You are here
Home > Blog > Family Practice Medicine > Long COVID Treatment Breakthrough- From Symptoms to Science-Backed Solutions

Long COVID Treatment Breakthrough- From Symptoms to Science-Backed Solutions

Long COVID Treatment Breakthrough: From Symptoms to Science-Backed Solutions


Long Covid Treatment




Defining Long COVID and Its Global Impact

As the acute phase of COVID-19 begins to resolve, a substantial proportion of patients continue to experience persistent symptoms that evolve into what is now recognized as Long COVID. Unlike the initial infection, Long COVID presents as a multifaceted condition affecting multiple organ systems, often with devastating impacts on quality of life, functionality, and economic stability. Understanding the definition, prevalence, and symptom clusters of this condition provides the foundation for developing effective treatment approaches.

WHO Clinical Case Definition (2021)

In October 2021, after an extensive Delphi consensus process involving 265 patients, clinicians, researchers, and WHO staff, the World Health Organization established the first formal clinical case definition for Long COVID, officially termed “post COVID-19 condition” [1]. According to this definition, post COVID-19 condition “occurs in individuals with a history of probable or confirmed SARS-CoV-2 infection, usually 3 months from the onset of COVID-19 with symptoms that last for at least 2 months and cannot be explained by an alternative diagnosis” [1]. This definition additionally acknowledges that symptoms may be new onset following initial recovery, persist from the initial illness, or fluctuate and relapse over time [1].

Before this standardized definition, various terms circulated including “long COVID,” “long-haul COVID,” “post-acute COVID-19,” and “post COVID syndrome” [2]. The WHO definition created much-needed consistency for clinical diagnosis, research protocols, and epidemiological tracking. Nevertheless, many experts recognize that as understanding evolves, this definition will likely require refinement to capture the full spectrum of post-infection sequelae.

Prevalence Estimates: 10–30% of Infected Individuals

The global burden of Long COVID continues to grow with each new wave of infections. Current estimates indicate that 10-30% of individuals who contract SARS-CoV-2 develop Long COVID [3]. However, prevalence rates vary considerably depending on hospitalization status, with estimates of 50-70% among hospitalized patients and 10-30% among non-hospitalized cases [3]. Moreover, even vaccinated individuals face a 10-12% risk of developing Long COVID following breakthrough infections [3].

Regional data provides additional insight into prevalence patterns. In the UK, approximately 21% of COVID-19 patients exhibited symptoms at 5 weeks post-infection, while 10% still experienced symptoms at 12 weeks [3]. At a safety-net hospital serving predominantly Black and Hispanic patients in Chicago, 34% reported at least one Long COVID symptom at a median of 255 days after discharge [4].

The impact extends beyond individual health concerns to create substantial economic consequences. At least 65 million individuals worldwide are estimated to have Long COVID [3], with significant proportions unable to return to work [3]. This workforce reduction contributes to ongoing labor shortages [3] and creates an annual global economic cost estimated at $1 trillion [3].

Common Symptom Clusters: Fatigue, Brain Fog, Dyspnea

Long COVID manifests through a diverse constellation of symptoms affecting multiple organ systems. The most frequently reported symptoms include:

  • Systemic symptoms: Fatigue/malaise reported by over 50% of patients [4], fever, and general weakness

  • Neurocognitive manifestations: Memory and concentration difficulties (brain fog) affecting 32% of patients [4], headache, sleep disturbances, and sensory alterations

  • Respiratory complaints: Shortness of breath (dyspnea) in over 50% of patients [4], persistent cough, chest pain

  • Psychological impacts: Anxiety (46%) and depression (44%) [4]

When analyzing symptom clusters, neurocognitive symptoms are most common, followed by respiratory symptoms [3]. These symptoms are not merely inconvenient but significantly disruptive to daily functioning. According to the CDC, Long COVID symptoms “may be difficult to recognize or diagnose, require comprehensive care, and result in disability” [3].

The timeline and progression of symptoms vary considerably. Many patients experience improvement within 3-6 months, while others face prolonged disability [3]. Notably, some symptoms emerge immediately following acute infection, while others develop weeks or months later, particularly neurological manifestations [3]. Additionally, symptoms can resolve and then reappear, creating a challenging pattern of relapse and remission [3].

