Fact-checked by Grok 2 weeks ago

Caloric restriction mimetic

Caloric restriction mimetics (CRMs) are compounds that mimic the biochemical and functional effects of caloric restriction (), a dietary involving a 10–30% reduction in calorie intake without causing , which has been shown to extend lifespan and delay the onset of age-related diseases in model organisms ranging from to . , first demonstrated in , activates cellular stress response pathways that enhance metabolic efficiency, reduce oxidative damage, and promote , leading to improved healthspan without compromising essential nutrient intake. CRMs aim to replicate these outcomes pharmacologically or through natural dietary agents, offering a practical alternative for individuals unable or unwilling to adhere to strict CR regimens. At the molecular level, CRMs primarily target conserved pathways such as the activation of (AMPK), sirtuins (e.g., SIRT1), and induction, while inhibiting the (mTOR) complex, which collectively mimic the nutrient-sensing adaptations triggered by CR. These mechanisms involve reducing by depleting , inhibiting acetyltransferases, or stimulating deacetylases, thereby promoting cellular repair and resilience against stressors like and genomic instability. In preclinical studies, CRMs have demonstrated benefits in mitigating age-related pathologies, including neurodegeneration, , and metabolic disorders, though effects can vary by , , and dosage. Prominent examples of CRMs include , a found in grapes and that activates sirtuins and extends lifespan in some models; rapamycin, an inhibitor that prolongs mouse lifespan but carries risks like ; metformin, an AMPK activator commonly used for with emerging evidence for healthspan extension; and spermidine, a natural linked to enhancement and reduced mortality in human cohort studies. Clinical trials are ongoing to evaluate CRMs for applications in aging, cancer immunotherapies, and chronic conditions, with compounds like D-glucosamine showing promise in lowering all-cause mortality and improving outcomes, though larger randomized controlled trials are needed to confirm efficacy and safety in humans.

Introduction

Definition and Concept

Caloric restriction (CR) is a dietary intervention characterized by a 20–40% reduction in calorie intake relative to ad libitum feeding, while ensuring adequate nutrition to prevent malnutrition. This approach has been demonstrated to extend lifespan and delay age-related diseases in various model organisms, including yeast (Saccharomyces cerevisiae), nematodes (Caenorhabditis elegans), fruit flies (Drosophila melanogaster), and rodents such as rats and mice. In these species, CR consistently promotes longevity by modulating physiological processes, with effects observed across evolutionary distant taxa, underscoring its broad applicability. Caloric restriction mimetics (CRMs) are pharmacological or dietary compounds designed to replicate the beneficial biochemical and physiological effects of without necessitating a reduction in overall consumption. These agents activate CR-like cellular responses, including enhanced , improved stress resistance, and shifts in energy metabolism, thereby conferring similar health benefits such as extended healthspan and reduced disease risk. True CRMs specifically emulate the metabolic, hormonal, and physiological adaptations of , distinguishing them from related interventions like mimetics, which primarily target temporal nutrient availability rather than sustained modulation. At their core, CRMs operate within a conceptual framework that leverages nutrient-sensing pathways to simulate a "famine-like" state at the cellular level, even in the presence of ample nutrients. By engaging pathways such as those involving AMPK and sirtuins, CRMs induce protective responses akin to those triggered by actual nutrient scarcity, fostering cellular resilience and without dietary hardship. This targeted mimicry positions CRMs as a practical alternative for translating CR's advantages into human applications.

