ArticlePDF Available

Fasting-mimicking diet and markers/risk factors for aging, diabetes, cancer, and cardiovascular disease

Authors:

Abstract

Calorie restriction or changes in dietary composition can enhance healthy aging, but the inability of most subjects to adhere to chronic and extreme diets, as well as potentially adverse effects, limits their application. We randomized 100 generally healthy participants from the United States into two study arms and tested the effects of a fasting-mimicking diet (FMD)—low in calories, sugars, and protein but high in unsaturated fats—on markers/risk factors associated with aging and age-related diseases. We compared subjects who followed 3 months of an unrestricted diet to subjects who consumed the FMD for 5 consecutive days per month for 3 months. Three FMD cycles reduced body weight, trunk, and total body fat; lowered blood pressure; and decreased insulin-like growth factor 1 (IGF-1). No serious adverse effects were reported. After 3 months, control diet subjects were crossed over to the FMD program, resulting in a total of 71 subjects completing three FMD cycles. A post hoc analysis of subjects from both FMD arms showed that body mass index, blood pressure, fasting glucose, IGF-1, triglycerides, total and low-density lipoprotein cholesterol, and C-reactive protein were more beneficially affected in participants at risk for disease than in subjects who were not at risk. Thus, cycles of a 5-day FMD are safe, feasible, and effective in reducing markers/risk factors for aging and age-related diseases. Larger studies in patients with diagnosed diseases or selected on the basis of risk factors are warranted to confirm the effect of the FMD on disease prevention and treatment.
METABOLIC DISEASE 2017 © The Authors,
some rights reserved;
exclusive licensee
American Association
for the Advancement
of Science.
Fasting-mimicking diet and markers/risk factors for aging,
diabetes, cancer, and cardiovascular disease
Min Wei,
1
* Sebastian Brandhorst,
1
* Mahshid Shelehchi,
1
Hamed Mirzaei,
1
Chia Wei Cheng,
1
Julia Budniak,
1
Susan Groshen,
2
Wendy J. Mack,
2
Esra Guen,
1
Stefano Di Biase,
1
Pinchas Cohen,
1
Todd E. Morgan,
1
Tanya Dorff,
3
Kurt Hong,
4
Andreas Michalsen,
5
Alessandro Laviano,
6
Valter D. Longo
1,7
Calorie restriction or changes in dietary composition can enhance healthy aging, but the inability of most
subjects to adhere to chronic and extreme diets, as well as potentially adverse effects, limits their application.
We randomized 100 generally healthy participants from the United States into two study arms and tested the
effects of a fasting-mimicking diet (FMD)low in calories, sugars, and protein but high in unsaturated fatson
markers/risk factors associated with aging and age-related diseases. We compared subjects who followed 3 months
of an unrestricted diet to subjects who consumed the FMD for 5 consecutive days per month for 3 months. Three
FMD cycles reduced body weight, trunk, and total body fat; lowered blood pressure; and decreased insulin-like
growth factor 1 (IGF-1). No serious adverse effects were reported. After 3 months, control diet subjects were crossed
over to the FMD program, resulting in a total of 71 subjects completing three FMD cycles. A post hoc analysis of
subjects from both FMD arms showed that body mass index, blood pressure, fasting glucose, IGF-1, triglycerides,
total and low-density lipoprotein cholesterol, and C-reactive protein were more beneficially affected in participants
at risk for disease than in subjects who were not at risk. Thus, cycles of a 5-day FMD are safe, feasible, and effective
in reducing markers/risk factors for aging and age-related diseases. Larger studies in patients with diagnosed dis-
eases or selected on the basis of risk factors are warranted to confirm the effect of the FMD on disease prevention
and treatment.
INTRODUCTION
Metabolic syndrome is defined by co-occurrence of three of five of
the following conditions: abdominal obesity, elevated fasting glucose,
elevated blood pressure, high serum triglycerides, and low levels of
high-density lipoprotein (HDL) cholesterol (1). Affecting 47 million
Americans (2), it is associated with a major increase in the risk of
cardiovascular disease (CVD) and all-cause mortality (3). Although
prolonged fasting or very low calorie fasting-mimicking diets (FMDs)
can ameliorate the incidence of diseases such as cancer and multiple
sclerosis in mice (46), randomized trials to assess fastingsabilityto
reduce markers/risk factors for aging and major age-related diseases
have not been carried out (79). Prolonged fasting, in which only water
is consumed for 2 or more days, reduces pro-growth signaling and acti-
vates cellular protection mechanisms in organisms ranging from single-cell
yeast to mammals (10). In mammals, this is achieved in part by tempora-
rily reducing glucose and circulating insulin-like growth factor 1 (IGF-1),
a hormone well studied for its role in metabolism, growth, and develop-
ment, as well as for its association with aging and cancer (1116). Severe
growth hormone receptor and IGF-1 deficiencies are associated with a
reduced risk of cancer, diabetes, and overall mortality in humans (17,18).
Mice fed periodically with the FMD show extended healthspan and
multisystem regeneration, reduced inflammation and cancer inci-
dence, and enhanced cognitive performance (5). Despite its potential
for disease prevention and treatment, prolonged fasting is difficult to
implement in human subjects and may exacerbate preexisting nutri-
tional deficiencies, making it not feasible and/or safe for children, the
elderly, frail individuals, and even most of the healthy adults. We have
investigated whether a dietary intervention more practical and safer
than fasting could affect markers or risk factors for aging and diseases.
To this end, we developed an FMD based on a diet previously tested
in animals and designed to achieve effects similar to those caused
by fasting on IGF-1, insulin-like growth factorbinding protein
1 (IGFBP-1), glucose, and ketone bodies (17). To prevent nutrient
deficiency, this FMD provided between 3000 and 4600 kJ per day, as
well as high micronutrient nourishment, to each human subject (5).
We also previously showed the safety and feasibility of this interven-
tion in 19 study participants who consumed three monthly cycles of
this FMD lasting 5 days each (5).
We now report the results of a randomized controlled trial of 100
subjects, 71 of whom completed three cycles of the FMD either in a
randomized phase (n=39)orafterbeingcrossed over from a control
diet group to the FMD group (n=32).Weevaluatedtheeffectsofthe
FMD on risk factors and markers for aging, cancer, metabolic syn-
drome, and CVDs in generally healthy participants ranging from 20
to 70 years of age.
RESULTS
Baseline data for all subjects
From April 2013 to July 2015, 100 study participants were randomized
and assigned to either arm 1 (n= 48) or arm 2 (n= 52). At enrollment,
independent of whether they completed the trial or not, subjects in the
two arms were comparable for age, sex, race, and body weight (Table 1).
Hispanics (27%) were underrepresented in the study population in
1
Longevity Institute, School of Gerontology, and Department of Biological Sciences,
University of Southern California, Los Angeles, CA 90089, USA.
2
Department of Preventive
Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
90033, USA.
3
Norris Comprehensive Cancer Center, Keck School of Medicine, University
of Southern California, Los Angeles, CA 90033, USA.
4
Department of Internal Medicine,
Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
5
Department of Internal and Complementary Medicine, Charité University Medical
Center, 10117 Berlin, Germany.
6
Department of Clinical Medicine, Sapienza University,
00185 Rome, Italy.
7
FIRC Institute of Molecular Oncology, Italian Foundation for Cancer
Research Institute of Molecular Oncology, 20139 Milan, Italy.
*These authors contributed equally to this work.
Corresponding author. Email: vlongo@usc.edu
SCIENCE TRANSLATIONAL MEDICINE |RESEARCH ARTICLE
Wei et al., Sci. Transl. Med. 9, eaai8700 (2017) 15 February 2017 1of12
by guest on October 20, 2017http://stm.sciencemag.org/Downloaded from
comparison to their representation (~45%) in the greater Los Angeles
area (California) (19). The participants in control arm 1 were asked to
continue their normal diet for 3 months, whereas participants in arm 2
started the FMD intervention. Two participants withdrew from arm
1 because of scheduling conflicts before completion of the informed
consent. In the randomized comparison (Fig. 1), 18 participants or 5
of 48 (10%) in the control arm and 13 of 52 participants in the
FMD arm (25%) were excluded or withdrew from the study. Of the
48 subjects enrolled in the control arm, two withdrew because of schedul-
ing conflicts, two because of unspecified personal issues, and one for un-
known reasons. Six of the 52 subjects enrolled in the FMD arm withdrew
from the study because of scheduling conflicts, five withdrew because of
unspecified personal issues, and two participants were excluded from the
study because of noncompliance to the FMD protocol.
Adverse effects and safety
Following the Common Terminology Criteria for Adverse Events
(CTCAE; v4.0), 54 to 100% (depending on the adverse event) of the
participants reported no adverse effects during the FMD cycles (fig.
S1). The most common self-reported grade 1 (mild) or grade 2 (mod-
erate) symptoms experienced by the participants were fatigue, weak-
ness, and headaches. No adverse effects of grade 3 or higher were
reported. A comprehensive metabolic panel that measured changes
in metabolic markers and liver and kidney function showed no neg-
ative effects of three cycles of the FMD (table S1). In summary, after
three cycles of the FMD, subjects reported only some mild and very
few moderate side effects.
Baseline risk factors and metabolic markers: Comparison of
randomized control and FMD subjects who completed the trial
At baseline, there were no significant differences in metabolic markers
or risk factors for age-related diseases and conditions between the
subjects who successfully completed the randomized trial in arm
1 (normal diet) and arm 2 (FMD), including body weight (P=0.39),
body mass index (BMI) (P= 0.24), total body fat (P=0.11),trunkfat
(P= 0.087), lean body mass (P= 0.15), waist circumference (P=0.34),
fasting glucose (P=0.55),IGF-1(P= 0.51), systolic and diastolic blood
pressure (P=0.60andP= 0.91, respectively), triglycerides (P=0.21),
and C-reactive protein (CRP) (P= 0.28). The notable exception was
that total cholesterol (P= 0.014) and low-density lipoprotein (LDL)
(P= 0.024), but not HDL (P= 0.99), were significantly lower at baseline
for subjects who were enrolled and completed arm 2 (Table 2). In sum-
mary, the values for disease markers and risk factors at baseline were
comparable between the control diet and FMD groups, with the excep-
tion of total and LDL cholesterol.
Changes in risk factors and metabolic markers: Comparison
of randomized control and FMD groups
Next, we evaluated the effects of the FMD by assessing the changes
in marker/risk factor values between baseline and 5 to 7 days after
the end of the third cycle of the FMD and compared them to those
occurring in the control arm within the same 3-month period (Fig. 2,
Table2,andtableS2).ParticipantsintheFMDarm(arm2)loston
average 2.6 ± 2.5 kg (±SD) (P< 0.0001) of weight, which was due in
part to a reduction in total body fat (absolute values and relative vol-
ume % of total mass) and trunk fat (absolute values) (Table 2 and
table S2). Subjects on the control diet did not lose body weight (0.1 ±
2.1 kg). After controlling the false discovery rate (FDR) of 0.05 between
the control and FMD groups, no change in the percentage of lean body
mass was observed (relative to the total mass; P= 0.07), although ab-
solute lean body mass was reduced in arm 2 (P=0.004)(Table2and
table S2). Waist circumference measured after three FMD cycles was
reduced by 4.1 ± 5.2 cm (P= 0.0035 between groups). The FMD cycles
also resulted in a decrease in IGF-1 concentrations of 21.7 ± 46.2 ng/ml
(P= 0.0017 between groups). Systolic blood pressure was reduced by 4.5
±6.0mmHg(P=0.023 between groups), and diastolic blood pressure
was reduced by 3.1 ± 4.7 mmHg (P= 0.053 between groups). Fasting
glucose (P= 0.27), triglycerides (P= 0.27), cholesterol (total, P= 0.81;
Table 1. Characteristics of all subjects at enrollment. Plus-minus values
are means ± SD rounded to the nearest 10th.
