Hyperfunction as a Driver of Aging. Rather than resulting from cellular failure, aging often stems from the chronic overactivation of growth pathways. Persistent anabolic signaling drives hypertrophy, senescence, and inflammation—hallmarks of aging that arise from hyperfunction, not decline.
mTORC1 as a Lifespan-Limiting Pathway. Rapamycin’s inhibition of mTORC1 extends lifespan by 15–20% in mice when used alone. It achieves this by suppressing protein synthesis, reactivating autophagy, and redirecting cellular priorities away from growth and toward repair, especially in metabolically active tissues like the liver and muscle.
Ras–ERK as a Pro-Inflammatory Amplifier. Chronic activation of the Ras–MEK–ERK pathway contributes to inflammaging by amplifying cytokine signaling. In mice, trametinib treatment reduced expression of pro-inflammatory chemokines such as Cxcl11, Cd5l, and Ccl8 across multiple tissues, especially in female mice.
Dual Pathway Targeting as a Systems-Level Strategy. The 2025 Nature Aging study showed that combining rapamycin and trametinib extended lifespan by 34.9% in females and 27.4% in males—significantly exceeding either drug alone. This additive effect supports the need to modulate multiple aging pathways simultaneously.
Autophagy Restored by mTOR Inhibition. Rapamycin restores autophagic flux suppressed by mTORC1. In aging models, this promotes clearance of damaged mitochondria and protein aggregates. In the RAPACAT trial, cats treated with rapamycin showed increased markers of autophagy alongside reductions in left ventricular wall thickness by 17–22%.
MAPK Inhibition Dampens Inflammaging. Trametinib reduced microglial and astrocyte activation in the striatum, a brain region tied to cognitive function. Brain glucose uptake, which typically increases with age and neuronal stress, was normalized in treated mice—indicating preserved metabolic balance and reduced neuroinflammation.
Lifespan Gains Reflect Pathway Complementarity. Mice receiving both drugs exhibited a 30% average increase in lifespan compared to controls. This additive benefit was not due to higher drug burden but to non-overlapping effects on mTORC1 and Ras–ERK signaling—highlighting the advantage of targeting converging, but distinct, pathways.
Healthspan Improvements Go Beyond Longevity. Combination therapy reduced tumor incidence in liver and spleen, lowered systemic cytokine levels, and preserved muscle, kidney, and brain function. These improvements support the idea that dual-pathway inhibition compresses morbidity and delays functional decline.
Translational Barriers Remain for Trametinib. While effective in mice, trametinib’s side effects—such as hepatic steatosis, splenomegaly, and testicular degeneration at higher doses (11.52 mg/kg)—limit its use in healthy humans. A lower dose (1.44 mg/kg) achieved benefits with fewer side effects, but human trials are needed.
Toward Safer MAPK Modulation. The success of trametinib invites development of alternative strategies to modulate MAPK signaling. Naturally derived compounds like curcumin, EGCG, and quercetin may offer mild, chronic Ras–ERK suppression with more favorable safety profiles—though controlled clinical trials are still needed.
Introduction
Why do we age?
For decades, the dominant view held that aging was simply the result of wear and tear—a gradual breakdown of biological systems as damage accumulates faster than the body can repair it. But a growing body of research is challenging this idea, suggesting that aging may stem not just from loss, but from too much of a good thing running too long.
The very molecular programs that drive growth, development, and regeneration in youth—once essential for building the body—don’t shut off when their primary job is done. Instead, they keep humming in the background, promoting activity and proliferation in cells that no longer need to expand. Over time, this persistent activity becomes maladaptive. Like a construction crew that keeps adding scaffolding long after the building is finished, these once-helpful programs begin to disrupt structure, fuel inflammation, and accelerate tissue dysfunction.
This shift in perspective has given rise to a powerful new idea in aging biology: that aging is not merely the result of declining function, but also of overfunction—a state of cellular hyperactivity that drives disease from within.
As researchers have dug deeper into the molecular underpinnings of this process, they’ve turned their attention to compounds that can gently dial down these overactive systems. Known as geroprotectors, these interventions aim to restore balance—to slow the pace of aging by nudging cells out of chronic growth mode and back into states of rest, repair, and maintenance.
