Rapamycin
The most powerful tool to stop the acceleration of aging caused by mTOR dysfunction and cellular senescence.
Long COVID, a condition characterized by persistent symptoms such as fatigue, cognitive impairments, and systemic inflammation, presents a significant global health challenge with limited treatment options. Recent interest in rapamycin as a potential therapeutic has been fueled by emerging anecdotal reports, including a notable viral social media account describing a patient’s recovery from severe post-exertional malaise (PEM) after using the drug. Rapamycin, traditionally studied for its geroprotective and cellular-modulating properties, inhibits the mechanistic target of rapamycin (mTOR) pathway, promoting autophagy, reducing immune dysregulation, mitigating chronic inflammation, and enhancing mitochondrial function. These mechanisms align with the pathophysiology of long COVID and are supported by insights from related conditions like Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). This research review aims to explore the scientific foundation for rapamycin’s use in addressing the multifactorial pathology of long COVID. By examining its mechanisms of action and insights from related conditions such as ME/CFS, the review provides a basis for understanding how rapamycin may offer a novel therapeutic approach for this debilitating condition.
rapamycin
Anti-Inflammation
long COVID
32 mins
By: Kristen Race, Shriya Bakhshi, Dr Ryan N. Marshall, PhD.
The COVID-19 pandemic, which emerged in 2020, introduced profound health, economic, and psychosocial challenges worldwide. As the acute phase of the pandemic receded, a new and complex challenge emerged: long COVID.
Long COVID is characterized by persistent symptoms, including dizziness, headaches, palpitations, chest pain, fatigue, and cognitive impairments. Affecting millions globally, the condition often severely impacts quality of life. Despite growing recognition of its prevalence, treatment options remain limited, leaving patients and healthcare providers grappling with uncertainty.
Amid this backdrop, social media has become an unexpected source of shared insights and anecdotal evidence. One particularly notable viral account on Reddit, titled "From bed bound with severe PEM to playing basketball in 6 weeks: a rapamycin success story," recounts a long COVID patient’s dramatic recovery. After being bedridden with severe post-exertional malaise (PEM), the individual attributed their improvement to the use of rapamycin. While rapamycin is well-documented as a geroprotective molecule targeting cellular pathways integral to healthspan, its application in treating long COVID and related chronic inflammatory conditions remains largely unexplored.
Emerging evidence suggests that long COVID shares mechanisms with other chronic inflammatory conditions, including systemic inflammation, immune dysregulation, and cellular dysfunction—pathways that rapamycin is known to target. Research into related conditions, such as Myalgic Encephalomyelitis and Chronic Fatigue Syndrome (ME/CFS), further supports its potential to modulate these processes and offers a scientific foundation for its reported benefits.
In this research review, Shriya Bakhshi and Kristen Race of the Healthspan clinical team explore the science behind rapamycin and its potential as a targeted treatment for long COVID. By examining its mechanisms of action—including the modulation of inflammation, immune responses, and cellular repair—this discussion seeks to provide insights into its potential as a therapeutic intervention for those grappling with the complexities of post-viral recovery.
Long COVID, also known as post-acute sequelae of SARS-CoV-2 infection (PASC), corresponds to a condition where individuals experience persistent symptoms and health issues for weeks or months after the acute phase of a COVID-19 infection has resolved. [1]
While many people recover from COVID-19 within a few weeks, some individuals, even those with initially mild cases, continue to experience a range of symptoms that can significantly impact their quality of life. It is worth noting that a person's susceptibility to long COVID is independent of the severity of their original COVID-19 infection [2]. Common symptoms of long COVID include fatigue, shortness of breath, chest pain, concentration impairments, joint pain, and persistent loss of taste or smell. Interestingly, long COVID can affect people of all ages, including those who had mild or asymptomatic cases of COVID-19 initially. [1]
Researchers have proposed several interrelated hypotheses to explain the underlying mechanisms of long COVID. Emerging evidence suggests that the condition is not confined to a single organ system but is instead systemic in nature, involving complex interactions among multiple physiological and psychological components. This multi-system involvement underscores the complexity of its etiology and clinical presentation.
A pivotal study conducted in the United Kingdom has provided valuable insights into the systemic nature of long COVID and its far-reaching effects on the body. Between April 1 and September 14, 2020, researchers assessed a cohort of individuals experiencing persistent symptoms following recovery from acute SARS-CoV-2 infection. Participants underwent comprehensive evaluations, including standardized symptom questionnaires, biochemical assays, and advanced quantitative magnetic resonance imaging (MRI), to determine the extent and scope of organ dysfunction.
The study revealed that 70% of individuals classified as having a low risk of mortality from COVID-19 exhibited measurable impairments in one or more organs. Multi-organ dysfunction—encompassing the heart, lungs, kidneys, liver, pancreas, and spleen—was identified in 29% of the cohort. These findings underscore the complex, multi-systemic etiology of long COVID, illustrating that its impact extends beyond a single organ system to encompass widespread physiological disruption.
To better understand the complex and persistent symptoms of long COVID, it is essential to examine the biological mechanisms driving this condition. When we analyze the underlying mechanisms, long COVID is not merely an acute infection but a multifaceted syndrome with systemic implications. Research increasingly points to several interconnected pathways, including viral persistence, immune system dysregulation, and vascular dysfunction, as key contributors to its pathology. In the following sections, we will examine these pathways in greater detail to uncover how they contribute to the chronic symptoms experienced by long COVID patients.
Viral Persistence
SARS-CoV-2, the virus responsible for COVID-19, gains entry into the human body by infecting cells in the respiratory system. These cells are equipped with specialized surface proteins known as receptors, which function as docking stations to bind specific molecules. The interaction between a receptor and its matching molecule initiates cellular processes that vary depending on the receptor type and the molecule involved.
