate Professor Partington Jonathan +44(0)113 343 5123 J.R.Partington@leeds.ac.uk Professor Pask Christopher +44(0)113 343 4658 C.M.Pask@leeds.ac.uk Experimental Officer/Sen.Teaching Fellow Paterson Samantha S.Paterson@leeds.ac.uk Research Fellow Peacock David D.C.Peacock@leeds.ac.uk Teaching Fellow Pegler Sam +44(0)113 343 0048 S.Pegler@leeds.ac.uk University Academic Fellow Pensabene Virginia V.Pensabene@leeds.ac.uk Associate Professor Pepper Max M.Pepper@leeds.ac.uk Technician Pessoa de Miranda Marcelo +44(0)113 343 6332 M.Miranda@leeds.ac.uk Lecturer Pessu Frederick Oritseweneye F.O.Pessu@leeds.ac.uk Lecturer in Corrosion engineering Peyman Sally +44(0)113 343 3747 S.Peyman@leeds.ac.uk University Academic Fellow Phillips Luke L.Phillips@leeds.ac.uk Additive Manufacturing/3D Printing Tech Phylaktou Herodotos +44(0)113 343 2505 H.N.Phylaktou@leeds.ac.uk Senior Lecturer Pickering Jonathan +44(0)113 343 5836 J.H.Pickering@leeds.ac.uk Research Fellow Pickering Andrew +44(0)113 343 2131 A.D.Pickering@leeds.ac.uk Robotics Manufacturing Technician Pimm Andrew A.J.Pimm@leeds.ac.uk Research Fellow Pittard Julian +44(0)113 343 3805 J.M.Pittard@leeds.ac.uk Reader in Theoretical Astrophysics Piya Afrina Khan A.K.Piya@leeds.ac.uk Marie Curie ESR Researcher Plane John +44(0)113 343 8044 J.M.C.Plane@leeds.ac.uk Professor of Atmospheric Chemistry Pollock Isobel I.A.Pollock@leeds.ac.uk Visiting Professor Ponjavic Aleks +44(0)113 343 3839 A.Ponjavic@leeds.ac.uk University Academic Fellow Pournaras Evangelos E.Pournaras@leeds.ac.uk Associate Professor Prato Carlo C.Prato@leeds.ac.uk Professor of Transportation Engineering Preston George G.W.Preston@leeds.ac.uk Research Fellow Price Victoria +44(0)113 343 7218 V.Price@leeds.ac.uk Head of Marketing Pryce Gregory G.M.Pryce@leeds.ac.uk Research Fellow Pugazhendi Navaneethakrishnan N.Pugazhendi@leeds.ac.uk Marie Curie ESR Researcher Pugh Samantha +44(0)113 343 2985 S.L.Pugh@leeds.ac.uk Associate Professor in STEM Education Purdy Robert R.Purdy@leeds.ac.uk Lecturer and Admissions Tutor Purnell Philip +44(0)113 343 0370 P.Purnell@leeds.ac.uk Professor of Materials and Structures Qidan Ahmad A.A.Qidan@leeds.ac.uk Postdoctoral Research Fellow Querin Ozz +44(0)113 343 2218 O.M.Querin@leeds.ac.uk Professor of Design Optimisation Rabbani Arash A.Rabbani@leeds.ac.uk Lecturer (Assistant Professor) Raffle-Edwards Shona +44(0)113 343 8598 S.Raffle@leeds.ac.uk Outreach Coordinator Rahman Ali A.Rahman1@leeds.ac.uk SWJTU Joint School staff member Rai Kiran +44(0)113 343 5116 K.K.Rai@leeds.ac.uk Research and Facilities Officer Raihan MM M.M.Raihan@leeds.ac.uk Marie Curie Early Stage Researcher Ramanatha Sachin S.P.H.Ramanatha@leeds.ac.uk Research Fellow Ramasse Quentin Q.M.Ramasse@leeds.ac.uk Chair in Advanced Electron Microscopy Ranathunga Ashani