One of the most studied geroprotectors is rapamycin, a compound that inhibits key nutrient-sensing machinery in the cell and has consistently extended lifespan in a range of model organisms. But rapamycin targets only one arm of the body’s growth machinery. Could we do more by targeting multiple pathways simultaneously?
That question took center stage in 2025, when a groundbreaking study published in Nature Aging by Gkioni and colleagues tested a bold hypothesis: that combining rapamycin with trametinib, a cancer drug that blocks another major growth signal, might more effectively slow aging. The results were striking. Mice given the combination lived significantly longer than their untreated peers—regardless of sex. They also showed reductions in systemic inflammation, tumor burden, and age-related metabolic and neurological decline. [1]
The findings raised tantalizing questions: Are we approaching a new era of combination therapies for aging? Could dialing down multiple pro-growth signals at once deliver a more powerful longevity effect? And what would it take to translate this strategy safely into humans?
In this review, we explore the science behind this dual approach, unpack the molecular logic for targeting these intersecting pathways, and assess the opportunities—and challenges—that lie ahead as we move from animal models to clinical application.
The mTOR Pathway: Command Center for Growth, Energy, and Aging
The mechanistic Target of Rapamycin Complex 1 (mTORC1) is not a solitary molecule, but a multi-protein complex that functions as one of the cell’s most sophisticated decision-making hubs. Operating like a central command center, mTORC1 continuously monitors nutrient levels, cellular energy reserves, and hormonal cues such as insulin and IGF-1. When conditions signal abundance—particularly when amino acids like leucine are elevated, along with glucose and growth factors—mTORC1 shifts the cell into a state of growth and construction. It stimulates the synthesis of proteins, lipids, and nucleotides needed to build and replicate cellular infrastructure.
This growth-promoting machinery plays a crucial role in development and tissue repair. From infancy to adulthood, mTORC1 helps orchestrate the body’s construction—fueling cell proliferation, organ growth, and the buildup of energy reserves. In skeletal muscle, it stimulates protein synthesis essential for recovery from injury and exercise. In the liver and adipose tissue, it steers metabolism toward energy storage, stockpiling glycogen and triglycerides when fuel is abundant. This anabolic mode, optimized for regeneration and expansion, conferred clear evolutionary advantages during periods of growth, fertility, and physical stress—times when rapid cellular building was critical for survival and reproduction. [2]
Yet in the modern world, where nutrient availability is near-constant, this once-beneficial pathway is rarely given the chance to rest. The Standard American Diet (SAD)—rich in refined carbohydrates, saturated fats, and animal protein—repeatedly activates mTORC1, mimicking the cellular signals of continuous feast. Chronic stimulation, especially from amino acids like leucine, glucose, and insulin surges, drives mTORC1 into sustained overactivity.
The result is not simply enhanced tissue maintenance, but the unintended growth of dysfunctional tissues: hypertrophic adipocytes, enlarged senescent cells, and hyperplastic lesions that set the stage for chronic disease. What was once a finely tuned system of adaptive growth has become, under conditions of excess, a molecular engine for aging.
So we have established that mTOR is critical for our development into adulthood. But what happens when the cellular growth machinery keeps running long after the job is done?
This question lies at the heart of a conceptual revolution in the biology of aging.
Traditionally, we have believed that aging results from wear and tear and the gradual loss of cellular function. This understanding has been rooted in the idea that living organisms have a limited ability to repair themselves, leading to the accumulation of damage over time.
However, recent research has proposed an entirely different perspective, challenging the notion that aging results from the loss of cellular function. According to this counterintuitive theory, known as cellular hyperfunctionality, age-related diseases are caused by excessive cellular activity, not by a loss of function or damage to tissues. This shift in understanding has significant implications for how we approach the aging process and has captured the attention of longevity scientists.
The theory of hyperfunction in aging is a phenomenon popularized by Dr. Mikhail Blagosklonny; it aims to explain much of the aging process and the success of drugs like rapamycin through the lens of cellular overactivity.
Generally speaking, diseases of aging are characterized by cellular 'hyper-functions,' not the loss of cellular function or 'wear and tear' of tissues. Blagosklonny outlines three hyperfunctional features to describe the morphology and pathology of a dysfunctional cell. These include hyperplasia, hypertrophy, and hyperfunctionality:
Rather than viewing aging as a loss of function, hyperfunction theory posits that aging-related diseases arise when once-beneficial growth programs persist past their developmental window, leading to cellular overactivity that damages tissues over time. mTORC1 is central to this perspective. Instead of slowing down in adulthood, cells driven by mTORC1 continue producing proteins, expanding organelles, and synthesizing lipids—essentially engaging in runaway construction. The result isn’t renewal, but pathological remodeling.