In the case of SARS-CoV-2, its spike protein—a prominent feature of the virus's outer surface—plays a critical role in infection. This spike protein contains a segment known as the receptor-binding domain (RBD), which is specifically designed to bind to the angiotensin-converting enzyme 2 (ACE2) receptor. ACE2 receptors are abundantly expressed on cells in the respiratory system, particularly in the nasal passages and upper respiratory tract. Once the RBD attaches to the ACE2 receptor, it triggers a conformational change in the spike protein, akin to unlocking a door, allowing the virus to fuse with the host cell membrane. This fusion facilitates viral entry into the cell, where the virus can hijack the host's machinery to replicate and propagate the infection.
Notably, ACE2 receptors are not limited to the respiratory tract. They are also present in diverse tissues, including the kidneys, gastrointestinal tract, esophagus, liver, and brain, rendering these organs susceptible to SARS-CoV-2 infection. This widespread distribution of ACE2 receptors helps explain the multi-organ involvement seen in both acute COVID-19 and long COVID.
Adding to the complexity, viral RNA—the genetic material of SARS-CoV-2—has been shown to persist in the body long after clinical recovery from the acute infection. This persistence is not limited to the respiratory system; viral RNA has been detected in the gastrointestinal tract, blood, and other tissues for months following initial infection. Such persistence raises concerns about its role in driving the prolonged symptoms characteristic of long COVID.
A meta-analysis published in The Lancet titled "SARS-CoV-2, SARS-CoV, and MERS-CoV viral load dynamics, duration of viral shedding, and infectiousness: a systematic review and meta-analysis" provides valuable insights into the dynamics of viral shedding—the process by which virus particles are released from infected cells and expelled from the body. The study quantified the duration of shedding across various anatomical sites, revealing that SARS-CoV-2 can persist in the body for extended periods even after clinical recovery. On average, viral shedding was observed for up to 17 days in the upper respiratory tract (nose and throat), approximately 14.6 days in the lower respiratory tract (lungs), around 17.2 days in the gastrointestinal tract, and about 1.6 days in the blood. Shedding mechanisms vary depending on the site, such as through coughing for the respiratory tract or excretion for the gastrointestinal tract. [4]
The persistence of viral RNA—the genetic blueprint of SARS-CoV-2—alongside prolonged viral shedding is emerging as a key factor in the pathophysiology of long COVID. Viral RNA remnants may not represent active infection but can nonetheless stimulate ongoing immune responses, potentially triggering low-grade inflammation and disrupting normal physiological processes. These interactions are thought to play a central role in sustaining the chronic symptoms associated with long COVID, such as fatigue, cognitive dysfunction, and multi-organ involvement. [4]
By demonstrating the prolonged presence of SARS-CoV-2 RNA across multiple organ systems, this meta-analysis underscores the systemic nature of long COVID. The findings suggest that viral remnants may act as a persistent irritant, perpetuating immune dysregulation and contributing to the protracted and diverse symptomatology observed in affected individuals. These insights highlight the importance of addressing viral persistence as part of a comprehensive strategy for understanding and treating long COVID. [4]
Immunological Factors
Long COVID might also be explained by changes in how the immune system works, specifically regarding T cells, which are crucial defenders of our immune system. T cells are critical components of the adaptive immune system, orchestrating targeted immune responses to infections. They can be broadly categorized into two main types based on their distinct roles:
Cytotoxic T cells (CD8+ cells): Often described as the immune system’s soldiers, these cells identify and destroy infected host cells, thereby halting the replication and spread of viruses like SARS-CoV-2.
Helper T cells (CD4+ cells): These cells function as commanders within the immune system, coordinating the response by activating and directing other immune cells, such as B cells (which produce antibodies) and macrophages (which engulf and destroy pathogens).
When T cells encounter a pathogen like SARS-CoV-2, they multiply and differentiate into specialized subsets, including memory T cells. Memory T cells are particularly critical because they "remember" the pathogen, enabling a faster and more efficient immune response if the body encounters the same virus again. They can quickly differentiate into either helper or cytotoxic T cells, guiding the immune system to the infection site to fight off the virus effectively.
However, studies on long COVID have identified significant changes in T cell functionality, particularly in memory T cells. These alterations may explain the prolonged and systemic symptoms seen in some patients. One study tracked T cell responses in individuals who had recovered from COVID-19 with varying levels of disease severity—mild, moderate, and severe—at two time points: 3 months and 6 months post-infection.
The researchers noticed that the T cell response varied depending on the severity of the COVID-19 case and the time since recovery. In severe cases, there was a significant change towards a state of 'T cell exhaustion' in both CD4+ and CD8+ T cells.
To understand 'T cell exhaustion,' imagine a long-distance runner who starts energetically but gets tired and less efficient over time. Similarly, T cells initially respond vigorously to a virus but can become exhausted and less effective if the virus or its remnants, like RNA, linger for a long time, as in long COVID. In the study, T cells in severe cases showed clear signs of this exhaustion, impairing their two main functions: activating other immune cells and directly killing infected cells.
Additionally, in severe cases, CD8+ T cells were found to produce more granzyme B and IFN-γ after six months. Granzyme B helps destroy virus-infected cells, while IFN-γ attracts other immune cells, like macrophages, to the infection site. Macrophages act like vacuums, engulfing and destroying virus-infected cells. This process is called phagocytosis. The increased production of these proteins in severe cases was linked to higher inflammation, which might be related to the prolonged symptoms of COVID-19.