A.S.Ranathunga@leeds.ac.uk Lecturer (Academic) Ranner Thomas +44(0)113 343 4697 T.Ranner@leeds.ac.uk Lecturer Rathjen Michael +44(0)113 343 5109 M.Rathjen@leeds.ac.uk Professor Rathore Rajesh +44(0)113 343 8984 R.S.Rathore@leeds.ac.uk Research Support Officer Ravi Manoj M.Ravi@leeds.ac.uk Lecturer Ravikumar Nishant N.Ravikumar@leeds.ac.uk Lecturer in Computer Science Raxworthy Mike +44(0)113 343 8775 M.J.Raxworthy@leeds.ac.uk Associate Professor in Engineering Management and Innovation Rayner Christopher +44(0)113 343 6579 C.M.Rayner@leeds.ac.uk Professor of Organic Chemistry Rayner Christopher +44(0)113 343 6579 C.M.Rayner@leeds.ac.uk Professor of Organic Chemistry Razavi Mohsen +44(0)113 343 9406 M.Razavi@leeds.ac.uk Professor Read Daniel +44(0)113 343 5124 D.J.Read@leeds.ac.uk Professor of Soft Matter Readioff Rosti R.Readioff@leeds.ac.uk Research Fellow Rees Amy +44(0)113 343 1032 A.L.Rees@leeds.ac.uk Research Administrator Rees Simon +44(0)113 343 1638 S.J.Rees@leeds.ac.uk Professor of Building Energy Systems Revill Charlotte C.H.Revill@leeds.ac.uk Research Fellow Rice Hugh H.P.Rice@leeds.ac.uk Postdoctoral research fellow Richards Megan M.K.Richards@leeds.ac.uk Demonstrator/Module Assistant Richards Daniel D.H.Richards@leeds.ac.uk Research Associate Richardson David +44(0)113 343 2101 D.Richardson@leeds.ac.uk Associate Professor of Structural Design Richardson Thomas T.Richardson1@leeds.ac.uk Teaching Assistant Richardson Ian +44(0)113 343 2331 I.G.Richardson@leeds.ac.uk Professor Richardson Robert +44(0)113 343 2156 R.C.Richardson@leeds.ac.uk Professor Richardson-Barlow Clare C.G.Richardson-Barlow@leeds.ac.uk Research Fellow Richter Ralf +44(0)113 343 1969 R.Richter@leeds.ac.uk Associate Professor Ries Michael +44(0)113 343 3859 M.E.Ries@leeds.ac.uk Professor Rigby Andrew A.J.Rigby@leeds.ac.uk Postdoctoral Research Fellow Rimmer Jo +44(0)113 343 6667 J.Rimmer1@leeds.ac.uk Administration Manager Rimmington Emma E.Rimmington@leeds.ac.uk General Support Assistant/ Receptionist Roberts Kevin +44(0)113 343 2408 K.J.Roberts@leeds.ac.uk Professor Robertson Ian +44(0)113 343 7076 I.D.Robertson@leeds.ac.uk Professor Robertson Clare +44(0)113 343 7805 C.Robertson1@leeds.ac.uk Senior Marketing Executive Robinson Amanda +44(0)113 343 2080 A.Robinson@leeds.ac.uk Personal Assistant Robinson Catherine C.L.Robinson1@leeds.ac.uk Research Support Officer (Pre-Award) Robinson Rachel +44(0)113 343 7680 R.A.Robinson@leeds.ac.uk Research Administrator Rooney Gabriel G.G.Rooney@leeds.ac.uk Visiting Research Fellow Roper Richard +44(0)113 343 6445 P.R.Roper@leeds.ac.uk Technician Rosamond Mark +44(0)113 343 7381 M.C.Rosamond@leeds.ac.uk Experimental Officer Ross Andrew +44(0)113 343 1017 A.B.Ross@leeds.ac.uk Associate Professor Rostami Javad J.Rostami@leeds.ac.uk KTP Associate Rucklidge Alastair +44(0)113 