Historical Development

The origins of research on caloric restriction (CR) and its mimetics trace back to the early , when experimental observations first linked reduced caloric intake to extended lifespan. In , and colleagues at reported that rats subjected to severe caloric restriction from weaning—limiting intake to about one-third of ad libitum-fed controls—exhibited significantly prolonged lifespans, up to 33% longer, without , establishing CR as a potent modulator of aging in mammals. This seminal work laid the groundwork for subsequent investigations into CR's mechanisms and potential pharmacological alternatives that could replicate its benefits without dietary austerity. Advancements in the and shifted focus toward genetic and molecular underpinnings, fueling interest in CR mimetics. In , the discovery of the SIR2 gene's role in lifespan regulation highlighted conserved pathways; a study showed that CR extended replicative lifespan via NAD+-dependent activation of Sir2, a , prompting exploration of drugs targeting similar pathways. Key milestones included the 2003 identification of , a from , as a potent activator of SIRT1 (the mammalian Sir2 homolog), which extended lifespan and mimicked CR effects . By 2009, rapamycin emerged as another mimetic through its inhibition of the pathway; late-life administration extended mouse lifespan by 9-14% in a genetically diverse , paralleling CR's suppression of nutrient-sensing signals. The and marked a transition toward clinical translation and broader candidates. Metformin, an antidiabetic drug acting as an AMPK activator and indirect inhibitor, gained prominence as a CR mimetic; the Targeting Aging with Metformin () trial, proposed in 2019, aims to test its effects on age-related outcomes in 3,000 nondiabetic adults aged 65-79 over six years (as of November 2025, the trial remains in the planning stage). Emerging compounds like spermidine and have shown promise in neuroprotective contexts; a 2025 study demonstrated spermidine's role in enhancing mitochondrial function and during protein restriction, protecting against brain aging in flies, while was reported to induce via ATG101 interaction in cellular models, with potential neuroprotective effects in neurodegenerative diseases. Pioneering researchers have shaped this field. Leonard Guarente's work at elucidated sirtuins' central role in CR-mediated , from yeast Sir2 to mammalian SIRT1, including brain-specific mechanisms during restriction. Valter Longo's development of fasting-mimicking diets (FMDs) in the 2010s further influenced CRM concepts by demonstrating periodic low-calorie regimens that activate similar protective pathways, such as IGF-1 reduction and regeneration, without full fasting.

Biological Mechanisms

Key Pathways Involved

Caloric restriction mimetics (CRMs) primarily engage nutrient-sensing pathways to replicate the adaptive responses of caloric restriction, with (AMPK) serving as a central energy sensor that detects low cellular energy states through an elevated AMP/ATP ratio. Upon activation by upstream kinases like LKB1, AMPK promotes catabolic processes such as and fatty acid oxidation while inhibiting anabolic pathways, thereby conserving energy and enhancing metabolic efficiency in a manner akin to nutrient scarcity. This activation fosters a shift toward cellular maintenance over growth, contributing to stress resistance and longevity promotion. The mammalian target of rapamycin (mTOR) pathway, which integrates signals from nutrients, growth factors, and energy status, is inhibited by CRMs to mimic the effects of reduced nutrient availability. mTOR complex 1 (mTORC1) normally drives protein synthesis and cell growth; its suppression reduces translation initiation and promotes the recycling of cellular components, thereby alleviating resource demands during perceived famine. This inhibition creates a metabolic state that prioritizes survival and repair over proliferation. Sirtuin pathways, particularly those involving NAD+-dependent deacetylases like , are upregulated by CRMs through increased NAD+ availability, enhancing the cell's resilience to oxidative and metabolic stress. These enzymes deacetylate key transcription factors and metabolic regulators, modulating to support mitochondrial function, , and responses, which collectively emulate caloric restriction's protective effects. activation thus coordinates a broad reprogramming of cellular toward . Autophagy induction represents a downstream consequence of CRM action, particularly via mTOR inhibition and AMPK activation, whereby damaged organelles and proteins are sequestered and degraded to maintain and . This self-cleaning process, involving formation and lysosomal fusion, clears cellular debris accumulated under stress, preventing and supporting renewal in a nutrient-limited context. CRMs thereby enhance autophagic flux to sustain cellular integrity without actual calorie reduction. Downregulation of insulin/insulin-like growth factor-1 (IGF-1) signaling by CRMs shifts metabolism away from growth-oriented states toward those favoring longevity, by attenuating downstream effectors like PI3K/Akt that promote anabolism. This suppression activates FOXO transcription factors, which orchestrate defenses and , mirroring the reduced insulinemic environment of caloric restriction. The pathway's modulation thus integrates nutrient sensing with hormonal regulation to optimize resource allocation. These pathways interconnect to generate a coordinated resistance phenotype: AMPK and sirtuins synergize to boost NAD+ levels and , while their actions converge on and insulin/IGF-1 signaling to amplify and suppress , creating an integrated response that enhances cellular resilience akin to caloric restriction. This ensures a holistic metabolic , where energy conservation and repair mechanisms predominate over growth and reproduction.