Interestingly, mild cases of COVID-19 exhibited a different pattern. Researchers observed an increase in CD4+ regulatory T cells (Tregs), which help modulate the immune response by suppressing overactive immune cells. While this suppression is generally protective, an overabundance of Tregs could lead to immune dysregulation, insufficient viral clearance, and lingering inflammation. This dysregulation may contribute to long COVID symptoms, such as fatigue and cognitive dysfunction, even in patients who experienced relatively mild acute infections. [5]
These findings highlight the complexity of immune responses in long COVID, particularly the role of T cell dysfunction. In severe cases, T cell exhaustion and persistent inflammation appear to drive chronic symptoms, while in milder cases, immune dysregulation may be the primary contributor.
Vascular and Microvascular Effects
In addition to its impact on the immune system, SARS-CoV-2 has significant effects on the circulatory system, particularly on the function of blood vessels. The virus enters human cells through the ACE2 receptor, which is expressed on the surface of various cell types in the body, including those in the lungs, blood vessels, and small intestine. [6]
ACE2 serves as a critical entry point for the virus, enabling SARS-CoV-2 to infect and replicate within host cells. This interaction disrupts normal ACE2 function, which plays a key role in regulating vascular homeostasis. The resulting dysregulation can lead to inflammation and damage to the endothelial cells lining the blood vessels, triggering a condition known as microvascular dysfunction. [7]
Microvascular dysfunction involves impaired blood flow within the smallest blood vessels (capillaries), which compromises oxygen and nutrient delivery to tissues. This condition can contribute to symptoms such as fatigue, shortness of breath, and cognitive impairments, which are hallmarks of long COVID. Furthermore, endothelial inflammation can exacerbate the pro-inflammatory environment, creating a feedback loop that sustains vascular damage and systemic inflammation.
Another significant issue related to COVID-19 is coagulopathy, which affects how blood clots [8, 9]. Under normal circumstances, blood clotting is a tightly regulated process that prevents excessive bleeding by forming clots at sites of vascular injury. However, in coagulopathy, this balance is disrupted, leading either to excessive bleeding or, more commonly in COVID-19, to a hypercoagulable state where the blood is abnormally prone to clotting. [8, 9]
Patients with long COVID often exhibit signs of hypercoagulability, which can result in various thrombotic conditions. One such condition is deep vein thrombosis (DVT), where blood clots form in deep veins, typically in the legs. Symptoms of DVT include swelling, pain, and redness in the affected area. If a clot dislodges and travels through the bloodstream to the lungs, it can cause a pulmonary embolism (PE)—a life-threatening condition characterized by shortness of breath, chest pain, and coughing up blood. [10]
Hypercoagulability in long COVID patients is believed to stem from a combination of factors, including persistent endothelial inflammation, microvascular damage, and immune activation. Elevated levels of procoagulant factors and dysregulated fibrinolysis—the process that dissolves blood clots—further contribute to the risk of abnormal clot formation. These processes not only increase the risk of acute thrombotic events but may also play a role in the chronic symptoms of long-term COVID by impairing oxygen delivery and tissue perfusion.
A detailed longitudinal study investigating the role of coagulopathy in long COVID tracked patients from hospital admission through discharge and at follow-up intervals of 3 and 6 months. The researchers focused on measuring D-dimer levels, a biomarker that reflects the breakdown of fibrin, the main protein involved in blood clot formation. Elevated D-dimer levels are indicative of recent or ongoing clotting activity, making them a valuable tool for assessing thrombotic risk.
The study revealed that D-dimer levels were persistently elevated during the recovery phase, often exceeding levels observed at the time of hospital admission. These findings suggest that the clotting abnormalities associated with acute COVID-19 do not resolve entirely and may persist well into the post-acute phase, contributing to the chronic symptoms of long COVID. [10]
The persistence of heightened D-dimer levels underscores the role of ongoing vascular and coagulatory dysfunction in long COVID. This prolonged thrombotic activity could impair microvascular circulation, reducing oxygen delivery and nutrient transport to tissues. Such disruptions may exacerbate symptoms like fatigue, cognitive impairments, and shortness of breath, which are hallmark features of long COVID. [10]
The multifaceted impact of the SARS-CoV-2 virus on blood vessel function and coagulation processes not only elucidates the complexities of its pathogenesis but also sheds light on the intricate mechanisms contributing to the persistent and diverse symptoms observed in long COVID cases.
Now that we have reviewed the intricate mechanisms underlying long COVID—encompassing immune dysregulation, inflammation, vascular dysfunction, and mitochondrial impairment—let’s examine how rapamycin addresses these underlying pathologies and its potential as a novel therapeutic for this complex condition.
Long COVID is a complex, multifactorial condition characterized by systemic inflammation, immune dysregulation, cellular damage, and energy deficits, presenting significant challenges for effective treatment. Rapamycin, a pharmacologic agent with well-established anti-aging and cellular-modulating properties, is gaining attention as a potential therapeutic intervention. Its ability to target key cellular pathways that regulate immune function and autophagy, particularly through inhibition of the mechanistic target of rapamycin (mTOR), positions it as a promising candidate for addressing the underlying pathophysiological mechanisms of long COVID.
By modulating the mTOR pathway, rapamycin exerts diverse therapeutic effects, including suppression of inflammatory signaling, restoration of immune homeostasis, enhancement of mitochondrial function, and promotion of cellular repair. This section provides a comprehensive analysis of these mechanisms—mTOR inhibition, anti-inflammatory activity, immune system modulation, and mitochondrial support—and examines their potential role in mitigating the clinical manifestations and pathologies associated with long COVID.
To begin, let’s review the very foundation of how rapamycin works—its regulation of the mTOR pathway.
