Skip to main content
American Journal of Lifestyle Medicine logoLink to American Journal of Lifestyle Medicine
. 2019 Mar 20;14(2):169–186. doi: 10.1177/1559827619834527

Precision Medicine in Lifestyle Medicine: The Way of the Future?

Ian D Gray 1,, Andrea R Kross 1, Melanie E Renfrew 1, Paul Wood 1
PMCID: PMC7092395  PMID: 32231483

Abstract

Precision medicine has captured the imagination of the medical community with visions of therapies precisely targeted to the specific individual’s genetic, biological, social, and environmental profile. However, in practice it has become synonymous with genomic medicine. As such its successes have been limited, with poor predictive or clinical value for the majority of people. It adds little to lifestyle medicine, other than in establishing why a healthy lifestyle is effective in combatting chronic disease. The challenge of lifestyle medicine remains getting people to actually adopt, sustain, and naturalize a healthy lifestyle, and this will require an approach that treats the patient as a person with individual needs and providing them with suitable types of support. The future of lifestyle medicine is holistic and person-centered rather than technological.

Keywords: precision medicine, genomics, epigenetics, microbiome, caloric restriction, genetic risk score, individual support, social connectedness


‘Lifestyle medicine has 3 simple goals for the individual: to remain healthy as long as possible, to remain independent as long as possible, and to live as long as possible.’

While “precision medicine” has been described as an approach that integrates individual differences in lifestyle, environment, and biology, in actual practice it is simply a rebranding of genomic medicine. Genomics dominates in almost all research papers pertaining to precision medicine with the underlying assumption that, at its root, disease primarily results from genetics. As we shall see, the use of the term “precision” is aspirational and prematurely hopeful rather than descriptive. Even when the alternative term “personalized medicine” is used it primarily refers to determining what subgroup an individual belongs to rather than to medicine that considers the personhood and individuality of the patient.1

Lifestyle medicine, while recognizing that genes may predispose to various diseases, nonetheless postulates, based on overwhelming evidence, that most chronic disease results from lifestyle factors. So, the question is, “Should the future of lifestyle be centered on genetics or on lifestyle as the core factor for everyone in remaining disease-free and independent as long as possible? Should it be technological or holistic and humanistic?”

Lifestyle medicine has 3 simple goals for the individual: to remain healthy as long as possible, to remain independent as long as possible, and to live as long as possible. In other words, the 3 things we are working against are disease, dependency, and death. Notably, we are always working against time since the longer an unhealthy lifestyle is left unchecked, the shorter the time until one or more of these 3 possibilities will be realized. Ideally, we do not want to simply increase life span; we want to increase health span2 and compress morbidity.3-5

Genomic medicine is still in its infancy and currently the preponderance of evidence favors the lifestyle approach. Thousands of studies demonstrate not only that poor lifestyle increases the risk of chronic disease but that healthy lifestyle changes can reduce the risk of chronic disease, in some cases slowing or even reversing its progression.

There are 3 areas in which precision medicine could potentially be of value to lifestyle medicine:

  • Establishing a causal basis for the known effectiveness of lifestyle recommendations

  • Earlier identification of risk, with a motivating effect for adopting lifestyle changes at an earlier age

  • Individualizing lifestyle recommendations to deal with differences in response

How Genomic Medicine Helps Explain the Effectiveness of Lifestyle Medicine

In recent decades, advances in genomics have helped explain precisely why lifestyle changes work.

The first discovery was epigenetic change and gene methylation in the mid-1970s.6 While it had been earlier recognized that every human cell contains the same genetic material, the question was how cells were able to differentiate during embryogenesis and how genes were able to be either expressed or silenced. This resulted in the discovery of heritable epigenetic changes and finally epigenetic changes as a result of diet and exercise. In turn this provided insight into how inflammation and oxidative stress could affect gene expression and provided a pathway to underpinning lifestyle medicine with fundamental science. Interest in this area has grown substantially since 2006. We now know that what is important is the complex interplay within the whole genome, with genes being turned on and off in response to cellular exposures to chemical gradients and physiological stressors.7

The second discovery was that of the human gut biome.8 Although the significance of the microbiome was first suspected in the mid-1980s,9 the advent of new genomic technologies in the 21st century made it possible to identify thousands of distinct species and families of bacteria populating the human gut. For the first time it was possible to see the effects of diet, exercise, and probiotics on the ecology of the gut and to see the effect of microbiomic diversity and composition on risk of chronic disease, including certain infectious diseases. Interest in this area has been rapidly increasing since 2013.

The third discovery was the effect of various forms of caloric restriction (CR),10,11 including fasting-mimicking diets12 and time-restricted feeding,13 on gene expression,14 on the composition and function of gut microbiota,15 and via differential stress response on cancerous cells.16,17 CR has been found to have benefits for autophagy induction (necessary to destroy dysfunctional cellular components),18-20 which has potential impacts on increasing healthy longevity.21,22 Interestingly, aspirin has been found to display similar features to CR.23 It has been hypothesized that moderate intermittent stressors, like CR, may mobilize body systems to work more effectively.24 Whereas the benefits of fasting had been proclaimed for more than 2000 years, the underlying mechanisms have only been placed on a firm scientific footing within the last 10 years.

Paradoxically, these discoveries diminish the importance of pure genetics as an explanatory factor in disease. Epigenetic change and microbiome composition and function are driven by diet and physical activity, which along with CR, are largely a matter of choice rather than genetic determinism. Studies of monozygotic twins who are genetically identical but disease-discordant have found epigenetic25,26 and microbiomic differences,27-29 which strongly suggest that lifestyle and environment may largely override genetics, at least for some diseases. Two further discoveries complicate the genetic picture: microchimerism and somatic mosaicism. In microchimerism, a woman’s body may contain fetal cells and alien genetic material from her child, which persist for decades in different tissues with the potential for both beneficial and adverse effects.30,31 Somatic mosaicism is the occurrence of genetically distinct populations of cells within an individual due primarily to mutations during embryogenesis and to mutations during cell division over the course of a lifetime,32-34 which may accumulate with ageing.35

In the face of epigenetics, microbiomics, microchimerism, and somatic mosaicism, the search for risky genes for chronic disease, rather than being a cost-saving fast track to accelerated medical progress, may instead turn out to be an expensive blind alley. As one review of the progress of genomic medicine put it, “Soccer is the sport of the future in America . . . and it always will be.”36 Claims made for precision medicine, which always appear to be just over the horizon, may well fall into the same category.37

While genomic medicine has had some successes in relation to targeting drugs and gene therapies for some rare genetic variants38 and therapies for some cancers,39 in general the results have been mixed.40 Even diseases such as cancers may be 70% to 90% non-genomic in genesis,41,42 which suggests it would be better to promote prevention than cure.

Animal experimentation has revealed effects of diet and activity, and specific dietary components that also demonstrably apply to humans. An extreme case is that of intermittent CR, which demonstrably increases longevity in species as diverse as yeast, nematodes, mice, and humans.43 If human genetic diversity were a key factor in chronic disease, animal models would be almost worthless. Changes in disease patterns when East Asian or indigenous peoples44 adopt a Western lifestyle, as well as the increases in chronic diseases since the second half of the 20th century,45 strongly suggest that chronic disease is primarily non-genetic in origin.

The Success of Lifestyle Medicine in the Absence of Genetic Information

The theoretical basis of lifestyle medicine has changed significantly over the past decade. Cholesterol has reduced importance as a risk factor for cardiovascular disease (CVD),46-49 with more emphasis on chronic inflammation50,51 (or metaflammation52) and oxidative stress53 and the interaction between the two54 (which have been referred to as oxy-flammation55 or as an oxidative-inflammatory cascade56) as key factors in the genesis of chronic diseases in general and in their complications.57,58 In the context of aging-related disease, this has been referred to as “inflammaging.”59-62

What integrates many aspects of a healthy lifestyle is mitochondrial dynamics and its relationship with inflammation, oxidative stress, and chronic disease.63-65 Poor lifestyle may cause mitochondrial dysregulation and dysfunction,66 while exercise67-69 and caloric restriction70 may improve mitochondrial function. Mitochondrial function has also been identified as a potential target for mitigating the effects of age-related chronic disease.71,72 It has been hypothesized that cancers, rather than being caused by somatic mutation, may be caused by or promoted by mitochondrial dysfunction (based in part on the role of mitochondrial cell signaling on apoptosis).73-78 If true, this would help explain how a healthy lifestyle reduces cancer risk.

Epigenetic mechanisms show that genes are not destiny. Instead, there is an interplay between genetic and lifestyle factors, both prenatal and over the life course, influencing gene expression and the potential for a given disease to become a reality.79 The ecology of the human gut and the makeup of the species with which it is populated also demonstrably play a role in human health.80 Lifestyle factors mediate the composition of and changes in gut flora, which in turn affect the risk of chronic disease. The microbiome also appears to be independent of host genetics81 but is affected by both diet82,83 and activity,84 independently of one another.85

Last, there is now greater emphasis on activity generally rather than just exercise as a key factor in maintaining a lifetime of health, with a role in reducing oxidative stress.86,87 Physical inactivity has been linked to multiple chronic diseases including coronary artery disease, type 2 diabetes (T2D), various cancers, mental illness, and dementia.88-95 Conversely, increasing physical activity may assist in secondary prevention or reversal of such diseases96 and reduce mortality in survivors of breast, bowel, and prostate cancers97 as well as increase brain volumes and improve memory in older adults,98 reduce depressive symptoms and the risk of relapse in depression sufferers.99-101 The latter is particularly important given the massive increase in anti-depressant use in the West and the association between anti-depressant use and increased risk of CVD.102 Yet between 2001 and 2015, physical inactivity rose from 27% to 37% in developed countries, placing a further 10% of the population at risk.103

In summary, we now have a more complex theoretical base for looking at chronic disease and a clearer perspective on the relative importance of different lifestyle factors, much of it derived from population and clinical studies or cell and molecular biology, rather than genetics. One complication in many studies is that lifestyle behaviors tend to cluster. People with a healthy diet also tend to be less likely to smoke and more likely to be physically active; those with a less healthy diet and in particular those who eat the most meat tend to have an less healthy lifestyle overall.104 A study that only looks at one lifestyle factor risks confounding from other unmeasured lifestyle factors. This in itself highlights the need for a holistic approach.

The power of the lifestyle approach is that despite the changes in how we explain chronic disease and its prevention, the theoretical changes have simply served to reinforce the same recommendations while providing ever deeper explanations for their effectiveness.

Major studies over the past few years have reinforced existing recommendations105-107 but also provided some surprises. A major Canadian study,108 centered on 4 lifestyle factors (smoking, alcohol consumption, physical activity, and diet), found that those who had a healthy lifestyle in relation to all 4 factors could have a life expectancy up to 18 years longer than those who scored poorly on all 4 factors. Most surprisingly, the reduction in life expectancy as a result of physical inactivity was just as high as the reduction from smoking, and both were twice as high as the effect of diet with minimal reduction in life expectancy from excessive alcohol consumption. A study of the risk factors for being metabolically obese normal-weight,109 using factor analysis, found not 1 but 2 different dietary approaches that reduced this risk: a “healthy” approach (high in fruit, vegetables, and low-fat dairy) and a “prudent” approach (high in fish and whole grains, low in refined grains, sweets, sugars, boiled potatoes, and cured meats), as well as 2 diets that increased the risk, designated as “fat, meat, and alcohol” and “coca cola, hard cheese, and French fries.” Thus, within the lifestyle paradigm there is still room for diversity both in how people stay healthy as well as how they become chronically ill. Two recent studies have found that a healthy lifestyle significantly reduces the risk of CVD and diabetes for both those who are genetically at risk and the general population.110,111

Michael Pollan’s advice, “Eat Food. Not too much. Mostly Plants,”112 is supported by a growing body of research. Predominantly plant-based dietary patterns, both vegetarian and Mediterranean, are associated with increased longevity and significant reductions in risk of chronic disease.113-116 Several small studies have even found evidence that broad-based intensive interventions, which include such dietary patterns, may slow and even reverse various chronic diseases, including coronary artery disease117-120 and age-related cognitive impairment.121,122 Community-based programs that encourage such eating patterns demonstrably result in significant reductions in risk factors for coronary artery disease among program participants, often within a very short period of time.123,124 Eating less red meat significantly reduces risk of type 2 diabetes,125 while predominantly plant-based eating126,127 may markedly improve glycemic control, reduce medication use, and potentially reverse complications.128-131

Would Knowing Genetic Risk Make a Difference?

In unpacking this question, there are 3 issues to consider: “knowing,” “risk,” and “make a difference.”

On the question of “knowing,” genomics produces ambiguous evidence at best for chronic disease (as opposed to rare genetic syndromes) and at worst spurious associations. One example is the association of over a thousand genes with educational attainment.132 Social disadvantage may be associated with race, so racial differences in gene frequency could spuriously suggest a causative association between genes and education level, health, or economic achievement. One example of such a racial difference is the APOE4 gene, which is found in 25% to 40% of indigenous people across the world, while only found in around 12% of non-indigenous people.133

A core concept in genomics is gene penetrance, the likelihood that carrying one or more copies of genes associated with a disease will actually result in that disease.134 However, genomic research involves populations in which the majority of people lead an unhealthy lifestyle. Thus, estimates of gene penetrance are contaminated by the effects of the gene-lifestyle interaction.135 With as much as 80% of chronic disease attributable to lifestyle,136 this interaction is likely to be significant. A large part of gene penetrance may be explicable purely in terms of lifestyle and actual absolute risk from such genetic risks may be grossly overestimated. Genetic risk may largely be vulnerability to the effects of an unhealthy lifestyle. Estimates of gene penetrance also require some matching between genes and diagnosed disease; however, the rate of medical misdiagnosis may be as high as 10% to 15%,137 significantly adding to the uncertainty of any association found.