Furthermore, Long COVID appears to disproportionately affect certain populations. Women, Hispanic individuals, those with severe acute COVID-19, patients with underlying health conditions, and unvaccinated individuals all face higher risk [3]. This demographic variability underscores the need for tailored clinical approaches addressing the unique challenges of different patient populations.

As our understanding of Long COVID continues to evolve, the refined definition, prevalence data, and symptom characterization provide the necessary framework for developing targeted treatment strategies to address this growing global health challenge.


Mapping Long COVID Symptoms to Organ Systems

Long COVID presents with diverse manifestations across multiple organ systems, complicating diagnosis and treatment approaches. The multi-systemic nature of this condition requires a thorough understanding of how different body systems are affected to develop effective management strategies. Recent research has documented distinct patterns of organ involvement that persist long after the acute phase of infection has resolved.

Cardiovascular: Myocarditis, Arrhythmias, Chest Pain

Cardiovascular complications rank among the most common extrapulmonary manifestations in Long COVID. Studies indicate that 10-20% of patients develop cardiac arrhythmias [3], with palpitations reported in 9% of patients and chest pain in 5% at six months post-infection [3]. Cardiac magnetic resonance imaging reveals inflammation in up to 60% of patients more than two months after diagnosis [3], with cardiac impairment detected in 78% of patients approximately 71 days after infection [3].

Research using the US Department of Veterans Affairs database identified significantly increased risk of various cardiovascular conditions regardless of initial COVID-19 severity, including heart failure, dysrhythmias, and stroke [3]. About 20-30% of hospitalized COVID-19 patients develop cardiac complications [3], and cardiac MRI studies conducted 12 months post-infection showed persistent cardiac abnormalities in 58% of Long COVID patients [3].

Specifically, patients frequently present with inappropriate sinus tachycardia syndrome and postural orthostatic tachycardia syndrome (POTS), with one study finding POTS in 67% of Long COVID patients [3]. Chest pain occurs in approximately 17% of Long COVID patients, whereas palpitations affect roughly 20% [3].

Neurological: Brain Fog, Headaches, Cognitive Impairment

Neurological and cognitive symptoms constitute a major component of Long COVID. Fatigue affects 32% of patients, whereas cognitive impairment occurs in 22% at 12 weeks post-infection [3]. Cognitive deficits in Long COVID patients can be debilitating—equivalent to the cognitive impact of aging 10 years or to intoxication at the UK drink driving limit [3].

Cognitive impairment appears to worsen over time in some patients, with one study documenting occurrence in 16% of patients at 2 months but increasing to 26% by 12 months [3]. Brain imaging studies reveal concerning changes, such as:

  • Reduced gray matter thickness in orbitofrontal cortex and parahippocampal gyrus

  • Overall reduction in brain size

  • Greater cognitive decline compared to controls [3]

Consequently, persistent neurocognitive symptoms often interfere with daily functioning. Notably, mental health conditions like anxiety and depression may improve over time, yet cognitive impairment, seizures, and dementia risks persist for at least two years [3]. These deficits exist independently of mental health conditions and occur at similar rates in both hospitalized and non-hospitalized patients [3].

Pulmonary: Dyspnea, Fibrosis, Reduced Lung Capacity

Respiratory conditions occur twice as frequently in COVID-19 survivors compared to the general population [3]. Shortness of breath persists in 40% of Long COVID patients for at least seven months, whereas chronic cough affects 20% [3]. Imaging studies of non-hospitalized Long COVID patients demonstrate abnormalities in air trapping and lung perfusion [3].

Pulmonary function tests reveal decreased diffusion capacity as the most commonly reported physiologic impairment, directly correlated with illness severity [3]. Persistent restrictive pulmonary physiology at 3-6 months mirrors findings seen in other acute respiratory distress syndrome survivors [3]. Immunological characterization has identified a correlation between decreased lung function, systemic inflammation, and SARS-CoV-2-specific T cells [3].