Rapamycin is a macrolide compound that has been shown to have a wide range of biological effects, including immunosuppressive, anti-cancer, and longevity properties [22]. Notably, rapamycin was 1 of only 10 (out of 64) therapeutics tested so far as part of the Interventions Testing Programme (ITP) at the National Institute on Aging (NIA) that improved lifespan [23]. Other successful therapeutics were Acarbose [24], Aspirin [25], Canagliflozin [26], Glycine [27], Protandim [28], 17α-estradiol [24] and nordihydroguaiaretic acid [25]. Since then, several research groups from around the world have begun to demonstrate that the off-label repurposing of rapamycinmay also have potential as a therapeutic for preventing many non-communicable diseases [22].
The primary mechanism in which rapamycin has a beneficial therapeutic effect is by inhibiting the activity of the mammalian target of rapamycin (mTOR), a key regulator of cellular metabolism, growth, and proliferation. Specifically, rapamycin binds to a protein called FK506-binding protein 12 (FKBP12), which then dephosphorylates mTOR, thereby 'turning off' downstream growth signals such as P70S6K & ribosomal complexes needed for cellular growth [29].
mTOR functions as a master regulator of cellular growth, acting like a switch that activates in response to signals of nutrient abundance or growth factors. When activated, mTOR directs cellular machinery to prioritize growth and division, processes essential for tissue repair and development in growing organisms.
However, when nutrients are scarce, mTOR activity decreases, which triggers the cell's autophagy machinery to assemble to break down and recycle its components to generate energy and nutrients. By initiating autophagy, the cell can maintain its energy balance and survive during times of nutrient scarcity.
Chronic overactivation of mTOR is implicated in the pathogenesis of many age-related diseases. As individuals age, mTOR activity may remain excessively high, fueling unchecked cellular growth, reducing repair capacity, and contributing to systemic inflammation and immune dysregulation. This hyperactivity is linked to the decline of healthy tissue, cancer proliferation, and the accumulation of cellular damage. In the context of long COVID, these same mechanisms—chronic inflammation, immune dysregulation, and cellular dysfunction—are key drivers of the persistent symptoms experienced by patients.
By inhibiting mTOR, rapamycin restores mTOR activity to more balanced, youthful levels and promotes autophagy. This process clears toxic cellular debris and enhances cellular repair, addressing critical hallmarks of aging, including:
Improved proteostasis: Reduced overactive protein synthesis and enhanced protein quality control.
Enhanced mitochondrial function: Lowered reactive oxygen species (ROS) production and improved energy dynamics. [20, 21]
Better metabolic health: Increased metabolic efficiency and reduced systemic inflammation. [22]
These effects collectively support cellular health and resilience, offering wide-ranging therapeutic benefits.
Rapamycin’s ability to inhibit mTORC1 and shift cellular priorities from growth to repair and maintenance makes it a compelling candidate for treating long COVID. By promoting autophagy, rapamycin enhances the clearance of damaged organelles, proteins, and other cellular debris. This process is particularly critical in mitigating the cellular damage caused by viral persistence and chronic inflammation—key contributors to the lingering symptoms of long COVID. Enhanced autophagy supports tissue repair and recovery, aiding in the restoration of normal cellular function.
Additionally, rapamycin addresses cellular senescence, a state in which non-dividing "zombie cells" accumulate and actively secrete pro-inflammatory molecules, perpetuating tissue damage and systemic inflammation. Senescent cells not only disrupt the surrounding cellular environment but also contribute to chronic inflammatory states, a hallmark of both aging and long COVID. By reducing the accumulation of these dysfunctional cells, rapamycin alleviates inflammation, promotes tissue homeostasis, and supports a healthier cellular landscape.
Chronic systemic inflammation is a prominent feature of long COVID, often manifested through elevated levels of pro-inflammatory cytokines such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-alpha), and C-reactive protein (CRP). This “cytokine storm” may persist long after the acute phase of infection, contributing to symptoms like fatigue, joint pain, and neuroinflammation. [13]
Rapamycin’s anti-inflammatory properties stem from its ability to modulate the immune response by inhibiting mTORC1. This inhibition reduces the production of pro-inflammatory cytokines and shifts the immune response toward a less inflammatory profile. [14] Moreover, rapamycin downregulates the activity of inflammasomes, which are protein complexes that drive inflammation in response to cellular stress. Rapamycin can decrease systemic inflammation by suppressing these pathways and potentially mitigating the progression of long-term organ damage associated with long COVID.
The anti-inflammatory effects of rapamycin may also extend to the vascular system. Long COVID has been associated with endothelial dysfunction, where inflammation damages the lining of blood vessels, impairing their ability to regulate blood flow. Rapamycin, through its anti-inflammatory actions, may help restore endothelial function and reduce the risk of vascular complications. [15]
Long COVID is frequently associated with immune dysregulation, encompassing chronic activation of immune cells, T-cell exhaustion, and, in some cases, the emergence of autoimmune phenomena. These disruptions are thought to be driven by multiple factors, including the persistence of viral antigens, residual inflammation, and molecular mimicry, where cross-reactivity between viral and self-antigens triggers inappropriate immune responses against the body’s own tissues.
Persistent immune activation not only contributes to systemic inflammation but can also lead to the depletion of immune resources, impairing the body’s ability to resolve infections and repair tissue damage. Additionally, the resulting cytokine imbalances and immune dysregulation may perpetuate the chronic symptoms observed in long COVID patients.