Characteristics Arm 1 (n= 48) Arm 2 (n= 52)
Sex, n(%)
Male 18 (37.5) 19 (36.5)
Female 30 (62.5) 33 (63.5)
Race or ethnic group, n(%)*
White 26 (54.2) 25 (48.1)
Black 2 (4.2) 5 (9.6)
Hispanic 13 (27.1) 14 (26.9)
Asian 6 (12.5) 7 (13.5)
Other 1 (2.1) 1 (1.9)
Age (years) 42.2 ± 12.5 43.3 ± 11.7
Weight (kg) 77.0 ± 15.9 74.3 ± 16.6
Education (years) 16.7 ± 2.8 16.6 ± 2.3
Smoking status, n(%)
Never smoked 29 (60.4) 39 (75.0)
Former smoker 13 (27.1) 9 (17.3)
Current smoker 6 (12.5) 4 (7.7)
BMI, n(%)
Mean 27.8 ± 5.1 26.6 ± 4.9
<25 17 (35.4) 20 (38.4)
2530 18 (37.5) 21 (40.4)
>30 13 (27.1) 11 (21.2)
Systolic blood pressure (mmHg) 117.2 ± 12.3 117.2 ± 13.0
Diastolic blood pressure (mmHg) 75.6 ± 9.2 75.2 ± 7.8
Triglycerides (mg/dl) 104.0 ± 64.6 84.7 ± 37.2
Cholesterol (mg/dl)
Total 197.5 ± 39.6 185.7 ± 36.6
LDL 114.5 ± 36.1 110.3 ± 61.6
HDL 62.2 ± 16.4 65.2 ± 18.1
*The race or ethnic group was assigned by the subjects themselves. The
BMI is the weight in kilograms divided by the square of the height in meters.
SCIENCE TRANSLATIONAL MEDICINE |RESEARCH ARTICLE
Wei et al., Sci. Transl. Med. 9, eaai8700 (2017) 15 February 2017 2of12
by guest on October 20, 2017http://stm.sciencemag.org/Downloaded from
LDL, P=0.50;HDL,P= 0.90), and the acute-phase inflammatory
marker CRP (P= 0.27) did not differ significantly between groups. A
graphical summary of these data is presented in Fig. 2. In conclusion,
three cycles of the FMD reduced body weight, trunk and total body
fat, blood pressure, and IGF-1 in comparison to a normal diet.
Changes in risk factors and metabolic markers of
age-related diseases and conditions: Observational
pre-post FMD comparison
After 3 months, 43 subjects from the control arm were crossed over
to the FMD intervention. Eleven (26%) of these subjects withdrew
before completing three FMD cycles (Fig. 1). Five of these participants
withdrew because of scheduling issues, and two subjects opted to leave
the trial for unspecificpersonal reasons. We also excluded four partic-
ipants based on nonadherence to the FMD protocol. The causes for
withdrawal/exclusion were comparable between the arms. Considering
both FMD treatment arms, 24 of the 95 participants (25%) were ex-
cluded or withdrew from the study before completion of the three
FMD cycles (arm 2, n=13FMDs;arm1,n= 11 after FMD crossover)
because of schedulingconflicts(total,n= 11: arm 2, n=6FMDs;arm
1, n= 5 after FMD crossover), personal issues (total, n=7:arm2,n=
5FMDs;arm1,n= 2 after FMD crossover), or dislike of the diet and/or
nonadherence to the dietary protocol (total, n=6:arm2,n=2FMD;
arm 1, n= 4 after FMD crossover) (Fig. 1). The 25% dropout rate for
participants during the FMD is higher than the 10% dropout rate ob-
served during control diet in arm 1, but this is expected consid-
ering that subjects in control diet group only dropped out because
of scheduling conflicts because they were allowed to remain on their
normal diet. Ninety-five (95%) subjects completed one cycle, and 71
(71%) subjects completed three cycles of the FMD. Compared to the
71 participants who completed the three FMD cycles in arms 1 and
2, the 24 subjects who dropped out were not different in age (42.5 ±
11.6 years versus 43.3 ± 13.1 years) or BMI (27.1 ± 4.9 versus 26.9 ±
4.7) but were mostly female (18% male versus 82% female; P= 0.0045,
Fishersexacttest;fig.S2).
Because the differential dropout rate during the FMD treatment
period (25% in FMD in the randomized arm 2 and/or after arm
1 crossover versus 10% in the randomized arm 1 control) may have
caused biases in estimates of the FMD treatment effect, we compared
the changes in trial outcomes between the two groups who completed
three FMD cycles (n=39FMDrandomizedarm2andn=32after
arm 1 crossover to FMD) using sensitivity analysis. Three FMD cycles
had comparable effects between subjects in arm 1 (after crossover) and
arm 2 (randomized) with the exception of HDL, which underwent
a greater reduction in arm 2 (P= 0.03), and the decrease in absolute
lean body mass, which was observed in arm 2 but not arm 1 (table
S2). Because the FMD had similar effects in both arms, we combined
the results from the two arms to assess the changes in metabolites
Fig. 1. CONSORT diagram. Consolidated Standards of Reporting Trials (CONSORT) diagram of 102 contacted subjects of which 100 were enrolled into the study two
arms. Arm 1 (n= 48), the controlgroup, maintained their normal caloric intake for a 3-month monitoring period. Data were collected at enrollment and again after 3 months.
Participants in arm 2 (n= 52) started the FMD after randomization. The FMD is provided for 5 days permonthforthreeconsecutivecycles.Data were collected at enrollment,
at the completion of the first FMD cycle but before resuming normal dietary intake, and also on average 5 days after subjects resumed their normal diet after the final FMD
cycle. After the initial 3-month period, subjects in arm 1 also started the FMD. An optional follow-up visit in the clinic for analysis was offered to all participants about 3 months
after the completion of the third FMD cycle.
SCIENCE TRANSLATIONAL MEDICINE |RESEARCH ARTICLE
Wei et al., Sci. Transl. Med. 9, eaai8700 (2017) 15 February 2017 3of12
by guest on October 20, 2017http://stm.sciencemag.org/Downloaded from
Table 2. Biomarker/risk factor changes in subjects who completed the trial. CI, confidence interval.
Variable n
Baseline* CTRL: 3 months after baseline Efficacy
(comparing D)
P
§
Mean ± SD (95% CI)
FMD: 5 days after third FMD cycle
Mean ± SD (95% CI) P
Difference: D
Body weight (kg)
Control diet, arm 1 43 77.2 ± 16.5 (72.182.2) 77.3 ± 17.0 (72.082.5) 0.72 0.1 ± 2.1 <0.0001
||
FMD, arm 2 39 74.1 ± 15.5 (69.378.9) 71.6 ± 14.6 (67.076.1) <0.0001 2.6 ± 2.5
BMI
Control diet, arm 1 43 27.4 ± 4.8 (25.928.9) 27.4 ± 5.0 (25.928.9) 0.82 0.0 ± 0.7 <0.0001
||
FMD, arm 2 39 26.2 ± 4.4 (24.827.6) 25.3 ± 4.3 (24.026.5) <0.0001 0.9 ± 0.9
Total body fat** (absolute volume)
Control diet, arm 1 43 23,651 ± 8,155 (21,14226,161) 23,607 ± 8,337 (21,04126,173) 0.83 44 ± 1,365 0.0002
||
FMD, arm 2 38 20,643 ± 8,459 (17,95323,332) 19,249 ± 7,792 (16,77221,726) <0.0001 1,393 ± 1,786
Trunk fat** (absolute volume)
Control diet, arm 1 43 8,429 ± 4,742 (6,9699,888) 8,395 ± 4,776 (6,9259,865) 0.83 33 ± 1,046 0.018
FMD, arm 2 38 6,573 ± 4,877 (5,0228,124) 5,938 ± 4,295 (4,5727,303) 0.0023 636 ± 1,198
Lean body mass** (relative volume %)
Control diet, arm 1 43 63.9 ± 8.2 (61.466.4) 64.0 ± 8.7 (61.366.7) 0.64 0.1 ± 1.5 0.070
FMD, arm 2 38 66.8 ± 9.6 (63.769.8) 67.6 ± 9.4 (64.670.6) 0.016 0.8 ± 2.0 0.8 ± 25
Waist circumference (cm)
Control diet, arm 1 28 95.4 ± 14.2 (89.9100.9) 94.6 ± 14.5 (88.9100.2) 0.10 0.8 ± 25 0.0035
||
FMD, arm 2 28 92.1 ± 11.2 (87.996.2) 87.9 ± 120 (83.592.4) 0.0003 4.1 ± 5.2
Fasting glucose (mg/dl)
Control diet, arm 1 41 88.1 ± 8.9 (85.390.9) 90.3 ± 9.7 (87.393.4) 0.14 2.2 ± 9.5 0.27
FMD, arm 2 36 89.7 ± 8.5 (86.592.1) 89.0 ± 8.0 (86.491.6) 0.87 0.8 ± 9.9
IGF-1 (ng/ml)
Control diet, arm 1 41 180.2 ± 84.5 (153.52,069) 188.9 ± 91.0 (160.2217.7) 0.14 8.7 ± 36.9 0.0017
||
FMD, arm 2 38 168.6 ± 69.1 (146.6190.5) 146.9 ± 62.3 (127.0166.7) 0.0063 21.7 ± 46.2
Systolic blood pressure (mmHg)
Control diet, arm 1 43 116.5 ± 12.3 (112.71,203) 115.8 ± 13.6 (111.6120.0) 0.60 0.7 ± 8.4 0.023
FMD, arm 2 38 118.0 ± 13.4 (113.71,222) 113.5 ± 13.2 (109.3117.7) <0.0001 4.5 ± 6.0
Diastolic blood pressure (mmHg)
Control diet, arm 1 43 75.5 ± 9.6 (72.578.5) 74.8 ± 10.0 (71.777.9) 0.45 0.7 ± 6.2 0.053
FMD, arm 2 38 75.7 ± 8.0 (73.278.3) 72.6 ± 8.7 (70.576.0) 0.0089 3.1 ± 4.7
Triglycerides (mg/dl)
Control diet, arm 1 37 100.5 ± 68.2 (77.7123.2) 101.5 ± 57.1 (82.5120.6) 0.85 1.0 ± 35.0 0.27
FMD, arm 2 30 83.0 ± 39.5 (69.196.9) 74.9 ± 37.6 (61.788.2) 0.19 8.1 ± 33.5
continued on next page
SCIENCE TRANSLATIONAL MEDICINE |RESEARCH ARTICLE
Wei et al., Sci. Transl. Med. 9, eaai8700 (2017) 15 February 2017 4of12
by guest on October 20, 2017http://stm.sciencemag.org/Downloaded from
and risk factors during the first FMD cycle (at day 5 of the FMD and
before refeeding; table S3) and after completion of three FMD cycles
(5 to 7 days after completing the third FMD cycle; table S2).
At the end of the first FMD cycle and before resuming the normal
diet, body weight (P< 0.0001), BMI (P< 0.0001), absolute lean body
mass (P< 0.0001), waist circumference (P< 0.0001), fasting glucose (P<
0.0001), IGF-1 (P< 0.0001), diastolic blood pressure (P< 0.0003), tri-
glycerides (P< 0.0001), and LDL (P< 0.0026) were significantly reduced
compared to baseline. In contrast, relative lean body mass (P=0.02),
b-hydroxybutyrate (P< 0.0001), and IGFBP-1 (P< 0.0001) were in-
creased. Both absolute and relative total body fat (P= 0.075 and P=
0.047, respectively), systolic blood pressure (P= 0.076), as well as CRP
(P= 0.75) were not significantly changed after completion of the first
FMD cycle compared to baseline (table S3). These results indicate that
subjects did follow the dietary changes imposed by the FMD and re-
sponded to them as anticipated.
In subjects who completed three FMD cycles (combining both
FMD arms) and who returned to the normal diet for 5 to 7 days, body
weight (P< 0.0001; n=71),BMI(P< 0.0001; n= 71), total body fat
(absolute, P< 0.0001; relative, P< 0.0001; n= 70), trunk fat (absolute,
P< 0.001; relative, P= 0.0002; n= 70), absolute lean body mass (P=
0.0001; n= 70), waist circumference (P< 0.0001; n= 52), IGF-1 (P<
0.0001; n= 69), systolic and diastolic blood pressure (P<0.0001and
P< 0.0004, respectively; n= 70), total cholesterol (P= 0.004; n= 55),
LDL (P< 0.0011; n=55),andHDL(
P=0
.02;n= 55) were signif-
icantly reduced, and relative lean body mass (P=0.0002;n= 70) was
increased. Fasting glucose (P=0.28;n=66),b-hydroxybutyrate (P=
0.23; n= 69), IGFBP-1 (P=0.84;n= 69), triglycerides (P=0.16;n=
55), and CRP (P= 0.052; n= 69) were not significantly changed
(table S2). In summary, the combined FMD groups from arms 1 and 2
confirmed that the FMD cycles promoted potent effects on many meta-
bolic markers and disease risk factors, which are maintained after
subjects return to their normal diet.