Some recent studies suggest that genetic risk is readily modifiable by lifestyle change. A large study found that women who were in the highest decile for nonmodifiable risk of breast cancer but who had low BMI, did not drink or smoke, and did not use menopausal hormone therapy had risks comparable to an average woman in the general population.138 Similarly, individuals in the top quintile of genetic risk for incident coronary events who had at least 3 of 4 healthy lifestyle factors (no smoking, BMI <30, physical activity at least once weekly, and a healthy diet) had a 46% lower relative risk of coronary events compared with those with a less favorable lifestyle.139 In both cases, even a moderately healthy lifestyle significantly reduced genetic risk. A study of genetic risk versus lifestyle factors in relation to colorectal cancer found that lifestyle factors had more weight than the genetic score.140 Other studies have further shown that lifestyle factors account for most of the risk in relation to CVD.141

A systematic review of the FTO genotype (a variant related to increased risk of obesity) and weight loss found that carriers responded equally well to weight-loss interventions as noncarriers.142 Another study, the DIETFITS study,143 looked at a low-carbohydrate and a low-fat diet to identify any difference in outcomes within groups as a result of genetic differences or in insulin dynamics. But the study found that at 12 months there was no significant difference in outcome and neither of the potential predisposing factors could identify which diet was better for whom.

This sampling of studies demonstrates that whatever the future may hold in relation to teasing out gene-disease links, a healthy lifestyle must still play the major role in mitigating risk. An unwarranted emphasis on genetic factors may simply dilute the message that taking responsibility for positive lifestyle behaviors may prevent, delay, or attenuate most premature disease. It may focus too much on individual genetic risk at the expense of the lifestyle risks that everyone faces.

The second aspect of genetic risk is to what extent it is a meaningful concept. It has been estimated that an individual may carry hundreds of genes associated with increased risk of various diseases144,145 for which they will never display any sign. So, what does it mean to say that the genes carry a risk? If each person has a unique genetic profile of several hundred variants associated with disease, how could this inform any clinical decision? Given that most people are healthy most of the time as are those around them, to what extent would this simply undermine genetic risk as a factor to be considered?146

A person may carry a gene associated with increased risk for a disease without any familial history of the disease. They (and/or their family) may also possess one or more genes that modify or nullify the effect of the first gene such that their risk of that disease is effectively nil.147 Not only genetics but familial patterns of disease may be important,148 and even then, the impact of shared lifestyle and environmental exposures cannot be dismissed. Complicating matters further, a SNP (single nucleotide polymorphism) protecting against one disease may be a risk factor for another disease.149 The danger in acting on such perceived risks is a higher likelihood of overtreatment or treatment of unclear value,150 carrying with it risks of its own.

Finally, would knowing genetic risks make a difference? In many cases, the answer is no.

Several studies have shown that being advised of an increased genetic risk does not result in any significant change in health behaviors.151,152 Nor does being diagnosed and treated for hypertension,153,154 coronary heart disease,155 type 2 diabetes,156-159 or chronic disease generally.155 A study of college athletes found that being advised of increased genetic risk of poor recovery from traumatic brain injury would not affect their playing behavior.160

Even surviving cancer makes little difference to adopting a healthy lifestyle.161-165 This is of particular concern given that cancer survivors are much more likely to suffer from comorbid chronic disease than the general population even where their lifestyle behaviors are the same.166,167 There is growing evidence that cancer treatments themselves significantly increase the risk of subsequent CVD.168,169 Adopting a healthy lifestyle may increase the likelihood of disease-free survival170,171 with higher levels of physical activity reducing the specific risk of CVD.172,173 The effect of chemo- and radiotherapy as cancer treatments on risk of CVD is itself a warning that technological approaches to health care (such as gene therapy) may have unforeseen adverse health consequences downstream.

With substantial evidence that knowing the risk of one disease does not motivate many people to change their behavior, what could we then expect of being advised of genetic risk of a hundred or more diseases? Would this be motivating, overwhelming, or simply unbelievable? Responses are likely to range from fatalism, panic, and tunnel vision to incredulity, leading to either inaction or to overreaction and unnecessary preemptive treatment. All of these responses could be dysfunctional, especially when making healthy lifestyle changes could provide broad-spectrum protection against almost all of these risks.

Carrying a gene that increases risk of one disease does not reduce risks of other diseases. A narrow focus on the one genetic risk may simply shift the risk to such other diseases instead. A meta-analysis by the Cochrane Collaboration on cancer screening found that “the trials with adequate randomization did not find an effect of screening on total cancer mortality, including breast cancer, after 10 years . . . or on all-cause mortality after 13 years.”174

Why Do Not People Adopt Healthy Lifestyle Behaviors?

We tend to make unjustified assumptions about human behavior including the assumptions that people are rational/irrational or that all people need is more information to motivate change.175 However, we sometimes overlook the fact that, for many people, chronic disease has low saliency and low perceived risk,176,177 both of which may need to be addressed if healthy lifestyle is to be promoted. There are at least 4 barriers to healthy people adopting a healthier lifestyle.

First, for much of its course, chronic disease is essentially invisible to other people. We do not know what medications the people around us may be taking for a chronic disease, and it is only when such a disease reaches a critical point such as requiring dialysis, or amputation or other surgery, or where a person visibly deteriorates or needs mobility or other functional aids that we actually see evidence of chronic disease. This may lead many people to underestimate the risk. In 2014-2015, a massive 50% of Australians reported having at least 1 of 8 chronic diseases.178 Yet in the mass media, there is virtual silence regarding the prevalence such diseases. Paradoxically, those at highest risk of chronic disease may perceive their risk to be low.179

Second, the normalization of obesity may reduce motivation to do anything about weight gain.180,181 While stigmatization of obesity is counterproductive182 and obese people may need additional emotional support for health behavior change,183 the validation of obesity by movements such as the “fat acceptance movement” potentially undermines public health efforts to combat obesity and its health consequences, by encouraging complacency and inaction.

Third, based on age-specific mortality rates for Australia, 90% of people in Western countries now live to at least the age of 65,184 85% to the age of 70, and 80% to the age of 75. So, while people are working, they are unlikely to see significant levels of mortality in coworkers or their age-cohort and would tend to associate chronic disease with aging, without drawing the connection between morbidity/mortality and the cumulative effect of lifestyle behaviors. When age-specific causes of death are considered for people under 45, the main causes are suicide and accidents, which in themselves do not directly relate to factors such as diet or activity levels.

Finally, the very success of modern medicine in stabilizing chronic diseases (without actually curing them) may reduce the perceived threat. Coupled with social safety nets for subsidized health care and disability payments in many Western countries, reduction in the perceived risk of unhealthy behaviors may lead to more rather than less unhealthy behavior, the so-called “Fence Paradox,”185 due to the reduced costs involved to the individual.186 One such example is HIV prophylaxis and treatment.187-189

Can Genetic Risk Actually Be Predicted With Precision?

A number of recent studies claim to be able to predict risk of CVD with accuracy as great as or better than conventional clinical measures. One study190 generated a genetic risk score (GRS) based on 49310 SNPs (single nucleotide polymorphisms); however, when applied to new data gave inconsistent results for different populations (Finnish vs British), with no overlap in 95% confidence intervals for odds ratios for the 2 populations. A second study191 used 1.7 million genetic variants to generate a genetic risk score, but only gave a marginal improvement over clinical measures. (Interestingly another study using only 31 variants yielded comparable accuracy,192 suggesting that almost all of the 1.7 million variants were redundant.) Both of the studies using large numbers of variants appear to have a number of methodological issues, including the assumption that including more variants of lower demonstrated association with CVD risk will somehow improve accuracy rather than simply adding noise.

But the greatest deficit in such studies is the lack of consideration of the false negative rate, the false positive rate, or specificity,193-195 any of which could have serious consequences196 for those whom a model predicts of being at high risk. Such models may result in overtesting, overdiagnosis, and overtreatment. In the process, more people will join the ranks of the “worried well,” anxious about a disease they will never get, hypervigilant for any associated symptoms, and perhaps less alert to symptoms of the genesis of an actual unrelated disease. There are already indications that some genetic associations may be spurious with the same SNP showing increased risk in some populations but not others.197-199 Some researchers argue for a more rigorous approach to determining causality200 and a greater focus on biological mechanisms,201 with one recent survey even casting doubt on whether extensive genetic data will ever be useful for making reliable causal inferences.202 In many studies, rather than all of the SNPs being verified as present, they are imputed algorithmically. In the UK Biobank of around 500 000 people, used in many studies, around 805 000 genetic markers have been collected that by imputation are increased to 95 million variants.203 Such high levels of imputation raise reasonable concerns about the results of such research.

The human genome is incredibly variable with the 1000 Genomes Project finding more than 88 million variants in just 2504 individuals.204 Such vast numbers of genetic variants or SNPs can only be accommodated into existing statistical methods by aggregating them and then stratifying the aggregated values, automatically resulting in loss of information.205 Different genes may promote heart disease via different pathways, for example, by increasing endogenous cholesterol or by moderating lipid metabolism, anti-inflammatory processes, or antioxidant defenses. But the grab-bag approach of throwing them all into a homogeneous category means that even if risk is established from the GRS it provides no guidance as to how it should be mitigated and thus has to fall back on blanket treatments, which could be ineffective for the gene variant the individual actually has. Unless the functional role of a SNP is established and how that function relates to increased risk of CVD, it may simply be a chance artefact of testing thousands or millions of variants. Extending a predictive model beyond a few dozen variants may not result in increased predictive power.206,207 One study that looked at the clinically confirmed severity of coronary artery disease and genotype data imputed to 2.5 million SNPs was only able to confirm a single, already known, locus as a risk for severity of coronary artery disease.208

Genomic prediction probably will not markedly improve in the future simply because the most common variants with moderate to high association with chronic disease have already been identified,209 that is, the low-hanging fruit have already been picked.210 As Fröhlich and coauthors state,

The lack of impact on clinical practice can largely be attributed to insufficient performance of predictive models, difficulties to interpret complex model predictions, and lack of validation via prospective clinical trials that demonstrate a clear benefit compared to the standard of care.211

A recent study, using only 48 SNPs, identified from genome-wide association studies, found that GRS and diet were independently associated with risk of T2D and concluded that everyone regardless of genetic risk would benefit from favorable food choices.212 Identifying increased genetic risk of CVD, T2D, or cancer would not significantly change recommendations for a healthy lifestyle. The massive increase in chronic disease since the mid-20th century has been driven, not by a massive change in the genetic make-up of the population but by changes in lifestyle and environmental exposures.

What Is the Future of Lifestyle Medicine?

Lifestyle medicine ultimately aims to make a healthy lifestyle the norm rather than the exception. This means finding better strategies to promote a healthy lifestyle, helping individuals adopt and sustain such a lifestyle, and combatting the detrimental effects of an obesogenic environment. The maximum gains to be made in reducing chronic disease still lie in a focus on improving health behaviors for people generally rather than a focus on outliers, simply because of the high prevalence of unhealthy lifestyles.

At the risk of seeming Luddite, the future of lifestyle medicine is humanistic rather than technological. It needs to focus on how more people can be induced to adopt a healthy lifestyle and how such a lifestyle can be sustained and become habitual.213 Whereas limited frequency health behaviors such as vaccinations and screening are relatively easy to promote, a healthy lifestyle requires repeated-occurrence health behaviors and continued abstention from unhealthy behaviors across the entire lifespan,214 a much tougher proposition.

The obesogenic environment is a continuing, if not rising, problem (with digital technology a contributor toward increased obesity).215 An analogous approach may need to be taken to unhealthy foods as has been successfully taken with smoking, including things such as banning advertising and promotion of unhealthy foods aimed at children, increasing sales taxes on unhealthy food items, or subsidies on fruit and vegetables to increase their affordability.216 However, we still need to make a distinction between the environment as a stimulus and individual responsibility for what people do in response to that stimulus. If individuals are not ultimately responsible for their own health behaviors, health promotion becomes irrelevant. Environmental triggers alone do not cause unhealthy lifestyles. On a more positive note, there is some evidence that healthy behaviors may be becoming more prevalent at least in the Asia-Pacific region (including Australia and New Zealand).217

Another area on which lifestyle medicine needs to focus is making better use of “teachable moments,” particularly those times where a patient is advised of a risk or diagnosis218-220 or where they have been successfully treated but face increased risk of co-morbidity.

Increasing our effectiveness in helping individuals sustain healthy behaviors requires acknowledging the ways in which they differ in the kinds of messages that they find sufficiently persuasive to result in action,221,222 in the misinformation and misperceptions they may have,223 in how they differ in their motivations224 and in their ability to implement and sustain changes, and in how their social environment can support or undermine change.

Lifestyle medicine also needs to more deeply explore how mental health risks may be reduced via health behavioral change.225 This is of increasing concern given the huge increase in the rate of anti-depressant use in Western societies. Finally, we may need to embrace ideas that have historically been considered to lie within the ambit of spirituality, with numerous studies finding positive associations between religious participation and physical and mental health226-228 as well as associations with particular components of religious attitudes such as generosity.196,229

Lifestyle medicine’s future may ultimately lie in individualizing support so that adopted lifestyle behaviors become permanent rather than transient.

Individualizing Support

There are several excellent resources dealing in detail with individualizing exercise recommendations for chronic disease.230-234 In addition, Minich and Bland’s coverage of issues relating to special dietary considerations is also wide-ranging.235 So, the issues concerning physical aspects of lifestyle will be covered only briefly here, followed by further discussion about personalization of lifestyle medicine in 3 areas: social connectedness, psychological skills and support, and basic practical skills.

Exercise and Activity

For physical activity, the most critical aspect is to start with activities that lie within the individual’s capabilities but which serve to extend those capabilities over time. This is particularly necessary where individuals suffer from chronic diseases, which may cause dysfunction at the cellular level, but which may improve over time with diet and activity. Individuals differ in response to exercise depending on the intensity, frequency, duration, and modality, as well as on the timing and composition of meals,236 so exercise needs to be tailored to the individual237 to elicit the best response for that person.238 For some people, exercise (not activity) may lead to adverse effects on blood pressure, high-density lipoprotein cholesterol or other biomarkers or symptoms239 so a more gradual approach, with more biometric monitoring, could be warranted for such individuals.

There is some evidence of nonresponse to particular kinds of exercise for some people,240 which could mean experimenting to see what works best at a given time for a given individual at a particular stage of chronic disease.241-243 Compensatory behavior, such as increased eating or reduced activity, may negate any benefits,244-246 so this may also need to be addressed.