Rapamycin has demonstrated a unique ability to rebalance immune responses by modulating both the innate and adaptive branches of the immune system. Its effects include:
Suppression of hyperactive effector T cells and macrophages: Effector T cells, particularly CD8+ cells, and macrophages play critical roles in immune defense but can become chronically overactivated in conditions like long COVID, leading to sustained tissue damage and inflammation. Rapamycin reduces their hyperactivity, helping to dampen excessive immune responses. [16]
Enhancement of regulatory T cell (Treg) function: Tregs are essential for maintaining immune tolerance—the ability to prevent the immune system from attacking healthy tissues. By promoting Treg activity, rapamycin mitigates autoimmune phenomena and helps resolve chronic inflammation. [16]
Downregulation of inflammatory cytokines: Through its inhibition of the mTORC1 pathway, rapamycin reduces the production of pro-inflammatory cytokines such as interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), which are often elevated in long COVID. This shift helps create a more regulated and less inflammatory immune environment. [16]
For long COVID patients experiencing symptoms linked to immune overactivation, such as persistent inflammation, tissue damage, or autoimmune-related effects, rapamycin’s immune-modulatory properties could prove beneficial. Rapamycin may alleviate lingering symptoms and reduce the likelihood of further immune-related complications by restoring a balanced immune response.
Mitochondrial dysfunction has emerged as a significant contributor to long COVID symptoms, particularly fatigue, cognitive impairments (commonly referred to as "brain fog"), and exercise intolerance. Mitochondria, often described as the powerhouses of the cell, generate energy through oxidative phosphorylation. In long COVID, mitochondrial function may be compromised due to viral damage, chronic inflammation, and oxidative stress, resulting in reduced energy production and systemic fatigue.
Rapamycin indirectly supports mitochondrial health by promoting autophagy and mitophagy—the selective degradation of damaged mitochondria. These processes are essential for clearing dysfunctional mitochondria and facilitating the generation of healthier, more efficient organelles. Additionally, rapamycin has been shown to increase mitochondrial biogenesis, the production of new mitochondria, further supporting cellular energy demands and overall metabolic health.
A pivotal 2022 study published in Cell Metabolism by Dr. Tom McWilliams elucidates rapamycin's role in mitophagy and mitochondrial quality control. Using a model of mitochondrial disease characterized by respiratory chain defects, “ragged-red fibers” (indicative of pathogenic mitochondrial DNA variants), and severe accumulation of damaged mitochondria, the study investigated the effects of rapamycin treatment. The administration of rapamycin at a dose of 8 mg/kg/day over 70 days resulted in a remarkable 125% increase in mitophagy. [28]
The study highlighted key findings, including the observation that patients with mitochondrial disease exhibited over an 80-fold increase in the autophagy adapter protein p62, a marker of dysfunctional autophagy. This hyper-autophagic state was effectively mitigated by rapamycin treatment, which led to a ~90% reduction in p62 levels, restoring autophagy to near-normal levels. These results underscore rapamycin’s ability to stimulate mitophagy, providing a critical quality control mechanism that maintains mitochondrial integrity by recycling dysfunctional mitochondria.
The implications of these findings are significant for long COVID. Improved mitochondrial function through rapamycin treatment could enhance physical stamina and cognitive performance, addressing some of the most debilitating aspects of the condition. By targeting the root causes of energy deficits, rapamycin offers a promising therapeutic avenue for alleviating fatigue and other energy-related impairments in long COVID patients.
The Simmaron Research Foundation is conducting a pivotal trial investigating the use of low-dose rapamycin in patients with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS), a condition with striking parallels to long COVID. Both ME/CFS and long COVID are characterized by chronic fatigue, immune dysregulation, and cellular dysfunction, suggesting shared underlying mechanisms. This trial is particularly relevant for understanding how rapamycin might address the challenges of post-viral syndromes. [11]
Autophagy is often impaired in both ME/CFS and long COVID. Dysregulated autophagy has been implicated in the persistence of chronic fatigue and immune imbalances observed in these patients, making it an important target for therapeutic intervention. The trial seeks to determine whether rapamycin, through its ability to inhibit the mechanistic target of rapamycin (mTOR), can restore autophagic activity and mitigate these dysfunctions.
By enhancing autophagy, rapamycin also reduces chronic inflammatory signaling, particularly in senescent cells, which disrupt cellular environments and release pro-inflammatory cytokines. Reducing the burden of senescent cells not only diminishes inflammation but also supports tissue repair and overall cellular function.
The study focuses on alleviating hallmark symptoms of ME/CFS, such as debilitating fatigue and immune dysregulation, which significantly impair patients’ quality of life. Given the similarities between ME/CFS and long COVID, findings from this trial could provide valuable insights into the potential of rapamycin to address the overlapping pathological features of these post-viral conditions. By targeting fundamental cellular dysfunctions, rapamycin offers a promising avenue for improving outcomes in ME/CFS and related disorders like long COVID. [11]
Preliminary findings have been encouraging. Patients treated with low-dose rapamycin report reductions in fatigue and overall symptom severity. These improvements are believed to be linked to enhanced autophagic activity, which clears damaged cellular components, and better-regulated immune responses. By addressing these underlying dysfunctions, rapamycin shows promise as a therapeutic option for managing chronic post-viral conditions. While these findings specifically relate to ME/CFS, the study provides valuable insights into how rapamycin might be applied to related conditions, such as long COVID.
Similarly, a pilot study supported by The ME Association has investigated rapamycin's potential to address autophagy impairments in patients with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). The study specifically examined elevated levels of pATG13, a protein associated with disrupted autophagy. As we mentioned, impaired autophagy is thought to play a central role in the fatigue and cellular dysfunction characteristic of ME/CFS. [12]
The trial aimed to evaluate whether low-dose rapamycin could inhibit mTOR activity and restore normal autophagic function. This intervention targeted hallmark symptoms of ME/CFS, including post-exertional malaise (PEM), a debilitating condition where physical or mental exertion significantly worsens symptoms. By improving autophagy, researchers hypothesized that rapamycin could enhance cellular repair, optimize energy dynamics, and alleviate the profound fatigue experienced by patients.