343 5161 A.M.Rucklidge@leeds.ac.uk Professor Ruddle Roy +44(0)113 343 1711 R.A.Ruddle@leeds.ac.uk Professor of Computing Rumble Olivia O.J.Rumble@leeds.ac.uk Student Ed Service Officer (Admissions) Ruprecht Daniel +44(0)113 343 2201 D.Ruprecht@leeds.ac.uk Visiting Professor Russo Mirko M.Russo@leeds.ac.uk Research Fellow in Design and Virtual Reality Sabini Luca L.Sabini@leeds.ac.uk Associate Professor in Project Management Sagar Catherine C.Sagar@leeds.ac.uk Research Support Assistant Saha Dipankar D.Saha@leeds.ac.uk Research Fellow Sainati Tristano T.Sainati@leeds.ac.uk Lecturer in project management Saleh Ehab +44(0)113 343 9336 E.Saleh@leeds.ac.uk Lecturer in Manufacturing Processes Salman Naveed N.Salman1@leeds.ac.uk Research Fellow Sanni Olujide O.S.Sanni@leeds.ac.uk Research Fellow/Experimental Officer Santos-Carballal David D.Santos-Carballal@leeds.ac.uk Research Fellow Sarhosis Vasilis +44(0)113 343 9343 V.Sarhosis@leeds.ac.uk Professor of Resilient Structures & Infrastructure Sarrami Foroushani Ali A.Sarrami@leeds.ac.uk Research Fellow Sasaki Satoshi +44(0)113 343 3578 S.Sasaki@leeds.ac.uk Lecturer (Academic) Saul Glyn +44(0)113 343 5528 G.Saul@leeds.ac.uk Faculty Head of Finance Savage Michael +44(0)113 343 3905 M.D.Savage@leeds.ac.uk Emeritus Professor Savy Claire +44(0)113 343 5449 C.Savy@leeds.ac.uk Centre Manager Scarabel Francesca F.Scarabel@leeds.ac.uk Lecturer in Mathematical Biology Schilhan Jonathan J.Schilhan@leeds.ac.uk Senior Research Associate Schneider Judith +44(0)113 343 2126 J.M.Schneider@leeds.ac.uk Administrative Support Officer - iTF Schroeder Sven S.L.M.Schroeder@leeds.ac.uk Centenary Chair-Engineering Applications Scott Andrew +44(0)113 343 2573 A.J.Scott@leeds.ac.uk Senior Lecturer Scott Mark +44(0)113 343 3819 M.Scott@leeds.ac.uk Technician Scott Stephen +44(0)113 343 6492 S.K.Scott@leeds.ac.uk Executive Dean (Interim) Seakins Paul +44(0)113 343 6568 P.W.Seakins@leeds.ac.uk Professor Seakins Paul +44(0)113 343 6568 P.W.Seakins@leeds.ac.uk Professor Selim Gehan +44(0)113 343 3082 G.Selim@leeds.ac.uk Hoffman Wood Professorof Architecture Sergeeva Natalia +44(0)113 343 7553 N.Sergeeva@leeds.ac.uk Lecturer Shafagh Ida I.Shafagh@leeds.ac.uk Research Fellow in Geothermal Energy Exc Shafer Paul +44(0)113 343 4843 P.E.Shafer@leeds.ac.uk Associate Professor Shalashilin Dmitry +44(0)113 343 7610 D.Shalashilin@leeds.ac.uk Professor of Computational Chemistry Shang Shang S.Shang@leeds.ac.uk Visiting Researcher Sharma Krishna K.Sharma1@leeds.ac.uk Research Fellow Sharp Benjamin +44(0)113 343 5487 B.G.Sharp@leeds.ac.uk Lecturer in Mathematical Analysis Shehzad Muhammad M.K.Shehzad@leeds.ac.uk Postdoctoral Research Fellow Shepherd Simon +44(0)113 343 6616 S.P.Shepherd@its.leeds.ac.uk Professor of Transport Modelling Shepley Philippa P.M.Shepley@leeds.ac.uk