Blagosklonny defines three morphological features of this dysfunction:
- Hypertrophy: cells grow larger in size due to sustained protein synthesis,
- Hyperplasia: senescent cells secrete mitogens that cause nearby cells to divide excessively,
- Hyperfunctionality: cells become metabolically overactive, secreting inflammatory and growth-promoting molecules well beyond what is needed.
Together, these states set the stage for many chronic diseases of aging. Consider the thickening of heart muscle in hypertrophic cardiomyopathy, the overactive osteoclasts in osteoporosis that accelerate bone loss, or the enlarged keratinocytes in aging skin that reduce elasticity and contribute to wrinkle formation. In each case, hyperfunction—not failure—drives dysfunction.
This framework helps explain why senescent cells, often thought of as “dead-end” cells due to their halted division, are in fact metabolically hyperactive. Unable to divide in response to growth stimuli, these cells instead swell in size and enter a state of persistent signaling, releasing a cocktail of pro-inflammatory and mitogenic factors known as the senescence-associated secretory phenotype (SASP). These secretions can stimulate nearby cells to divide uncontrollably, disrupt tissue architecture, and inflame surrounding environments. And because the immune system’s ability to clear senescent cells declines with age, these harmful messengers accumulate.
In this light, rapamycin’s ability to inhibit mTORC1 takes on new significance. By putting the brakes on this hyperfunctional overdrive, rapamycin doesn’t just slow aging—it may restore cellular self-regulation, reviving the capacity for rest, repair, and balanced responsiveness. It re-engages processes like autophagy, the cellular recycling program suppressed under mTORC1’s watch. This shift from unchecked growth to deliberate maintenance appears to be a common denominator in the lifespan-extending effects of rapamycin observed in model organisms ranging from yeast to mice.
Blagosklonny uses an elegant metaphor to illustrate this dynamic: the aging organism is like a car that has accelerated through development but never let off the gas. The engine is still revving long after the freeway ends—now careening through side streets, damaging its surroundings with every turn. The solution, then, is not to repair what’s broken, but to slow down before damage occurs.
By reframing aging as a consequence of overactivity of cellular functions, rather than inevitable breakdown, this view opens new therapeutic possibilities. mTORC1 emerges not merely as a growth controller, but as a molecular dimmer switch—one that can be turned down pharmacologically to prevent overgrowth, preserve function, and extend the period of life spent in good health.
The Ras–MEK–ERK Pathway: The Cellular Communication Highway
If mTORC1 is the foreman of the cell’s construction crew—deciding when to build and store—then the Ras–MEK–ERK pathway (also known as the MAPK/ERK cascade) is the messenger system, relaying information from the external world to the command center in the nucleus. It functions like a high-speed communication highway, transmitting real-time updates from growth factors, hormones, and stress cues directly to the cell’s genetic decision-makers.
The pathway begins at the cell surface. When external signals like epidermal growth factor (EGF) or insulin bind to receptors on the plasma membrane, they activate Ras, a molecular switch that flips the pathway into the “on” position. From there, the signal flows through a tightly controlled relay race of kinases: first to RAF, then to MEK (Mitogen-Activated Protein Kinase Kinase), and finally to ERK (Extracellular Signal-Regulated Kinase). At each step, phosphorylation—the addition of phosphate tags—activates the next runner in the chain, efficiently conveying the message from the outer membrane into the depths of the nucleus.
Once it reaches the nucleus, ERK modifies the activity of transcription factors—proteins that determine which genes are expressed. These gene expression changes guide some of the most fundamental cellular decisions: when to grow, divide, repair, or rest. During early development and tissue repair, the pathway is indispensable. It helps stem cells differentiate into specialized types, directs wound healing, and even prevents premature cell death (apoptosis) when survival is necessary. In short, it ensures that the cell’s responses are appropriately matched to the environment—dynamic, adaptive, and life-sustaining.[3]
When the Signal Won’t Shut Off: Ras–ERK and the Aging Process
Yet as with many of biology’s most elegant systems, balance is key. When the Ras–MEK–ERK pathway becomes chronically activated, its once-precise messaging turns into a blaring alarm—flooding the nucleus with signals that say “grow,” “divide,” and “stay alive,” even when those responses are no longer appropriate. This dysregulation is especially problematic with age, when tissue renewal must be carefully balanced with the need for cellular stability.