Preliminary findings from the study indicate a promising trend. Participants reported reductions in post-exertional malaise and improvements in energy regulation—key outcomes for ME/CFS patients. These results suggest that rapamycin’s ability to enhance autophagic processes directly addresses the mechanisms underlying the fatigue and cellular dysfunction observed in the condition. [12]
Together, these studies highlight the potential of rapamycin to address chronic fatigue syndromes. While additional research is needed, early results underscore its capacity to improve energy regulation, reduce fatigue, and restore cellular balance—key considerations that could shape future treatment approaches for conditions like long COVID.
Chronic inflammation is a hallmark of long COVID, contributing to debilitating symptoms such as fatigue, joint pain, and neuroinflammation. As we’ve discussed, low-dose rapamycin offers a promising approach to addressing this inflammation by targeting mTOR, a key regulator of inflammatory signaling. By inhibiting mTORC1, rapamycin downregulates pro-inflammatory cytokines such as interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α), both of which are commonly elevated in long COVID patients. This reduction in systemic inflammation not only alleviates symptom burden but may also facilitate broader immune system restoration.
Recent findings from a meta-analysis published in the Journal of Geroscience further support rapamycin’s immunomodulatory potential. The study, titled “Rapamycin not dietary restriction improves resilience against pathogens: a meta-analysis,” demonstrated that rapamycin treatment significantly increased post-infection survival rates in mice, suggesting its ability to enhance immune resilience. Importantly, rapamycin was shown to delay immunosenescence, the gradual decline of immune function associated with aging. This immunomodulatory effect is particularly relevant to chronic inflammatory conditions like long COVID, where persistent immune dysregulation exacerbates inflammation and tissue damage. [17]
Unlike dietary restriction, which the meta-analysis associated with reduced post-infection survival, rapamycin improved resilience against pathogens, highlighting its unique capacity to bolster immune function while addressing inflammation. By mitigating immunosenescence, rapamycin offers a dual benefit: reducing chronic inflammation and enhancing the immune system’s ability to respond to threats, a critical advantage for long COVID patients facing prolonged immune dysregulation.
These preclinical findings align with human studies led by Dr. Joan Mannick, who explored the effects of mTOR inhibition on immunosenescence in elderly populations. In a randomized, observer-blind, placebo-controlled trial involving 218 elderly participants aged 65 years and older, three dosing regimens of RAD001 (a rapamycin variant) were tested: 0.5 mg daily, 5 mg weekly, and 20 mg weekly. Participants were treated for six weeks, followed by a two-week break, after which they received a seasonal flu vaccine. [18, 19]
The study revealed that low-dose RAD001 (0.5 mg daily or 5 mg weekly) significantly enhanced immune responses to the flu vaccine, as measured by increases in hemagglutination inhibition (HI) titers. Notably, these lower doses were as effective as higher doses, underscoring that partial mTOR inhibition may achieve optimal immune benefits without the risks associated with complete mTOR suppression. This is particularly relevant for conditions like long COVID, where balancing immune modulation and avoiding excessive immunosuppression is critical. [18, 19]
The trial also demonstrated broader immune benefits beyond the vaccine strains. Participants treated with RAD001 showed enhanced immune responses to heterologous influenza strains not included in the vaccine, suggesting a more generalized improvement in immune function. Mechanistically, RAD001 reduced the percentage of exhausted T cells (characterized by PD-1 expression), highlighting its role in rejuvenating immune cell function. [18, 19]
Compared to conventional anti-inflammatory treatments, such as corticosteroids or non-steroidal anti-inflammatory drugs (NSAIDs), rapamycin presents distinct advantages. While corticosteroids and NSAIDs primarily suppress inflammation, rapamycin targets the underlying mechanisms of cellular dysfunction by promoting autophagy and cellular repair. This dual action addresses the root causes of inflammation and immune dysregulation, rather than merely alleviating symptoms. Furthermore, its low-dose regimen minimizes the risk of significant immunosuppression, making it a safer long-term option for managing chronic inflammation in Long COVID.
The findings from both preclinical and clinical studies underscore rapamycin’s potential to reduce chronic inflammation, delay immunosenescence, and rejuvenate immune function. By targeting both inflammation and immunosenescence, rapamycin offers a scientifically grounded, multi-faceted potential approach to alleviating the persistent symptoms of long COVID.
Chronic fatigue is a hallmark of long COVID, often linked to impaired cellular energy production and mitochondrial dysfunction. As we've discussed, rapamycin, by inhibiting the mTOR pathway, enhances autophagy—a cellular process that removes damaged components and restores normal function. This promotion of autophagy facilitates the clearance of dysfunctional mitochondria, leading to the generation of healthier, more efficient energy-producing organelles.
In the context of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS), which shares similarities with Long COVID, studies have shown that rapamycin can improve mitochondrial function. A recent study titled "Mitophagy Activation by Rapamycin Enhances Mitochondrial Function and Cognition" provides valuable insights into rapamycin's potential benefits for conditions like long COVID. The research demonstrates that rapamycin activates mitophagy—the selective removal of damaged mitochondria—thereby improving mitochondrial function and cognitive performance.
This enhancement is particularly relevant for long COVID patients, who often experience fatigue and cognitive impairments due to mitochondrial dysfunction. By promoting the clearance of dysfunctional mitochondria, rapamycin may help restore cellular energy production and alleviate some of the debilitating symptoms associated with long COVID. [20]
Another study published in the Journal of Gerontology: Biological Sciences investigated rapamycin’s effects on brain protein synthesis rates in genetically heterogeneous mice. The study found that rapamycin treatment increased mitochondrial protein synthesis rates in older female mice, indicating enhanced mitochondrial biogenesis. These findings suggest that rapamycin not only promotes the clearance of damaged mitochondria but also supports the generation of new, functional mitochondria. For long COVID patients, this dual action could address systemic fatigue and improve cognitive function by bolstering overall cellular energy production. [21]
Evidence from related conditions, such as Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS), further supports rapamycin’s potential. Studies indicate that rapamycin improves mitochondrial function and increases mitochondrial efficiency, leading to better energy dynamics. In ME/CFS, rapamycin has been observed to improve physical endurance and mental clarity—benefits that could extend to long COVID patients, helping them increase daily activity levels, reduce brain fog, and improve overall quality of life. [11, 12]
While direct studies on rapamycin's effects in long COVID patients are still emerging, the existing research highlights its potential to enhance cellular health, optimize mitochondrial function, and address the root causes of fatigue and cognitive impairments.