FMD effects stratified by baseline risk factor values: A post
hoc observational pre-post FMD comparison
Age-related physiological changes that lead to increased risk factors
occur before diseases can be diagnosed (20,21). We used the aggregated
FMD data of both study arms and performed a post hoc analysis of the
FMD effect on risk factors for CVD and metabolic syndrome, defined
as three of five of the following conditions: abdominal obesity, elevated
fasting glucose, elevated blood pressure, high serum triglycerides, and
low HDL cholesterol (1). We selected clinically relevant cutoffs and
compared normal and at-risk subjects for each risk factor: total choles-
terol>199mg/dlandLDLcholesterollevels >130 mg/dl are associated
with an increased risk for CVD (22), a fasting glucose >99 mg/dl indi-
cates impaired fasting glucose/prediabetes (23), and triglyceride
levels >100 mg/dl (24) as well as CRP >1 mg/liter are associated
with increased risk for CVD (25). For serum IGF-1, no clinically re-
levant risk level has been established, but a number of epidemiological
studies have associated IGF-1 levels above 200 ng/ml with various
cancers (17,26). We therefore compared the effect of FMD cycles
on subjects in the highest quartile of IGF-1 expression (>225 ng/ml)
with that on subjects with IGF-1 levels 225 ng/ml.
In a post hoc analysis, we tested how the changes in the FMD
normal and at-risk subgroups compared to those in the control diet
normal and at-risk subgroups, as defined by their baseline levels of var-
ious risk factors (Table 3). We observed a benefit of the FMD, but not in
the control arm, on BMI in all BMI subgroups (Pvalue for interaction =
0.03), although the FMD was particularly beneficial among subjects
Variable n
Baseline* CTRL: 3 months after baseline
Efficacy
(comparing D)
P
§
Mean ± SD (95% CI)
FMD: 5 days after third FMD cycle
Mean ± SD (95% CI) P
Difference: D
Total cholesterol (mg/dl)
Control diet, arm 1 37 195.9 ± 38.9 (182.92,089) 183.9 ± 35.2 (172.1195.6) 0.0015 12.0 ± 21.3 0.81
FMD, arm 2 30 175.3 ± 25.3 (166.41,842) 164.4 ± 23.4 (156.1172.6) 0.0012 10.9 ± 17.0
LDL cholesterol (mg/dl)
Control diet, arm 1 37 111.2 ± 35.6 (99.4123.1) 104.0 ± 31.8 (93.4114.6) 0.018 7.2 ± 17.7 0.50
FMD, arm 2 30 94.1 ± 23.0 (86.0102.2) 89.7 ± 22.8 (81.797.7) 0.13 4.4 ± 16.0
HDL cholesterol (mg/dl)
Control diet, arm 1 37 64.3 ± 16.1 (5.9.269.9) 59.3 ± 14.9 (54.364.3) 0.0002 5.3 ± 7.8 0.90
FMD, arm 2 30 64.8 ± 17.2 (58.670.6) 59.6 ± 12.8 (55.164.2) 0.0097 5.0 ± 10.0
C-reactive protein (mg/liter)
Control diet, arm 1 42 1.5 ± 1.9 (0.922.11) 1.9 ± 2.7 (1.072.75) 0.31 0.4 ± 2.5 0.27
FMD, arm 2 38 1.1 ± 1.3 (0.711.52) 1.0 ± 1.2 (0.611.37) 0.61 0.1 ± 1.5
*No significant differences at baseline between arm 1 and arm 2 (FMD), with exception of total cholesterol (P= 0.014) and LDL (P= 0.024). Pvalues
comparing within-group changes were calculated using paired two-tailed Studentsttest. Plus-minus values are means ± SD rounded to the nearest
tenth. §Between-arm comparison was calculated using two-tailed two-sample equal variance ttests. Using the Benjamini-Hochberg method for controlling
the FDR of 0.05. ||Pvalues indicate that the null hypothesis of no difference between the control diet (arm 1) to FMD (arm 2) can be rejected. ¶The BMI
is the weight in kilograms divided by the square of the height in meters. **Analyzed by DEXA.
SCIENCE TRANSLATIONAL MEDICINE |RESEARCH ARTICLE
Wei et al., Sci. Transl. Med. 9, eaai8700 (2017) 15 February 2017 5of12
by guest on October 20, 2017http://stm.sciencemag.org/Downloaded from
who were obese (BMI >30) at baseline. The FMD-dependent reduction
in IGF-1 was also larger in participants with baseline IGF-1 225 ng/ml
(Pvalue for interaction = 0.018).
Next,weevaluatedtheeffectsize,that is, efficacy in normal and at-
risk subjects (Table 4) in subjects stratified by risk factor. Subjects with
a BMI of greater than 30 (obese) experienced a greater reduction in
BMI by the end of the three FMD cycles than those with a BMI of less
than 25 (P= 0.011 between groups) and BMI of 25 to 30 (P= 0.0011
between groups). Systolic blood pressure was reduced by 2.4 ± 6.3 mmHg
in subjects with baseline systolic blood pressure 120 mmHg but by
6.7 ± 6.9 mmHg in subjects with systolic blood pressure >120 mmHg
(P= 0.013 between groups), and diastolic blood pressure was re-
duced by 1.5 ± 5.1 mmHg in subjects with diastolic blood pressure
80 mmHg but by 5.5 ± 6.4 mmHg in those with baseline levels above
80 mmHg (P= 0.01 between groups). Fasting glucose did not change
for participants with baseline levels 99 mg/dl but was reduced by 11.8
± 6.9 mg/dl in participants with baseline fasting glucose >99 mg/dl (P<
0.0001 between groups); notably, this reduction brought glucose in
these subjects within the healthy range. IGF-1 levels in subjects with
baseline levels above 225 ng/ml were reduced by 55.1 ± 45.6 ng/ml,
nearly four times more (P< 0.001 between groups) than the 14.1 ±
39.9 ng/ml reduction observed in participants with IGF-1 concentrations
below 225 ng/ml. Triglyceride levels were reduced more in participants
with baseline levels >100 mg/dl (P= 0.0035 between groups). Total
cholesterol was reduced significantly more in participants with total
cholesterol higher than 199 mg/dl at baseline (P= 0.015 between
groups). LDL was reduced by 14.9 ± 21.7 mg/dl in those with total
cholesterol baseline levels above 199 mg/dl but was not reduced by
Fig. 2. Change analysis of metabolic variables during the randomization. Effects on aging/disease markers and risk factors in all subjects who completed the
randomized analysis in either the control arm or the FMD arm (5 to 7 days after the third cycle of FMD). (A) Body weight, (B) BMI, (C) total body fat, (D) trunk fat, (E) lean
body mass, (F) waist circumference, (G) serum glucose level, (H) insulin-like growth factor 1, (I) systolic blood pressure, (J) diastolic blood pressure, (K) triglycerides, (L)total
cholesterol, (M)LDL,(N)HDL,and(O) CRP were measured in both cohorts as described. The Dchange represents a comparison to baseline. All data are means ± SD. Between-
arm comparisons were calculated using two-tailed two-sample equal variance ttests. For some of the 100 enrolled participants, the nurses were unable to collect all the
samples/measurements from all subjects. We therefore excluded subjects with incomplete measurements from a particular marker group (see Table 2 for details). Abs,
absolute; Rel, relative; BP, blood pressure.
SCIENCE TRANSLATIONAL MEDICINE |RESEARCH ARTICLE
Wei et al., Sci. Transl. Med. 9, eaai8700 (2017) 15 February 2017 6of12
by guest on October 20, 2017http://stm.sciencemag.org/Downloaded from
FMD cycles in normal-range subjects (P= 0.013 between groups).
There was no reduction (P= 0.094 between groups) in HDL levels
for those study participants with HDL levels below or above 50 mg/
dl at baseline. CRP was not reduced for subjects with levels below
1 mg/liter but was reduced by 1.6 ± 1.3 mg/liter and returned to
the normal levels in most subjects with baseline CRP higher than
1mg/liter(P= 0.0003 between groups). A graphical summary of these
data is presented in Fig. 3; before-after dot plots of individual subjects
in the control cohort as well as in normal and at-risk subjects in the
FMD cohort are presented in fig. S3.
ThisposthocanalysisindicatesthattheFMDhadmorepronounced
effects in at-risk participants than in those subjects with risk factor
values within the normal range, with the exception of HDL. Larger ran-
domized trials are necessary to confirm the results on the efficacy of the
FMD in the treatment of patients at risk for diseases.
Voluntary follow-up 3 months after FMD
We invited participants to return on a voluntary basis about 3 months
(actual mean follow-up time, 3.2 ± 1.3 months; n= 50) after their
third and final FMD cycle. In these subjects, the FMDs effects on
body weight, BMI, waist circumference, glucose (in at-risk subjects),
IGF-1, and systolic (in at-risk subjects) and diastolic blood pressure
persisted for at least 3 months after the final FMD cycle (table S4).
Subjects with low HDL levels at baseline displayed increased HDL
levels at the 3-month follow-up, whereas CRP levels remained signif-
icantly lower in study participants with baseline CRP levels above
1 mg/liter. Notably, some of the at-risk groups include only a few sub-
jects, and thus, larger studies are needed to establish long-term effects
of the FMD on disease risk factors.
These results indicate that some of the beneficial effects of multiple
cycles of the FMD may last for several months. Although subjects were
not advised to change their diet or exercise regimen after the FMD
cycles ended, we cannot rule out that some of the changes after the ad-
ditional 3 months may be a result of lifestyle changes such as healthier
diets and/or improved physical activity after the completion of this trial.
DISCUSSION
This randomized phase 2 trial indicates that three cycles of a 5-day FMD
per month are feasible, safe, and effective in reducing body weight, waist
circumference and BMI, absolute total body and trunk fat, systolic blood
pressure, as well as IGF-1. Metabolic markers such as fasting glucose,
triglycerides, and CRP, as well as total, HDL, and LDL cholesterol, which
were within the normal range at baseline, were not significantly affected
in the randomized comparison after three FMD cycles. After 3 months,
subjects from the control arm were crossed over to the FMD interven-
tion. Our post hoc analysis of the aggregated data from all 71 subjects
who completed three FMD cycles confirmed the effects of the FMD on
trunk and total body fat, blood pressure, and IGF-1. A post hoc analysis
also allowed us to analyze subjects with elevated risk factors or metabolic
markers associated with metabolic syndrome and age-related diseases,
such as high BMI, blood pressure, fasting glucose, triglycerides, CRP,
cholesterol, and IGF-1. The FMD had more pronounced effects on all
these markers in at-risk participants than in those subjects who had risk
factor values within the normal range. Some of these metabolic markers,
namely, CRP, systolic/diastolic blood pressure, and serum lipids, have
been proposed as markers of biological aging (27). However, other mar-
kers affected by the FMD, including IGF-1 and glucose, have been
strongly implicated in aging and age-related diseases (5,18,28).
Study participants were instructed not to alter their lifestyle for
the duration of the trial and were allowed to consume food of their
choice during the normal diet periods, that is, subjects were not placed
on a prespecified or calorie-restricted diet. We observed changes that
were both positive (total cholesterol and LDL) and negative (HDL) in
arm 1 subjects during the control diet period, potentially explained by
Table 3. Post hoc comparisons for changes in risk factors for age-related
diseases and conditions by baseline subgroups.
Subgroup
Group
differences
(FMD Control)
Mean (95% CI)
Within
subgroup
P
Interaction
P
BMI
<25 0.6 (1.2 to 0.05) 0.03 0.03
2530 0.8 (1.4 to 0.3) 0.003
>30 1.9 (2.6 to 1.1) 0.0009
Systolic blood pressure (mmHg)
<120 3.4 (7.2 to 0.5) 0.086 0.80
120 4.3 (10.4 to 1.8) 0.17
Diastolic blood pressure (mmHg)
<80 2.5 (5.3 to 0.3) 0.08 0.87
80 3.0 (8.2 to 2.3) 0.26
Fasting glucose (mg/dl)
<99 0.8 (5.2 to 3.6) 0.72 0.12
99 11.7 (25.0 to 1.5) 0.08
IGF-1 (ng/ml)
<225 18.7 (38.6 to 1.2) 0.065 0.018
225 70.9 (109.3 to 32.6) 0.0004
Triglycerides (mg/dl)
<100 4.6 (24.1 to 15.0) 0.64 0.38
100 19.1 (45.8 to 7.6) 0.16
Cholesterol (mg/dl)
Total, <199 1.8 (12.6 to 9.0) 0.73 0.88
Total, 199 0.2 (18.2 to 17.7) 0.98
LDL, <199 total
cholesterol
1.0 (8.8 to 10.8) 0.84 0.60
LDL, 199 total
Cholesterol
6.2 (11.2 to 23.6) 0.48
HDL, <50 1.2 (9.0 to 0.5) 0.75 0.70
HDL, 50 0.5 (4.2 to 5.2) 0.83
CRP (mg/liter)
<1 0.4 (1.5 to 8.7) 0.47 0.59
10.9 (2.4 to 0.6) 0.24
SCIENCE TRANSLATIONAL MEDICINE |RESEARCH ARTICLE
Wei et al., Sci. Transl. Med. 9, eaai8700 (2017) 15 February 2017 7of12
by guest on October 20, 2017http://stm.sciencemag.org/Downloaded from
dietary habit changes in anticipation for the FMD, despite no change
inweight,BMI,bodyfat,orleanmass.Similarly,thepersistenteffects
of the FMD observed 3 months after study completion may be
affected by changes in dietary habits and/or physical activity. The
composition of the diet tested in this trial was based on the FMD that
isknowntoextendhealthspaninmice.Similarlytothestudyinmice
(5), we expect the FMD effects to be mostly independent of an overall
caloric restriction, because both groups likely consumed similar levels
of calories per month: For example, estimating a 9200 kJ diet for each
of the 25 to 26 nonrestricted days and about 19,200 kJ kcal for the
Table 4. Post-hoc analysis of risk factors for age-related diseases and conditions in at-risk subjects who completed the FMD trial.