Persistent Ras–ERK activation is one of the most commonly observed features in cancer, where mutations lock Ras or upstream receptors in the “on” position, resulting in uncontrolled proliferation. But beyond tumor biology, the pathway’s overactivation has far-reaching implications for aging. It promotes a state of chronic, low-grade inflammation—known as inflammaging—by amplifying the production of cytokines and inflammatory mediators. It also encourages cellular senescence, not by halting growth, but paradoxically by driving hyperfunctional cells into a stressed, inflammatory state that contributes to tissue damage.
In neurons, for instance, aberrant MAPK signaling has been linked to excessive tau phosphorylation, a key feature in Alzheimer’s disease pathology. In the vasculature, it contributes to arterial stiffening, endothelial dysfunction, and vascular remodeling, all of which raise the risk of hypertension and cardiovascular disease. In essence, what was once an adaptive survival mechanism becomes, with time and overuse, a molecular contributor to degeneration.
Targeting the Pathway: Trametinib as a Molecular Brake
Enter trametinib, a selective MEK inhibitor originally developed for the treatment of melanoma. Trametinib binds to and inhibits MEK1/2, effectively interrupting the kinase relay before it can activate ERK. This targeted intervention allows researchers to dial down the volume on MAPK signaling without fully silencing it—attenuating harmful overactivity while preserving essential cellular functions.
Recent research suggests that trametinib’s benefits may extend far beyond oncology. In preclinical aging studies, inhibiting MEK in aged mice not only reduced tumor burden but also improved metabolic parameters, lowered systemic inflammation, and preserved cognitive function. The idea is not to stop communication altogether, but to filter the noise—restoring a level of signaling precision that resembles a younger, more responsive system. [4]
The Synergy of Rapamycin and Trametinib in Drosophila Studies
Before the mouse studies drew broader attention, foundational insights into the synergy between rapamycin and trametinib emerged from research in Drosophila melanogaster, a well-established model organism in aging science. Owing to their short lifespans, conserved genetic pathways, and ease of genetic manipulation, fruit flies have long been instrumental in identifying mechanisms that regulate longevity.
A pivotal study titled "A triple drug combination targeting components of the nutrient-sensing network maximizes longevity" examined the effects of rapamycin, trametinib, and lithium (individually, in pairs, and all together) on fruit fly lifespan and healthspan. [5] Each of these compounds targets a distinct arm of cellular signaling: rapamycin inhibits mTORC1, trametinib blocks MEK in the Ras–MEK–ERK pathway, and lithium inhibits glycogen synthase kinase-3 (GSK-3), a key enzyme in metabolism and cellular regulation.
The researchers found that all three drugs extended lifespan when administered alone, resulting in an average increase of approximately 11%. However, the combinations were notably more potent. Pairing rapamycin with trametinib, lithium with rapamycin, or lithium with trametinib led to roughly a 30% increase in lifespan.
The most striking result came from the triple-drug combination. When all three compounds were used together, the median lifespan of fruit flies increased by 48%, far surpassing the effects of individual or dual-drug treatments. This finding underscored the value of targeting multiple aging-related pathways simultaneously and showed that the drugs contributed independently to the overall benefit.
Figure 1. Median and maximum lifespan (in days) of Drosophila melanogaster across different treatment groups. The combination of trametinib, rapamycin, and lithium produced the longest lifespan extension, followed by the dual combination of trametinib and rapamycin, and then trametinib and lithium.
To rule out alternative explanations, the researchers evaluated whether behavioral or metabolic artifacts could explain the longevity gains. Food intake, feeding behavior, and drug absorption remained unchanged across groups, confirming that the lifespan extension was not due to caloric restriction or altered nutrient uptake. They also explored potential trade-offs with reproduction, a common concern in aging interventions. While trametinib and trametinib-containing combinations did reduce egg-laying, the triple combination did not further suppress fecundity beyond what trametinib alone caused, suggesting that the extended lifespan was not simply a consequence of reduced reproductive investment.