Molecular Targets

Caloric restriction mimetics (CRMs) primarily target key molecular components within nutrient-sensing pathways to replicate the beneficial effects of caloric restriction, such as enhanced cellular and metabolic efficiency. These targets include enzymes and transcription factors that regulate , , and . By modulating these elements, CRMs promote longevity-associated processes without necessitating dietary intervention. Sirtuins, a family of NAD+-dependent deacetylases (SIRT1-7), play a central role in mediating effects by deacetylating histones and transcription factors, thereby altering to favor resistance and metabolic . SIRT1, in particular, deacetylates substrates like and FOXO proteins to enhance and defenses, while SIRT3-5 localize to mitochondria to regulate fatty acid oxidation and scavenging. CRMs such as NAD+ boosters activate sirtuins by increasing NAD+ availability, mimicking the elevated NAD+/NADH ratio observed during caloric restriction. AMP-activated protein kinase (AMPK), an energy-sensing kinase, is another primary target activated by CRMs through allosteric modulation or upstream signals like increased AMP/ATP ratios. Upon activation, AMPK phosphorylates downstream effectors such as tuberous sclerosis complex 2 (TSC2), which inhibits the mechanistic target of rapamycin complex 1 () to suppress anabolic processes and promote . This enhances and mitochondrial function, core mechanisms underlying caloric restriction benefits. Direct AMPK activators, such as certain polyphenols, replicate this effect independently of energy status. The complex, comprising , , and other regulatory proteins, is inhibited by CRMs to shift cellular from growth to maintenance. Rapalogs, a class of allosteric inhibitors, bind to the intracellular protein FKBP12, forming a complex that disrupts the interaction between and , thereby preventing activation by and growth factors. This inhibition reduces protein synthesis and promotes , paralleling the nutrient scarcity signals of caloric restriction. Epigenetic modifications targeted by CRMs include reductions in lysine acetylation on histones and non-histone proteins, which compact and repress pro-aging while activating longevity-promoting genes. Sirtuin-mediated deacetylation of H4 at 16, for instance, correlates with caloric restriction-induced lifespan extension in model organisms, leading to altered transcriptional profiles that enhance and reduce . CRMs achieve this by boosting activity or directly inhibiting acetyltransferases like EP300. Additional targets encompass FOXO transcription factors, which coordinate stress responses by translocating to the upon dephosphorylation and deacetylation, upregulating genes for arrest and detoxification. Similarly, peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) is activated via sirtuin-dependent deacetylation to drive and oxidative metabolism, amplifying caloric restriction's protective effects on energy production. Despite their specificity, CRMs face challenges from off-target effects, such as unintended modulation of interconnected pathways like , which can alter insulin signaling and promote compensatory growth responses that diminish therapeutic efficacy. These interactions highlight the need for precise targeting to avoid disrupting balanced nutrient sensing.