Immune dysregulation is another key feature of long COVID, with patients often experiencing chronic immune activation or even autoimmune-like symptoms. Low-dose rapamycin has been shown to effectively modulate immune responses. It suppresses hyperactive immune cells while enhancing the function of regulatory T cells, which helps maintain immune balance and prevent excessive inflammation or autoimmunity.
Unlike traditional immunosuppressants, rapamycin prevents immune overactivation without excessively dampening the body’s ability to fight infections. This balanced immune modulation makes it an attractive therapeutic option for long COVID, particularly for patients who experience symptoms linked to persistent immune system dysfunction, such as lingering inflammation or tissue damage.
Beyond its direct effects on inflammation, energy production, and immune response, rapamycin may offer additional benefits for long COVID patients. Rapamycin’s neuroprotective properties, including its ability to reduce neuroinflammation and enhance synaptic plasticity, could also address neurological symptoms like brain fog and memory impairment.
Although these observations are not yet supported by large-scale clinical trials, ongoing research, and real-world experiences continue to shed light on rapamycin’s broader benefits. These findings underscore the need for further studies to fully understand its potential and optimize its use for long COVID patients.
While rapamycin shows promise as a treatment for long COVID, it is not without potential side effects. Commonly reported issues include gastrointestinal discomfort such as nausea, diarrhea, or abdominal pain, as well as mouth sores, which are particularly common at higher doses. Additionally, rapamycin can cause temporary increases in blood glucose and lipid levels, raising concerns for individuals with pre-existing metabolic conditions. These side effects underscore the importance of careful monitoring and individualized dosing to ensure patient safety. The low-dose regimens proposed for long COVID aim to mitigate these risks by reducing exposure while maintaining therapeutic benefits. Regular blood tests to monitor metabolic and immune parameters are crucial for identifying and addressing potential issues early.
Despite its potential, there is a notable gap in research specifically focusing on rapamycin’s application in long COVID. While preliminary findings from related conditions such as ME/CFS are encouraging, a limited number of clinical trials directly assess rapamycin's efficacy for long COVID. This lack of data highlights the need for larger, placebo-controlled studies to establish its therapeutic role and determine best practices for its use. Without such studies, the long-term efficacy of rapamycin remains uncertain, particularly in younger populations and individuals with varying degrees of immune function. Understanding how rapamycin impacts these groups over extended periods will be essential for evaluating its broader applicability.
Additionally, there is significant potential for exploring combination therapies to enhance rapamycin’s effectiveness. Pairing rapamycin with agents such as antioxidants or mitochondrial support supplements may offer synergistic effects, particularly in addressing the multifactorial nature of long COVID. These strategies and rigorous clinical trials could pave the way for optimized treatment protocols that maximize benefits while minimizing risks.
Long COVID represents an intricate medical challenge, affecting millions worldwide with a constellation of persistent and debilitating symptoms. As researchers continue to unravel the mechanisms behind this condition, it has become clear that addressing long COVID requires innovative and systemic therapeutic strategies. Rapamycin, with its multifaceted effects on inflammation, immune modulation, cellular repair, and mitochondrial function, emerges as a promising candidate for treatment. Early research and clinical observations suggest it may reduce chronic inflammation, enhance energy production, and optimize immune responses, offering meaningful relief for many patients grappling with the long-term impacts of COVID-19.
However, further investigation is essential to solidify its role and ensure its safe and effective use. Rigorous clinical trials, broader patient cohorts, and the exploration of combination therapies are necessary to establish best practices for implementing rapamycin in long COVID care. By integrating these insights, we can move closer to delivering impactful, evidence-based solutions for this complex and evolving condition.
TAKE HOME POINTS
Long COVID as a Systemic Condition: Long COVID, also known as post-acute sequelae of SARS-CoV-2 infection (PASC), affects individuals of all ages, including those with mild or asymptomatic initial infections. It is characterized by persistent symptoms such as fatigue, shortness of breath, chest pain, and cognitive impairments, stemming from systemic and multi-organ dysfunction involving complex physiological and psychological interactions. A UK study revealed that 70% of individuals in a low-mortality-risk group exhibited impairments in one or more organs, with multi-organ dysfunction observed in 29% of participants.
Mechanisms of SARS-CoV-2 Infection and Multi-Organ Involvement: SARS-CoV-2 infects host cells by utilizing its spike protein to bind to the angiotensin-converting enzyme 2 (ACE2) receptor, which is abundantly expressed in the respiratory system and other tissues, including the kidneys, liver, gastrointestinal tract, and brain. This interaction facilitates viral entry and replication, explaining the multi-organ involvement observed in acute and long COVID.
Viral Persistence and Its Role in Long COVID Pathophysiology: Viral RNA from SARS-CoV-2 has been detected in multiple organ systems, including the respiratory and gastrointestinal tracts, long after clinical recovery. A Lancet meta-analysis revealed extended viral shedding durations, such as up to 17 days in the upper respiratory tract and 17.2 days in the gastrointestinal tract. While not indicative of active infection, these viral remnants may trigger ongoing immune responses and low-grade inflammation, contributing to chronic symptoms like fatigue, cognitive dysfunction, and systemic inflammation in long COVID.