Experimental Officer Shi Wenyuan W.Shi1@leeds.ac.uk Associate Professor of Electronics and Electrical Engineering Shim Jung-uk +44(0)113 343 3903 J.Shim@leeds.ac.uk Lecturer Shires Andrew +44(0)113 343 5457 A.Shires@leeds.ac.uk Associate Professor Shuttleworth Matthew M.P.Shuttleworth@leeds.ac.uk Research Fellow Simpson Robert +44(0)113 343 2362 R.J.Simpson@leeds.ac.uk Research Technician Skene Calum C.S.Skene@leeds.ac.uk Research Fellow Slade Fiona +44(0)113 343 2202 F.R.Slade@leeds.ac.uk Administrative Support Officer - iFS Sleigh Andrew +44(0)113 343 2398 P.A.Sleigh@leeds.ac.uk Senior Lecturer Smith Ryan R.J.Smith1@leeds.ac.uk Trainee Technician Smith Nigel +44(0)113 343 2301 N.J.Smith@leeds.ac.uk Professor Smith Colin +44(0)113 343 3765 C.C.Smith@leeds.ac.uk Goods Inward Technician Smith Neil N.W.Smith@leeds.ac.uk Technician Smye Stephen S.W.Smye@leeds.ac.uk Professor Soltanahmadi Siavash +44(0)113 343 2107 S.Soltanahmadi@leeds.ac.uk Research Fellow Somjit Nutapong N.Somjit@leeds.ac.uk Associate Professor in Microwave and Wireless Engineering Speight Martin +44(0)113 343 5169 J.M.Speight@leeds.ac.uk Professor of Mathematics Spraggs Rachael +44(0)113 343 3057 R.E.Spraggs@leeds.ac.uk Research and Innovation Development Manager Squires David +44(0)113 343 2183 D.Squires@leeds.ac.uk Estates & Fabrics Officer Staggs John +44(0)113 343 2495 J.E.J.Staggs@leeds.ac.uk Senior Lecturer Steenson Paul +44(0)113 343 2024 D.P.Steenson@leeds.ac.uk Senior Lecturer Steenson Karen +44(0)113 343 2057 K.A.Steenson@leeds.ac.uk ERIS Manager Stell John +44(0)113 343 1076 J.G.Stell@leeds.ac.uk Senior Lecturer Stevens Karen +44(0)113 343 2255 K.Stevens@leeds.ac.uk Lead Technician Stewart Doug +44(0)113 343 2287 D.I.Stewart@leeds.ac.uk Professor Stewart Todd +44(0)113 343 2133 T.D.Stewart@leeds.ac.uk Professor of Mechanical and Medical Engineering Stockdale Andrew A.Stockdale2@leeds.ac.uk Research Support Technician Stone Daniel +44(0)113 343 6508 D.Stone@leeds.ac.uk Associate Professor Straw Philip P.A.Straw@leeds.ac.uk Research Assistant Strohmaier Alexander +44(0)113 343 8884 A.Strohmaier@leeds.ac.uk Chair in Analysis Sturman Rob +44(0)113 343 5139 R.Sturman@leeds.ac.uk Associate Professor Sturman Rob +44(0)113 343 5139 R.Sturman@leeds.ac.uk Associate Professor Subramanian Priya +44(0)113 343 2930 P.Subramanian@leeds.ac.uk Research Fellow Summers Jon +44(0)113 343 2151 J.L.Summers@leeds.ac.uk Senior Lecturer Sutherland Joanne +44(0)113 343 8308 J.Sutherland@leeds.ac.uk SOFI CDT Administrator Sweetman Adam +44(0)113 343 3808 A.M.Sweetman@leeds.ac.uk Royal Society University Research Fellow Szamuk Emil +44(0)113 343 3765 E.Szamuk@leeds.ac.uk Senior Estates and Fabric Technician Szumilo Karol K.Szumilo@leeds.ac.uk Research Fellow Talbot Paula +44(0)113 343 3862 P.Talbot@leeds.ac.uk PA to