Prominent Candidates

Pharmacological Agents

Pharmacological agents as caloric restriction mimetics (CRMs) encompass synthetic or semi-synthetic compounds designed to replicate the beneficial effects of caloric restriction on aging and healthspan by targeting key metabolic pathways, such as and AMPK, without requiring dietary intervention. These drugs often originate from treatments for other conditions, like or , and are being repurposed for applications through preclinical and . Rapamycin, a macrolide compound and potent inhibitor of the mechanistic target of rapamycin (mTOR) pathway, serves as a prototypical CRM by suppressing mTORC1 activity, which mimics the nutrient-sensing effects of caloric restriction to promote autophagy and extend lifespan. Originally isolated from soil bacteria and FDA-approved in 1999 for immunosuppression in organ transplant patients, rapamycin has been repurposed for anti-aging due to its ability to delay age-related diseases. In mice, chronic low-dose administration starting in mid-life extends median lifespan by 9%–14%, with 2025 studies confirming robust longevity benefits comparable to dietary restriction across vertebrates, including reduced cancer incidence and improved metabolic health. Rapalogs, such as —a of rapamycin with improved —similarly inhibit and are FDA-approved for cancer and , showing potential as CRMs in aging models. has demonstrated safety and immune-modulating effects in human trials for age-related immune decline, with ongoing studies exploring low-dose regimens (0.5 mg/day or 5 mg/week) to enhance responses and delay without significant toxicity. Metformin, a drug and activator of (AMPK), is another cornerstone CRM that enhances cellular akin to caloric restriction by reducing hepatic glucose production and improving insulin sensitivity. Widely prescribed since the 1950s for management and FDA-approved for this indication, metformin is under investigation for anti-aging effects, with mixed preclinical data on lifespan extension in , potentially via AMPK-dependent pathways, though recent meta-analyses indicate inconsistent effects across vertebrates. As of 2025, the Targeting Aging with Metformin () trial—a six-year, multi-site study involving over 3,000 participants aged 65–79—remains ongoing, aiming to establish metformin's efficacy in delaying age-related as a primary outcome, building on positive preclinical and observational evidence. Other pharmacological CRMs include NAD+ precursors like (NMN) and (NR), which boost activity by elevating NAD+ levels, thereby mimicking caloric restriction's enhancement of and mitochondrial function. These semi-synthetic compounds, administered orally, have shown safety and NAD+ elevation in human trials, with potential to counteract age-related decline in energy metabolism. Additionally, 17α-estradiol, a non-feminizing stereoisomer, acts as a sex-specific CRM in , extending lifespan by up to 19% through metabolomic shifts that reduce and , with no significant effects in females. The development history of these agents traces from their initial approvals for non-aging indications—such as rapamycin's immunosuppressant role and metformin's antidiabetic use—to their recognition as CRMs following landmark studies in the demonstrating lifespan extension in model organisms. Repurposing efforts gained momentum with the identification of shared pathways like inhibition and AMPK activation, but regulatory hurdles persist, as agencies like the FDA do not yet recognize aging as a treatable indication, complicating approvals for longevity claims and necessitating trials like to prove clinical benefits. Achieving CRM effects at safe, chronic doses poses challenges related to dosing regimens and ; for instance, rapamycin's narrow therapeutic window requires intermittent low-dose protocols (e.g., weekly administration) to minimize side effects like while sustaining anti-aging benefits in mice. Similarly, metformin's gastrointestinal is limited (around 50%), prompting research into optimized formulations, though its established safety profile in supports long-term use. These issues underscore the need for tailored to translate preclinical efficacy into human applications.