Variable
(at baseline) n
Baseline FMD: 5 days after third FMD Efficacy
(comparing D)P
Mean ± SD (95% CI) Mean ± SD (95% CI) P*D
BMI
§
<25 27 22.4 ± 1.7 (21.6823.03) 21.9 ± 1.6 (21.2422.52) 0.0014 0.5 ± 0.7 0.0020
||
2530 30 27.1 ± 1.4 (26.5827.60) 26.4 ± 1.6 (25.7626.99) <0.0001 0.7 ± 0.7 0.21
>30 14 34.4 ± 3.5 (32.3736.36) 33.0 ± 3.5 (30.9534.95) 0.0001 1.4 ± 1.0 0.011
Systolic blood pressure (mmHg)
<120 49 110.3 ± 6.9 (108.5112.4) 107.9 ± 7.7 (105. 7110.1) 0.0089 2.4 ± 6.3 0.013
>120 21 133.4 ± 9.0 (129.3137.5) 126.7 ± 11.3 (121.5131.8) 0.0002 6.7 ± 6.9
Diastolic blood pressure (mmHg)
<80 53 71.8 ± 5.0 (70.4373.21) 70.3 ± 5.9 (68.6471.89) 0.032 1.5 ± 5.1 0.010
>80 17 87.7 ± 6.9 (84.2091.27) 82.2 ± 10.0 (77.0987.38) 0.0026 5.5 ± 6.4
Fasting glucose (mg/dl)
<99 53 87.7 ± 6.4 (85.9789.50) 89.0 ± 8.2 (86. 7591.25) 0.29 1.3 ± 8.6 <0.0001
>99 13 104.2 ± 4.4 (101.5106.9) 92.4 ± 8.2 (87.4297.35) <0.0001 11.8 ± 6.9
IGF-1 (ng/ml)
<225 52 145.1 ± 38.4 (134.4155.8) 131.0 ± 46.8 (118.0144.0) 0.014 14.1 ± 39.9 0.00088
>225 17 286.6 ± 47.0 (261.6311.7) 231.5 ± 64.7 (197.0265.9) 0.0002 55.1 ± 45.6
Triglycerides (mg/dl)
<100 34 65.8 ± 18.4 (59.4572.09) 69.5 ± 28.9 (59.5879.45) 0.33 3.7 ± 22.3 0.0035
>100 21 149.3 ± 41.2 (127.9160.8) 123.7 ± 52.7 (97.54 148.0) 0.058 25.6 ± 50.2
Cholesterol (mg/dl)
Total, <199 40 170.3 ± 18.3 (164.4176.2) 164.2 ± 20.2 (157. 7170.6) 0.016 6.1 ± 15.3 0.015
Total, >199 15 228.1 ± 23.0 (215.4240.9) 208.2 ± 26.3 (193.6222.8) 0.0088 19.9 ± 25.4
LDL, <199 total
cholesterol
40 91.4 ± 20.2 (85.0 97.73) 88.8 ± 21.0 (82.1395.38) 0.22 2.6 ± 13.4 0.013
LDL, >199 total
cholesterol
15 141.1 ± 29.1 (125.0157.2) 126.2 ± 24.7 (112.2139.9) 0.018 14.9 ± 21.7
HDL, <50 17 44.1 ± 4.1 (41.9446.17) 44.2 ± 4.6 (41.8746.6) 0.82 0.1 ± 3.1 0.094
HDL, >50 38 69.6 ± 14.1 (65.0674.17) 65.6 ± 11.6 (61.8169.32) 0.015 4.0 ± 10.0
CRP (mg/liter)
<1 43 0.4 ± 0.3 (0.280.46) 0.6 ± 0.9 (0.270.85) 0.20 0.2 ± 1.0 0.0003
>1 26 3.3 ± 2.8 (2.224.44) 1.6 ± 1.2 (1.062.15) 0.0085 1.7 ± 3.1
*Pvalues comparing within-group changes were calculated using paired two-tailed Studentsttest. Plus-minus values are means ± SD rounded to the
nearest tenth. Between-group comparison was calculated using two-tailed two-sample equal variance ttests. §The BMI is the weight in kilograms
divided by the square of the height in meters. ||One-way analysis of variance for the BMI groups.
SCIENCE TRANSLATIONAL MEDICINE |RESEARCH ARTICLE
Wei et al., Sci. Transl. Med. 9, eaai8700 (2017) 15 February 2017 8of12
by guest on October 20, 2017http://stm.sciencemag.org/Downloaded from
5 days of FMD per month, the between-group difference in consumed
calories is expected to be about 10%. In addition, this difference may
be overestimated because it is likely that subjects have an elevated calorie
intake after the FMD period, as we have shown for mice (5). Day 1 of
the FMD supplies ~4600 kJ (11% protein, 46% fat, and 43% carbohy-
drate), whereas days 2 to 5 provide ~3000 kJ per day (9% protein, 44%
fat, and 47% carbohydrate); thus, fat and complex carbohydrates are
the major source of calories in the FMD.
Our studies in cells and mice indicate that both glucose and pro-
teins interfere with the protective and regenerative effects of fasting
(29). Because our previous data indicate that dietary composition
can be equally or more important than calorie restriction, it will be
important to test the effects of a similarly restricted diet that provides
higher proportions of carbohydrates and/or proteins. It remains to be
established whether part of the effects of FMD that we observed are
mediated by stem cellbased regeneration or rejuvenation, as indicated
by our mouse studies (5).
The FMD-induced reduction in serum glucose and IGF-1 is of
interest given their role in pro-aging signaling pathways and cancer
(17,3033). In addition to a marker for insulin resistance and a
metabolicinputforcancercells,glucose is associated with cellular sen-
sitization to toxins and senescence (28,34,35). Growth hormone re-
ceptor deficiency, resulting in reduced IGF-1 levels, is associated with
a major reduction in pro-aging signaling, cancer, and diabetes in
humans (18). The observed reduction in IGF-1 in our study, but not
either after 6 months of intermittent energy restriction (IER) (36)or
after 6 years of 20% caloric restriction (37), is probably related to the
long-lasting effects of the low protein/amino acid content of the FMD
(average 5 days of FMD; 11.5% versus 21% IER or 24% long-term CR). In
fact, twenty eight vegans consuming a moderately protein-restricted (10%)
diet for about 5 years had reduced IGF-1 levels compared to a group that
consumed a chronic 20% calorie-restricted diet (37). We also previously
showed that reduced IGF-1 levels and reduced cancer risk were asso-
ciated with low protein consumption in participants of the National
Health and Nutrition Examination Survey cohort (17). Specific ingre-
dients, for example, high levels of unsaturated fats and micronutrients,
may also positively contribute to some of the beneficial effects of the
FMD.
Note that 25% of the subjects who tested the FMD dropped out
of the trial, whereas 10% of the participants opted out of the control
arm. This indicates that, despite our efforts to reduce the burden of
low-calorie/protein diets, adherence to this dietary regimen requires
committed study participants. Further, compared to the control diet
arm, the FMD arm imposed an additional daylong visit to the clinic,
which may have contributed to reduced compliance. Compliance with
prescribed therapies, even placebo, may be an identifiable marker for
an overall healthy behavior of study participants (38). Thus, this kind
of volunteer, who is observing a benefit and thus not dropping out,
could introduce potential bias into the analysis of our trial. The overall
comparability at baseline between the control and both FMD arms, as
wellasthecomparableresponsetotheFMD(arm2andarm1after
crossover) suggests no major differences in response for those subjects
who completed the trial. Further, those subjects who dropped out of
this trial were not different in age or BMI compared to those who
completed the trial. It remains to be established why we experienced
a gender difference (82% of dropouts were female). The 25% overall
Fig. 3. Post hoc analysis of metabolic variables in subgroups identified by severity of risk factors. Subjects from both study arms who completed three FMD
cycles were post hoc stratified on the basis of being in either normal-risk or at-risk subgroups for factors associated with age-related diseases and conditions. The Dchange
shown represents comparisons to baseline. All data are means ± SD. Between-arm comparisons were calculated using two-tailed two-sample equal variance ttests. One-way
analysis of variance was used for the BMI groups (see Table 4 for details).
SCIENCE TRANSLATIONAL MEDICINE |RESEARCH ARTICLE
Wei et al., Sci. Transl. Med. 9, eaai8700 (2017) 15 February 2017 9of12
by guest on October 20, 2017http://stm.sciencemag.org/Downloaded from
dropout rate (all causes) of study participants before the completion of
the third FMD cycle is in the range observed in other trials aimed at
evaluating dietary interventions in adults. For example, 16 weeks of
dieting in combination with physical exercise yielded a discontinuation
rate of about 30% (39), and a hypocaloric diet in 28 overweight/obese
womenresultedinadropoutrateof40%after6months(40). In a trial
assessing the effect of intermittent energy/carbohydrate restriction and
daily energy restriction on weight loss and metabolic disease risk markers
in overweight women, Harvie et al. reported a 23% dropout rate (41).
Nonetheless, there are limitations of our trial that should be considered:
(i) A relatively small number of subjects in the randomized comparison;
(ii) despite providing nourishment and calories for the duration of the
FMD, we experienced a higher dropout rate during the FMD interven-
tion than in the control arm; (iii) the findings that the FMD reduced
metabolic markers more effectively in at-risk subjects are based on a
non-randomized post hoc analysis of the individual factors in generally
healthy participants, and thus, further evaluation in subjects with diag-
nosed disease is needed.
Otherlessrestrictivedietssuchasthoserequiringaverylowcalorie
intake twice a week would impose 8 days per month of a severe restric-
tion compared to the 5 days per month or per several months of a less
restrictive intervention tested here (41). However, an advantage of these
diets is that they may not require as much medical supervision as the
longer FMD. FMDs or any type of prolonged fasting interventions last-
ing more than 12 hours, particularly those lasting several days, require
supervision, preferably from a health care professional familiar with pro-
longed fasting. Although our results suggestthatcyclesoftheplant-based
FMD might be safe for elderly individuals, additional studies are neces-
sary to determine its safety for subjects who are 70 years and older.
In summary, and with the limitations outlined above, these results
indicate that the periodic FMD cycles are effective in improving the
levels of an array of metabolic markers/risk factors associated with
poor health and aging and with multiple age-related diseases. As sug-
gested by preclinical studies, interventions that promote longevity
should also extend healthspan. Further investigations in larger clinical
trials focused on subjects with diagnosed metabolic syndrome, diabe-
tes, and CVDs as well as subjects at high risk for developing cancer
and other age-related diseases are needed.
MATERIALS AND METHODS
Subjects
One-hundred participants without a diagnosed medical condition in the
previous 6 months were enrolled (ClinicalTrials.gov; NCT02158897).
All participants provided written informed consent, and the University
of Southern California (USC) Institutional Review Board (IRB) ap-
proved the protocol. Recruitment of subjects was based on fliers, the
ClinicalTrials.gov and usc.com websites, and/or word of mouth. Be-
cause this was a dietary intervention study, it was not possible for par-
ticipants or all study personnel to be blinded to group assignment.
However, study personnel involved in data collection and specimen
analysis were blinded to group assignments.