Gastrointestinal: IBS, Nausea, Appetite Loss

Gastrointestinal symptoms frequently manifest in Long COVID. Common presentations include nausea, abdominal pain, loss of appetite, heartburn, and constipation [5]. The gut microbiota composition undergoes significant alteration during and after COVID-19 infection [5], with dysbiosis persisting for at least 14 months [3].

Higher levels of Ruminococcus gnavus and Bacteroides vulgatus alongside lower levels of Faecalibacterium prausnitzii have been documented in Long COVID patients compared to controls [3]. Low levels of butyrate-producing bacteria strongly correlate with Long COVID at 6 months [3]. Respiratory and neurological symptoms associate with specific gut pathogens, suggesting complex gut-brain-lung interactions [3].

Crucially, persisting respiratory and neurological symptoms each correlate with specific gut pathogens [3]. SARS-CoV-2 RNA persists in stool samples of 12.7% of patients at 4 months post-diagnosis and in 3.8% at 7 months [3]. This viral persistence may explain why 29% of patients report persistent GI symptoms six months after acute infection [5].

Musculoskeletal: Myalgia, Joint Pain, Fatigue

Musculoskeletal involvement represents a prominent feature of Long COVID. Fatigue ranks as the most common complaint (97.7%), followed by myalgia/arthralgia (69.2%), back pain (43.6%), low back pain (33.1%), and chest pain (25%) [6]. Joint pain primarily affects wrists, ankles, and knees [6].

The pathogenesis of musculoskeletal symptoms in Long COVID involves multiple mechanisms, primarily:

  1. Direct viral effects via ACE2 receptors, which are expressed in smooth muscle, cartilage, and kidneys [6]

  2. Inflammation due to cytokine storm, particularly involving C-reactive protein, IL-1β, IL-2, IL-6, IL-8, IL-10, IL-17, and TNF-α [6]

  3. Hypoxia triggering muscle catabolism and dysfunction [6]

  4. Muscle wasting due to inflammation, malnutrition, and impaired metabolism [6]

In a follow-up study of patients who recovered from COVID-19 pneumonia, researchers found weakness in biceps and quadriceps muscles in 73% of patients [6]. Muscle strength measurements revealed quadriceps and biceps performance at just 54% and 69% of predicted normal values, respectively [6].


Immune Dysregulation and Viral Persistence Mechanisms Top Of Page

Autoimmunity and Endothelial Dysfunction in Post COVID Syndrome

Emerging evidence points to a complex interplay between autoimmune responses and vascular abnormalities as central mechanisms underlying persistent symptoms in Long COVID. These pathophysiological processes create a foundation for understanding why some patients experience prolonged illness and offer potential targets for therapeutic intervention.

Autoantibodies Against ACE2 and β2-Adrenergic Receptors

The development of autoantibodies following SARS-CoV-2 infection represents a critical aspect of Long COVID pathophysiology. In fact, research has identified IgG autoantibodies against angiotensin-converting enzyme 2 (ACE2) in approximately 1.5% of patients who had recovered from SARS-CoV-2 infection [3]. Interestingly, these autoantibodies were more prevalent in males (76.5%) and were found at higher titers in patients who had experienced severe COVID-19 [3]. The presence of these autoantibodies can potentially disrupt the protective effects of ACE2, including its role in converting angiotensin II into the vasoprotective angiotensin (1-7).

Beyond ACE2, functionally active autoantibodies against G-protein-coupled receptors (GPCR-AAbs) have been detected in patients with persistent Long COVID symptoms. These include:

  • β2-adrenergic receptor autoantibodies (found in 93.98% of Long COVID patients)

  • Muscarinic M2-receptor autoantibodies (93.98%)

  • Angiotensin II AT1-receptor autoantibodies (92.77%)

  • MAS-receptor autoantibodies (92.77%)

  • α1-adrenergic receptor autoantibodies (24.1%)

  • Nociceptin receptor autoantibodies (31.33%) [7]

These autoantibodies contribute to dysregulation of autonomic functions and may explain symptoms like tachycardia, blood pressure fluctuations, and fatigue. Upon closer examination, studies have demonstrated that seropositivity for β2-adrenergic, α1-adrenergic, and MAS receptor autoantibodies correlates with impaired microcirculation in Long COVID patients [7]. The presence of these autoantibodies appears to trigger a vicious cycle where ischemia-induced GPCR-AAb activity causes harmful pathogenic impact via lack of tachyphylaxia (ability to develop tolerance to repeated drug doses) [7].