Natural Compounds

Natural compounds, derived from and foods, have been investigated as caloric restriction mimetics (CRMs) due to their ability to activate similar cellular pathways without requiring reduced intake. These substances, including polyphenols and polyamines, often influence sirtuins, , and metabolic regulation, offering potential benefits for and health. Preliminary evidence from preclinical and early human studies highlights their roles in mimicking the protective effects of caloric restriction, though challenges like low persist. Resveratrol, a stilbenoid polyphenol found in grapes, , peanuts, and berries, acts as a sirtuin activator, particularly SIRT1, which mediates pathways akin to caloric restriction. Early enthusiasm stemmed from a seminal study in , where resveratrol supplementation improved healthspan and survival in mice on a high-calorie by enhancing mitochondrial function and insulin sensitivity. However, its clinical translation has been limited by poor , as rapid results in low systemic levels after oral intake, necessitating higher supplemental doses for efficacy. Spermidine, a naturally occurring abundant in wheat germ, soybeans, mushrooms, and , induces , a mechanism in caloric restriction that promotes cellular cleanup and renewal. As an autophagy inducer, it inhibits acetyltransferases to enhance autophagic flux, potentially extending lifespan in model organisms. Recent 2025 human studies have linked higher spermidine intake to improved cardiovascular health, including reduced cardiac remodeling and inflammation in elderly patients with , supporting its role as a dietary . Fisetin, a present in strawberries, apples, and onions, and chlorogenic acid, a in coffee, apples, and potatoes, exhibit and effects that mimic caloric restriction by clearing senescent cells and reducing . Fisetin selectively eliminates senescent cells, alleviating age-related , while chlorogenic acid modulates metabolic pathways to improve glucose handling and . A 2025 study highlighted their neuroprotective potential in age-related neurodegenerative models, where they activated and reduced without caloric reduction. D-glucosamine, an found in and produced endogenously, acts as a by inhibiting (via ) and inducing , mimicking caloric restriction's metabolic effects. Preclinical studies show it extends lifespan in nematodes, flies, and mice (e.g., up to 15% in mice), with mechanisms involving stress resistance and reduced . In humans, observational data from large cohorts indicate higher intake correlates with 15–39% lower all-cause mortality, and randomized trials support benefits for symptom relief, though larger studies are needed for anti-aging confirmation. Broader classes of polyphenols, such as those in fruits, , and teas, exert CRM-like effects by regulating and , thereby influencing energy and . Hydroxycitric acid, derived from the rind of cambogia fruit, inhibits ATP-citrate lyase to suppress fat synthesis and appetite, mimicking caloric restriction's metabolic shifts in preclinical models. Dietary intake of these natural CRMs varies widely, with typical food sources providing low milligram levels—such as 0.1–5 mg of per glass of or 10–20 mg of spermidine per serving of wheat germ—compared to supplemental doses of 100–500 mg, which aim to achieve therapeutic effects. Human absorption is highly variable due to factors like and , often resulting in only 1–10% for compounds like and , underscoring the need for optimized formulations.

Physiological Effects

Lifespan and Healthspan Extension

Caloric restriction mimetics (CRMs) have been shown to extend lifespan across diverse model organisms by mimicking the longevity-promoting effects of caloric restriction without requiring dietary changes. In mice, rapamycin administration starting in increases median lifespan by 15-20%, with effects persisting even after short-term treatment. Similarly, extends lifespan in nematodes by up to 14% and in fruit flies by approximately 30% under certain conditions. These extensions highlight CRMs' potential to modulate aging processes at the organismal level. Beyond lifespan, CRMs enhance healthspan by postponing functional declines associated with aging. In models, rapamycin improves physical , as evidenced by enhanced rotarod performance and forelimb following transient treatment. Cognitive function benefits through delayed age-related impairments, while resilience is bolstered, reducing susceptibility to infections in later life. These improvements contribute to a prolonged period of , allowing organisms to maintain and physiological longer. The lifespan-extending effects of CRMs exhibit conservation from unicellular to nonhuman , underscoring evolutionary preservation of underlying pathways, though data remain preliminary. Notable organism-specific variations include differences; for example, 17α-estradiol extends median lifespan in male mice by 19% when initiated late in life, with minimal impact on females. Such disparities emphasize the need for tailored applications in . Key biomarkers of CRM-induced include attenuated , such as reduced circulating IL-6 levels, and optimized mitochondrial function, which supports and reduces . Compared to traditional caloric restriction, which can extend lifespan by 30-40%, CRMs like rapamycin achieve roughly 50% of these benefits while avoiding caloric deficits and associated nutritional risks.