Study design
Flow of participant enrollment and participation was prepared
following the CONSORT standards for randomized clinical trials
with crossover design. All data were collected at the USC Diabetes
and Obesity Research Institute. Subjects were recruited from April 2013
to July 2015 under protocols approved by the USC IRB (HS-12-00391)
based on established inclusion (generally healthy adult volunteers and
18 to 70 years of age; BMI, 18.5 and up) and exclusion [any major med-
ical condition or chronic diseases, mental illness, drug dependency, hor-
mone replacement therapy (dehydroepiandrosterone, estrogen, thyroid,
and testosterone), pregnant or nursing female, special dietary require-
ments or food allergies, alcohol dependency, and medications known to
affect body weight] criteria. Intention to treat analysis was performed by
including all available observations. Eligible participants were randomly
assigned using a random-number generator to either arm 1 or arm 2 of
the study. All participants completed a health habits questionnaire. Pre-
specified outcome measures included safety and feasibility, and evalua-
tion of changes in metabolic risk factors for diabetes and CVD and
metabolic markers associated with age-related diseases and mortality;
these outcomes were measured at baseline during and after completion
of the intervention. Laboratory examinations included height, weight,
body composition (including total and trunk body fat, soft lean tissue,
and bone mineral content) measured by dual-energy x-ray absorptiom-
etry (DEXA), oscillometric blood pressure measurements, and
overnight fasting blood draw through venipuncture.
Arm 1 (control).
Participants completed anthropometric measurements and blood collec-
tion at enrollment and after 3 months to provide an estimate of non
diet-related changes (Fig. 1). Participants were instructed to maintain
their regular eating habits. After 3 months, subjects were crossed over
to the experimental FMD group (Fig. 1).
Arm 2 (FMD).
Participants were instructed to consume the FMD, which was provided
in a box, for 5 continuous days, and to return to their normal diet after
completion until the next cycle that was initiated about 25 days later.
Participants completed three cycles of this 5-day FMD (Fig. 1). Partic-
ipants completed baseline and follow-up examinations at the end of the
first FMD (before resuming normal diet to measure the acute FMD
effects) and after a washout period of 5 to 7 days of normal caloric
intake after the third FMD cycle. An optional follow-up assessment
3 months after the third FMD cycle was offered.
Experimental FMD
The FMD is a plant-based diet designed to attain fasting-like effects on
the serum levels of IGF-1, IGFBP-1, glucose, and ketone bodies while
providing both macro- and micronutrients to minimize the burden of
fasting and adverse effects (5). Day 1 of the FMD supplies ~4600 kJ
(11% protein, 46% fat, and 43% carbohydrate), whereas days 2 to 5 pro-
vide ~3000 kJ (9% protein, 44% fat, and 47% carbohydrate) per day.
The FMD comprises proprietary formulations belonging to USC and
L-Nutra (www.prolonfmd.com) of vegetable-based soups, energy bars,
energy drinks, chip snacks, tea, and a supplement providing high levels
of minerals, vitamins, and essential fattyacids(fig.S4).Allitemstobe
consumed per day were individually boxed to allow the subjects to
choose when to eat while avoiding accidentally consuming components
of the following day.
Common Terminology Criteria for Adverse Events
Study participants were asked about adverse events at each study visit;
events were graded according to the general CTCAE guidelines (see
the Supplementary Materials for details).
Blood tests and serum markers
Complete metabolic and lipid panels (overnight fasting) were completed
at the Clinical Laboratories at the Keck Medical Center of USC and
SCIENCE TRANSLATIONAL MEDICINE |RESEARCH ARTICLE
Wei et al., Sci. Transl. Med. 9, eaai8700 (2017) 15 February 2017 10 of 12
by guest on October 20, 2017http://stm.sciencemag.org/Downloaded from
analyzed immediately after the blood draw of each visit (see the Sup-
plementary Materials for details).
Statistical analysis
The primary comparisons of randomized groups involved changes in
outcomes observed in the control period of arm 1 versus the changes
observed in the FMD group (arm 2) after completion of three FMD
cycles. Secondary observational analyses involved (i) comparing the
FMD effects in arm 2 (randomized to FMD) versus arm 1 (receiving
FMD after completion of the randomized control period) and (ii)
summarizing the changes for arms 1 and 2 combined after comple-
tion of the first and third FMD cycles. Changes from baseline were
normally distributed. Comparison of changes from baseline within
the treatment arms was performed using paired two-tailed Students
ttests, and Pvalues <0.05 were considered significant. The between-
arm comparison of treatment changes from baseline was performed
using two-tailed two-sample equal variance ttests, and Pvalues <0.05
were considered significant. To control for multiple testing, we used
the Benjamini-Hochberg FDR method. All reported Pvalues are
nominal two-sided Pvalues; those that met the FDR criteria and re-
mained significantat P< 0.05 are indicated with an asterisk.
M.W. generated the random allocation sequence and enrolled and
assigned participants to interventions. M.W. was not involved in out-
come assessments. For this initial randomized trial, the sample size of
100 total subjects was based on detection of a 25% reduction in mean
IGF-1, with a two-sided aof 0.05 and 70% power. The estimated con-
trol group mean (SD) IGF-1 of 194 (97) used published data on males
and females aged 26 to 40 years (42). Statistical analyses were per-
formed on deidentified data. Baseline information and changes from
baseline were summarized using means ± SDs for subjects randomized
to the control (arm 1, n=48)andthedietgroup(arm2,n=52).All
subjects are included in the arm assigned regardless of treatment adher-
ence (intention to treat); no attempt was made to impute missing values
(primarily because if data after completion of the third FMD cycle were not
available, then other measurement time points were usually unavailable).
In post hoc subgroup analyses, we compared FMD-control group
differences over the randomized trial period (three FMD cycles versus
control) within high/lower-risk subgroups and tested whether those
treatment effects differed in the higher-risk versus lower-risk groups.
This subgroup analysis was completed using analysis of variance, with
main effects of treatment (FMD and control) and risk group (high and
low); the interaction of treatment-by-risk group tested whether the
randomized FMD effect differed in high-risk versus low-risk groups.
In observational analyses of the pre-post FMD changes combining the
two treatment arms, pre-post changes in markers within risk subgroups
were tested using paired ttest; pre-post changes over risk subgroups
were compared using two-sample ttest or analysis of variance.
SUPPLEMENTARY MATERIALS
www.sciencetranslationalmedicine.org/cgi/content/full/9/377/eaai8700/DC1
Materials and Methods
Table S1. Complete metabolic panel.
Table S2. Arm-specific markers of adherence and changes in risk factors, including arm 1 after
crossover to FMD, and summary of FMD arms 1 and 2.
Table S3. Changes in risk factors and metabolic markers of adherence after the first FMD.
Table S4. Changesin risk factors and metabolic markers of adherence 3 months after intervention.
Fig. S1. Subject self-reported adverse effects based on CTCAE.
Fig. S2. Comparison of participants who completed the trial versus dropouts.
Fig. S3. Baseline to 3 months before/after comparison of individual subjects in the control
cohort and all subjects who completed the FMD.
Fig. S4. Nutritional information of the FMD.
CONSORT checklist
Trial protocols
REFERENCES AND NOTES
1. K.G.Alberti,R.H.Eckel,S.M.Grundy,P.Z.Zimmet,J.I.Cleeman,K.A.Donato,
J.-C. Fruchart, W. P. T. James, C. M. Loria, S. C. Smith Jr.; International Diabetes
Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and
Blood Institute; American Heart Association; World Heart Federation; International
Atherosclerosis Society; International Association for the Study of Obesity,
Harmonizing the metabolic syndrome: A joint interim statement of the International
Diabetes Federation Task Force on Epidemiology and Prevention; National Heart,
Lung, and Blood Institute; American Heart Association; World Heart Federation;
International Atherosclerosis Society; and International Association for the Study of
Obesity. Circulation 120,16401645 (2009).
2. E. S. Ford, W. H. Giles, W. H. Dietz, Prevalence of the metabolic syndrome among
US adults: Findings from the third National Health and Nutrition Examination Survey.
JAMA 287, 356359 (2002).
3. A.S.Gami,B.J.Witt,D.E.Howard,P.J.Erwin,L.A.Gami,V.K.Somers,V.M.Montori,
Metabolic syndrome and risk of incident cardiovascular events and death: A
systematic review and meta-analysis of longitudinal studies. J. Am. Coll. Cardiol. 49,
403414 (2007).
4. I. Y. Choi, L. Piccio, P. Childress, B. Bollman, A. Ghosh, S. Brandhorst, J. Suarez,
A. Michalsen, A. H. Cross, T. E. Morgan, M. Wei, F. Paul, M. Bock, V. D. Longo, A diet
mimicking fasting promotes regeneration and reduces autoimmunity and multiple
sclerosis symptoms. Cell Rep. 15, 21362146 (2016).
5. S. Brandhorst, I. Y. Choi, M. Wei, C. W. Cheng, S. Sedrakyan, G. Navarrete, L. Dubeau,
L. P. Yap, R. Park, M. Vinciguerra, S. D. Biase, H. Mirzaei, M. G. Mirisola, P. Childress, L. Ji,
S. Groshen, F. Penna, P. Odetti, L. Perin, P. S. Conti, Y. Ikeno, B. K. Kennedy, P. Cohen,
T. E. Morgan, T. B. Dorff, V. D. Longo, A periodic diet that mimics fasting promotes
multi-system regeneration, enhanced cognitive performance, and healthspan. Cell Metab.
22,8699 (2015).
6. V. D. Longo, S. Panda, Fasting, circadian rhythms, and time-restricted feeding in healthy
lifespan. Cell Metab. 23, 10481059 (2016).
7. A. J. Bruce-Keller, G. Umberger, R. McFall, M. P. Mattson, Food restriction reduces
brain damage and improves behavioral outcome following excitotoxic and metabolic
insults. Ann. Neurol. 45,815 (1999).
8. A. L. Hartman, J. E. Rubenstein, E. H. Kossoff, Intermittent fasting: A newhistorical
strategy for controlling seizures? Epilepsy Res. 104, 275279 (2013).
9. H. Müller, F. W. de Toledo, K. L. Resch, Fasting followed by vegetarian diet in patients with
rheumatoid arthritis: A systematic review. Scand. J. Rheumatol. 30,110 (2001).
10. V. D. Longo, M. P. Mattson, Fasting: Molecular mechanisms and clinical applications.
Cell Metab. 19, 181192 (2014).
11. C.-W. Cheng, G. B. Adams, L. Perin, M. Wei, X. Zhou, B. S. Lam, S. D. Sacco, M. Mirisola,
D. I. Quinn, T. B. Dorff, J. J. Kopchick, V. D. Longo, Prolonged fasting reduces IGF-1/PKA
to promote hematopoietic-stem-cell-based regeneration and reverse immunosuppression.
Cell Stem Cell 14,810823 (2014).
12. W.E.Sonntag,D.Lynch,W.T.Cefalu,R.L.Ingram,S.A.Bennett,P.L.Thornton,
A. S. Khan, Pleiotropic effects of growth hormone and insulin-like growth factor (IGF)-1 on
biological aging: Inferences from moderate caloric-restricted animals. J. Gerontol. A Biol.
Sci. Med. Sci. 54, B521B538 (1999).
13. Y. Ikeno, R. T. Bronson, G. B. Hubbard, S. Lee, A. Bartke, Delayed occurrence of fatal
neoplastic diseases in Ames dwarf mice: Correlation to extended longevity. J. Gerontol.
A Biol. Sci. Med. Sci. 58, 291296 (2003).
14. K. Flurkey, J. Papaconstantinou, R. A. Miller, D. E. Harrison, Lifespan extension and
delayed immune and collagen aging in mutant mice with defects in growth hormone
production. Proc. Natl. Acad. Sci. U.S.A. 98, 67366741 (2001).
15. S. E. Dunn, F. W. Kari, J. French, J. R. Leininger, G. Travlos, R. Wilson, J. C. Barrett, Dietary
restriction reduces insulin-like growth factor I levels, which modulates apoptosis, cell
proliferation, and tumor progression in p53-deficient mice. Cancer Res. 57, 46674672
(1997).
16. M. S. Bonkowski, F. P. Dominici, O. Arum, J. S. Rocha, K. A. Al Regaiey, R. Westbrook,
A. Spong, J. Panici, M. M. Masternak, J. J. Kopchick, A. Bartke, Disruption of growth
hormone receptor prevents calorie restriction from improving insulin action and
longevity. PLOS ONE 4, e4567 (2009).
17. M. E. Levine, J. A. Suarez, S. Brandhorst, P. Balasubramanian, C.-W. Cheng, F. Madia,
L. Fontana, M. G. Mirisola, J. Guevara-Aguirre, J. Wan, G. Passarino, B. K. Kennedy, M. Wei,
P. Cohen, E. M. Crimmins, V. D. Longo, Low protein intake is associated with a major
reduction in IGF-1, cancer, and overall mortality in the 65 and younger but not older
population. Cell Metab. 19, 407417 (2014).