While there is evidence that personalized exercise prescription may enhance response,237 at this point the specific use of genetic information to inform exercise prescription may be premature. A 2017 review of genetic testing for exercise prescription and injury prevention found that “the predictive value of such tests is too low to warrant clinical application.”247 A systematic review of VO2-max trainability found that of 97 genes identified as possible predictors only 13 were reproduced in more than 2 studies and that heterogeneity in the studies limited the conclusions that could be drawn.248 The META-PREDICT study, which involved developing predictors, based largely on genetics, for the health benefits of exercise for individuals appears to have quietly died following its final report in 2016.249

Individuals vary considerably in their affective response to exercise intensity. Most people have a positive response to moderate-intensity exercise while having an aversive response to higher intensities.250 Additionally, people who have more positive feelings about exercise are more likely to engage in it.250 So for an individual to continue to want to exercise they need to feel good as a result of the exercise,251,252 and it needs to be set at a level that best balances effectiveness and affective response, with an initial focus on increasing enjoyment of physical activity.253 Taking individual differences into account is crucial for effective physical activity interventions.254

Nutrition

The effects of some nutrients may differ in people with different gene variants, although the evidence is often mixed. While increased requirements for certain micronutrients have been established beyond doubt for some people (eg, folate for pregnant women to prevent neural tube defects and anencephaly),255 most findings that relate genetics to nutrient requirements find either small effect sizes or conflicting evidence for the direction of the effect. For example, an examination of genetic variations and zinc requirements256 concluded that “the data extracted confirmed a connection between genetics and zinc requirements, although the direction and magnitude of the dietary modification for carriers of specific genotypes could not be defined.”

In 3 studies (all by the same researchers) of interaction of DHA with the APOE4 gene (a risk factor for Alzheimer’s disease [AD]),257 one study found limited transfer of DHA to cerebrospinal fluid,258 another study using a different measurement method found increased brain-uptake of DHA for the same gene,259 while a third study suggested that high-dose DHA in early stages of AD dementia could decrease prevalence in APOE4 carriers.260 Other studies have found improved cognitive function with fish oil supplementation only in APOE4 carriers261 or conversely no benefit only for APOE4 carriers.262 The bottom line is that we simply do not know what the interaction is, if any.

One difficulty in linking micronutrients with chronic disease is that if an individual has a high-energy, nutrient-poor diet, then, rather than nutritional deficiency being the cause of the disease (eg, obesity), both the disease and nutritional deficiency may be attributable to diet quality. Accurately measuring nutrient intake and nutritional needs for micronutrients for individuals is extremely challenging,263 so rather than focusing on specific nutrients, the safest approach is a varied diet of healthy foods,264 adjusting for particular food sensitivities. However, including unhealthy foods in a varied diet may actually increase risk of abdominal obesity and T2D.265

Social Connectedness

Social isolation and loneliness have been recognized as detrimental to health for more than 30 years.266 Growing numbers of people report social isolation or loneliness, while others experience dysfunctional or undermining relationships that can also be detrimental to health or the success of a lifestyle medicine intervention.267 Negative social experiences correlate with poorer health behaviors268 while loneliness tends to be associated with poorer social skills.269 Conversely, support from family, friends, or workmates may all contribute to a person making and sustaining healthy lifestyle changes.270 Belonging to a cohesive, stable, and homogeneous community may in itself have positive health benefits (the so-called Roseto Effect271), something that modernity seems to have undermined. Blue Zones notably involve groups who, whether by reason of ethnicity, isolation, or religious participation, constitute such cohesive communities.

Addressing social isolation may be a core factor in improving lifestyle behaviors, whether this involves helping people to improve social skills or facilitating participation in a stable social group. Face-to-face support groups272 that persist beyond the intervention and peer mentoring/support (the buddy system)273-275 may be effective means of both supporting behavior change and reducing the negative impact of social isolation by providing new social ties and support, other than that of a paid health professional. They can also be more cost-effective,276 an important consideration in an era of skyrocketing health costs.

Social skills training and opportunities to practice these growing skills may help overcome some of the more detrimental emotional effects of loneliness that for some underpin dysfunctional health behaviors. Finally, the health benefits of volunteering277,278 may in part lie in increased social contact with less focus on self and could form part of a lifestyle intervention for people lacking social support.

Psychological Factors

Individual psychological differences may affect their capacity to adopt and sustain healthy lifestyle behaviors. How people deal with failure279 may influence abandonment of health behaviors, and it is possible that similar strategies for dealing with relapse could be utilized as for addiction.280 One possible future research direction may be how individuals deal with micro-temporal factors such as temporal and situational cues, as well as transient thoughts and feelings.214 Individuals also differ on multiple dimensions on how they approach goal setting and achievement,281 so finding the best approach for the individual may be essential for long-term success. One key strategy may be planning in advance how to deal with obstacles or setbacks282 and using implementation intentions, which has shown promise in terms of reducing meat consumption283 and increasing physical activity.284 Individuals may have chronic diseases as a result of past self-regulatory failure and may need training in a range of skills such as planning, mental contrasting, distracting, and reframing.285 Motivational interviewing and health coaching have proven effective in assisting individuals in meeting their health goals286-288 and may help individuals build self-efficacy.

For individuals with multiple comorbidities, regimen factors,289 burden of treatment,290 and patient capacity291 may all need to be considered in deciding what approach to take with promoting lifestyle changes for individuals who may already be struggling to cope. An approach known as “minimally disruptive medicine” may be needed.292 In some cases, implementing small changes may be the best approach to take293 with a focus on progress rather than perfection.

Skills Training

For many individuals, just knowing what they should be eating is not enough, they need to be given the skills to put those recommendations into practice. In order to be able to eat healthily, an individual may need to learn basic cooking and shopping skills and strategies. Teaching basic cooking skills has been shown to encourage healthy eating,294-297 with home-cooked meals associated with better dietary quality.298 Community interventions to improve cooking skills have been shown to increase food literacy,299 while incorporating cooking demonstrations and opportunities to taste healthier foods as part of a health promotion program could help encourage healthier eating.300 Using a grocery list when shopping is also associated with a healthier diet among high risk adults301 and healthy shopping tours are being increasingly offered by health organizations.

Conclusion

Precision or genomic medicine is not the enemy.302 There may be some scope for cross-fertilization between the 2 specializations. For individuals who conscientiously adopt a healthy lifestyle but show no improvement in biomarkers, genetic research could help identify whether there is a genetic explanation or whether there is some previously unknown lifestyle or environmental factor that needs to be considered. Conversely longitudinal research on populations who live a healthy lifestyle could help sharpen estimates of gene penetrance paving the way to better predictive models. Such research could possibly even help identify new lifestyle factors by looking at differences in outcome between genetically similar people following the same healthy lifestyle.

However, chronic disease is simply not the primary target for a genetic approach and extending it to the broad mass of people at low genetic risk is overkill. Precision medicine is best targeted at gene therapy for gene variants with proven etiology, identifying genetic factors in variations in drug effectiveness and identifying high penetrance genes for disease screening.209 Primary genetic research may also identify links between diseases and the functions of genes and gene networks that may lead to novel insights into the genesis of disease.303,304

While this article has identified technical barriers to a genuinely “precision” medicine, there are also numerous ethical issues305-307 and regulatory protections that would need to be ironed out should such an approach become the dominant paradigm.308 These include things such as informed consent, continued ownership of one’s own genetic information309 and the right to have it destroyed, privacy (especially in an era where data leaks are so common and where depersonalized data can be re-personalized310), genetic discrimination, the right to refuse genetic testing, and the potential for future abuse by governments.311 Precision medicine has been described as “drowning in a regulatory soup.”312 The demand for ever bigger genomic data sets and ever more personal medical information with which to match it, combined with the rush by governments to accommodate these demands, is likely to lead to fundamental human rights and freedoms being overridden. There are already calls for every newborn to be genetically sequenced313 with the consequent medicalization of life.

None of these concerns apply to lifestyle medicine.

We currently seem to be at the “Peak of Inflated Expectations,”314 and it may be some years before the limited utility of precision medicine is recognized and that projected cost savings are illusory.315 The financial resources being allocated, for what is effectively a promissory note, may ultimately divert resources from the more acute problem: How can we persuade most people to adopt a healthy lifestyle?316

Lifestyle medicine now possesses a much deeper scientific foundation but the actual recommendations have not markedly changed as a result. The fundamental problem for lifestyle medicine remains: How people can be motivated to adopt, sustain, and ultimately naturalize a healthy lifestyle. Rather than delving ever more deeply into physical mechanisms, instead we need to look at the psychological and social factors that either encourage or obstruct healthy lifestyle behaviors. Interventions need to be personalized to the individual and their embodied experience of the world. This does not necessarily mean changing what we recommend, but it does mean changing how we support the individual in their efforts to live healthier.

Footnotes

Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.

Ethical Approval: Not applicable, because this article does not contain any studies with human or animal subjects.

Informed Consent: Not applicable, because this article does not contain any studies with human or animal subjects.

Trial Registration: Not applicable, because this article does not contain any clinical trials.