One particularly interesting insight was lithium’s ability to counteract some of rapamycin’s metabolic side effects. In fruit flies, as in other organisms, rapamycin can cause hypertriglyceridemia and fat accumulation. However, when lithium was co-administered with rapamycin, these adverse effects were reversed. While lithium alone did not alter fat levels, it appeared to buffer against rapamycin-induced metabolic disruption—a finding with potential translational relevance for minimizing side effects in humans.
Figure 2. Triglyceride levels in Drosophila under different treatment conditions. While rapamycin alone led to elevated triglycerides, simultaneous treatment with lithium and rapamycin reversed this dyslipidemia. This suggests lithium may help mitigate one of rapamycin’s potential metabolic side effects.
Together, the results from this study highlight a central tenet of modern aging biology: the complexity of aging likely cannot be addressed by a single intervention. Instead, targeting multiple, conserved signaling pathways through carefully selected combinations may provide a more effective and balanced strategy for extending lifespan.
Combination Treatment with Rapamycin and Trametinib Extends Lifespan and Healthspan in Mice
Building on the promising findings from Drosophila, researchers turned to more complex mammalian models to assess whether the synergy between rapamycin and trametinib would hold. A 2025 study published in Nature Aging, titled "The geroprotectors trametinib and rapamycin combine additively to extend mouse healthspan and lifespan" by Gkioni et al., took this next step by examining the effects of these two compounds, both individually and in combination, in mice. [1]
Individually, both rapamycin and trametinib have already shown the ability to extend lifespan in murine models. Rapamycin consistently increases average lifespan by approximately 15–20% when administered alone. Trametinib, initially validated in Drosophila, was also found to extend lifespan in mice by approximately 5–10% on its own.
The most striking outcome from the Gkioni study, however, emerged when the two drugs were used in combination. Mice treated with both rapamycin and trametinib experienced an average lifespan extension of around 30%, with female mice seeing a 34.9% increase and male mice a 27.4% increase. These results provide compelling evidence of an additive and potentially synergistic effect—one that exceeds the sum of the individual treatments. Importantly, the enhanced longevity was not attributed merely to a higher overall drug burden, but rather to distinct and complementary effects that emerged only in the combined treatment condition.
Figure 3. Survival curves of male and female mice fed a control diet, rapamycin alone, trametinib alone, or a combination of both, beginning at 6 months of age. As illustrated in Figure 2, both rapamycin and trametinib extended lifespan when administered individually. However, their combined use yielded the most significant benefits.
The findings from this study that targeting both the mTORC1 and Ras–MEK–ERK pathways simultaneously may engage a broader and more effective anti-aging response than inhibiting either pathway alone. It also supports a larger strategic insight in longevity science: multi-pathway modulation may be necessary to capture the full complexity of aging biology.
Beyond lifespan extension, the combination therapy also meaningfully improved healthspan—the length of time an organism remains healthy and free from serious disease or functional decline. One of the most notable benefits observed was a reduction in systemic, age-related inflammation. Often referred to as “inflammaging,” this persistent low-grade inflammation plays a central role in the progression of age-related diseases, including cardiovascular disease, cancer, and neurodegeneration. [6] The study found that inflammation was reduced across multiple organ systems, including the brain, kidney, spleen, and muscle.
This effect was reflected at the molecular level. Pro-inflammatory cytokines and chemokines—including CD5 antigen-like (Cd5l), C-C motif chemokine ligand 8 (Ccl8 or Mcp2), and various C-X-C motif chemokines like Cxcl7, Cxcl11, and Cxcl13—were all downregulated; however, this effect was significantly more pronounced in female mice. These signaling molecules normally promote immune cell recruitment and inflammatory amplification; therefore, their reduction suggests a more balanced and less inflammatory tissue environment in treated mice.
The combination therapy also offered notable protection against age-related cancers. As animals age, their risk of tumor formation rises due to cumulative genetic and cellular damage. Mice treated with both drugs had significantly fewer liver tumors—among the most common spontaneous tumors in aging mice—and a marked reduction in spleen tumors, particularly in males. This suggests that dual-pathway targeting may help suppress both the initiation and progression of cancer by simultaneously modulating cell proliferation and inflammation.
Brain health was another area of improvement. Typically, aging is associated with increased brain glucose uptake, which can reflect metabolic inefficiency or neuronal stress. In the treated mice, this age-related rise in brain glucose uptake was blocked, suggesting improved metabolic stability. Moreover, the combination therapy reduced activation of microglia and astrocytes, key support cells in the brain that, when overactivated, contribute to neuroinflammation and cognitive decline. These changes were particularly notable in the striatum, a brain region involved in motor control and cognitive processing.