SCIENCE TRANSLATIONAL MEDICINE |RESEARCH ARTICLE
Wei et al., Sci. Transl. Med. 9, eaai8700 (2017) 15 February 2017 11 of 12
by guest on October 20, 2017http://stm.sciencemag.org/Downloaded from
18. J. Guevara-Aguirre, P. Balasubramanian, M. Guevara-Aguirre, M. Wei, F. Madia,
C.-W. Cheng, D. Hwang, A. Martin-Montalvo, J. Saavedra, S. Ingles, R. de Cabo,
P. Cohe n, V. D. Longo, Growth hormone receptor deficiency is associated with a major
reduction in pro-aging signaling, cancer, and diabetes in humans. Sci. Transl. Med. 3,
70ra13 (2011).
19. www.census.gov
20. M. W. Gillman, Developmental origins of health and disease. N. Engl. J. Med. 353,
18481850 (2005).
21. L. Fontana, B. K. Kennedy, V. D. Longo, D. Seals, S. Melov, Medical research: Treat ageing.
Nature 511, 405407 (2014).
22. M. Nayor, R. S. Vasan, Recent update to the US cholesterol treatment guidelines:
A comparison with international guidelines. Circulation 133, 17951806 (2016).
23. American Diabetes Association, Diagnosis and classification of diabetes mellitus.
Diabetes Care 37 (suppl. 1), S81S90 (2014).
24. M. Miller, N. J. Stone, C. Ballantyne, V. Bittner, M. H. Criqui, H. N. Ginsberg, A. C. Goldberg,
W. J. Howard, M. S. Jacobson, P. M. Kris-Etherton, T. A. Lennie, M. Levi, T. Mazzone,
S. Pennathur, Triglycerides and cardiovascular disease: A scientific statement from the
American Heart Association. Circulation 123, 22922333 (2011).
25. T. A. Pearson, G. A. Mensah, R. W. Alexander, J. L. Anderson, R. O. Cannon, M. Criqui,
Y. Y. Fadl, S. P. Fortmann, Y. Hong, G. L. Myers, N. Rifai, S. C. Smith, K. Taubert, R. P. Tracy,
F. Vinicor; Centers for Disease Control and Prevention, American Heart Association,
Markers of inflammation and cardiovascular disease: Application to clinical and
public health practice: A statement for healthcare professionals from the Centers
for Disease Control and Prevention and the American Heart Association. Circulation
107,499511 (2003).
26. M. N. Pollack, Insulin, insulin-like growth factors, insulin resistance, and neoplasia. Am. J.
Clin. Nutr. 86, s820s822 (2007).
27. M. E. Levine, Modeling the rate of senescence: Can estimated biological age predict
mortality more accurately than chronological age? J. Gerontol. A Biol. Sci. Med. Sci. 68,
667674 (2013).
28. L. Fontana, L. Partridge, V. D. Longo, Extending healthy life spanFrom yeast to humans.
Science 328, 321326 (2010).
29. S. Brandhorst, M. Wei, S. Hwang, T. E. Morgan, V. D. Longo, Short-term calorie and protein
restriction provide partial protection from chemotoxicity but do not delay glioma
progression. Exp. Gerontol. 48, 11201128 (2013).
30. A. G. Renehan, M. Zwahlen, C. Minder, S. T. ODwyer, S. M. Shalet, M. Egger, Insulin-like
growth factor (IGF)-I, IGF binding protein-3, and cancer risk: Systematic review and
meta-regression analysis. Lancet 363, 13461353 (2004).
31. J. M. Chan, M. J. Stampfer, J. Ma, P. Gann, J. M. Gaziano, M. Pollak, E. Giovannucci,
Insulin-like growth factor-I (IGF-I) and IGF binding protein-3 as predictors of
advanced-stage prostate cancer. J. Natl. Cancer Inst. 94, 10991106 (2002).
32. N.E.Allen,A.W.Roddam,D.S.Allen,I.S.Fentiman,I.dosSantosSilva,J.Peto,
J. M. P. Holly, T. J. Key, A prospective study of serum insulin-like growth factor-I
(IGF-I), IGF-II, IGF-binding protein-3 and breast cancer risk. Br. J. Cancer 92,
12831287 (2005).
33. O. Fletcher, L. Gibson, N. Johnson, D. R. Altmann, J. M. P. Holly, A. Ashworth, J. Peto,
I. dos Santos Silva, Polymorphisms and circulating levels in the insulin-like growth factor
system and risk of breast cancer: A systematic review. Cancer Epidemiol. Biomarkers Prev.
14,219 (2005).
34. K.Rapp,J.Schroeder,J.Klenk,H.Ulmer,H.Concin,G.Diem,W.Oberaigner,
S. K. Weiland, Fasting blood glucose and cancer risk in a cohort of more than 140,000
adults in Austria. Diabetologia 49, 945952 (2006).
35. T. Stocks, K. Rapp, T. Bjørge, J. Manjer, H. Ulmer, R. Selmer, A. Lukanova, D. Johansen,
H. Concin, S. Tretli, G. Hallmans, H. Jonsson, P. Stattin, Blood glucose and risk of incident
and fatal cancer in the metabolic syndrome and cancer project (Me-Can): Analysis of
six prospective cohorts. PLOS Med. 6, e1000201 (2009).
36. M. N. Harvie, M. Pegington, M. P. Mattson, J. Frystyk, B. Dillon, G. Evans, J. Cuzick,
S. A. Jebb, B. Martin, R. G. Cutler, T. G. Son, S. Maudsley, O. D. Carlson, J. M. Egan,
A. Flyvbjerg, A. Howell, The effects of intermittent or continuous energy restriction on
weight loss and metabolic disease risk markers: A randomized trial in young overweight
women. Int. J. Obes. 35, 714727 (2011).
37. L. Fontana, E. P. Weiss, D. T. Villareal, S. Klein, J. O. Holloszy, Long-term effects of calorie or
protein restriction on serum IGF-1 and IGFBP-3 concentration in humans. Aging Cell 7,
681687 (2008).
38. R. D. Hays, R. L. Kravitz, R. M. Mazel, C. D. Sherbourne, M. R. DiMatteo, W. H. Rogers,
S. Greenfield, The impact of patient adherence on health outcomes for patients with
chronic disease in the medical outcomes study. J. Behav. Med. 17, 347360 (1994).
39. T. P. Wycherley, M. Noakes, P. M. Clifton, X. Cleanthous, J. B. Keogh, G. D. Brinkworth,
A high-protein diet with resistance exercise training improves weight loss and body
composition in overweight and obese patients with type 2 diabetes. Diabetes Care 33,
969976 (2010).
40. D. Florakis, E. Diamanti-Kandarakis, I. Katsikis, G. P. Nassis, A. Karkanaki, N. Georgopoulos,
D. Panidis, Effect of hypocaloric diet plus sibutramine treatment on hormonal and
metabolic features in overweight and obese women with polycystic ovary syndrome:
A randomized, 24-week study. Int. J. Obes. 32, 692699 (2008).
41. M. Harvie, C. Wright, M. Pegington, D. McMullan, E. Mitchell, B. Martin, R. G. Cutler,
G. Evans, S. Whiteside, S. Maudsley, S. Camandola, R. Wang, O. D. Carlson, J. M. Egan,
M. P. Mattson, A. Howell, The effect of intermittent energy and carbohydrate restriction v.
daily energy restriction on weight loss and metabolic disease risk markers in overweight
women. Br. J. Nutr. 110, 15341547 (2013).
42. G. Brabant, A. von zur Mühlen, C. Wüster, M. B. Ranke, J. Kratzsch, W. Kiess, J.-M. Ketelslegers,
L. Wilhelmsen, L. Hulthén, B. Saller, A. Mattsson, J. Wilde, R. Schemer, P. Kann, Serum
insulin-like growth factor I reference values for an automated chemiluminescence
immunoassay system: Results from a multicenter study. Horm. Res. 60,5360 (2003).
Acknowledgments: We thank Y. Guan for help in data curation and acknowledge the
technical expertise of R. Buono (USC Leonard Davis School of Gerontology). Funding: Funding
was provided by the USC Edna Jones chair fund to V.D.L. W.J.M.s contributions were provided
through the Southern California Clinical and Translational Science Institute supported by
NIH UL1TR001855. Author contributions: V.D.L., M.W., T.E.M., and T.D. designed the clinical
trial. M.W., K.H., and T.D. supervised the clinical trial. J.B. and M.S. contributed to patient
recruitment and supervision. S.B., H.M., C.W.C., E.G., S.G., and W.J.M. performed data analysis.
S.B. and V.D.L. wrote the paper. All authors discussed the r esults and commente d on the
manuscript. Co mpeting intere sts: The experimen tal FMD was provided by L-Nutra Inc. The
funding sources had no involvement in study des ign; collection, analysis, and interpretation
of data; writing of t he report; or decisi on to submit the article for publicati on. The USC has
licensed intell ectual property to L-Nutra that is under stu dy in this research.
As part of this license agreeme nt, the University has the potential to receive royalty
payments from L-Nutra. V.D.L. a nd T.E.M., who have equity interes t in L-Nutra, did no t
participate in the collection and analysis of the data. One-hundred percent of V.D.L.s equity
will be assigne d to the nonprofit foundation Create Cures. U.S. pa tents 20140227 373A1
(Methods and diets to protec t against chemot oxic ity and age related illnesses)and
20140112909A1 (Methods and formulations promoting tissue regeneration, longevity and
healthspan) related to the work described here have been filed. Data and materialsavailability:
Use of the FMD may require a material transfer agreement.
Submitted 27 April 2016
Resubmitted 23 August 2016
Accepted 20 December 2016
Published 15 February 2017
10.1126/scitranslmed.aai8700
Citation: M. Wei, S. Brandhorst, M. Shelehchi, H. Mirzaei, C. W. Cheng, J. Budniak, S. Groshen,
W. J. Mack, E. Guen, S. Di Biase, P. Cohen, T. E. Morgan, T. Dorff, K. Hong, A. Michalsen,
A. Laviano, V. D. Longo, Fasting-mimicking diet and markers/risk factors for aging, diabetes,
cancer, and cardiovascular disease. Sci. Transl. Med. 9, eaai8700 (2017).
SCIENCE TRANSLATIONAL MEDICINE |RESEARCH ARTICLE
Wei et al., Sci. Transl. Med. 9, eaai8700 (2017) 15 February 2017 12 of 12
by guest on October 20, 2017http://stm.sciencemag.org/Downloaded from
cardiovascular disease
Fasting-mimicking diet and markers/risk factors for aging, diabetes, cancer, and
Michalsen, Alessandro Laviano and Valter D. Longo
Wendy J. Mack, Esra Guen, Stefano Di Biase, Pinchas Cohen, Todd E. Morgan, Tanya Dorff, Kurt Hong, Andreas
Min Wei, Sebastian Brandhorst, Mahshid Shelehchi, Hamed Mirzaei, Chia Wei Cheng, Julia Budniak, Susan Groshen,
DOI: 10.1126/scitranslmed.aai8700
, eaai8700.9Sci Transl Med
healthy metabolic system.
study is needed to replicate these results, but they raise the possibility that fasting may be a practical road to a
effects were generally larger in the subjects who were at greater risk of disease at the start of the study. A larger
decreased BMI, glucose, triglycerides, cholesterol, and C-reactive protein (a marker for inflammation). These
been implicated in aging and disease. A post hoc analysis replicated these results and also showed that fasting
diet reduced body weight and body fat, lowered blood pressure, and decreased the hormone IGF-1, which has
months or maintained their normal diet for 3 months and then switched to the fasting schedule. The fasting-like
studied 71 people who either consumed a fasting-mimicking diet for 5 days each month for 3et al.as well, Wei
Mice that fast periodically are healthier, metabolically speaking. To explore whether fasting can help people
Fasting: More than a fad
ARTICLE TOOLS http://stm.sciencemag.org/content/9/377/eaai8700
MATERIALS
SUPPLEMENTARY http://stm.sciencemag.org/content/suppl/2017/02/13/9.377.eaai8700.DC1
CONTENT
RELATED
http://stm.sciencemag.org/content/scitransmed/9/407/eaad4000.full
http://science.sciencemag.org/content/sci/357/6350/455.full
http://science.sciencemag.org/content/sci/357/6350/507.full
http://stm.sciencemag.org/content/scitransmed/9/394/eaah4477.full
REFERENCES http://stm.sciencemag.org/content/9/377/eaai8700#BIBL
This article cites 41 articles, 12 of which you can access for free
PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions
Terms of ServiceUse of this article is subject to the
is a registered trademark of AAAS.Science Translational Medicinetitle
licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. The
Science, 1200 New York Avenue NW, Washington, DC 20005. 2017 © The Authors, some rights reserved; exclusive
(ISSN 1946-6242) is published by the American Association for the Advancement ofScience Translational Medicine
by guest on October 20, 2017http://stm.sciencemag.org/Downloaded from
... Various methods of intermittent and periodic energy restriction have shown variable effects on glycaemic control in people with type 2 diabetes [7]. Limiting (formula) dietary intake to 850 kcal/day for [12][13][14][15][16][17][18][19][20] weeks, followed by structural support for weight loss maintenance, facilitates disease remission in people with type 2 diabetes [8][9][10]. However, severely restricting calorie intake for extended periods is burdensome for many people and reduces energy expenditure [11], rendering weight maintenance a challenge in the long term [12]. ...