Head of School Taleb Wassim W.Taleb@leeds.ac.uk Teaching and Research Fellow in Electrochemistry and Corrosion Tang Yuzhou Y.Tang@leeds.ac.uk Research Fellow Tange Rudolf +44(0)113 343 9246 R.H.Tange@leeds.ac.uk Lecturer Tapley Kelvin +44(0)113 343 6732 K.Tapley@leeds.ac.uk Senior Lecturer in Colour Science Tarn Mark +44(0)113 343 5605 M.D.Tarn@leeds.ac.uk Research Fellow Tate James +44(0)113 343 6608 J.E.Tate@its.leeds.ac.uk Associate Professor Taylor Zeike +44(0)113 343 0767 Z.Taylor@leeds.ac.uk Associate Professor Taylor Peter +44(0)113 343 7169 P.G.Taylor@leeds.ac.uk Chair in Sustainable Energy Systems Taylor Paul +44(0)113 343 6529 P.C.Taylor@leeds.ac.uk Professor of Chemical Education Taylor Charles +44(0)113 343 5168 C.C.Taylor@leeds.ac.uk Professor Tedd Christopher C.F.Tedd@leeds.ac.uk Teaching Fellow and Digital Transformation Champion Thomas Briony +44(0)113 343 9694 B.G.Thomas@leeds.ac.uk Associate Professor in Design Science Thomas Jordan J.Thomas@leeds.ac.uk Lead Technician Thompson Peter +44(0)113 343 2471 P.R.Thompson@leeds.ac.uk Technical Officer Thompson Mark M.A.Thompson@leeds.ac.uk Head of School Thompson Harvey +44(0)113 343 2136 H.M.Thompson@leeds.ac.uk Professor of Computational Fluid Dynamics Thomson Neil +44(0)113 343 7289 N.H.Thomson@leeds.ac.uk Reader Thomson Tamaryn-Lee T.L.Thomson@leeds.ac.uk Electronics Engineer in Medical Dev. Thornton Phillip +44(0)113 343 3832 P.Thornton@leeds.ac.uk Electronics Engineer Thornton Paul +44(0)113 343 2935 P.D.Thornton@leeds.ac.uk Lecturer Thorpe Benjamin +44(0)113 343 2785 B.Thorpe@leeds.ac.uk Teaching Fellow in Statistics Thrush Julie +44(0)113 343 7046 J.Thrush@leeds.ac.uk Education Service Officer Tillotson Martin +44(0)113 343 2295 M.R.Tillotson@leeds.ac.uk Chair in Water Management Titarenko Sofya S.Titarenko@leeds.ac.uk Lecturer Tobias Steven +44(0)113 343 5172 S.M.Tobias@leeds.ac.uk Professor Tobias Steven +44(0)113 343 5172 S.M.Tobias@leeds.ac.uk Professor Tomlin Alison +44(0)113 343 2500 A.S.Tomlin@leeds.ac.uk Professor Tomlinson Laura +44(0)113 343 0667 L.E.Tomlinson@leeds.ac.uk Finance Assistant Tomlinson Gordon +44(0)113 343 2035 G.M.Tomlinson@leeds.ac.uk Education Service Officer Toppaladoddi Srikanth S.Toppaladoddi@leeds.ac.uk Lecturer in Applied Mathematics Torres Sebastian +44(0)113 343 5131 S.D.Torres@leeds.ac.uk Student Education Service Officer (Admissions) Trembath Roy +44(0)113 343 2310 R.Trembath@leeds.ac.uk Technician Trigg Mark +44(0)113 343 2265 M.Trigg@leeds.ac.uk Associate Professor of Water Risk Trowsdale Dan +44(0)113 343 8120 D.B.Trowsdale@leeds.ac.uk Excellence and Innovation Fellowship Tsang Yue-Kin +44(0)113 343 9628 Y.Tsang@leeds.ac.uk Research Fellow Turnbull Rory R.P.Turnbull@leeds.ac.uk Research Fellow Turnbull Bruce +44(0)113 343 7438 W.B.Turnbull@leeds.ac.uk Professor of