Safety and Translational Considerations: Why Trametinib Is Not Yet Fit for Human Geroprotection
While the benefits of trametinib and rapamycin in extending both lifespan and healthspan in animal studies are compelling, it is essential to weigh these findings against their limitations, especially when considering translation to human use. One of the primary concerns is the safety profile of trametinib, a drug developed for oncology with well-documented adverse effects. [7]
Trametinib is currently FDA-approved for treating several aggressive cancers, most notably melanoma, a fast-growing and potentially fatal skin cancer that originates in melanocytes, the pigment-producing cells of the skin. [8] In approximately half of melanoma cases, mutations occur in the BRAF gene, most commonly the BRAF V600E variant, which drives uncontrolled cell proliferation through aberrant activation of the MAPK pathway. While trametinib does not directly inhibit BRAF, it targets MEK, a downstream kinase in the same pathway, thereby effectively disrupting the proliferative signaling cascade.
In oncology settings, trametinib is administered at relatively high doses and often for extended durations, conditions under which a variety of adverse effects are common. These include dermatologic issues such as acneiform rashes, gastrointestinal symptoms like diarrhea, nausea, and vomiting, as well as systemic effects including fatigue, peripheral edema, hypertension, hepatic enzyme elevations, and, in some cases, cardiotoxicity. [2] As a result, patients on trametinib typically undergo regular clinical monitoring to manage these toxicities.
This raises an important question in the context of geroscience: even if a drug shows potential to extend lifespan, is it tolerable enough for long-term use in otherwise healthy individuals? The risk-benefit ratio becomes fundamentally different when the goal is disease prevention rather than treatment.
To address these concerns, researchers evaluating trametinib in mouse aging studies closely monitored for signs of toxicity across dosing ranges. At higher doses (e.g., 11.52 mg/kg of food), mice exhibited notable side effects, including body weight loss, splenomegaly, and hepatic steatosis—commonly referred to as fatty liver disease. This condition, characterized by the abnormal accumulation of fat within liver cells, is increasingly common in aging populations and is associated with metabolic dysfunction, insulin resistance, and type 2 diabetes. [9] Additionally, high-dose trametinib was linked to testicular degeneration, suggesting potential impairment of reproductive function.
However, the study also identified a lower, optimized dose—1.44 mg/kg of food—that achieved partial inhibition of the Ras/MAPK pathway without inducing widespread toxicity. At this dose, mice maintained normal behavior and physiology, with no significant impairments in liver function or overt signs of distress. These findings suggest that low-dose trametinib may have a therapeutic window in which aging-related benefits can be achieved with manageable side effects, at least in preclinical models.
Still, it must be emphasized: mice are not humans. [10] While murine models are indispensable for identifying biological mechanisms of aging, they do not always predict human responses to drugs. The human body is shaped by a vastly more complex interplay of genetics, environment, lifestyle, and disease history. A treatment that extends mouse lifespan by 30% might not translate to decades of human life extension, nor will the safety margins be the same. This point is especially salient when considering long-term use in healthy populations.
Early clinical trials are now underway to evaluate rapamycin and related compounds in healthy older adults. These studies will be essential for assessing long-term safety, refining dosage strategies, and determining the extent to which the benefits observed in animal models translate to humans. While trametinib has shown potential to enhance the effects of rapamycin in preclinical settings, it remains an adjunct, one with a far more limited safety profile and no established role in preventive medicine. For now, rapamycin stands on firmer ground as a candidate for human geroprotection.
Conclusion: Dual Pathway Targeting Holds Promise, But Translation Remains Complex
The combination of rapamycin and trametinib represents a compelling example of how modulating multiple, conserved aging pathways can produce additive benefits in preclinical models. By jointly targeting mTORC1 and the Ras–MEK–ERK cascade, this dual-drug strategy not only extended lifespan in mice and flies but also improved markers of systemic inflammation, brain health, and tumor suppression.
However, trametinib’s side effect profile, rooted in its role as a cancer therapeutic, limits its suitability for preventive use in otherwise healthy individuals. While low-dose regimens may hold theoretical promise, substantial translational barriers remain.