... In mice, periodic FMD cycles ameliorate the metabolic anomalies of type 2 diabetes, reverse defects in insulin production [13], and prevent premature death caused by high-fat/high-calorie diets [14]. In healthy (non-diabetic) humans, three 5-day cycles of FMD monthly were shown to reduce fat mass, blood pressure, triglyceride levels, and fasting glucose, particularly in people with high levels of these risk factors at baseline [15]. ...
... The results of three previous studies are in line with our findings. Three 5-day cycles of similar composition and timing as used in our trial improved (average) anthropometric measures and metabolic control particularly in obese people with metabolic anomalies at baseline [15], as well as in people with type 2 diabetes [28]. Six cycles improved markers of metabolic control in the FMD group but not in a group with similarly timed cycles of a Mediterranean diet [27] in people with type 2 diabetes. ...
Preprint
Full-text available
Aims/hypothesis The aim of this study was to evaluate the impact on metabolic control of the periodic use of a 5-day fasting-mimicking diet (FMD) program as an adjunct to usual care in people with type 2 diabetes under regular primary care surveillance. Methods In this randomised, controlled, assessor-blinded trial, people with type 2 diabetes using metformin only and/or diet alone for glycaemic control were randomised to receive 5-day cycles of FMD monthly as adjunct to regular care by their general practitioner or regular care only. Primary outcomes were changes in glucose-lowering medication and HbA1c levels after 12 months. Moreover, changes in use of glucose-lowering medication and/or HbA1c levels in individual participants were combined to yield a clinically relevant primary outcome measure (glycaemic management), categorized as improved, stable or deteriorated after one year of follow-up. Results 100 individuals with type 2 diabetes, age 18-75 years, and BMI > 27 kg/m2, were randomised to the FMD (n=51) or control group (n=49). Eight FMD participants and ten controls were lost to follow-up. In complete case intention-to-treat analyses, the mean medication effect score (MES) significantly declined in patients receiving FMD as compared to controls (FMD -0.2 ± 0.3 vs controls +0.2 ± 0.4, p<0.0001) in the face of similar changes of HbA1c adjusted for MES (FMD -0.4 ± 0.8 % vs controls +0.2 ± 0.8 %, p=0.0021). Glycaemic management improved in 53% of participants using FMD vs 8% of controls, remained stable in 23% vs 33%, and deteriorated in 23% vs 59% (p<0.0001). Conclusions/interpretation Integration of a monthly FMD program in regular primary care for people with type 2 diabetes who use metformin only and/or diet alone for glycaemic control reduces the need for glucose-lowering medication and appears to be safe in routine clinical practice. Trial registration ClinicalTrials.gov: NCT03811587
... The three meal pattern is not necessarily based upon hunger or optimal health, but is likely to be driven by prevailing environmental and sociocultural norms [6]. Fasting-foregoing food or calorie containing drinks for long periods-has a growing body of evidence supporting health-promoting benefits, but fasting may be difficult to implement [5,[7][8][9][10][11]. ...
Article
Full-text available
Background Eating frequency may affect body weight and cardiometabolic health. Intervention trials and observational studies have both indicated that high- and low-frequency eating can be associated with better health outcomes. There are currently no guidelines to inform how to advise healthy adults about how frequently to consume food or beverages. Aim To establish whether restricted- (≤ three meals per day) frequency had a superior effect on markers of cardiometabolic health (primary outcome: weight change) compared to unrestricted-eating (≥ four meals per day) frequency in adults. Methods We searched Medline (Ovid), Embase, CINAHL (EBSCO), Cochrane Central Register of Controlled Trials (CENTRAL), CAB Direct and Web of Science Core Collection electronic databases from inception to 7 June 2022 for clinical trials (randomised parallel or cross-over trials) reporting on the effect of high or low-frequency eating on cardiometabolic health (primary outcome: weight change). Trial interventions had to last for at least two weeks, and had to have been conducted in human adults. Bias was assessed using the Cochrane Risk of Bias tool 2.0. Standardized mean differences (SMD) and 95% confidence intervals were calculated for all outcomes. Certainty of the evidence was assessed using the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) approach. Results Seventeen reports covering 16 trials were included in the systematic review. Data from five trials were excluded from meta-analysis due to insufficient reporting. 15 of 16 trials were at high risk of bias. There was very low certainty evidence of no difference between high- and low-frequency eating for weight-change (MD: -0.62 kg, CI⁹⁵: -2.76 to 1.52 kg, p = 0.57). Conclusions There was no discernible advantage to eating in a high- or low-frequency dietary pattern for cardiometabolic health. We cannot advocate for either restricted- or unrestricted eating frequency to change markers of cardiometabolic health in healthy young to middle-aged adults. Protocol registration CRD42019137938.
... To overcome this limitation, a fasting-mimicking diet (FMD) has been developed. This diet, which is low in calories and protein yet high in fats, is specifically designed to mimic the effects of fasting while still providing essential nutrients to the body [4]. The safety and feasibility of FMD have been demonstrated in various solid-tumor patients, indicating its potential for use in combination with anti-tumor treatment [5][6][7][8][9]. ...
Article
Full-text available
Background Recent research shows that tumor-associated macrophages (TAMs) are the primary consumers of glucose in tumor tissue, surpassing that of tumor cells. Our previous studies revealed that inhibiting glucose uptake impairs the survival and tumor-promoting function of hypoxic TAMs, suggesting that glucose reduction by energy restriction (calorie restriction or short-term fasting) may has a significant impact on TAMs. The purpose of this study is to verify the effect of fasting-mimicking diet (FMD) on TAMs, and to determine whether FMD synergizes with anti-angiogenic drug apatinib via TAMs. Methods The effect of FMD on TAMs and its synergistic effects with apatinib were observed using an orthotopic mouse breast cancer model. An in vitro cell model, utilizing M2 macrophages derived from THP-1 cell line, was intended to assess the effects of low glucose on TAMs under hypoxic and normoxic conditions. Bioinformatics was used to screen for potential mechanisms of action, which were then validated both in vivo and in vitro. Results FMD significantly inhibit the pro-tumor function of TAMs in vivo and in vitro, with the inhibitory effect being more pronounced under hypoxic conditions. Additionally, the combination of FMD-mediated TAMs inhibition with apatinib results in synergistic anti-tumor activity. This effect is partially mediated by the downregulation of CCL8 expression and secretion by the mTOR-HIF-1α signaling pathway. Conclusions These results support further clinical combination studies of FMD and anti-angiogenic therapy as potential anti-tumor strategies. Graphical Abstract
... Mouse models of diabetes and multiple sclerosis have shown considerable physiological improvements upon intermittent FMD, including extended longevity, increased cognitive performance, improved immune function, and reduced overall morbidity [18][19][20][21]. In humans, a clinical trial with recurrent FMD resulted in reduced IGF-1 and blood glucose levels and lowered blood pressure [22]. The first clinical trial to determine the effect of FMD in cancer patients showed promising preliminary results [23]. ...
... Understanding the complex relationship between metabolic diseases and healthy aging is crucial for promoting wellbeing and preventing disease burden. While genetics play a role in metabolic disease development, environmental and behavioral factors also contribute significantly (6)(7)(8)(9)(10). An obesogenic environment, characterized by air pollution, pesticides and exposure to environmental toxins, correlates strongly with the rising prevalence of obesity and its associated comorbidities (8,(10)(11)(12)(13)(14). ...
Article
Full-text available
Aging is a progressive and irreversible pathophysiological process that manifests as the decline in tissue and cellular functions, along with a significant increase in the risk of various aging-related diseases, including metabolic diseases. While advances in modern medicine have significantly promoted human health and extended human lifespan, metabolic diseases such as obesity and type 2 diabetes among the older adults pose a major challenge to global public health as societies age. Therefore, understanding the complex interaction between risk factors and metabolic diseases is crucial for promoting well-being and healthy aging. This review article explores the environmental and behavioral risk factors associated with metabolic diseases and their impact on healthy aging. The environment, including an obesogenic environment and exposure to environmental toxins, is strongly correlated with the rising prevalence of obesity and its comorbidities. Behavioral factors, such as diet, physical activity, smoking, alcohol consumption, and sleep patterns, significantly influence the risk of metabolic diseases throughout aging. Public health interventions targeting modifiable risk factors can effectively promote healthier lifestyles and prevent metabolic diseases. Collaboration between government agencies, healthcare providers and community organizations is essential for implementing these interventions and creating supportive environments that foster healthy aging.
... Interestingly, a consistent trend emerges across these tables. Higher body weight and BMI are negatively correlated with HDL cholesterol levels, suggesting that increased body weight is associated with a decrease in HDL cholesterol, which is considered a risk factor for cardiovascular health [17][18]. Similarly, higher body weight and BMI are negatively correlated with LDL cholesterol levels. ...
Article
Background: The escalating prevalence of overweight and obesity has heightened concerns about cardiovascular health. The intricate interplay between lipid profile variables and weight-related outcomes plays a pivotal role in shaping cardiovascular risks among individuals with excess body weight. This study investigates the associations between lipid profile variables and weight-related indicators in a cohort of healthy, overweight adults, shedding light on potential implications for cardiovascular risk management. Methods: The prevalence of metabolic syndrome, risk factors, and related lifestyle in adult Myanmar citizens were examined in the Inter-University Consortium for Political and Social Research Study (ICPSR146521) using prospective data. The patient’s lipid profiles and demographic data were statistically analyzed using the statistical MedCalc tool. Statistical significance was determined using a 0.05 p-value. Results: Results demonstrate that while BMI is influenced by multiple factors beyond lipid profile variables, waist circumference exhibits moderate association with these variables. Notably, triglycerides significantly correlated with waist circumference, suggesting a potential role in visceral fat accumulation. Additionally, the study highlights the impact of high-density lipoprotein (HDL) cholesterol levels on body weight, indicating that higher HDL levels are associated with lower body weights among healthy overweight adults. Conclusion: This study provides comprehensive insights into the complex relationship between lipid profile variables and weight-related outcomes among healthy, overweight adults. These findings underscore the importance of weight management strategies for cardiovascular health and suggest potential avenues for targeted interventions. By understanding the interplay between lipid profiles and obesity, clinicians and public health professionals can develop more effective strategies to mitigate cardiovascular risks in this demographic
Article
Fasting has been grown in popularity with multiple potential benefits. However, very few studies dynamically monitor physiological and pathological changes during long‐term fasting using noninvasive methods. In the present study, we recruited 37 individuals with metabolic syndrome to engage in a 5‐day water‐only fasting regimen, and simultaneously captured the molecular alterations through urinary proteomics and metabolomics. Our findings reveal that water‐only fasting significantly mitigated metabolic syndrome‐related risk markers, such as body weight, body mass index, abdominal circumference, blood pressure, and fasting blood glucose levels in metabolic syndrome patients. Indicators of liver and renal function remained within the normal range, with the exception of uric acid. Notably, inflammatory response was inhibited during the water‐only fasting period, as evidenced by a decrease in the human monocyte differentiation antigen CD14. Intriguingly, glycolysis, tricarboxylic acid cycle, and oxidative phosphorylation underwent a sex‐dependent reprogramming throughout the fasting period, whereby males exhibited a greater upregulation of carbohydrate metabolism‐related enzymes than females. This disparity may be attributed to evolutionary pressures. Collectively, our study sheds light on the beneficial physiological effects and novel dynamic molecular features associated with fasting in individuals with metabolic syndrome using noninvasive methods.