References

  • 1. Feiler T, Gaitskell K, Maughan T, Hordern J. Personalised medicine: the promise, the hype and the pitfalls. New Bioeth. 2017;23:1-12. doi: 10.1080/20502877.2017.1314895 [DOI] [PubMed] [Google Scholar]
  • 2. Olshansky SJ. From lifespan to healthspan. JAMA. 2018;320:1323-1324. doi: 10.1001/jama.2018.12621 [DOI] [PubMed] [Google Scholar]
  • 3. Allen NB, Zhao L, Liu L, et al. Favorable cardiovascular health, compression of morbidity and healthcare costs: 40-year follow up of the CHA Study (Chicago Heart Association Detection Project in Industry). Circulation. 2017;135:1693-1701. doi: 10.1161/CIRCULATIONAHA.116.026252 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Botes R, Vermeulen KM, Correia J, Buskens E, Janssen F. Relative contribution of various chronic diseases and multi-morbidity to potential disability among Dutch elderly. BMC Health Serv Res. 2018;18:24. doi: 10.1186/s12913-017-2820-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Jacob ME, Yee LM, Diehr PH, et al. Can a healthy lifestyle compress the disabled period in older adults? J Am Geriatr Soc. 2016;64:1952-1961. doi: 10.1111/jgs.14314 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Felsenfeld G. A brief history of epigenetics. Cold Spring Harb Perspect Biol. 2014;6:a018200. doi: 10.1101/cshperspect.a018200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Davies JA. Life Unfolding How the Human Body Creates Itself. 1st ed. Oxford, England: Oxford University Press; 2015. [Google Scholar]
  • 8. Valdes AM, Walter J, Segal E, Spector TD. Role of the gut microbiota in nutrition and health. BMJ. 2018;361:k2179. doi: 10.1136/bmj.k2179 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Prescott S, Nowak-Węgrzyn A. Strategies to prevent or reduce allergic disease. Ann Nutr Metab. 2011;59(suppl 1):28-42. doi: 10.1159/000334150 [DOI] [PubMed] [Google Scholar]
  • 10. Trepanowski JF, Bloomer RJ. The impact of religious fasting on human health. Nutr J. 2010;9:57. doi: 10.1186/1475-2891-9-57 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Longo VD, Mattson MP. Fasting: molecular mechanisms and clinical applications. Cell Metab. 2014;19:181-192. doi: 10.1016/j.cmet.2013.12.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Wei M, Brandhorst S, Shelehchi M, et al. Fasting-mimicking diet and markers/risk factors for aging, diabetes, cancer, and cardiovascular disease. Sci Transl Med. 2017;9:eaai8700. doi: 10.1126/scitranslmed.aai8700 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Chaix A, Lin T, Le HD, Chang MW, Panda S. Time-restricted feeding prevents obesity and metabolic syndrome in mice lacking a circadian clock. Cell Metab. 2019;29:303-319. doi: 10.1016/j.cmet.2018.08.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Swindell WR. Genes and gene expression modules associated with caloric restriction and aging in the laboratory mouse. BMC Genomics. 2009;10:585. doi: 10.1186/1471-2164-10-585 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Fabbiano S, Suárez-Zamorano N, Chevalier C, et al. Functional gut microbiota remodeling contributes to the caloric restriction-induced metabolic improvements. Cell Metab. 2018;28:907-921 doi: 10.1016/j.cmet.2018.08.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Buono R, Longo VD. Starvation, stress resistance, and cancer. Trends Endocrinol Metab. 2018;29:271-280. doi: 10.1016/j.tem.2018.01.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Lee C, Raffaghello L, Brandhorst S, et al. Fasting cycles retard growth of tumors and sensitize a range of cancer cell types to chemotherapy. Sci Transl Med. 2012;4:124ra27. doi: 10.1126/scitranslmed.3003293 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Bagherniya M, Butler AE, Barreto GE, Sahebkar A. The effect of fasting or calorie restriction on autophagy induction: a review of the literature. Ageing Res Rev. 2018;47:183-197. doi: 10.1016/j.arr.2018.08.004 [DOI] [PubMed] [Google Scholar]
  • 19. Ntsapi C, Loos B. Caloric restriction and the precision-control of autophagy: a strategy for delaying neurodegenerative disease progression. Exp Gerontol. 2016;83:97-111. doi: 10.1016/j.exger.2016.07.014 [DOI] [PubMed] [Google Scholar]
  • 20. Abdellatif M, Sedej S, Carmona-Gutierrez D, Madeo F, Kroemer G. Autophagy in cardiovascular aging. Circ Res. 2018;123:803-824. [DOI] [PubMed] [Google Scholar]
  • 21. Nakamura S, Yoshimori T. Autophagy and longevity. Mol Cells. 2018;41:65-72. doi: 10.14348/molcells.2018.2333 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Carmona JJ, Michan S. Biology of healthy aging and longevity. Rev Invest Clin. 2016;68:7-16. [PubMed] [Google Scholar]
  • 23. Pietrocola F, Castoldi F, Markaki M, et al. Aspirin recapitulates features of caloric restriction. Cell Rep. 2018;22:2395-2407. doi: 10.1016/j.celrep.2018.02.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Pruimboom L, Muskiet FAJ. Intermittent living; the use of ancient challenges as a vaccine against the deleterious effects of modern life—a hypothesis. Med Hypotheses. 2018;120:28-42. doi: 10.1016/j.mehy.2018.08.002 [DOI] [PubMed] [Google Scholar]
  • 25. Castillo-Fernandez JE, Spector TD, Bell JT. Epigenetics of discordant monozygotic twins: implications for disease. Genome Med. 2014;6:60. doi: 10.1186/s13073-014-0060-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Demir AB, Demir N. Epigenetic basis of twin discordance in diseases: future benefits. Gynecol Obstet Reprod Med. 2018;24:108-118. doi: 10.21613/GORM.2017.741 [DOI] [Google Scholar]
  • 27. Turnbaugh PJ, Hamady M, Yatsunenko T, et al. A core gut microbiome in obese and lean twins. Nature. 2009;457:480-484. doi: 10.1038/nature07540 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Xie H, Guo R, Zhong H, et al. Shotgun metagenomics of 250 adult twins reveals genetic and environmental impacts on the gut microbiome. Cell Syst. 2016;3:572-584. doi: 10.1016/j.cels.2016.10.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Zierer J, Jackson MA, Kastenmüller G, et al. The fecal metabolome as a functional readout of the gut microbiome. Nat Genet. 2018;50:790-795. doi: 10.1038/s41588-018-0135-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Cheng SB, Davis S, Sharma S. Maternal fetal cross-talk through, cell-free fetal DNA, telomere shortening, microchimerism, and inflammation. Am J Reprod Immunol. 2018;79:e12851. doi: 10.1111/aji.12851 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Gammill HS, Nelson JL. Naturally acquired microchimerism. Int J Dev Biol. 2010;54:531-543. doi: 10.1387/ijdb.082767hg [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Freed D, Stevens EL, Pevsner J. Somatic mosaicism in the human genome. Genes (Basel). 2014;5:1064-1094. doi: 10.3390/genes5041064 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Frank SA. Somatic mosaicism and disease. Curr Biol. 2014;24:R577-R581. doi: 10.1016/j.cub.2014.05.021 [DOI] [PubMed] [Google Scholar]
  • 34. Campbell IM, Shaw CA, Stankiewicz P, Lupski JR. Somatic mosaicism: implications for disease and transmission genetics. Trends Genet. 2015;31:382-392. doi: 10.1016/j.tig.2015.03.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Risques RA, Kennedy SR. Aging and the rise of somatic cancer-associated mutations in normal tissues. PLoS Genetics. 2018;14:e1007108. doi: 10.1371/journal.pgen.1007108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Evans JP, Meslin EM, Marteau TM, Caulfield T. Deflating the genomic bubble. Science. 2011;331:861-862. doi: 10.1126/science.1198039 [DOI] [PubMed] [Google Scholar]
  • 37. Comfort N. The Science of Human Perfection: How Genes Became the Heart of American Medicine: New Haven, CT: Yale University Press; 2014. [Google Scholar]
  • 38. Psaty BM, Dekkers OM, Cooper RS. Comparison of 2 treatment models: precision medicine and preventive medicine. JAMA. 2018;320:751-752. doi: 10.1001/jama.2018.8377 [DOI] [PubMed] [Google Scholar]
  • 39. Mennel RG. Precision medicine: hype or hope? Proc (Bayl Univ Med Cent). 2015;28:397-400. [PMC free article] [PubMed] [Google Scholar]
  • 40. Joyner MJ. Precision medicine: a second opinion. https://www.acc.org/latest-in-cardiology/articles/2017/11/20/14/29/precision-medicine-a-second-opinion. Published November 20, 2017. Accessed February 26, 2019.
  • 41. Wu S, Zhu W, Thompson P, Hannun YA. Evaluating intrinsic and non-intrinsic cancer risk factors. Nat Commun. 2018;9:3490. doi: 10.1038/s41467-018-05467-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Wu S, Powers S, Zhu W, Hannun YA. Substantial contribution of extrinsic risk factors to cancer development. Nature. 2016;529:43-47. doi: 10.1038/nature16166 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Fontana L, Partridge L. Promoting health and longevity through diet: from model organisms to humans. Cell. 2015;161:106-118. doi: 10.1016/j.cell.2015.02.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Gracey M, King M. Indigenous health part 1: determinants and disease patterns. Lancet. 2009;374:65-75. doi: 10.1016/S0140-6736(09)60914-4 [DOI] [PubMed] [Google Scholar]
  • 45. Remington PL, Brownson RC. Fifty years of progress in chronic disease epidemiology and control. MMWR Morb Mortal Wkly Rep. 2011;60:70-77. [PubMed] [Google Scholar]
  • 46. Soliman GA. Dietary cholesterol and the lack of evidence in cardiovascular disease. Nutrients. 2018;10:E780. doi: 10.3390/nu10060780 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Tsoupras A, Lordan R, Zabetakis I. Inflammation, not cholesterol, is a cause of chronic disease. Nutrients. 2018;10:E604. doi: 10.3390/nu10050604 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Lopez-Candales A, Hernández Burgos PM, Hernandez-Suarez DF, Harris D. Linking chronic inflammation with cardiovascular disease: from normal aging to the metabolic syndrome. J Nat Sci. 2017;3:e341. [PMC free article] [PubMed] [Google Scholar]
  • 49. Ridker PM. C-reactive protein and the prediction of cardiovascular events among those at intermediate risk: moving an inflammatory hypothesis toward consensus. J Am Coll Cardiol. 2007;49:2129-2138. doi: 10.1016/j.jacc.2007.02.052 [DOI] [PubMed] [Google Scholar]
  • 50. Chung HY, Kim DH, Lee EK, et al. Redefining chronic inflammation in aging and age-related diseases: proposal of the senoinflammation concept [published online February 22, 2017]. Aging Dis. doi: 10.14336/AD.2018.0324 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Goosby BJ, Cheadle JE, McDade T. Birth weight, early life course BMI, and body size change: chains of risk to adult inflammation? Soc Sci Med. 2016;148:102-109. doi: 10.1016/j.socscimed.2015.11.040 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu Rev Immunol. 2011;29:415-445. doi: 10.1146/annurev-immunol-031210-101322 [DOI] [PubMed] [Google Scholar]
  • 53. Liguori I, Russo G, Curcio F, et al. Oxidative stress, aging, and diseases. Clin Interv Aging. 2018;13:757-772. doi: 10.2147/CIA.S158513 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Biswas SK. Does the interdependence between oxidative stress and inflammation explain the antioxidant paradox? Oxid Med Cell Longev. 2016;2016:5698931. doi: 10.1155/2016/5698931 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Vikram A, Tripathi DN, Kumar A, Singh S. Oxidative stress and inflammation in diabetic complications. Int J Endocrinol. 2014;2014:679754. doi: 10.1155/2014/679754 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Lamb RE, Goldstein BJ. Modulating an oxidative-inflammatory cascade: potential new treatment strategy for improving glucose metabolism, insulin resistance, and vascular function. Int J Clin Pract. 2008;62:1087-1095. doi: 10.1111/j.1742-1241.2008.01789.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Pickering RJ, Rosado CJ, Sharma A, Buksh S, Tate M, de Haan JB. Recent novel approaches to limit oxidative stress and inflammation in diabetic complications. Clin Transl Immunology. 2018;7:e1016. doi: 10.1002/cti2.1016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Sharma A, Tate M, Mathew G, Vince JE, Ritchie RH, de Haan JB. Oxidative stress and NLRP3-inflammasome activity as significant drivers of diabetic cardiovascular complications: therapeutic implications. Front Physiol. 2018;9:114. doi: 10.3389/fphys.2018.00114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Calder PC, Bosco N, Bourdet-Sicard R, et al. Health relevance of the modification of low grade inflammation in ageing (inflammageing) and the role of nutrition. Ageing Res Rev. 2017;40:95-119. doi: 10.1016/j.arr.2017.09.001 [DOI] [PubMed] [Google Scholar]
  • 60. Prattichizzo F, De Nigris V, Spiga R, et al. Inflammageing and metaflammation: the yin and yang of type 2 diabetes. Ageing Res Rev. 2018;41:1-17. doi: 10.1016/j.arr.2017.10.003 [DOI] [PubMed] [Google Scholar]