Microclots and Capillary Rarefaction

Within the vascular system, Long COVID patients exhibit persistent abnormalities in microcirculation. Strikingly, studies using sidestream dark field imaging have revealed significant decreases in vascular density that exclusively affect very small capillaries, with reductions of 45.16% in 5μm diameter vessels, 35.60% in 6μm vessels, and 22.79% in 7μm vessels [8]. This capillary rarefication persists even 18 months after infection [8], suggesting long-lasting vascular damage.

Concurrently, fibrinoid microclots have been identified in the plasma of Long COVID patients [5]. These microclots are resistant to fibrinolysis and can block capillaries, thereby causing tissue hypoxia [5]. The isolated SARS-CoV-2 spike protein S1 subunit acts as a proinflammatory inflammagen that can directly interact with platelets and fibrinogen to induce hypercoagulability [5]. Essentially, these microclots contain trapped inflammatory molecules including alpha 2-antiplasmin (α2AP), von Willebrand factor (VWF), platelet factor 4 (PF4), serum amyloid A (SAA), and various fibrinogen chains [5].

The combination of capillary rarefication and microclots leads to impaired oxygen exchange and tissue hypoxia that can persist for months [5]. Likewise, plotting red blood cell velocity in capillaries and feed vessels showed that the number of capillaries perfused in Long COVID patients was comparable to that of critically ill COVID-19 patients and did not respond adequately to local variations of tissue metabolic demand [8].

Endothelial Inflammation via NF-κB Signaling

Endothelial inflammation, or endothelialitis, serves as another fundamental mechanism in Long COVID pathophysiology. SARS-CoV-2 can affect the endothelium either directly through infection or indirectly through cytokine storms, leading to endothelial dysfunction [9]. Subsequently, this dysfunction manifests as reduced nitric oxide (NO) bioavailability, oxidative stress, endothelial injury, glycocalyx/barrier disruption, hyperpermeability, inflammation, senescence, and endothelial-to-mesenchymal transition [9].

Despite debates about whether SARS-CoV-2 directly infects endothelial cells, research has demonstrated that the virus can activate the NF-κB inflammatory pathway in these cells [10]. This activation occurs when SARS-CoV-2 spike proteins bind to TMPRSS-1 receptors and extracellular vimentin [11]. Following this binding, there is increased expression of chemokines, cell adhesion molecules, and other proteins governing cell-cell interactions [5].

Therefore, the ongoing NF-κB signaling perpetuates a state of chronic endothelial inflammation. In Long COVID patients, endothelial biomarkers such as VWF, Factor VIII, ET-1, and angiopoietin-2 remain dysregulated even eight months after mild-to-moderate COVID-19 infection [5]. Furthermore, the endothelial glycocalyx, a protective layer on the luminal surface of blood vessels, shows damage in Long COVID, contributing to increased vascular permeability and inflammatory cell adhesion [9].

The combination of autoimmune phenomena, microclots, and endothelial inflammation creates a multi-faceted pathophysiological framework that helps explain the diverse and persistent symptoms experienced by Long COVID patients. Understanding these mechanisms offers promising avenues for developing targeted therapies to address the underlying causes of this challenging condition.

Long Covid Treatment


Symptom-Specific Treatment Approaches

The management of long COVID requires a tailored, symptom-specific approach given its heterogeneous nature and varying presentation among patients. Treatment strategies continue to evolve as clinical understanding of the condition improves, though many approaches remain experimental with limited high-quality evidence supporting their efficacy.

Cognitive Dysfunction: Low-Dose Naltrexone, CBT

Cognitive dysfunction or “brain fog” presents substantial challenges for long COVID patients, impacting daily functioning and quality of life. Low-dose naltrexone (LDN), an opioid antagonist administered at doses of 1-4.5mg, has emerged as a promising intervention. A single-center pre-post study found that LDN improved well-being in 6 of 7 parameters measured among patients with post-COVID syndrome, including recovery from COVID-19, limitation in activities of daily living, energy levels, and levels of concentration (p ≤ 0.001) [6]. The proposed mechanism involves immunomodulation, potentially through antagonism of opioid growth factor receptors or inhibition of toll-like receptor-4 inflammatory signaling.