Disease Prevention and Treatment

Caloric restriction mimetics (CRMs) have shown promise in preventing and treating age-related neurodegenerative diseases by targeting pathological protein accumulation and enhancing cellular clearance mechanisms. In Alzheimer's disease models, rapamycin, an mTOR inhibitor, has been demonstrated to reduce amyloid-beta levels and abolish cognitive deficits in mice by inhibiting mTOR signaling, thereby lowering the production of toxic amyloid species. Similarly, metformin attenuates amyloid-beta and tau pathology in transgenic mouse models of Alzheimer's, promoting amyloid clearance through activation of AMPK pathways and reducing neuroinflammation. For Parkinson's disease, recent 2025 studies indicate that CRMs like resveratrol, metformin, and rapamycin enhance autophagy to mitigate alpha-synuclein aggregation and dopaminergic neuron loss in preclinical models, potentially slowing disease progression by improving protein degradation. In the context of cancer, CRMs contribute to disease prevention by bolstering immunosurveillance and suppressing tumor progression through induction and metabolic reprogramming. Spermidine, a natural CRM, inhibits tumor cell proliferation and in preclinical colorectal cancer models by interfering with the and enhancing autophagic flux, which limits invasive potential without promoting tumor growth. These effects extend to improved antitumor responses, where spermidine augments efficacy in models by fostering an autophagy-dependent immune microenvironment that enhances T-cell activity against tumors. For metabolic disorders, CRMs play a key role in diabetes prevention and management by modulating insulin signaling and glucose homeostasis. Metformin reduces the incidence of type 2 diabetes in high-risk individuals by improving insulin sensitivity and suppressing hepatic glucose production, as evidenced in large-scale prevention trials. Resveratrol enhances insulin sensitivity in diabetic subjects by activating SIRT1 and reducing inflammation in adipose tissue, leading to better glycemic control without adverse effects on nondiabetic individuals. Combined metformin-resveratrol therapy further ameliorates insulin resistance in preclinical models of metabolic syndrome by preventing lipolysis and inflammatory responses in hypoxic tissues. CRMs also mitigate cardiovascular and inflammatory diseases by reducing vascular damage and plaque formation. lowers risk in hyperlipidemic mouse models by activating FXR-mediated cholesterol efflux and inhibiting in endothelial cells, thereby decreasing plaque burden and . protects against by attenuating vascular and in endothelial cells, improving bioavailability and reducing adhesion molecule expression in preclinical studies. These compounds collectively diminish levels, supporting their potential in preventing age-related cardiovascular events. Therapeutically, CRMs hold adjunctive value in and brain aging mitigation. Spermidine enhances the efficacy of chemotherapeutic agents in preclinical tumor models by promoting , which selectively clears damaged cells and boosts immune-mediated tumor regression. Emerging 2025 evidence highlights rapamycin's role in mitigating brain aging pathologies, including reduced burden in established Alzheimer's models, suggesting its utility as an adjunct to standard therapies for neurodegenerative conditions.

Research Evidence

Preclinical Studies

Preclinical studies on caloric restriction mimetics (CRMs) have primarily utilized animal models and cultures to evaluate their effects on , healthspan, and underlying cellular processes. In models, rapamycin, an inhibitor of the pathway, has been shown to extend median lifespan in genetically heterogeneous mice by 9-14% when administered late in life, starting at 600 days of age, with similar benefits observed in both sexes. This extension was associated with delayed age-related pathologies, including reduced incidence of cancer and improved immune function. Similarly, metformin, an activator of AMPK, delayed the onset of frailty and extended healthspan in male mice when provided at 0.1% in the from , enhancing physical and insulin without altering food intake. In female SHR mice, chronic metformin treatment (100 mg/kg in drinking water) increased mean lifespan by 37.8% and postponed spontaneous tumorigenesis. Invertebrate models, such as , have provided insights into CRM mechanisms paralleling dietary restriction. Resveratrol, a activator, extended the lifespan of C. elegans by up to 30% in a dose-dependent manner when added to the food from early adulthood, mimicking caloric restriction through enhanced stress resistance and metabolic shifts. This effect was mediated by activation of SIR-2.1, the C. elegans ortholog of SIRT1, and required insulin/IGF-1 signaling pathways. In vitro evidence from cell culture assays supports CRM induction of protective cellular responses. promotes in mammalian cell lines, such as HEK293 cells, by inhibiting , leading to increased formation and clearance of damaged organelles at concentrations as low as 100 nM. similarly induces in cell lines via AMPK activation and mTOR suppression, reducing markers of like β-galactosidase activity and expression. These effects mimic caloric restriction by enhancing lysosomal degradation and limiting replicative without inducing at therapeutic doses. Studies on dose-response and chronic administration highlight the importance of low-dose regimens to replicate caloric restriction benefits while avoiding . For instance, low-dose (2-DG, 0.4% in diet), a glycolytic inhibitor, mimicked caloric restriction in rats by inducing mitohormesis—mild elevation that upregulated antioxidant enzymes like and —without the or mortality seen at higher doses (>1%). This regimen improved metabolic profiles and brain defense mechanisms in accelerated models over 12 months of administration. Recent 2025 updates include new mouse studies on , a , demonstrating efficacy against cerebral pathologies. In an model using Aβ(25–35)-injected mice, nanoformulated (10 mg/kg intraperitoneally) reduced , amyloid plaque burden, and cognitive deficits, preserving neuronal integrity in the hippocampus and over 28 days.