  • 61. Martucci M, Ostan R, Biondi F, et al. Mediterranean diet and inflammaging within the hormesis paradigm. Nutr Rev. 2017;75:442-455. doi: 10.1093/nutrit/nux013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Olivieri F, Prattichizzo F, Grillari J, Balistreri CR. Cellular senescence and inflammaging in age-related diseases. Mediators Inflamm. 2018;2018:9076485. doi: 10.1155/2018/9076485 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Picard M, Wallace DC, Burelle Y. The rise of mitochondria in medicine. Mitochondrion. 2016;30:105-116. doi: 10.1016/j.mito.2016.07.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Herst PM, Rowe MR, Carson GM, Berridge MV. Functional mitochondria in health and disease. Front Endocrinol (Lausanne). 2017;8:296. doi: 10.3389/fendo.2017.00296 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Hernández-Aguilera A, Rull A, Rodríguez-Gallego E, et al. Mitochondrial dysfunction: a basic mechanism in inflammation-related non-communicable diseases and therapeutic opportunities. Mediators Inflamm. 2013;2013:135698. doi: 10.1155/2013/135698 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Rasool S, Geetha T, Broderick TL, Babu JR. High fat with high sucrose diet leads to obesity and induces myodegeneration. Front Physiol. 2018;9:1054. doi: 10.3389/fphys.2018.01054 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Trewin AJ, Berry BJ, Wojtovich AP. Exercise and mitochondrial dynamics: keeping in shape with ROS and AMPK. Antioxidants (Basel). 2018;7:E7. doi: 10.3390/antiox7010007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Distefano G, Standley RA, Zhang X, et al. Physical activity unveils the relationship between mitochondrial energetics, muscle quality, and physical function in older adults. J Cachexia Sarcopenia Muscle. 2018;9:279-294. doi: 10.1002/jcsm.12272 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Heo JW, No MH, Park DH, et al. Effects of exercise on obesity-induced mitochondrial dysfunction in skeletal muscle. Korean J Physiol Pharmacol. 2017;21:567-577. doi: 10.4196/kjpp.2017.21.6.567 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Lettieri-Barbato D, Cannata SM, Casagrande V, Ciriolo MR, Aquilano K. Time-controlled fasting prevents aging-like mitochondrial changes induced by persistent dietary fat overload in skeletal muscle. PLoS One. 2018;13:e0195912. doi: 10.1371/journal.pone.0195912 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Vitetta L, Anton B. Lifestyle and nutrition, caloric restriction, mitochondrial health and hormones: scientific interventions for anti-aging. Clin Interv Aging. 2007;2:537-543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Labbadia J, Brielmann RM, Neto MF, Lin YF, Haynes CM, Morimoto RI. Mitochondrial stress restores the heat shock response and prevents proteostasis collapse during aging. Cell Rep. 2017;21:1481-1494. doi: 10.1016/j.celrep.2017.10.038 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Porporato PE, Filigheddu N, Pedro JMB-S, Kroemer G, Galluzzi L. Mitochondrial metabolism and cancer. Cell Res. 2018;28:265-280. doi: 10.1038/cr.2017.155 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Hsu CC, Tseng LM, Lee HC. Role of mitochondrial dysfunction in cancer progression. Exp Biol Med (Maywood). 2016;241:1281-1295. doi: 10.1177/1535370216641787 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Zong WX, Rabinowitz JD, White E. Mitochondria and cancer. Mol Cell. 2016;61:667-676. doi: 10.1016/j.molcel.2016.02.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Seyfried TN. Cancer as a mitochondrial metabolic disease. Front Cell Dev Biol. 2015;3:43. doi: 10.3389/fcell.2015.00043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Chattopadhyay E, Roy B. Altered mitochondrial signalling and metabolism in cancer. Front Oncol. 2017;7:43. doi: 10.3389/fonc.2017.00043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Sreedhar A, Zhao Y. Dysregulated metabolic enzymes and metabolic reprogramming in cancer cells. Biomed Rep. 2018;8:3-10. doi: 10.3892/br.2017.1022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Moosavi A, Ardekani AM. Role of epigenetics in biology and human diseases. Iran Biomed J. 2016;20:246-258. doi: 10.22045/ibj.2016.01 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Liang D, Leung RKK, Guan W, Au WW. Involvement of gut microbiome in human health and disease: brief overview, knowledge gaps and research opportunities. Gut Pathog. 2018;10:3. doi: 10.1186/s13099-018-0230-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Rothschild D, Weissbrod O, Barkan E, et al. Environment dominates over host genetics in shaping human gut microbiota. Nature. 2018;555:210-215. doi: 10.1038/nature25973 [DOI] [PubMed] [Google Scholar]
  • 82. Graf D, Di Cagno R, Fåk F, et al. Contribution of diet to the composition of the human gut microbiota. Microb Ecol Health Dis. 2015;26:26164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Bowyer RCE, Jackson MA, Pallister T, et al. Use of dietary indices to control for diet in human gut microbiota studies. Microbiome. 2018;6:77. doi: 10.1186/s40168-018-0455-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Monda V, Villano I, Messina A, et al. Exercise modifies the gut microbiota with positive health effects. Oxid Med Cell Longev. 2017;2017:3831972. doi: 10.1155/2017/3831972 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Kang SS, Jeraldo PR, Kurti A, et al. Diet and exercise orthogonally alter the gut microbiome and reveal independent associations with anxiety and cognition. Mol Neurodegener. 2014;9:36. doi: 10.1186/1750-1326-9-36 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Carraro E, Schilirò T, Biorci F, et al. Physical activity, lifestyle factors and oxidative stress in middle age healthy subjects. Int J Environ Res Public Health. 2018;15:E1152. doi: 10.3390/ijerph15061152 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Pesta D, Roden M. The Janus head of oxidative stress in metabolic diseases and during physical exercise. Curr Diab Rep. 2017;17:41. doi: 10.1007/s11892-017-0867-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Lee IM, Shiroma EJ, Lobelo F, et al. Effect of physical inactivity on major non-communicable diseases worldwide: an analysis of burden of disease and life expectancy. Lancet. 2012;380:219-229. doi: 10.1016/S0140-6736(12)61031-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Booth FW, Roberts CK, Laye MJ. Lack of exercise is a major cause of chronic diseases. Compr Physiol. 2012;2:1143-1211. doi: 10.1002/cphy.c110025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Warburton DER, Bredin SSD. Health benefits of physical activity: a systematic review of current systematic reviews. Curr Opin Cardiol. 2017;32:541-556. doi: 10.1097/HCO.0000000000000437 [DOI] [PubMed] [Google Scholar]
  • 91. Marques A, Santos T, Martins J, Matos MGD, Valeiro MG. The association between physical activity and chronic diseases in European adults. Eur J Sport Sci. 2018;18:140-149. doi: 10.1080/17461391.2017.1400109 [DOI] [PubMed] [Google Scholar]
  • 92. DeFina LF, Willis BL, Radford NB, et al. The association between midlife cardiorespiratory fitness levels and later-life dementia: a cohort study. Ann Intern Med. 2013;158:162-168. doi: 10.7326/0003-4819-158-3-201302050-00005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Zhou Z, Fu J, Hong YA, Wang P, Fang Y. Association between exercise and the risk of dementia: results from a nationwide longitudinal study in China. BMJ Open. 2017;7:e017497. doi: 10.1136/bmjopen-2017-017497 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Mavros Y, Gates N, Wilson GC, et al. Mediation of cognitive function improvements by strength gains after resistance training in older adults with mild cognitive impairment: outcomes of the study of mental and resistance training. J Am Geriatr Soc. 2017;65:550-559. doi: 10.1111/jgs.14542 [DOI] [PubMed] [Google Scholar]
  • 95. Morris JK, Vidoni ED, Johnson DK, et al. Aerobic exercise for Alzheimer’s disease: a randomized controlled pilot trial. PLoS One. 2017;12:e0170547. doi: 10.1371/journal.pone.0170547 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Leon AS, Bronas UG. Pathophysiology of coronary heart disease and biological mechanisms for the cardioprotective effects of regular aerobic exercise. Am J Lifestyle Med. 2009;3:379-385. doi: 10.1177/1559827609338145 [DOI] [Google Scholar]
  • 97. Sabiston CM, Brunet J. Reviewing the benefits of physical activity during cancer survivorship. Am J Lifestyle Med. 2012;6:167-177. doi: 10.1177/1559827611407023 [DOI] [Google Scholar]
  • 98. Erickson KI, Voss MW, Prakash RS, et al. Exercise training increases size of hippocampus and improves memory. Proc Natl Acad Sci U S A. 2011;108:3017-3022. doi: 10.1073/pnas.1015950108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Sarris J, O’Neil A, Coulson CE, Schweitzer I, Berk M. Lifestyle medicine for depression. BMC Psychiatry. 2014;14:107. doi: 10.1186/1471-244X-14-107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Martinsen EW, Raglin JS. Themed review: anxiety/depression: lifestyle medicine approaches. Am J Lifestyle Med. 2007;1:159-166. doi: 10.1177/1559827606298713 [DOI] [Google Scholar]
  • 101. Lavebratt C, Herring MP, Liu JJ, et al. Interleukin-6 and depressive symptom severity in response to physical exercise. Psychiatry Res. 2017;252:270-276. doi: 10.1016/j.psychres.2017.03.012 [DOI] [PubMed] [Google Scholar]
  • 102. Maslej MM, Bolker BM, Russell MJ, et al. The mortality and myocardial effects of antidepressants are moderated by preexisting cardiovascular disease: a meta-analysis. Psychother Psychosom. 2017;86:268-282. doi: 10.1159/000477940 [DOI] [PubMed] [Google Scholar]
  • 103. Guthold R, Stevens GA, Riley LM, Bull FC. Worldwide trends in insufficient physical activity from 2001 to 2016: a pooled analysis of 358 population-based surveys with 1.9 million participants. Lancet Global Health. 2018;6:e1077-e1086. doi: 10.1016/S2214-109X(18)30357-7 [DOI] [PubMed] [Google Scholar]
  • 104. Sinha R, Cross AJ, Graubard BI, Leitzmann MF, Schatzkin A. Meat intake and mortality: a prospective study of over half a million people. Arch Intern Med. 2009;169:562-571. doi: 10.1001/archinternmed.2009.6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Micha R, Peñalvo JL, Cudhea F, Imamura F, Rehm CD, Mozaffarian D. Association between dietary factors and mortality from heart disease, stroke, and type 2 diabetes in the United States. JAMA. 2017;317:912-924. doi: 10.1001/jama.2017.0947 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Schwingshackl L, Schwedhelm C, Hoffmann G, et al. Food groups and risk of all-cause mortality: a systematic review and meta-analysis of prospective studies. Am J Clin Nutr. 2017;105:1462-1473. doi: 10.3945/ajcn.117.153148 [DOI] [PubMed] [Google Scholar]
  • 107. Ford ES, Bergmann MM, Kröger J, Schienkiewitz A, Weikert C, Boeing H. Healthy living is the best revenge: findings from the European Prospective Investigation into Cancer and Nutrition-Potsdam study. Arch Intern Med. 2009;169:1355-1362. doi: 10.1001/archinternmed.2009.237 [DOI] [PubMed] [Google Scholar]
  • 108. Manuel DG, Perez R, Sanmartin C, et al. Measuring burden of unhealthy behaviours using a multivariable predictive approach: life expectancy lost in Canada attributable to smoking, alcohol, physical inactivity, and diet. PLoS Med. 2016;13:e1002082. doi: 10.1371/journal.pmed.1002082 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Suliga E, Kozieł D, Cieśla E, Głuszek S. Association between dietary patterns and metabolic syndrome in individuals with normal weight: a cross-sectional study. Nutr J. 2015;14:55. doi: 10.1186/s12937-015-0045-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Tikkanen E, Gustafsson S, Ingelsson E. Associations of fitness, physical activity, strength, and genetic risk with cardiovascular disease: longitudinal analyses in the UK Biobank study. Circulation. 2018;137:2583-2591. doi: 10.1161/CIRCULATIONAHA.117.032432 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Said MA, Verweij N, van der Harst P. Associations of combined genetic and lifestyle risks with incident cardiovascular disease and diabetes in the UK Biobank study. JAMA Cardiol. 2018;3:693-702. doi: 10.1001/jamacardio.2018.1717 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Pollan M. In Defense of Food: An Eater’s Manifesto. New York, NY: Penguin; 2009. [Google Scholar]
  • 113. Fraser GE, Shavlik DJ. Ten years of life: is it a matter of choice? Arch Intern Med. 2001;161:1645-1652. [DOI] [PubMed] [Google Scholar]
  • 114. Orlich MJ, Singh PN, Sabaté J, et al. Vegetarian dietary patterns and mortality in Adventist Health Study 2. JAMA Intern Med. 2013;173:1230-1238. doi: 10.1001/jamainternmed.2013.6473 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Martinez-Gonzalez MA, Martin-Calvo N. Mediterranean diet and life expectancy; beyond olive oil, fruits, and vegetables. Curr Opin Clin Nutr Metab Care. 2016;19:401-407. doi: 10.1097/MCO.0000000000000316 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Sofi F, Cesari F, Abbate R, Gensini GF, Casini A. Adherence to Mediterranean diet and health status: meta-analysis. BMJ. 2008;337:a1344. doi: 10.1136/bmj.a1344 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Shai I, Spence JD, Schwarzfuchs D, et al. ; DIRECT Group. Dietary intervention to reverse carotid atherosclerosis. Circulation. 2010;121:1200-1208. doi: 10.1161/CIRCULATIONAHA.109.879254 [DOI] [PubMed] [Google Scholar]
  • 118. Ornish D, Scherwitz LW, Billings JH, et al. Intensive lifestyle changes for reversal of coronary heart disease. JAMA. 1998;280:2001-2007. [DOI] [PubMed] [Google Scholar]
  • 119. Ornish D, Brown SE, Scherwitz LW, et al. Lifestyle changes and heart disease. Lancet. 1990;336:741-742. [DOI] [PubMed] [Google Scholar]
  • 120. Esselstyn CB, Jr, Gendy G, Doyle J, Golubic M, Roizen MF. A way to reverse CAD? J Fam Pract. 2014;63:356b-364b. [PubMed] [Google Scholar]
  • 121. Bredesen DE, Amos EC, Canick J, et al. Reversal of cognitive decline in Alzheimer’s disease. Aging (Albany NY). 2016;8:1250-1258. doi: 10.18632/aging.100981 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Ngandu T, Lehtisalo J, Solomon A, et al. A 2 year multidomain intervention of diet, exercise, cognitive training, and vascular risk monitoring versus control to prevent cognitive decline in at-risk elderly people (FINGER): a randomised controlled trial. Lancet. 2015;385:2255-2263. doi: 10.1016/S0140-6736(15)60461-5 [DOI] [PubMed] [Google Scholar]
  • 123. Rankin P, Morton DP, Diehl H, Gobble J, Morey P, Chang E. Effectiveness of a volunteer-delivered lifestyle modification program for reducing cardiovascular disease risk factors. Am J Cardiol. 2012;109:82-86. doi: 10.1016/j.amjcard.2011.07.069 [DOI] [PubMed] [Google Scholar]
  • 124. Daubenmier JJ, Weidner G, Sumner MD, et al. The contribution of changes in diet, exercise, and stress management to changes in coronary risk in women and men in the Multisite Cardiac Lifestyle Intervention Program. Ann Behav Med. 2007;33:57-68. doi: 10.1207/s15324796abm3301_7 [DOI] [PubMed] [Google Scholar]
  • 125. Pan A, Sun Q, Bernstein AM, Manson JE, Willett WC, Hu FB. Changes in red meat consumption and subsequent risk of type 2 diabetes mellitus: three cohorts of US men and women. JAMA Intern Med. 2013;173:1328-1335. doi: 10.1001/jamainternmed.2013.6633 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. McMacken M, Shah S. A plant-based diet for the prevention and treatment of type 2 diabetes. J Geriatr Cardiol. 2017;14:342-354. doi: 10.11909/j.issn.1671-5411.2017.05.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Snowdon DA, Phillips RL. Does a vegetarian diet reduce the occurrence of diabetes? Am J Public Health. 1985;75:507-512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Barnard ND, Cohen J, Jenkins DJA, et al. A low-fat vegan diet and a conventional diabetes diet in the treatment of type 2 diabetes: a randomized, controlled, 74-wk clinical trial. Am J Clin Nutr. 2009;89:1588S-1596S. doi: 10.3945/ajcn.2009.26736H [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Yokoyama Y, Barnard ND, Levin SM, Watanabe M. Vegetarian diets and glycemic control in diabetes: a systematic review and meta-analysis. Cardiovasc Diagn Ther. 2014;4:373-382. doi: 10.3978/j.issn.2223-3652.2014.10.04 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Bunner AE, Wells CL, Gonzales J, Agarwal U, Bayat E, Barnard ND. A dietary intervention for chronic diabetic neuropathy pain: a randomized controlled pilot study. Nutr Diabetes. 2015;5:e158. doi: 10.1038/nutd.2015.8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Crane MG, Sample C. Regression of diabetic neuropathy with total vegetarian (vegan) diet. J Nutr Med. 1994;4:431-439. doi: 10.3109/13590849409003592 [DOI] [Google Scholar]