Cognitive behavioral therapy (CBT) demonstrates moderate certainty evidence for reducing fatigue and improving concentration in long COVID patients. One trial showed that a 17-week online CBT program called “fit after covid” reduced fatigue (mean difference −8.4, 95% CI −13.11 to −3.69) and improved concentration (mean difference −5.2, 95% CI −7.97 to −2.43) compared to usual care [12]. This program addressed disruptive sleep-wake patterns, unhelpful beliefs about fatigue, and pain coping mechanisms.

Contrary to these findings, a recent randomized trial evaluating interventions for cognitive symptoms in 328 patients found no differential benefits from online cognitive training (BrainHQ), a structured cognitive rehabilitation program, or transcranial direct current stimulation [13]. All five study arms showed some improvement over time, suggesting a potential role for general engagement or natural recovery.

Fatigue and PEM: Pacing and Energy Management

Post-exertional malaise (PEM)—the worsening of symptoms after physical or mental exertion—represents a cardinal feature of long COVID. Pacing, an activity management strategy, helps mitigate PEM by balancing rest with activity. This approach involves determining individual limits for mental and physical activities, often referred to as staying within one’s “energy envelope” [14].

Heart rate monitoring (HRM) offers direct biofeedback to enhance symptom awareness during pacing. For those with ME/CFS and long COVID, adding 15 beats per minute to the resting heart rate provides a conservative estimate of the ventilatory/anaerobic threshold (V/AT), above which PEM risk increases [15]. Patients should aim to maintain heart rates below this threshold and rest when exceeding it.

In practice, pacing may include:

  • Activity planning with careful attention to intensity

  • Breaking down tasks into shorter, less strenuous segments

  • Monitoring physical and cognitive exertion

  • Avoiding “push and crash” cycles where patients overexert on “good days”

  • Incorporating scheduled rest periods

Clinical evidence indicates that patients who can pace effectively have fewer symptoms, better quality of life, and are more likely to improve physical functioning and fatigue severity [16].

Cardiac Symptoms: β-Blockers, Anticoagulants

Cardiac manifestations of long COVID frequently include arrhythmias, tachycardia, and potential thromboembolic complications. For postural orthostatic tachycardia syndrome (POTS), which affects approximately 40% of long COVID patients [17], beta-blockers represent a first-line treatment option. These medications reduce heart rate, thereby improving symptoms of palpitations and tachycardia, though they should be used cautiously in patients with concurrent hypotension.

For patients with evidence of hypercoagulability or microclotting, anticoagulant therapy might provide benefit. Currently, triple anticoagulant therapy has been reported to improve symptoms in a majority of patients with long COVID [18]. A nonrandomized retrospective study also reported lower risk of sudden cardiac arrest in patients discharged on rivaroxaban after hospitalization for COVID-19 [17], although this approach requires additional study before widespread implementation.

Importantly, interactions between COVID-19 medications and cardiovascular drugs must be considered. Nirmatrelvir-ritonavir (Paxlovid) can increase plasma levels of several antiarrhythmics and anticoagulants, potentially requiring dose adjustments or temporary discontinuation [17].

Triple therapy  Top Of Page

In the context of long COVID, the phrase “triple anticoagulant therapy” refers to a three-part blood-thinning regimen that some clinicians and researchers have used experimentally to try to reduce microvascular clotting and improve persistent symptoms. This approach has been reported in observational cohorts to improve symptoms in a majority of treated patients, but it is not an established, guideline-approved therapy, and carries significant bleeding risks.

Specifically, the triple therapy typically consists of:

  • A direct oral anticoagulant (DOAC) — most commonly apixaban, which inhibits factor Xa and reduces clot formation.
  • Dual antiplatelet therapy (DAPT) — usually aspirin plus clopidogrel.
  • Gastric protection with a proton-pump inhibitor (PPI) — such as pantoprazole, used to reduce the risk of gastrointestinal bleeding that can occur with aggressive antithrombotic therapy.