Biomolecular Chemistry Turner Thomas T.D.Turner@leeds.ac.uk Research Fellow Turner Rodney R.J.Turner1@leeds.ac.uk Professor Turner Gavin +44(0)113 343 2348 G.B.Turner@leeds.ac.uk Senior Administration Assistant Turner Amanda A.Turner5@leeds.ac.uk Professor of Statistics Tutesigensi Apollo +44(0)113 343 4678 A.Tutesigensi@leeds.ac.uk Associate Professor Unterhitzenberger Christine C.Unterhitzenberger@leeds.ac.uk Associate Professor in Project Management Valavanis Alexander +44(0)113 343 3224 A.Valavanis@leeds.ac.uk Associate Professor Valdastri Pietro +44(0)113 343 3706 P.Valdastri@leeds.ac.uk Chair in Robotics & Autonomous Systems Valera Sachin S.J.Valera@leeds.ac.uk Visiting Researcher Van de Sande Marie M.VandeSande@leeds.ac.uk Marie Skodowska-Curie Individual Fellow Van Loo Sven S.VanLoo@leeds.ac.uk Lecturer in Astrophysics Varcoe Ben +44(0)113 343 8290 B.Varcoe@leeds.ac.uk Professor Vaughan Matthew M.T.Vaughan@leeds.ac.uk Research Fellow Velenturf Anne A.Velenturf@leeds.ac.uk Research Impact Fellow in Circular Economy Velis Costas +44(0)113 343 2327 C.Velis@leeds.ac.uk Lecturer Vermeeren Mats M.Vermeeren@leeds.ac.uk DFG Research Fellow Vickers Eleanor E.H.Vickers@leeds.ac.uk Tutorial Assistant Virtanen Seppo S.Virtanen@leeds.ac.uk Lecturer in Statistics Voice Alison +44(0)113 343 6647 A.M.Voice@leeds.ac.uk Senior Lecturer Voss Jochen +44(0)113 343 5125 J.Voss@leeds.ac.uk Lecturer Vuskovic Kristina +44(0)113 343 5443 K.Vuskovic@leeds.ac.uk Professor of Algorithms and Combinatorics Wadud Zia +44(0)113 343 7733 Z.Wadud@leeds.ac.uk Associate Professor Walder Carol +44(0)113 343 6494 C.A.Walder@leeds.ac.uk School Administrator Walker Nicole N.Walker@leeds.ac.uk Administration Support Assistant Walker Philip +44(0)113 343 7585 P.Walker@leeds.ac.uk Senior Teaching Fellow Walkley Mark +44(0)113 343 5684 M.A.Walkley@leeds.ac.uk Lecturer Walko Martin M.Walko@leeds.ac.uk Lecturer Walsh Catherine +44(0)113 343 0958 C.Walsh1@leeds.ac.uk Associate Professor; UKRI Future Leader Fellow Walti Christoph +44(0)113 343 2023 C.Walti@leeds.ac.uk Professor Wanatowski Dariusz D.Wan@leeds.ac.uk Professor of Geomechanics Wang Zheng +44(0)113 343 1077 Z.Wang5@leeds.ac.uk Professor of Intelligent Software Technology Wang Yongxing +44(0)113 343 4874 Y.Wang3@leeds.ac.uk Lecturer Wang Judith +44(0)113 343 3259 J.Y.T.Wang@leeds.ac.uk Associate Professor Wang He +44(0)113 343 5767 H.E.Wang@leeds.ac.uk Associate Professor Wang Chun +44(0)113 343 2198 C.Wang@leeds.ac.uk Research Fellow Wang Lin L.Wang2@leeds.ac.uk Visiting Research Fellow Wang Xue +44(0)113 343 2427 X.Z.Wang@leeds.ac.uk Professor Wang Mi +44(0)113 343 2435 M.Wang@leeds.ac.uk Professor Wang Zhaobin Z.Wang4@leeds.ac.uk Visiting Researcher Ward Keeran K.R.Ward@leeds.ac.uk Lecturer Ward Jonathan +44(0)113 343 5157 J.A.Ward@leeds.ac.uk Lecturer