  • 132. Lee JJ, Wedow R, Okbay A, et al. Gene discovery and polygenic prediction from a genome-wide association study of educational attainment in 1.1 million individuals. Nat Genet. 2018;50:1112-1121. doi: 10.1038/s41588-018-0147-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Corbo RM, Scacchi R. Apolipoprotein E (APOE) allele distribution in the world. Is APOE*4 a “thrifty” allele? Ann Hum Genet. 1999;63(pt 4):301-310. doi: 10.1046/j.1469-1809.1999.6340301.x [DOI] [PubMed] [Google Scholar]
  • 134. Cooper DN, Krawczak M, Polychronakos C, Tyler-Smith C, Kehrer-Sawatzki H. Where genotype is not predictive of phenotype: towards an understanding of the molecular basis of reduced penetrance in human inherited disease. Hum Genet. 2013;132:1077-1130. doi: 10.1007/s00439-013-1331-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Rappaport SM. Genetic factors are not the major causes of chronic diseases. PLoS One. 2016;11:e0154387. doi: 10.1371/journal.pone.0154387 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Katz DL, Frates EP, Bonnet JP, Gupta SK, Vartiainen E, Carmona RH. Lifestyle as medicine: the case for a true health initiative. Am J Health Promot. 2018;32:1452-1458. doi: 10.1177/0890117117705949 [DOI] [PubMed] [Google Scholar]
  • 137. Graber ML. The incidence of diagnostic error in medicine. BMJ Qual Saf. 2013;22(suppl 2):ii21-ii27. doi: 10.1136/bmjqs-2012-001615 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Maas P, Barrdahl M, Joshi AD, et al. Breast cancer risk from modifiable and nonmodifiable risk factors among white women in the United States. JAMA Oncol. 2016;2:1295-1302. doi: 10.1001/jamaoncol.2016.1025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Khera AV, Emdin CA, Drake I, et al. Genetic risk, adherence to a healthy lifestyle, and coronary disease. N Engl J Med. 2016;375:2349-2358. doi: 10.1056/NEJMoa1605086 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Ibáñez-Sanz G, Díez-Villanueva A, Alonso MH, et al. Risk model for colorectal cancer in Spanish population using environmental and genetic factors: results from the MCC-Spain study. Sci Rep. 2017;7:43263. doi: 10.1038/srep43263 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Buttar HS, Li T, Ravi N. Prevention of cardiovascular diseases: role of exercise, dietary interventions, obesity and smoking cessation. Exp Clin Cardiol. 2005;10:229-249. [PMC free article] [PubMed] [Google Scholar]
  • 142. Livingstone KM, Celis-Morales C, Papandonatos GD, et al. FTO genotype and weight loss: systematic review and meta-analysis of 9563 individual participant data from eight randomised controlled trials. BMJ. 2016;354:i4707. doi: 10.1136/bmj.i4707 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Gardner CD, Trepanowski JF, Del Gobbo LC, et al. Effect of low-fat vs low-carbohydrate diet on 12-month weight loss in overweight adults and the association with genotype pattern or insulin secretion: the DIETFITS Randomized Clinical Trial. JAMA. 2018;319:667-679. doi: 10.1001/jama.2018.0245 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Xue Y, Chen Y, Ayub Q, et al. Deleterious- and disease-allele prevalence in healthy individuals: insights from current predictions, mutation databases, and population-scale resequencing. Am J Hum Genet. 2012;91:1022-1032. doi: 10.1016/j.ajhg.2012.10.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. 1000 Genomes Project Consortium; Abecasis GR, Altshuler D, et al. A map of human genome variation from population-scale sequencing. Nature. 2010;467:1061-1073. doi: 10.1038/nature09534 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Lautenbach DM, Christensen KD, Sparks JA, Green RC. Communicating genetic risk information for common disorders in the era of genomic medicine. Annu Rev Genomics Hum Genet. 2013;14:491-513. doi: 10.1146/annurev-genom-092010-110722 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Riordan JD, Nadeau JH. From peas to disease: modifier genes, network resilience, and the genetics of health. Am J Hum Genet. 2017;101:177-191. doi: 10.1016/j.ajhg.2017.06.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Castel SE, Cervera A, Mohammadi P, et al. Modified penetrance of coding variants by cis-regulatory variation contributes to disease risk. Nat Genet. 2018;50:1327-1334. doi: 10.1038/s41588-018-0192-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Tan H. On the protective effects of gene SNPs against human cancer. EBioMedicine. 2018;33:4-5. doi: 10.1016/j.ebiom.2018.06.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Vassy JL, Christensen KD, Schonman EF, et al. The impact of whole-genome sequencing on the primary care and outcomes of healthy adult patients: a pilot randomized trial. Ann Intern Med. 2017;167:159-169. doi: 10.7326/M17-0188 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Hollands GJ, French DP, Griffin SJ, et al. The impact of communicating genetic risks of disease on risk-reducing health behaviour: systematic review with meta-analysis. BMJ. 2016;352:i1102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Godino JG, van Sluijs EMF, Marteau TM, Sutton S, Sharp SJ, Griffin SJ. Lifestyle advice combined with personalized estimates of genetic or phenotypic risk of type 2 diabetes, and objectively measured physical activity: a randomized controlled trial. PLoS Med. 2016;13:e1002185. doi: 10.1371/journal.pmed.1002185 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Andjelkovic M, Mitrovic M, Nikolic I, et al. Older hypertensive patients’ adherence to healthy lifestyle behaviors. Serbian J Exp Clin Res. 2018;19:51-56. doi: 10.1515/sjecr-2016-0083 [DOI] [Google Scholar]
  • 154. Hamer M. Adherence to healthy lifestyle in hypertensive patients: ample room for improvement? J Hum Hypertens. 2010;24:559-560. doi: 10.1038/jhh.2010.61 [DOI] [PubMed] [Google Scholar]
  • 155. Newson JT, Huguet N, Ramage-Morin PL, et al. Health behaviour changes after diagnosis of chronic illness among Canadians aged 50 or older. Health Rep. 2012;23:49-53. [PMC free article] [PubMed] [Google Scholar]
  • 156. Olofsson C, Discacciati A, Åkesson A, Orsini N, Brismar K, Wolk A. Changes in fruit, vegetable and juice consumption after the diagnosis of type 2 diabetes: a prospective study in men. Br J Nutr. 2017;117:712-719. doi: 10.1017/S0007114516002257 [DOI] [PubMed] [Google Scholar]
  • 157. Chong S, Ding D, Byun R, Comino E, Bauman A, Jalaludin B. Lifestyle changes after a diagnosis of type 2 diabetes. Diabetes Spectr. 2017;30:43-50. doi: 10.2337/ds15-0044 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Strain WD, Cos X, Hirst M, et al. Time to do more: addressing clinical inertia in the management of type 2 diabetes mellitus. Diabetes Res Clin Pract. 2014;105:302-312. doi: 10.1016/j.diabres.2014.05.005 [DOI] [PubMed] [Google Scholar]
  • 159. van de Laar FA, van de Lisdonk EH, Lucassen PL, et al. Eating behaviour and adherence to diet in patients with type 2 diabetes mellitus. Diabet Med. 2006;23:788-794. doi: 10.1111/j.1464-5491.2006.01885.x [DOI] [PubMed] [Google Scholar]
  • 160. Hercher LS, Caudle M, Griffin J, Herzog M, Matviychuk D, Tidwell J. Student-athletes’ views on APOE genotyping for increased risk of poor recovery after a traumatic brain injury. J Genet Couns. 2016;25:1267-1275. doi: 10.1007/s10897-016-9965-6 [DOI] [PubMed] [Google Scholar]
  • 161. Park JG, Kim YA, Lee JW, Kim S, Ko YJ. Unhealthy eating habits among cancer survivors. Korean J Fam Pract. 2018;8:25-31. doi: 10.21215/kjfp.2018.8.1.25 [DOI] [Google Scholar]
  • 162. Kanera IM, Bolman CAW, Mesters I, Willems RA, Beaulen AAJM, Lechner L. Prevalence and correlates of healthy lifestyle behaviors among early cancer survivors. BMC Cancer. 2016;16:4. doi: 10.1186/s12885-015-2019-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Naik H, Qiu X, Brown MC, et al. Socioeconomic status and lifestyle behaviours in cancer survivors: smoking and physical activity. Curr Oncol. 2016;23:e546-e555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. O’Neill SC, DeFrank JT, Vegella P, et al. Engaging in health behaviors to lower risk for breast cancer recurrence. PLoS One. 2013;8:e53607. doi: 10.1371/journal.pone.0053607 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Blanchard CM, Courneya KS, Stein K; American Cancer Society’s SCS-II. Cancer survivors’ adherence to lifestyle behavior recommendations and associations with health-related quality of life: results from the American Cancer Society’s SCS-II. J Clin Oncol. 2008;26:2198-2204. doi: 10.1200/JCO.2007.14.6217 [DOI] [PubMed] [Google Scholar]
  • 166. Berry NM, Miller MD, Woodman RJ, et al. Differences in chronic conditions and lifestyle behaviour between people with a history of cancer and matched controls. Med J Aust. 2014;201:96-100. doi: 10.5694/mja13.10701 [DOI] [PubMed] [Google Scholar]
  • 167. Edgington A, Morgan M. Looking beyond recurrence: comorbidities in cancer survivors. Clin J Oncol Nurs. 2011;15:E3-E12. doi: 10.1188/11.CJON.E3-E12 [DOI] [PubMed] [Google Scholar]
  • 168. Aleman BMP, Moser EC, Nuver J, et al. Cardiovascular disease after cancer therapy. EJC Suppl. 2014;12:18-28. doi: 10.1016/j.ejcsup.2014.03.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Hooning MJ, Botma A, Aleman BMP, et al. Long-term risk of cardiovascular disease in 10-year survivors of breast cancer. J Natl Cancer Inst. 2007;99:365-375. doi: 10.1093/jnci/djk064 [DOI] [PubMed] [Google Scholar]
  • 170. Jochems SHJ, Van Osch FHM, Bryan RT, et al. Impact of dietary patterns and the main food groups on mortality and recurrence in cancer survivors: a systematic review of current epidemiological literature. BMJ Open. 2018;8:e014530. doi: 10.1136/bmjopen-2016-014530 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Rabin C. Promoting lifestyle change among cancer survivors: when is the teachable moment? Am J Lifestyle Med. 2009;3:369-378. doi: 10.1177/1559827609338148 [DOI] [Google Scholar]
  • 172. Keats MR, Cui Y, Grandy SA, Parker LN. Cardiovascular disease and physical activity in adult cancer survivors: a nested, retrospective study from the Atlantic PATH cohort. J Cancer Surviv. 2017;11:264-273. [DOI] [PubMed] [Google Scholar]
  • 173. Garcia DO, Thomson CA. Physical activity and cancer survivorship. Nutr Clin Pract. 2014;29:768-779. doi: 10.1177/0884533614551969 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Gøtzsche PC, Jørgensen KJ. Screening for breast cancer with mammography. Cochrane Database Syst Rev. 2013;(6):CD001877. doi: 10.1002/14651858.CD001877.pub5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175. Kelly MP, Barker M. Why is changing health-related behaviour so difficult? Public Health. 2016;136:109-116. doi: 10.1016/j.puhe.2016.03.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Hamilton JG, Lobel M. Psychosocial factors associated with risk perceptions for chronic diseases in younger and middle-aged women. Women Health. 2015;55:921-942. doi: 10.1080/03630242.2015.1061094 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Wang C, O’Neill SM, Rothrock N, et al. ; Family Healthware Impact Trial (FHITr) Group. Comparison of risk perceptions and beliefs across common chronic diseases. Prev Med. 2009;48:197-202. doi: 10.1016/j.ypmed.2008.11.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Australian Institute of Health and Welfare. Chronic disease: overview. https://www.aihw.gov.au/reports-statistics/health-conditions-disability-deaths/chronic-disease/overview. Accessed September 12, 2018.
  • 179. Brawarsky P, Eibensteiner K, Klinger EV, et al. Accuracy of self-perceived risk for common conditions. Cogent Med. 2018;5:1463894. doi: 10.1080/2331205X.2018.1463894 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Muttarak R. Normalization of plus size and the danger of unseen overweight and obesity in England. Obesity (Silver Spring). 2018;26:1125-1129. doi: 10.1002/oby.22204 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Robinson E. Overweight but unseen: a review of the underestimation of weight status and a visual normalization theory. Obes Rev. 2017;18:1200-1209. doi: 10.1111/obr.12570 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Puhl R, Suh Y. Health consequences of weight stigma: implications for obesity prevention and treatment. Curr Obes Rep. 2015;4:182-190. doi: 10.1007/s13679-015-0153-z [DOI] [PubMed] [Google Scholar]
  • 183. Rand K, Vallis M, Aston M, et al. “It is not the diet; it is the mental part we need help with.” A multilevel analysis of psychological, emotional, and social well-being in obesity. Int J Qual Stud Health Well-being. 2017;12:1306421. doi: 10.1080/17482631.2017.1306421 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Australian Institute of Health and Welfare. Deaths in Australia. https://www.aihw.gov.au/reports/life-expectancy-death/deaths-in-australia/data. Accessed September 12, 2018.
  • 185. The Triz Journal. The fence paradox. https://triz-journal.com/the-fence-paradox/. Accessed September 12, 2018.
  • 186. Stanciole AE. Health insurance and lifestyle choices: identifying ex ante moral hazard in the US market. Geneva Pap Risk Insur Issues Pract. 2008;33:627-644. doi: 10.1057/gpp.2008.27 [DOI] [Google Scholar]
  • 187. Traeger MW, Schroeder SE, Wright EJ, et al. Effects of pre-exposure prophylaxis for the prevention of human immunodeficiency virus infection on sexual risk behavior in men who have sex with men: a systematic review and meta-analysis. Clin Infect Dis. 2018;67:676-686. doi: 10.1093/cid/ciy182 [DOI] [PubMed] [Google Scholar]
  • 188. Kalichman SC, Price D, Eaton LA, et al. Diminishing perceived threat of AIDS and increasing sexual risks of HIV among men who have sex with men, 1997-2015. Arch Sex Behav. 2017;46:895-902. doi: 10.1007/s10508-016-0934-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Holt M, Murphy DA. Individual versus community-level risk compensation following preexposure prophylaxis of HIV. Am J Public Health. 2017;107:1568-1571. doi: 10.2105/AJPH.2017.303930 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190. Abraham G, Havulinna AS, Bhalala OG, et al. Genomic prediction of coronary heart disease. Eur Heart J. 2016;37:3267-3278. doi: 10.1093/eurheartj/ehw450 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Inouye M, Abraham G, Nelson CP, et al. Genomic risk prediction of coronary artery disease in nearly 500 000 adults: implications for early screening and primary prevention [published online January 19, 2018]. bioRxiv. doi: 10.1101/250712 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. Pereira A, Mendonça MI, Borges S, et al. Genetic risk analysis of coronary artery disease in a population-based study in Portugal, using a genetic risk score of 31 variants. Arq Bras Cardiol. 2018;111:50-61. doi: 10.5935/abc.20180107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193. Burke W. Genetic tests: clinical validity and clinical utility. Curr Protoc Hum Genet. 2014;81:9.15.1-8. doi: 10.1002/0471142905.hg0915s81 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Jostins L, Barrett JC. Genetic risk prediction in complex disease. Hum Mol Genet. 2011;20(R2):R182-R188. doi: 10.1093/hmg/ddr378 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195. Mihaescu R, Moonesinghe R, Khoury M, Janssens ACJW. Predictive genetic testing for the identification of high-risk groups: a simulation study on the impact of predictive ability. Genome Med. 2011;3:51. doi: 10.1186/gm267 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Whillans AV, Dunn EW, Sandstrom GM, Dickerson SS, Madden KM. Is spending money on others good for your heart? Health Psychol. 2016;35:574-583. doi: 10.1037/hea0000332 [DOI] [PubMed] [Google Scholar]