IIn many reports the combination has been used for several weeks to months with the aim of reducing fibrin amyloid microclots and platelet hyperactivation, which some researchers theorize contribute to long COVID symptoms such as fatigue, “brain fog,” and shortness of breath.

Respiratory Symptoms: Pulmonary Rehab, Inhaled Steroids

Respiratory sequelae remain among the most persistent symptoms of long COVID. Pulmonary rehabilitation programs show promise in addressing dyspnea and reduced exercise capacity. Moderate certainty evidence indicates that intermittent aerobic exercise 3-5 times weekly for 4-6 weeks probably improves physical function compared with continuous exercise (mean difference 3.8, 95% CI 1.12 to 6.48) [12].

For patients with airway inflammation or hyperresponsiveness, inhaled corticosteroids (ICS) may provide relief. A recent study examining ICS in patients with post-COVID chronic cough found that while ICS did not significantly impact symptom management, it improved impaired lung function parameters such as FEV1/FVC ratio and maximal mid-expiratory flow [19]. Among patients with airway hyperresponsiveness—identified in 82.6% of patients with long COVID-related cough—ICS treatment showed greater improvement in pulmonary function than in those not receiving ICS [19].

Respiratory rehabilitation should be introduced gradually, with careful monitoring for symptom exacerbation. For instance, inspiratory muscle training and active cycle of breathing techniques have been investigated, although evidence supporting these approaches remains limited [12].


Emerging Therapies Under Clinical Investigation Top Of Page

As researchers continue to unravel the complex pathophysiology of long COVID, several promising therapeutic approaches are currently undergoing clinical evaluation. These experimental treatments target distinct pathways implicated in symptom persistence and may eventually provide relief for millions affected by this debilitating condition.

Montelukast for Respiratory Symptoms (E-SPERANZA Trial)

The E-SPERANZA COVID Project represents an important advancement in targeting respiratory manifestations of long COVID. This double-blind, placebo-controlled randomized clinical trial is evaluating montelukast, a leukotriene receptor antagonist traditionally used for asthma management. The protocol involves daily administration of 10 mg oral montelukast for 28 days to patients with mild to moderate respiratory symptoms persisting more than 4 weeks after SARS-CoV-2 infection [20].

The trial primarily assesses quality of life improvements associated with respiratory symptoms using the COPD Assessment Test (CAT) questionnaire. Secondary endpoints include changes in exercise capacity measured by the 1-minute sit-to-stand test, oxygen saturation during exertion, and resolution of other symptoms including asthenia, headache, mental confusion, ageusia, and anosmia [20]. Given montelukast’s established safety profile and anti-inflammatory properties, it presents a readily available repurposing opportunity for long COVID management.

Leronlimab for Immune Modulation

Leronlimab, a CCR5-binding humanized IgG4 monoclonal antibody, has emerged as a candidate for addressing immune dysregulation in long COVID. In an exploratory trial involving 55 individuals, weekly subcutaneous doses of leronlimab (700 mg) were administered for 8 weeks. Intriguingly, researchers observed significantly increased blood cell surface CCR5 levels from baseline to week 8 among leronlimab-treated participants who experienced symptom improvement, but not in non-responders or placebo recipients [21].

This finding suggests an unexpected mechanism whereby leronlimab may normalize abnormal immune downmodulation rather than persistent immune activation. In fact, leronlimab treatment was associated with increases in key adaptive immune cell populations including T cells [21]. Furthermore, preliminary results indicated numerically higher percentages of participants with reduced symptom scores across 18 of 24 COVID symptoms when compared with placebo, yet these differences did not reach statistical significance in this small exploratory study [22].

Hyperbaric Oxygen Therapy for Fatigue

Hyperbaric oxygen therapy (HBOT) has demonstrated promising results for addressing persistent fatigue and cognitive dysfunction. A randomized controlled trial evaluating HBOT’s efficacy in long COVID patients reported significant improvements in global cognitive function, attention, executive function, and psychiatric symptoms [23]. The intervention typically involves administering 100% oxygen at 2.0 ATA for 90-minute sessions [24].