T Cell Dysfunction in Long COVID: Long COVID is associated with significant alterations in T cell functionality, particularly in memory T cells, which are critical for an efficient and targeted immune response. Severe cases exhibit 'T cell exhaustion,' characterized by diminished capacity of CD4+ and CD8+ T cells to activate other immune cells and directly kill infected cells. This exhaustion, marked by elevated levels of inflammatory proteins like granzyme B and IFN-γ, contributes to prolonged inflammation and chronic symptoms such as fatigue and cognitive dysfunction.
Impact on Vascular Function and Microvascular Health: SARS-CoV-2 disrupts ACE2 receptor function, a critical regulator of vascular homeostasis, leading to endothelial inflammation and microvascular dysfunction. This condition impairs capillary blood flow, reducing oxygen and nutrient delivery to tissues and contributing to hallmark symptoms of long COVID, including fatigue, shortness of breath, and cognitive impairments. Endothelial damage perpetuates a pro-inflammatory environment, creating a feedback loop that sustains systemic inflammation.
Coagulopathy and Its Role in Long COVID: Hypercoagulability is a significant issue in long COVID, marked by abnormal blood clotting and elevated D-dimer levels, a biomarker of ongoing clotting activity. This state increases the risk of thrombotic events like deep vein thrombosis (DVT) and pulmonary embolism (PE). Persistently elevated D-dimer levels suggest that clotting abnormalities persist into the post-acute phase, impairing microvascular circulation and exacerbating long COVID symptoms, including fatigue and cognitive dysfunction.
Rapamycin Reduces Chronic Overactivation of mTOR Observed in Long Covid Patients: Chronic mTOR overactivation contributes to inflammation, immune dysregulation, and cellular dysfunction—key drivers of long COVID symptoms. By inhibiting mTOR, rapamycin restores a balanced state, enhances autophagy, and mitigates cellular senescence. These actions reduce pro-inflammatory signals, support tissue repair, and improve mitochondrial function, addressing critical hallmarks of aging and long COVID pathology. Rapamycin’s ability to shift cellular focus from growth to repair underscores its potential as a targeted intervention for long COVID.
Anti-Inflammatory Effects of Rapamycin: Chronic systemic inflammation in long COVID is characterized by elevated pro-inflammatory cytokines like IL-6 and TNF-alpha, contributing to fatigue, joint pain, and neuroinflammation. Rapamycin mitigates these effects by inhibiting mTORC1, reducing cytokine production, and downregulating inflammasome activity. Its anti-inflammatory actions also extend to the vascular system, where rapamycin helps restore endothelial function, reducing the risk of vascular complications and long-term organ damage.
Rapamycin and Mitochondrial Health: Mitochondrial dysfunction, a driver of fatigue, brain fog, and exercise intolerance in long COVID, stems from viral damage, chronic inflammation, and oxidative stress. Rapamycin enhances mitochondrial health by promoting autophagy and mitophagy, clearing damaged mitochondria, and stimulating mitochondrial biogenesis. A 2022 study demonstrated that rapamycin increased mitophagy by 125% and normalized autophagy dysfunction in a mitochondrial disease model, highlighting its role in maintaining mitochondrial quality control. These benefits could improve energy production, alleviating systemic fatigue and cognitive impairments in long COVID patients.
Human Trial of Rapamycin to Treat Long Covid: A pivotal trial by the Simmaron Research Foundation is investigating low-dose rapamycin in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS), focusing on its ability to restore impaired autophagy, reduce senescent cell burden, and alleviate hallmark symptoms like chronic fatigue and immune dysregulation. Given the shared mechanisms between ME/CFS and Long COVID, the trial's findings may provide insights into rapamycin’s potential for addressing overlapping pathologies in post-viral syndromes.
Preliminary Findings and ME Association Study on Rapamycin for ME/CFS: Early findings from human trials, including a pilot study by The ME Association, show that low-dose rapamycin improves autophagic function, reduces post-exertional malaise, and enhances energy regulation in ME/CFS patients. These results highlight rapamycin’s promise in managing chronic fatigue and cellular dysfunction, with implications for its application in Long COVID.
Reduction of Chronic Inflammation and Immunosenescence: Rapamycin targets mTORC1 to reduce pro-inflammatory cytokines like IL-6 and TNF-α, commonly elevated in long COVID, thereby alleviating symptoms and restoring immune function. Preclinical studies and meta-analyses show rapamycin's potential to delay immunosenescence, enhancing immune resilience and addressing persistent immune dysregulation central to long COVID pathology.
Advantages over Conventional Treatments: Unlike corticosteroids and NSAIDs, which suppress inflammation without addressing underlying cellular dysfunction, rapamycin promotes autophagy and cellular repair. Its dual action mitigates chronic inflammation and immune dysregulation, with a low-dose regimen minimizing risks of immunosuppression, making it a safer and more effective long-term option for managing long COVID.
Rapamycin Promotes Mitochondrial Biogenesis: Studies demonstrate that rapamycin increases mitochondrial protein synthesis and supports the creation of new, functional mitochondria, enhancing cellular energy production and addressing fatigue and cognitive impairments often seen in long COVID.
Careful Dosing and Monitoring are Critical for Rapamycin Use: While rapamycin shows potential as a treatment for long COVID, its side effects, including gastrointestinal discomfort and temporary metabolic changes, highlight the importance of individualized dosing and regular monitoring to ensure safety and therapeutic efficacy.
Latest Longevity Research Straight to your Inbox
Sign up for The Longevity Blueprint, a weekly newsletter from Healthspan analyzing the latest longevity research.
Sign up for The Longevity Blueprint, a weekly newsletter from Healthspan analyzing the latest longevity research.