That said, the MAPK pathway need not be abandoned altogether. Several naturally derived compounds, such as curcumin, EGCG, and quercetin, have been shown to modulate Ras/MAPK signaling indirectly and at lower intensities. While their effects are modest compared to pharmacologic inhibitors like trametinib, they offer favorable safety profiles and potential for long-term use, especially as part of a broader healthspan strategy. Still, these compounds require more rigorous evaluation in controlled trials before they can be confidently positioned as geroprotective agents.
Ultimately, while the percentage gains seen in model organisms are striking, translating these findings to humans is far from straightforward. The complexity of human physiology, combined with the ethical and safety constraints of long-term interventions, means that enthusiasm must be matched by caution and rigorous research. Rapamycin, with its expanding human trial data, remains the most viable candidate for now. But as our understanding of interconnected aging pathways deepens, more nuanced, multi-target approaches will likely emerge—and with them, the possibility of safely targeting the Ras/MAPK pathway directly, using more selective molecules and carefully optimized dosing strategies.
- Gkioni, L., Nespital, T., Baghdadi, M., Monzó, C., Bali, J., Nassr, T., Cremer, A. L., Beyer, A., Deelen, J., Backes, H., Grönke, S., & Partridge, L. (2025). The geroprotectors trametinib and rapamycin combine additively to extend the healthspan and lifespan of mice. Nature aging, 10.1038/s43587-025-00876-4. Advance online publication. https://doi.org/10.1038/s43587-025-00876-4
- Wang, G., Chen, L., Qin, S., Zhang, T., Yao, J., Yi, Y., & Deng, L. (2022). Mechanistic Target of Rapamycin Complex 1: From a Nutrient Sensor to a Key Regulator of Metabolism and Health. Advances in nutrition (Bethesda, Md.), 13(5), 1882–1900. https://doi.org/10.1093/advances/nmac055
- Li, L., Zhao, G. D., Shi, Z., Qi, L. L., Zhou, L. Y., & Fu, Z. X. (2016). The Ras/Raf/MEK/ERK signaling pathway and its role in the occurrence and development of HCC. Oncology letters, 12(5), 3045–3050. https://doi.org/10.3892/ol.2016.5110
- Lugowska, I., Koseła-Paterczyk, H., Kozak, K., & Rutkowski, P. (2015). Trametinib: a MEK inhibitor for management of metastatic melanoma. OncoTargets and therapy, 8, 2251–2259. https://doi.org/10.2147/OTT.S72951
- Castillo-Quan, J. I., Tain, L. S., Kinghorn, K. J., Li, L., Grönke, S., Hinze, Y., Blackwell, T. K., Bjedov, I., & Partridge, L. (2019). A triple drug combination targeting components of the nutrient-sensing network maximizes longevity. Proceedings of the National Academy of Sciences of the United States of America, 116(42), 20817–20819. https://doi.org/10.1073/pnas.1913212116
- Ferrucci, L., & Fabbri, E. (2018). Inflammageing: chronic inflammation in ageing, cardiovascular disease, and frailty. Nature Reviews. Cardiology, 15(9), 505–522. https://doi.org/10.1038/s41569-018-0064-2
- Ureña, E., Xu, B., Regan, J. C., Atilano, M. L., Minkley, L. J., Filer, D., Lu, Y. X., Bolukbasi, E., Khericha, M., Alic, N., & Partridge, L. (2024). Trametinib ameliorates aging-associated gut pathology in Drosophila females by reducing Pol III activity in intestinal stem cells. Proceedings of the National Academy of Sciences of the United States of America, 121(4), e2311313121. https://doi.org/10.1073/pnas.2311313121
- Hoffner, B., & Benchich, K. (2018). Trametinib: A Targeted Therapy in Metastatic Melanoma. Journal of the advanced practitioner in oncology, 9(7), 741–745. https://pmc.ncbi.nlm.nih.gov/articles/PMC6570520/
- Nassir, F., Rector, R. S., Hammoud, G. M., & Ibdah, J. A. (2015). Pathogenesis and Prevention of Hepatic Steatosis. Gastroenterology & hepatology, 11(3), 167–175. https://pubmed.ncbi.nlm.nih.gov/27099587/
- Fischer M. (2021). Mice Are Not Humans: The Case of p53. Trends in cancer, 7(1), 12–14. https://doi.org/10.1016/j.trecan.2020.08.007