  • 197. Phani NM, Guddattu V, Bellampalli R, et al. Population specific impact of genetic variants in KCNJ11 gene to type 2 diabetes: a case-control and meta-analysis study. PLoS One. 2014;9:e107021. doi: 10.1371/journal.pone.0107021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198. Qin LJ, Lv Y, Huang QY. Meta-analysis of association of common variants in the KCNJ11-ABCC8 region with type 2 diabetes. Genet Mol Res. 2013;12:2990-3002. doi: 10.4238/2013.August.20.1 [DOI] [PubMed] [Google Scholar]
  • 199. Curtis D. Polygenic risk score for schizophrenia is more strongly associated with ancestry than with schizophrenia. bioRxiv. 2018;28:85-89. doi: 10.1101/287136 [DOI] [PubMed] [Google Scholar]
  • 200. MacArthur DG, Manolio TA, Dimmock DP, et al. Guidelines for investigating causality of sequence variants in human disease. Nature. 2014;508:469-476. doi: 10.1038/nature13127 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201. Gallagher MD, Chen-Plotkin AS. The post-GWAS era: from association to function. Am J Hum Genet. 2018;102:717-730. doi: 10.1016/j.ajhg.2018.04.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202. Burgess S, Foley CN, Zuber V. Inferring causal relationships between risk factors and outcomes from genome-wide association study data. Annu Rev Genomics Hum Genet. 2018;19:303-327. doi: 10.1146/annurev-genom-083117-021731 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203. Bycroft C, Freeman C, Petkova D, et al. Genome-wide genetic data on ~500 000 UK Biobank participants [published online July 20, 2017]. bioRxiv. doi: 10.1101/166298 [DOI] [Google Scholar]
  • 204. 1000 Genomes Project Consortium; Auton A, Brooks LD, et al. A global reference for human genetic variation. Nature. 2015;526:68-74. doi: 10.1038/nature15393 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205. Institute of Medicine (US) Committee on Assessing Interactions Among Social, Behavioral, and Genetic Factors in Health; Hernandez LM, Blazer DG, eds. Genetics and health. In: Genes, Behavior, and the Social Environment: Moving Beyond the Nature/Nurture Debate. Washington, DC: National Academies Press; 2006. https://www.ncbi.nlm.nih.gov/books/NBK19932/. [PubMed] [Google Scholar]
  • 206. Yoo W, Smith SA, Coughlin SS. Evaluation of genetic risk scores for prediction of dichotomous outcomes. Int J Mol Epidemiol Genet. 2015;6:1-8. [PMC free article] [PubMed] [Google Scholar]
  • 207. Kisiel B, Kisiel K, Szymański K, et al. The association between 38 previously reported polymorphisms and psoriasis in a Polish population: high predicative accuracy of a genetic risk score combining 16 loci. PLoS One. 2017;12:e0179348. doi: 10.1371/journal.pone.0179348 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208. Zeller T, Seiffert M, Müller C, et al. Genome-wide association analysis for severity of coronary artery disease using the Gensini Scoring System. Front Cardiovasc Med. 2017;4:57. doi: 10.3389/fcvm.2017.00057 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209. Torkamani A, Wineinger NE, Topol EJ. The personal and clinical utility of polygenic risk scores. Nat Rev Genet. 2018;19:581-590. doi: 10.1038/s41576-018-0018-x [DOI] [PubMed] [Google Scholar]
  • 210. Salisbury M. Why we still can’t rely on genomic medicine. https://techonomy.com/2017/09/still-cant-rely-genomic-medicine/. Accessed September 24, 2018.
  • 211. Fröhlich H, Balling R, Beerenwinkel N, et al. From hype to reality: data science enabling personalized medicine. BMC Med. 2018;16:150. doi: 10.1186/s12916-018-1122-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212. Ericson U, Hindy G, Drake I, et al. Dietary and genetic risk scores and incidence of type 2 diabetes. Genes Nutr. 2018;13:13. doi: 10.1186/s12263-018-0599-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213. Middleton KR, Anton SD, Perri MG. Long-term adherence to health behavior change. Am J Lifestyle Med. 2013;7:395-404. doi: 10.1177/1559827613488867 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Dunton GF. Sustaining health-protective behaviors such as physical activity and healthy eating. JAMA. 2018;320:639-640. doi: 10.1001/jama.2018.6621 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215. Cooper M, Morton J. Digital health and obesity: how technology could be the culprit and solution for obesity. In: Rivas H, Wac K, eds. Digital Health: Scaling Healthcare to the World. Cham, Switzerland: Springer International; 2018:169-178. doi: 10.1007/978-3-319-61446-5_12 [DOI] [Google Scholar]
  • 216. Rao M, Afshin A, Singh G, Mozaffarian D. Do healthier foods and diet patterns cost more than less healthy options? A systematic review and meta-analysis. BMJ Open. 2013;3:e004277. doi: 10.1136/bmjopen-2013-004277 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217. AIA Group. Seeking the path to healthier living: the AIA Healthy Living Index 2018. https://www.aia.com/content/dam/group/en/docs/healthy-living-pdf/Whitepaper.pdf. Accessed September 16, 2018.
  • 218. Frazelle ML, Friend PJ. Optimizing the teachable moment for health promotion for cancer survivors and their families. J Adv Pract Oncol. 2016;7:422-433. [PMC free article] [PubMed] [Google Scholar]
  • 219. Stevens C, Vrinten C, Smith SG, Waller J, Beeken RJ. Determinants of willingness to receive healthy lifestyle advice in the context of cancer screening. Br J Cancer. 2018;119:251-257. doi: 10.1038/s41416-018-0160-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220. Hackshaw-McGeagh LE, Sutton E, Persad R, et al. Acceptability of dietary and physical activity lifestyle modification for men following radiotherapy or radical prostatectomy for localised prostate cancer: a qualitative investigation. BMC Urol. 2017;17:94. doi: 10.1186/s12894-017-0284-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221. Kaptein M, Lacroix J, Saini P. Individual differences in persuadability in the health promotion domain. In: Ploug T, Hasle P, Oinas-Kukkonen H, eds. Persuasive Technology. Vol 6137. Berlin, Germany: Springer; 2010:94-105. doi: 10.1007/978-3-642-13226-1_11 [DOI] [Google Scholar]
  • 222. Williams-Piehota P, Latimer AE, Katulak NA, et al. Tailoring messages to individual differences in monitoring-blunting styles to increase fruit and vegetable intake. J Nutr Educ Behav. 2009;41:398-405. doi: 10.1016/j.jneb.2008.06.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223. Petr EJ, Ayers CR, Pandey A, et al. Perceived lifetime risk for cardiovascular disease (from the Dallas Heart Study). Am J Cardiol. 2014;114:53-58. doi: 10.1016/j.amjcard.2014.04.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224. Sebire SJ, Toumpakari Z, Turner KM, et al. “I’ve made this my lifestyle now”: a prospective qualitative study of motivation for lifestyle change among people with newly diagnosed type two diabetes mellitus. BMC Public Health. 2018;18:204. doi: 10.1186/s12889-018-5114-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225. Morton DP. Combining lifestyle medicine and positive psychology to improve mental health and emotional well-being. Am J Lifestyle Med. 2018;12:370-374. doi: 10.1177/1559827618766482 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226. Koenig HG. Religion, spirituality, and health: the research and clinical implications. ISRN Psychiatry. 2012;2012:78730. doi: 10.5402/2012/278730 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227. Strawbridge WJ, Shema SJ, Cohen RD, Kaplan GA. Religious attendance increases survival by improving and maintaining good health behaviors, mental health, and social relationships. Ann Behav Med. 2001;23:68-74. doi: 10.1207/S15324796ABM2301_1 [DOI] [PubMed] [Google Scholar]
  • 228. Idler E, Blevins J, Kiser M, Hogue C. Religion, a social determinant of mortality? A 10-year follow-up of the Health and Retirement Study. PLoS One. 2017;12:e0189134. doi: 10.1371/journal.pone.0189134 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229. Oman D, Thoresen CE, Mcmahon K. Volunteerism and mortality among the community-dwelling elderly. J Health Psychol. 1999;4:301-316. doi: 10.1177/135910539900400301 [DOI] [PubMed] [Google Scholar]
  • 230. Pasanen T, Tolvanen S, Heinonen A, Kujala UM. Exercise therapy for functional capacity in chronic diseases: an overview of meta-analyses of randomised controlled trials. Br J Sports Med. 2017;51:1459-1465. doi: 10.1136/bjsports-2016-097132 [DOI] [PubMed] [Google Scholar]
  • 231. Hoffmann TC, Maher CG, Briffa T, et al. Prescribing exercise interventions for patients with chronic conditions. CMAJ. 2016;188:510-518. doi: 10.1503/cmaj.150684 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Pedersen BK, Saltin B. Exercise as medicine—evidence for prescribing exercise as therapy in 26 different chronic diseases. Scand J Med Sci Sports. 2015;25(suppl 3):1-72. doi: 10.1111/sms.12581 [DOI] [PubMed] [Google Scholar]
  • 233. Moore GE, Durstine JL, Painter PL. ACSM’s Exercise Management for Persons With Chronic Diseases and Disabilities. 4th ed. Champaign, IL: Human Kinetics; 2016. [Google Scholar]
  • 234. Durstine JL, Gordon B, Wang Z, Luo X. Chronic disease and the link to physical activity. J Sport Health Sci. 2013;2:3-11. doi: 10.1016/j.jshs.2012.07.009 [DOI] [Google Scholar]
  • 235. Minich DM, Bland JS. Personalized lifestyle medicine: relevance for nutrition and lifestyle recommendations. ScientificWorldJournal. 2013;2013:129841. doi: 10.1155/2013/129841 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236. Mann TN, Lamberts RP, Lambert MI. High responders and low responders: factors associated with individual variation in response to standardized training. Sports Med. 2014;44:1113-1124. doi: 10.1007/s40279-014-0197-3 [DOI] [PubMed] [Google Scholar]
  • 237. Dalleck LC, Haney DE, Buchanan CA, Weatherwax RM. Does a personalised exercise prescription enhance training efficacy and limit training unresponsiveness? A randomised controlled trial. J Fit Res. 2016;5:15-27. [Google Scholar]
  • 238. Böhm A, Weigert C, Staiger H, Häring HU. Exercise and diabetes: relevance and causes for response variability. Endocrine. 2016;51:390-401. doi: 10.1007/s12020-015-0792-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239. Bouchard C, Blair SN, Church TS, et al. Adverse metabolic response to regular exercise: is it a rare or common occurrence? PLoS One. 2012;7:e37887. doi: 10.1371/journal.pone.0037887 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. Stephens NA, Sparks LM. Resistance to the beneficial effects of exercise in type 2 diabetes: are some individuals programmed to fail? J Clin Endocrinol Metab. 2015;100:43-52. doi: 10.1210/jc.2014-2545 [DOI] [PubMed] [Google Scholar]
  • 241. Ishiguro H, Kodama S, Horikawa C, et al. In search of the ideal resistance training program to improve glycemic control and its indication for patients with type 2 diabetes mellitus: a systematic review and meta-analysis. Sports Med. 2016;46:67-77. doi: 10.1007/s40279-015-0379-7 [DOI] [PubMed] [Google Scholar]
  • 242. Pesta DH, Goncalves RLS, Madiraju AK, Strasser B, Sparks LM. Resistance training to improve type 2 diabetes: working toward a prescription for the future. Nutr Metab (Lond). 2017;14:24. doi: 10.1186/s12986-017-0173-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243. Montero D, Lundby C. Refuting the myth of non-response to exercise training: “non-responders” do respond to higher dose of training. J Physiol. 2017;595:3377-3387. doi: 10.1113/JP273480 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244. Melanson EL, Keadle SK, Donnelly JE, Braun B, King NA. Resistance to exercise-induced weight loss: compensatory behavioral adaptations. Med Sci Sports Exerc. 2013;45:1600-1609. doi: 10.1249/MSS.0b013e31828ba942 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245. Herrmann SD, Willis EA, Honas JJ, Lee J, Washburn RA, Donnelly JE. Energy intake, nonexercise physical activity, and weight loss in responders and nonresponders: the Midwest Exercise Trial 2. Obesity (Silver Spring). 2015;23:1539-1549. doi: 10.1002/oby.21073 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246. Hopkins M, Blundell JE, King NA. Individual variability in compensatory eating following acute exercise in overweight and obese women. Br J Sports Med. 2014;48:1472-1476. doi: 10.1136/bjsports-2012-091721 [DOI] [PubMed] [Google Scholar]
  • 247. Vlahovich N, Hughes DC, Griffiths LR, et al. Genetic testing for exercise prescription and injury prevention: AIS-Athlome consortium-FIMS joint statement. BMC Genomics. 2017;18(suppl 8):818. doi: 10.1186/s12864-017-4185-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248. Williams CJ, Williams MG, Eynon N, et al. Genes to predict VO2max trainability: a systematic review. BMC Genomics. 2017;18(suppl 8):831. doi: 10.1186/s12864-017-4192-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 249. CORDIS, European Commission. Final report summary—META-PREDICT (developing predictors of the health benefits of exercise for individuals). https://cordis.europa.eu/result/rcn/201521_en.html. Accessed August 2, 2018.
  • 250. Ekkekakis P, Hall EE, Petruzzello SJ. Variation and homogeneity in affective responses to physical activity of varying intensities: an alternative perspective on dose-response based on evolutionary considerations. J Sports Sci. 2005;23:477-500. doi: 10.1080/02640410400021492 [DOI] [PubMed] [Google Scholar]
  • 251. Kwan BM, Bryan AD. Affective response to exercise as a component of exercise motivation: attitudes, norms, self-efficacy, and temporal stability of intentions. Psychol Sport Exerc. 2010;11:71-79. doi: 10.1016/j.psychsport.2009.05.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 252. Williams DM, Dunsiger S, Ciccolo JT, Lewis BA, Albrecht AE, Marcus BH. Acute affective response to a moderate-intensity exercise stimulus predicts physical activity participation 6 and 12 months later. Psychol Sport Exerc. 2008;9:231-245. doi: 10.1016/j.psychsport.2007.04.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253. Lewis BA, Williams DM, Frayeh A, Marcus BH. Self-efficacy versus perceived enjoyment as predictors of physical activity behaviour. Psychol Health. 2016;31:456-469. doi: 10.1080/08870446.2015.1111372 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254. Yanovski SZ, Yanovski JA. Toward precision approaches for the prevention and treatment of obesity. JAMA. 2018;319:223-224. doi: 10.1001/jama.2017.20051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255. Pitkin RM. Folate and neural tube defects. Am J Clin Nutr. 2007;85:285S-288S. doi: 10.1093/ajcn/85.1.285S [DOI] [PubMed] [Google Scholar]