Clinical Trials and Human Data

Observational studies have established epidemiological links between metformin use and reduced mortality in patients with . A and of over 78,000 patients demonstrated that metformin is associated with a 27% lower risk of all-cause mortality compared to other antidiabetic treatments, independent of control, and even lower rates than in non-diabetic controls. Similar findings from cohort studies indicate up to a 30% reduction in cardiovascular mortality among metformin users with and . Interventional clinical trials on caloric restriction mimetics (CRMs) in humans remain limited but are expanding, focusing primarily on metformin and emerging compounds like spermidine. The Targeting Aging with Metformin (TAME) trial, a landmark randomized controlled study involving approximately 3,000 adults aged 65-79, is ongoing as of 2025 and aims to evaluate metformin's impact on delaying multiple age-related diseases, including cardiovascular events, cancer, and cognitive decline, over six years. Preliminary data from the related Metformin in Study (MILES), a crossover in 14 older adults, revealed that short-term metformin treatment induces changes consistent with anti-aging effects, such as altered and pathways. For spermidine, a natural CRM, multiple randomized s have investigated its effects on ; a 2025 review of four interventional studies found that three reported positive outcomes, including improved performance and in older adults with subjective cognitive decline after 3-12 months of supplementation at doses of 1-3 mg/day, though one longer-term showed no significant changes in biomarkers. Safety profiles of CRMs in trials are generally favorable at therapeutic doses, though dose-dependent risks exist. Metformin, widely used for , commonly causes mild gastrointestinal side effects such as and diarrhea in 20-30% of users, which often resolve with dose adjustment or extended-release formulations. Rapamycin (), another CRM targeting , demonstrates good tolerability in low-dose intermittent regimens for aging-related trials, with the PEARL trial reporting no serious adverse events over one year in healthy adults, but higher doses are associated with risks, including increased infection rates. Human studies of CRMs have identified changes in key aging biomarkers, providing translational evidence from preclinical models. Metformin treatment in older adults reduces circulating IGF-1 levels by 20-30%, mirroring caloric restriction effects and potentially contributing to metabolic health improvements. Similarly, both metformin and spermidine supplementation lower inflammation markers, such as and IL-6, by 10-25% in short-term trials, correlating with enhanced and reduced . Despite these advances, significant gaps persist in CRM research for human aging. Long-term trials with aging-specific endpoints, such as or healthspan, are scarce, with most data derived from or short-term interventions lasting under two years. The U.S. (FDA) has not approved any CRM for anti-aging indications as of 2025, citing challenges in defining aging as a treatable condition and the need for robust evidence on delayed disease onset.