Preprint
Full-text available
BACKGROUND/OBJECTIVES: We investigated whether dietary interventions, i.e. a fasting mimicking diet (FMD, Prolon®) or glycocalyx mimetic supplementation (EndocalyxTM) could stabilize microvascular function in Surinamese South-Asian patients with type 2 diabetes (SA-T2DM) in the Netherlands, a patient population more prone to develop vascular complications. SUBJECTS/METHODS: A randomized, placebo controlled, 3-arm intervention study was conducted in 56 SA-T2DM patients between 18 and 75 years old, for 3 consecutive months, with one additional follow up measurement 3 months after the last intervention. Linear mixed models and interaction analysis were used to investigate the effects the interventions had on microvascular function. RESULTS: Despite a temporal improvement in BMI and HbA1c after FMD the major treatment effect on microvascular health was worsening for RBC-velocity independent PBRdynamic, especially at follow-up. Glycocalyx supplementation, however, reduced urinary MCP-1 presence and improved both PBRdynamic and MVHSdynamic, which persisted at follow-up. CONCLUSIONS: We showed that despite temporal beneficial changes in BMI and HbA1c after FMD, this intervention is not able to preserve microvascular endothelial health in Dutch South-Asian patients with T2DM. In contrast, glycocalyx mimetics preserves the microvascular endothelial health and reduces the inflammatory cytokine MCP-1.
Article
Objective Emerging evidence suggests that glucose depletion (GD)‐induced cell death depends on system Xc ⁻ , a glutamate/cystine antiporter extensively studied in ferroptosis. However, the underlying mechanism remains debated. Our study confirmed the correlation between system Xc ⁻ and GD‐induced cell death and provided a strategic treatment for oral squamous cell carcinoma (OSCC). Methods qPCR and Western blotting were performed to detect changes in xCT and CD98 expression after glucose withdrawal. Then, the cell viability of OSCCs under the indicated conditions was measured. To identify the GD‐responsible transcriptional factors of SLC7A11, we performed a luciferase reporter assay and a ChIP assay. Further, metabolomics was conducted to identify changes in metabolites. Finally, mitochondrial function and ATP production were evaluated using the seahorse assay, and NADP ⁺ /NADPH dynamics were measured using a NADP ⁺ /NADPH kit. Results In OSCCs, system Xc ⁻ promoted GD‐induced cell death by increasing glutamate consumption, which promoted NADPH exhaustion and TCA blockade. Moreover, GD‐induced xCT upregulation was governed by the p‐eIF2α/ATF4 axis. Conclusions System Xc ⁻ overexpression compromised the metabolic flexibility of OSCC under GD conditions, and thus, glucose starvation therapy is effective for killing OSCC cells.
Article
Full-text available
Dietary interventions have not been effective in the treatment of multiple sclerosis (MS). Here, we show that periodic 3-day cycles of a fasting mimicking diet (FMD) are effective in ameliorating demyelination and symptoms in a murine experimental autoimmune encephalomyelitis (EAE) model. The FMD reduced clinical severity in all mice and completely reversed symptoms in 20% of animals. These improvements were associated with increased corticosterone levels and regulatory T (Treg) cell numbers and reduced levels of pro-inflammatory cytokines, TH1 and TH17 cells, and antigen-presenting cells (APCs). Moreover, the FMD promoted oligodendrocyte precursor cell regeneration and remyelination in axons in both EAE and cuprizone MS models, supporting its effects on both suppression of autoimmunity and remyelination. We also report preliminary data suggesting that an FMD or a chronic ketogenic diet are safe, feasible, and potentially effective in the treatment of relapsing-remitting multiple sclerosis (RRMS) patients (NCT01538355).
Article
Full-text available
Prolonged fasting (PF) promotes stress resistance, but its effects on longevity are poorly understood. We show that alternating PF and nutrient-rich medium extended yeast lifespan independently of established pro-longevity genes. In mice, 4 days of a diet that mimics fasting (FMD), developed to minimize the burden of PF, decreased the size of multiple organs/systems, an effect followed upon re-feeding by an elevated number of progenitor and stem cells and regeneration. Bi-monthly FMD cycles started at middle age extended longevity, lowered visceral fat, reduced cancer incidence and skin lesions, rejuvenated the immune system, and retarded bone mineral density loss. In old mice, FMD cycles promoted hippocampal neurogenesis, lowered IGF-1 levels and PKA activity, elevated NeuroD1, and improved cognitive performance. In a pilot clinical trial, three FMD cycles decreased risk factors/biomarkers for aging, diabetes, cardiovascular disease, and cancer without major adverse effects, providing support for the use of FMDs to promote healthspan.
Article
Full-text available
Mice and humans with growth hormone receptor/IGF-1 deficiencies display major reductions in age-related diseases. Because protein restriction reduces GHR-IGF-1 activity, we examined links between protein intake and mortality. Respondents aged 50-65 reporting high protein intake had a 75% increase in overall mortality and a 4-fold increase in cancer death risk during the following 18 years. These associations were either abolished or attenuated if the proteins were plant derived. Conversely, high protein intake was associated with reduced cancer and overall mortality in respondents over 65, but a 5-fold increase in diabetes mortality across all ages. Mouse studies confirmed the effect of high protein intake and GHR-IGF-1 signaling on the incidence and progression of breast and melanoma tumors, but also the detrimental effects of a low protein diet in the very old. These results suggest that low protein intake during middle age followed by moderate to high protein consumption in old adults may optimize healthspan and longevity.
Article
Full-text available
Fasting has been practiced for millennia, but, only recently, studies have shed light on its role in adaptive cellular responses that reduce oxidative damage and inflammation, optimize energy metabolism, and bolster cellular protection. In lower eukaryotes, chronic fasting extends longevity, in part, by reprogramming metabolic and stress resistance pathways. In rodents intermittent or periodic fasting protects against diabetes, cancers, heart disease, and neurodegeneration, while in humans it helps reduce obesity, hypertension, asthma, and rheumatoid arthritis. Thus, fasting has the potential to delay aging and help prevent and treat diseases while minimizing the side effects caused by chronic dietary interventions.
Article
Most animals alternate periods of feeding with periods of fasting often coinciding with sleep. Upon >24 hr of fasting, humans, rodents, and other mammals enter alternative metabolic phases, which rely less on glucose and more on ketone body-like carbon sources. Both intermittent and periodic fasting result in benefits ranging from the prevention to the enhanced treatment of diseases. Similarly, time-restricted feeding (TRF), in which food consumption is restricted to certain hours of the day, allows the daily fasting period to last >12 hr, thus imparting pleiotropic benefits. Understanding the mechanistic link between nutrients and the fasting benefits is leading to the identification of fasting-mimicking diets (FMDs) that achieve changes similar to those caused by fasting. Given the pleiotropic and sustained benefits of TRF and FMDs, both basic science and translational research are warranted to develop fasting-associated interventions into feasible, effective, and inexpensive treatments with the potential to improve healthspan.
Article
The 2013 American College of Cardiology/American Heart Association (ACC/AHA) cholesterol guideline advocated several changes from the previous Adult Treatment Panel III guidelines. Assuming full implementation, the 2013 ACC/AHA guideline would identify ≈13 million Americans as newly eligible for consideration of statin therapy. Three features of the 2013 ACC/AHA guideline primarily responsible for these differences are the specific risk assessment tool endorsed, the risk threshold considered sufficient to warrant primary prevention statin therapy, and the decision not to include cholesterol treatment targets. There is no consensus among international guidelines on the optimal approach to these 3 components. The 2013 ACC/AHA guideline recommends assessing absolute risk with the Pooled Cohort equations, which were developed to improve on previous risk assessment models by including stroke as an outcome and by broadening racial and geographic diversity. Each of the leading international guidelines recommends a different equation for absolute risk assessment. The 2013 ACC/AHA guideline advises consideration of statin therapy for an estimated 10-year risk of atherosclerotic vascular disease of ≥7.5%, which is lower than the thresholds recommended by other leading international guidelines. Lastly, the 2013 ACC/AHA guideline does not endorse a treat-to-target strategy but instead specifies the appropriate intensity of statin for each risk category. This approach is shared by the National Institute for Health and Care Excellence guidelines but differs from other international guidelines. In this review, we summarize the 2013 ACC/AHA cholesterol guideline recommendations and compare them with recommendations from Adult Treatment Panel III and other leading international guidelines.
Article
Background: Plasma levels of insulin-like growth factor-I (IGF-I) have been associated with the risk of prostate cancer. However, the association of IGF-I with specific tumor stage and grade at diagnosis, which correlate with risk of recurrence and mortality, has not been studied rigorously. To determine whether plasma levels of IGF-I and its main circulating binding protein, IGF binding protein-3 (IGFBP-3), predict more aggressive forms of prostate cancer, we investigated the association between plasma levels of each and specific stages and grades of prostate cancer. Methods: We examined 530 case patients and 534 control subjects in a nested case–control study in the prospective Physicians' Health Study. Patients with prostate cancer diagnosed from 1982 through 1995 were matched to control subjects by age and smoking status. IGF-I and IGFBP-3 were measured in prospectively collected plasma samples. Conditional logistic regression models were used to estimate the relative risks (RRs) for prostate cancer associated with IGF-I and IGFBP-3, stratified on grade (Gleason score ≥7 versus <7) and stage (early = stage A or B prostate cancer versus advanced = stage C or D prostate cancer). All statistical tests were two-sided. Results: Plasma levels of IGF-I and IGFBP-3 were predictors of advanced-stage prostate cancer (RR = 5.1, 95% confidence interval [CI] = 2.0 to 13.2 for highest versus lowest quartiles of IGF-I; RR = 0.2, 95% CI = 0.1 to 0.6 for highest versus lowest quartiles of IGFBP-3) but not of early-stage prostate cancer. Neither was differentially associated with Gleason score. Men with high IGF-I levels and low IGFBP-3 levels had an RR for advanced-stage prostate cancer of 9.5 (95% CI = 1.9 to 48.4) compared with men with low levels of both. Combining IGF-I and IGFPB-3 measurements with a standard prostate-specific antigen (PSA) measurement for prostate cancer screening increased the specificity (from 91% to 93%) but decreased sensitivity (from 40% to 36%) compared with measurement of PSA alone. Conclusions: Circulating levels of IGF-I and IGFBP-3 may predict the risk of developing advanced-stage prostate cancer, but their utility for screening patients with incident prostate cancer may be limited.
Article
Context The Third Report of the National Cholesterol Education Program Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (ATP III) highlights the importance of treating patients with the metabolic syndrome to prevent cardiovascular disease. Limited information is available about the prevalence of the metabolic syndrome in the United States, however.Objective To estimate the prevalence of the metabolic syndrome in the United States as defined by the ATP III report.Design, Setting, and Participants Analysis of data on 8814 men and women aged 20 years or older from the Third National Health and Nutrition Examination Survey (1988-1994), a cross-sectional health survey of a nationally representative sample of the noninstitutionalized civilian US population.Main Outcome Measures Prevalence of the metabolic syndrome as defined by ATP III (≥3 of the following abnormalities): waist circumference greater than 102 cm in men and 88 cm in women; serum triglycerides level of at least 150 mg/dL (1.69 mmol/L); high-density lipoprotein cholesterol level of less than 40 mg/dL (1.04 mmol/L) in men and 50 mg/dL (1.29 mmol/L) in women; blood pressure of at least 130/85 mm Hg; or serum glucose level of at least 110 mg/dL (6.1 mmol/L).Results The unadjusted and age-adjusted prevalences of the metabolic syndrome were 21.8% and 23.7%, respectively. The prevalence increased from 6.7% among participants aged 20 through 29 years to 43.5% and 42.0% for participants aged 60 through 69 years and aged at least 70 years, respectively. Mexican Americans had the highest age-adjusted prevalence of the metabolic syndrome (31.9%). The age-adjusted prevalence was similar for men (24.0%) and women (23.4%). However, among African Americans, women had about a 57% higher prevalence than men did and among Mexican Americans, women had about a 26% higher prevalence than men did. Using 2000 census data, about 47 million US residents have the metabolic syndrome.Conclusions These results from a representative sample of US adults show that the metabolic syndrome is highly prevalent. The large numbers of US residents with the metabolic syndrome may have important implications for the health care sector.
Article
By 2050, the number of people over the age of 80 will triple globally. These demographics could come at great cost to individuals and economies.
Article
Immune system defects are at the center of aging and a range of diseases. Here, we show that prolonged fasting reduces circulating IGF-1 levels and PKA activity in various cell populations, leading to signal transduction changes in long-term hematopoietic stem cells (LT-HSCs) and niche cells that promote stress resistance, self-renewal, and lineage-balanced regeneration. Multiple cycles of fasting abated the immunosuppression and mortality caused by chemotherapy and reversed age-dependent myeloid-bias in mice, in agreement with preliminary data on the protection of lymphocytes from chemotoxicity in fasting patients. The proregenerative effects of fasting on stem cells were recapitulated by deficiencies in either IGF-1 or PKA and blunted by exogenous IGF-1. These findings link the reduced levels of IGF-1 caused by fasting to PKA signaling and establish their crucial role in regulating hematopoietic stem cell protection, self-renewal, and regeneration.