  • 256. Day KJ, Adamski MM, Dordevic AL, Murgia C. Genetic variations as modifying factors to dietary zinc requirements—a systematic review. Nutrients. 2017;9:E148. doi: 10.3390/nu9020148 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257. Brandon JA, Farmer BC, Williams HC, Johnson LA. APOE and Alzheimer’s disease: neuroimaging of metabolic and cerebrovascular dysfunction. Front Aging Neurosci. 2018;10:180. doi: 10.3389/fnagi.2018.00180 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258. Yassine HN, Rawat V, Mack WJ, et al. The effect of APOE genotype on the delivery of DHA to cerebrospinal fluid in Alzheimer’s disease. Alzheimers Res Ther. 2016;8:25. doi: 10.1186/s13195-016-0194-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259. Yassine HN, Croteau E, Rawat V, et al. DHA brain uptake and APOE4 status: a PET study with [1-11C]-DHA. Alzheimers Res Ther. 2017;9:23. doi: 10.1186/s13195-017-0250-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 260. Yassine HN, Braskie MN, Mack WJ, et al. Association of docosahexaenoic acid supplementation with Alzheimer disease stage in apolipoprotein E ε4 carriers: a review. JAMA Neurol. 2017;74:339-347. doi: 10.1001/jamaneurol.2016.4899 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261. Daiello LA, Gongvatana A, Dunsiger S, Cohen RA, Ott BR; Alzheimer’s Disease Neuroimaging Initiative. Association of fish oil supplement use with preservation of brain volume and cognitive function. Alzheimers Dement. 2015;11:226-235. doi: 10.1016/j.jalz.2014.02.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262. Chouinard-Watkins R, Plourde M. Fatty acid metabolism in carriers of apolipoprotein E epsilon 4 allele: is it contributing to higher risk of cognitive decline and coronary heart disease? Nutrients. 2014;6:4452-4471. doi: 10.3390/nu6104452 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263. Monteiro JP, Kussmann M, Kaput J. The genomics of micronutrient requirements. Genes Nutrition. 2015;10:19. doi: 10.1007/s12263-015-0466-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264. Vadiveloo M, Parekh N, Mattei J. Greater healthful food variety as measured by the US Healthy Food Diversity Index is associated with lower odds of metabolic syndrome and its components in US adults. J Nutr. 2014;145:564-571. doi: 10.3945/jn.114.199125 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265. Otto MC, Padhye NS, Bertoni AG, Jacobs DR, Jr, Mozaffarian D. Everything in moderation—dietary diversity and quality, central obesity and risk of diabetes. PLoS One. 2015;10:e0141341. doi: 10.1371/journal.pone.0141341 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266. House JS, Landis KR, Umberson D. Social relationships and health. Science. 1988;241:540-545. [DOI] [PubMed] [Google Scholar]
  • 267. Holt-Lunstad J. The potential public health relevance of social isolation and loneliness: prevalence, epidemiology, and risk factors. Public Policy Aging Rep. 2017;27:127-130. doi: 10.1093/ppar/prx030 [DOI] [Google Scholar]
  • 268. Croezen S, Picavet HSJ, Haveman-Nies A, Verschuren WM, de Groot LC, van’t Veer P. Do positive or negative experiences of social support relate to current and future health? Results from the Doetinchem Cohort Study. BMC Public Health. 2012;12:65. doi: 10.1186/1471-2458-12-65 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269. Segrin C. Indirect effects of social skills on health through stress and loneliness. Health Commun. 2019;34:118-124. doi: 10.1080/10410236.2017.1384434 [DOI] [PubMed] [Google Scholar]
  • 270. Wang ML, Pbert L, Lemon SC. The influence of family, friend, and coworker social support and social undermining on weight gain prevention among adults. Obesity (Silver Spring). 2014;22:1973-1980. doi: 10.1002/oby.20814 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271. Egolf B, Lasker J, Wolf S, Potvin L. The Roseto effect: a 50-year comparison of mortality rates. Am J Public Health. 1992;82:1089-1092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272. Jamal SN, Moy FM, Mohamed MNA, Mukhtar F. Effectiveness of a Group Support Lifestyle Modification (GSLiM) programme among obese adults in workplace: a randomised controlled trial. PLoS One. 2016;11:e0160343. doi: 10.1371/journal.pone.0160343 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273. Fisher EB, Boothroyd RI, Elstad EA, et al. Peer support of complex health behaviors in prevention and disease management with special reference to diabetes: systematic reviews. Clin Diabetes Endocrinol. 2017;3:4. doi: 10.1186/s40842-017-0042-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274. Petosa RL, Smith DLH. Peer mentoring for health behavior change: a systematic review. Am J Health Educ. 2014;45:351-357. doi: 10.1080/19325037.2014.945670 [DOI] [Google Scholar]
  • 275. Embuldeniya G, Veinot P, Bell E, et al. The experience and impact of chronic disease peer support interventions: a qualitative synthesis. Patient Educ Couns. 2013;92:3-12. doi: 10.1016/j.pec.2013.02.002 [DOI] [PubMed] [Google Scholar]
  • 276. Wingate L, Graffy J, Holman D, Simmons D. Can peer support be cost saving? An economic evaluation of RAPSID: a randomized controlled trial of peer support in diabetes compared to usual care alone in East of England communities. BMJ Open Diabetes Res Care. 2017;5:e000328. doi: 10.1136/bmjdrc-2016-000328 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277. Yeung JWK, Zhang Z, Kim TY. Volunteering and health benefits in general adults: cumulative effects and forms. BMC Public Health. 2017;18:8. doi: 10.1186/s12889-017-4561-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278. Burr JA, Han SH, Tavares JL. Volunteering and cardiovascular disease risk: does helping others get “under the skin?” Gerontologist. 2016;56:937-947. doi: 10.1093/geront/gnv032 [DOI] [PubMed] [Google Scholar]
  • 279. Kangovi S, Asch DA. Behavioral phenotyping in health promotion: embracing or avoiding failure. JAMA. 2018;319:2075-2076. doi: 10.1001/jama.2018.2921 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280. Melemis SM. Relapse prevention and the five rules of recovery. Yale J Biol Med. 2015;88:325-332. [PMC free article] [PubMed] [Google Scholar]
  • 281. Mann T, de Ridder D, Fujita K. Self-regulation of health behavior: social psychological approaches to goal setting and goal striving. Health Psychol. 2013;32:487-498. doi: 10.1037/a0028533 [DOI] [PubMed] [Google Scholar]
  • 282. Gollwitzer P. Implementation intentions: strong effects of simple plans. Am Psychol. 1999;54:493-503. doi: 10.1037/0003-066X.54.7.493 [DOI] [Google Scholar]
  • 283. Rees JH, Bamberg S, Jäger A, Victor L, Bergmeyer M, Friese M. Breaking the habit: on the highly habitualized nature of meat consumption and implementation intentions as one effective way of reducing it. Basic Appl Soc Psychol. 2018;40:136-147. doi: 10.1080/01973533.2018.1449111 [DOI] [Google Scholar]
  • 284. Epton T, Armitage CJ. Does situation-specificity affect the operation of implementation intentions? Behav Ther. 2017;48:860-869. doi: 10.1016/j.beth.2017.08.003 [DOI] [PubMed] [Google Scholar]
  • 285. Castonguay A, Miquelon P, Boudreau F. Self-regulation resources and physical activity participation among adults with type 2 diabetes. Health Psychol Open. 2018;5:2055102917750331. doi: 10.1177/2055102917750331 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 286. McNeil DW, Addicks SH, Randall CL. Motivational Interviewing and Motivational Interactions for Health Behavior Change and Maintenance. Oxford, England: Oxford University Press; 2017. doi: 10.1093/oxfordhb/9780199935291.013.21 [DOI] [Google Scholar]
  • 287. DeJesus RS, Clark MM, Rutten LJF, et al. Impact of a 12-week wellness coaching on self-care behaviors among primary care adult patients with prediabetes. Prev Med Rep. 2018;10:100-105. doi: 10.1016/j.pmedr.2018.02.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288. Britt E, Hudson SM, Blampied NM. Motivational interviewing in health settings: a review. Patient Educ Couns. 2004;53:147-155. doi: 10.1016/S0738-3991(03)00141-1 [DOI] [PubMed] [Google Scholar]
  • 289. Williams SL, Haskard-Zolnierek KB, DiMatteo MR. Psychosocial predictors of behavior change. In: Riekert KA, Ockene JK, Pbert L, eds. The Handbook of Health Behavior Change. 4th ed. New York, NY: Springer; 2013:69-86. [Google Scholar]
  • 290. Rosbach M, Andersen JS. Patient-experienced burden of treatment in patients with multimorbidity—a systematic review of qualitative data. PLoS One. 2017;12:e0179916. doi: 10.1371/journal.pone.0179916 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 291. Boehmer KR, Gionfriddo MR, Rodriguez-Gutierrez R, et al. Patient capacity and constraints in the experience of chronic disease: a qualitative systematic review and thematic synthesis. BMC Fam Pract. 2016;17:127. doi: 10.1186/s12875-016-0525-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292. Dabrh AMA, Gallacher K, Boehmer KR, Hargraves IG, Mair FS. Minimally disruptive medicine: the evidence and conceptual progress supporting a new era of healthcare. J R Coll Physicians Edinb. 2015;45:114-117. doi: 10.4997/JRCPE.2015.205 [DOI] [PubMed] [Google Scholar]
  • 293. Hills AP, Byrne NM, Lindstrom R, Hill JO. “Small changes” to diet and physical activity behaviors for weight management. Obes Facts. 2013;6:228-238. doi: 10.1159/000345030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294. Utter J, Larson N, Laska MN, Winkler M, Neumark-Sztainer D. Self-perceived cooking skills in emerging adulthood predict better dietary behaviors and intake 10 years later: a longitudinal study. J Nutr Educ Behav. 2018;50:494-500. doi: 10.1016/j.jneb.2018.01.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295. Garcia AL, Reardon R, Hammond E, Parrett A, Gebbie-Diben A. Evaluation of the “Eat Better Feel Better” cooking programme to tackle barriers to healthy eating. Int J Environ Res Public Health. 2017;14:E380. doi: 10.3390/ijerph14040380 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 296. Raber M, Chandra J, Upadhyaya M, et al. An evidence-based conceptual framework of healthy cooking. Prev Med Rep. 2016;4:23-28. doi: 10.1016/j.pmedr.2016.05.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 297. Eisenberg DM, Burgess JD. Nutrition education in an era of global obesity and diabetes: thinking outside the box. Acad Med. 2015;90:854-860. doi: 10.1097/ACM.0000000000000682 [DOI] [PubMed] [Google Scholar]
  • 298. Mills S, Brown H, Wrieden W, White M, Adams J. Frequency of eating home cooked meals and potential benefits for diet and health: cross-sectional analysis of a population-based cohort study. Int J Behav Nutr Phys Act. 2017;14:109. doi: 10.1186/s12966-017-0567-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 299. Garcia AL, Reardon R, McDonald M, Vargas-Garcia EJ. Community interventions to improve cooking skills and their effects on confidence and eating behaviour. Curr Nutr Rep. 2016;5:315-322. doi: 10.1007/s13668-016-0185-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 300. Goh LML, Wong AXY, Ang GY, Tan ASL. Effectiveness of nutrition education accompanied by cooking demonstration. Br Food J. 2017;119:1052-1066. doi: 10.1108/BFJ-10-2016-0464 [DOI] [Google Scholar]
  • 301. Dubowitz T, Cohen DA, Huang CY, Beckman RA, Collins RL. Using a grocery list is associated with a healthier diet and lower BMI among very high risk adults. J Nutr Educ Behav. 2015;47:259-264. doi: 10.1016/j.jneb.2015.01.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 302. de Leon J. Evidence-based medicine versus personalized medicine are they enemies? J Clin Psychopharmacol. 2012;32:153-164. doi: 10.1097/JCP.0b013e3182491383 [DOI] [PubMed] [Google Scholar]
  • 303. Altshuler D, Daly MJ, Lander ES. Genetic mapping in human disease. Science. 2008;322:881-888. doi: 10.1126/science.1156409 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 304. Timmons JA, Atherton PJ, Larsson O, et al. A coding and non-coding transcriptomic perspective on the genomics of human metabolic disease. Nucleic Acids Res. 2018;46:7772-7792. doi: 10.1093/nar/gky570 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 305. Heeney C, Kerr SM. Balancing the local and the universal in maintaining ethical access to a genomics biobank. BMC Med Ethics. 2017;18:80. doi: 10.1186/s12910-017-0240-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 306. De Souza YG, Greenspan JS. Biobanking past, present and future: responsibilities and benefits. AIDS. 2013;27:303-312. doi: 10.1097/QAD.0b013e32835c1244 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 307. Budimir D, Polašek O, Marušić A, et al. Ethical aspects of human biobanks: a systematic review. Croat Med J. 2011;52:262-279. doi: 10.3325/cmj.2011.52.262 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 308. Ormond KE, Cho MK. Translating personalized medicine using new genetic technologies in clinical practice: the ethical issues. Per Med. 2014;11:211-222. doi: 10.2217/pme.13.104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 309. Harvey A. Opinion leaders’ views on issues in personalized medicine. Per Med. 2012;9:127-131. doi: 10.2217/pme.12.14 [DOI] [PubMed] [Google Scholar]
  • 310. Gymrek M, McGuire AL, Golan D, Halperin E, Erlich Y. Identifying personal genomes by surname inference. Science. 2013;339:321-324. doi: 10.1126/science.1229566 [DOI] [PubMed] [Google Scholar]
  • 311. Kushner J. The ethics of personalized medicine. Per Med Universe. 2014;3:42-45. doi: 10.1016/j.pmu.2014.03.001 [DOI] [Google Scholar]
  • 312. Nicol D, Bubela T, Chalmers D, et al. Precision medicine: drowning in a regulatory soup? J Law Biosci. 2016;3:281-303. doi: 10.1093/jlb/lsw018 [DOI] [Google Scholar]
  • 313. Johnston J, Lantos JD, Goldenberg A, et al. ; Members of the NSIGHT Ethics and Policy Advisory Board. Sequencing newborns: a call for nuanced use of genomic technologies. Hastings Cent Rep. 2018;48(suppl 2):S2-S6. doi: 10.1002/hast.874 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 314. Korthals M, ed. Expectations and disappointments of the human genome: the genomics health card and anti obesity pills. In: Genomics, Obesity and the Struggle over Responsibilities. Vol 18. Dordrecht, Netherland: Springer Nature; 2011:207-214. doi: 10.1007/978-94-007-0127-4_14 [DOI] [Google Scholar]
  • 315. Vollmann J. Personalised medicine: priority setting and opportunity costs in European public health care systems. JAHR. 2015;6:255-266. [Google Scholar]
  • 316. Sturdy S. Personalised medicine and the economy of biotechnological promise. New Bioeth. 2017;23:30-37. doi: 10.1080/20502877.2017.1314892 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from American Journal of Lifestyle Medicine are provided here courtesy of SAGE Publications

RESOURCES