Challenges and Future Directions

Limitations and Side Effects

While caloric restriction mimetics (CRMs) hold promise for mimicking the benefits of caloric restriction, their clinical application is hindered by various side effects and limitations. For instance, metformin, a widely studied CRM that activates AMPK pathways, carries a risk of , a rare but potentially fatal condition particularly in individuals with renal impairment or during acute illnesses, with incidence rates estimated at 4.3 cases per 100,000 patient-years. Similarly, rapamycin, an inhibitor considered a CRM, induces by promoting regulatory T cells and inhibiting effector immune responses, thereby increasing susceptibility to infections such as and viral illnesses, as observed in transplant patients receiving the drug. , another natural CRM targeting sirtuins, is generally well-tolerated at doses up to 1 g/day but can cause mild gastrointestinal discomfort in some users. Pharmacokinetic challenges further complicate CRM efficacy. Resveratrol exhibits poor oral , often less than 1% due to rapid in the liver and intestines, necessitating high doses (up to 5 g/day in some studies) to achieve therapeutic plasma levels, which may exacerbate tolerability issues and limit practical use. These absorption barriers are not unique to resveratrol; many natural CRMs face similar stability and delivery problems, reducing their reliability in human applications. Population variability introduces additional constraints, with responses to CRMs differing by , , and . For example, preclinical studies indicate that caloric restriction and its mimetics produce weaker lifespan extension effects in female mice compared to males in certain strains, potentially due to sex-specific hormonal influences on metabolic pathways like IGF-1 signaling. Age-related differences also play a role, as older individuals may exhibit diminished CRM responsiveness owing to accumulated physiological declines, while genetic polymorphisms in drug-metabolizing enzymes can alter efficacy across diverse populations. Long-term unknowns pose significant risks. Moreover, in specific contexts, CRMs like metformin have been associated with elevated risk based on genetic evidence from studies, highlighting a possible dual role in oncogenesis despite overall protective trends against other cancers. Ethical and regulatory limitations restrict CRM deployment for aging. Agents like metformin and rapamycin are approved only for and , respectively, making their use for anti-aging off-label and subject to legal and ethical scrutiny in nondiabetic populations, with calls for personalized dosing to mitigate risks. The absence of FDA approval for indications underscores the need for rigorous trials to address these barriers before widespread adoption.

Emerging Research Areas

Recent investigations into caloric restriction mimetics (CRMs) emphasize combination therapies to amplify their effects on key aging pathways, particularly . For instance, pairing synthetic agents like metformin, which activates AMPK and inhibits to induce , with natural compounds such as spermidine, which inhibits acetyltransferases to promote mitophagy, may yield complementary benefits by optimizing cellular recycling and reducing . Such multi-drug approaches are proposed to enhance neuroprotective outcomes in neurodegenerative disorders, though longitudinal studies are required to validate safety and efficacy. Advancements in are tailoring CRM applications through genetic screening to identify responders. Studies in genetically diverse mouse models reveal that genetic background accounts for approximately % of lifespan variation under caloric restriction, with a on influencing and . This variability underscores the need for screening to predict individual responses, enabling customized CRM regimens based on haplotypes like CAST/EiJ, which shorten lifespan. Novel CRM candidates in 2025 focus on senolytics like for health and modulators. , administered intravenously at 100 mg/kg twice weekly for eight weeks in aged sheep, significantly reduced senescent cells in the and , including neurons, , and , while lowering plasma S100B levels—a marker of aging—demonstrating its role as a CRM in mitigating neurodegeneration. Concurrently, -modulating CRMs such as metformin and enhance beneficial gut bacteria like , increasing short-chain production to strengthen the gut- axis and alleviate age-related cognitive decline. similarly boosts Firmicutes and Actinobacteria populations, activating Nrf2 for protection. In 2025, additional emerging areas include high-fiber diets as novel CRMs that mimic aging-related signatures of caloric restriction by altering metabolic and pathways, as shown in mouse models. CRMs are also being explored as therapeutics for autoimmune diseases, with preclinical evidence suggesting benefits in reducing without the adherence challenges of actual . Efforts to address translational gaps include studies and integration with . In grey mouse lemurs, a nonhuman model, chronic treatment (200 mg/kg/day) extended median lifespan by 1.5 years and improved cognitive and motor performance, yet it did not reduce age-related pathologies and induced atrophy, highlighting the need for dose optimization before human translation. Integrating CRMs with is emerging as a strategy for , where pharmacological mimetics prime cells for genetic interventions targeting pathways, potentially enhancing durable expression of protective factors like sirtuins. Broader applications of CRMs extend to extreme environments, including space travel for and to environmental stress. Pre-irradiation caloric restriction, mimicked pharmacologically, has shown potential to mitigate radiation-induced damage in mice by enhancing and reducing , offering a non-nutritional strategy for astronauts facing galactic cosmic rays. In environmental stress contexts, CRMs activate adaptive responses like , improving to stressors such as heat or through upregulated and antioxidant defenses, as evidenced in models of chronic exposure.