Wareing Christopher C.J.Wareing@leeds.ac.uk Research Fellow Warner Katie +44(0)113 343 8104 K.E.Warner@leeds.ac.uk CPD Course and Events Co-ordinator Warren Nicholas N.Warren@leeds.ac.uk Associate Professor Warren James J.P.Warren@leeds.ac.uk Research Fellow Warriner Stuart +44(0)113 343 6437 S.L.Warriner@leeds.ac.uk Senior Lecturer Watson Alan A.A.Watson@leeds.ac.uk Emeritus Professor Webb Michael +44(0)113 343 6423 M.E.Webb@leeds.ac.uk Associate Professor Webster Clair +44(0)113 343 6149 C.Webster@leeds.ac.uk Deputy Faculty Research Manager Wei Lijun L.J.Wei@leeds.ac.uk Research Fellow Wen Dongsheng +44(0)113 343 1299 D.Wen@leeds.ac.uk Chair in Petroleum Engineering Weston Stuart +44(0)113 343 3819 S.Weston@leeds.ac.uk Technician Westwood Aidan +44(0)113 343 2555 A.V.K.Westwood@leeds.ac.uk Lecturer Wetherill Lee +44(0)113 343 2171 L.Wetherill@leeds.ac.uk Technician Whalley Lisa +44(0)113 343 6594 L.K.Whalley@leeds.ac.uk Senior Research Fellow Whitaker Becky +44(0)113 343 0827 R.J.Whitaker@leeds.ac.uk Education Service Functional Manager Whitefoot Hayley +44(0)113 343 9901 H.Whitefoot@leeds.ac.uk Blended Learning Enhancement Officer Whiteley Alison +44(0)113 343 3220 A.J.Whiteley@leeds.ac.uk Manager, CPD Conference & Events Unit Whitley Antonia +44(0)113 343 2411 A.Whitley@leeds.ac.uk School Administration Officer Widrascu Karl K.Widrascu@leeds.ac.uk Analytical Technician Wiese Tony +44(0)113 343 2187 A.M.Wiese@leeds.ac.uk Lead Technician Wijayathunga Nagitha +44(0)113 343 2125 V.N.Wijayathunga@leeds.ac.uk Senior Research Fellow Wilcox Ruth +44(0)113 343 7980 R.K.Wilcox@leeds.ac.uk Professor Wilkins Terry +44(0)113 343 2570 T.A.Wilkins@leeds.ac.uk Professor Wilkins Simon +44(0)113 343 3039 S.A.Wilkins@leeds.ac.uk R&I Development Manager Wilkinson Adam A.J.Wilkinson@leeds.ac.uk Student Education Service Assistant Manager Willans Charlotte +44(0)113 343 5868 C.E.Willans@leeds.ac.uk Associate Professor and Director of Research and Innovation Williams Alan +44(0)113 343 2507 A.Williams@leeds.ac.uk Research Professor Williams Paul +44(0)113 343 2504 P.T.Williams@leeds.ac.uk Professor Williams Jeanine J.Williams4@leeds.ac.uk Experimental Officer: Chromatography and Analysis Williams Gwenllian G.M.Williams@leeds.ac.uk Postdoctoral research fellow Williams Simon S.P.Williams@leeds.ac.uk Research Assistant Williams Sophie +44(0)113 343 2214 S.D.Williams@leeds.ac.uk Professor in Medical Engineering Wills Harriet +44(0)113 343 2494 H.Wills1@leeds.ac.uk CPD Conference and Events Co-ordinator Wilman Marvin +44(0)113 343 9451 M.Wilman@leeds.ac.uk Technician Wilson Samuel +44(0)113 343 5474 S.S.Wilson@leeds.ac.uk Lecturer of Computing Wilson Andrew +44(0)113 343 1409 A.J.Wilson@leeds.ac.uk Professor Wilson Mark +44(0)113 343 2177 M.Wilson@leeds.ac.uk Associate Professor Wilson Sarah +44(0)113 343 0515 S.Wilson3@leeds.ac.uk Marketing Executive Winyard Thomas +44(0)113 343 9628 T.Winyard@leeds.ac.uk Research Fellow Wood Ann +44(0)113 343 2355 A.R.Wood@leeds.ac.uk Education Service Assistant Wood Phillip +44(0)113 343 2188 P.M.Wood@leeds.ac.uk Laboratory Manager Wood Christopher +44(0)113 343 8335 C.D.Wood@leeds.ac.uk Associate Professor Wood David +44(0)113 343 6192 D.J.Wood@leeds.ac.uk Professor Wood John +44(0)113 343 5106 J.C.Wood@leeds.ac.uk Professor Woodhouse Ed +44(0)113 343 2387 E.Woodhouse@leeds.ac.uk Lead Technician Woodin Sarah S.L.Woodin@leeds.ac.uk Senior Research Fellow Woodward Peter P.K.Woodward@leeds.ac.uk Chair in High Speed Rail Engineering Wright Kathleen +44(0)113 343 1755 K.E.Wright@leeds.ac.uk Project Administrator Wright Megan +44(0)113 343 3196 M.H.Wright@leeds.ac.uk University Academic Fellow Wright Nigel +44(0)113 343 0350 N.G.Wright@leeds.ac.uk Visiting Professor Wu Guizhi G.Z.Wu@leeds.ac.uk Experimental Officer Xia Yan Y.Xia@leeds.ac.uk Research Fellow Xie Shane +44(0)113 343 4896 S.Q.Xie@leeds.ac.uk Chair in Robotics+Autonomous Systems Xu Nan N.Xu@leeds.ac.uk EPSRC Research Fellow Xu Bao Hua +44(0)113 343 2423 B.H.Xu@leeds.ac.uk Lecturer Xu Jie +44(0)113 343 5193 J.Xu@leeds.ac.uk Professor Yagmur Ahmet A.Yagmur@leeds.ac.uk Dr. Yamashita Naoto N.Yamashita@leeds.ac.uk Visiting Research Fellow Yang Junfeng +44(0)113 343 2151 J.Yang@leeds.ac.uk Associate Professor Yang Xuebin +44(0)113 343 6162 X.B.Yang@leeds.ac.uk Associate Professor Yang Renyu +44(0)113 343 5236 R.Yang1@leeds.ac.uk Research Fellow Yang Liuquan L.Q.Yang@leeds.ac.uk Lecturer in Surface Engineering Yenigul Beril B.S.Yenigul@leeds.ac.uk Marie Curie (ESR) Researcher Yildirim Kaygun Emine E.YildirimKaygun@leeds.ac.uk Research Fellow Yu Hai-Sui +44(0)113 343 6703 DVC@leeds.ac.uk Deputy Vice-Chancellor Yuan Jiachen J.Yuan@leeds.ac.uk Senior research associate Yuksel Gamze G.Yuksel@leeds.ac.uk Visiting Researcher Yusuf Muhammad M.Yusuf@leeds.ac.uk Research Fellow Yuval Omer O.Yuval@leeds.ac.uk Research assistant Zaidi Syed S.A.Zaidi@leeds.ac.uk Associate professor Zakaria Fiona F.Zakaria@leeds.ac.uk Research Fellow Zakeri Arezoo +44(0)113 343 5254 A.Zakeri@leeds.ac.uk Research Fellow Zhang Zhiqiang Z.Zhang3@leeds.ac.uk Associate Professor Zhang Zhaopeng Z.Zhang1@leeds.ac.uk Research Fellow Zhang Li X +44(0)113 343 2005 L.X.Zhang@leeds.ac.uk Associate Professor Zhang Xinyou X.Zhang5@leeds.ac.uk SWJTU Staff Zhang Li +44(0)113 343 2020 L.Zhang@leeds.ac.uk Senior Lecturer Zhao Hongyuan H.Zhao@leeds.ac.uk Visiting Researcher Zhou Dejian +44(0)113 343 6230 D.Zhou@leeds.ac.uk Professor of Nanochemistry Zhou Chengxu +44(0)113 343 2832 C.X.Zhou@leeds.ac.uk Lecturer in Mobile Robotics

Read the original:
People | Faculty of Engineering and Physical Sciences - University of Leeds