HBOT’s potential mechanisms involve several pathways: it increases tissue oxygenation in areas of local hypoxia, induces a hyperoxia-hypoxia paradox that activates HIF-1 signaling, and generates various cellular responses that contribute to angiogenesis and wound healing [23]. Additionally, the alternation between hyperoxia and normoxia produces anti-inflammatory and anti-edematous effects through multiple target gene pathways [23]. Currently, HBOT represents the only scientifically proven treatment in a prospective randomized controlled trial to demonstrate cognitive improvement, brain network regeneration, and enhanced cardiac function in long COVID patients [23].

Adaptogens and Nicotinamide Riboside Trials

Alternative approaches utilizing NAD+ precursors have garnered interest given mitochondrial dysfunction observations in long COVID. A randomized controlled trial investigating nicotinamide riboside (NR), a form of vitamin B3, examined its effects on NAD+ levels, cognition, and symptom recovery [25]. In this 24-week trial, participants received either 2,000 mg of NR daily for 20 weeks or placebo for 10 weeks followed by NR for 10 weeks [25].

While primary outcomes comparing the two groups showed no major differences in thinking or memory scores, exploratory analyzes revealed improvements in self-reported fatigue, sleep quality, depressive symptoms, and executive functioning after 10 weeks of NR supplementation compared to baseline [25]. This suggests potential benefits for some individuals, possibly through NAD+’s role in cellular energy production and inflammation control [25].

Collectively, these emerging therapies represent diverse approaches to long COVID treatment, ranging from repurposed anti-inflammatory medications to novel immune modulators and cellular energy enhancers. As clinical investigations continue, physicians may soon have evidence-based options for addressing this challenging post-viral syndrome.

Long Covid Treatment


Microbiome and Gut-Brain Axis in Long COVID

Growing evidence reveals the profound impact of gut microbiome alterations on long COVID pathophysiology. The intricate relationship between intestinal microbiota and central nervous system function through the gut-brain axis offers valuable insights into persistent symptoms and potential therapeutic approaches.

Reduced Faecalibacterium prausnitzii and Butyrate Producers

The gut microbiome of long COVID patients exhibits marked dysbiosis characterized by diminished bacterial diversity and richness [26]. Metagenomic analyzes demonstrate substantial depletion of short-chain fatty acid (SCFA)-producing bacteria from the Lachnospiraceae, Ruminococcaceae and Eubacteriaceae families [26]. In particular, levels of Faecalibacterium prausnitzii, a key butyrate producer with anti-inflammatory properties, remain markedly reduced in patients with persistent symptoms [26]. This microbiome disruption persists even after viral clearance, with studies documenting altered composition at 6 months post-infection [27].

Specific bacterial signatures correlate with distinct symptom clusters. Respiratory symptoms associate with elevated levels of Streptococcus anginosus and Streptococcus vestibularis, whereas neurological manifestations correlate with increased Clostridium innocuum and Actinomyces naeslundii[28]. Patients experiencing hair loss show marked reduction in butyrate-producing bacteria, including Bifidobacterium pseudocatenulatum[28].

Fungal Translocation and NF-κB Activation

Beyond bacterial dysbiosis, emerging research identifies fungal translocation as a crucial mechanism in long COVID pathophysiology [29]. Patients with PASC demonstrate elevated plasma levels of β-glucan, a fungal cell wall polysaccharide [29]. These elevated β-glucan levels correlate with increased inflammatory markers and dysregulated tryptophan catabolism pathways [29].

Mechanistically, β-glucan binds to Dectin-1 receptors on myeloid cells, subsequently activating Syk/NF-κB signaling pathways [29]. Experimental models confirm this connection, as plasma from individuals with PASC induces heightened NF-κB signaling compared with controls—an effect neutralized by piceatannol, a Syk inhibitor [29]. This inflammatory cascade may contribute to both systemic and neurological manifestations through persistent immune activation.

Probiotic Interventions: Bifidobacterium longum

Targeted microbiota modulation represents a promising therapeutic avenue. When gut bacteria from long COVID patients were transferred to healthy mice, the animals developed cognitive impairments and lung defense deficiencies—effects partially ameliorated by administration of Bifidobacterium longum[30].

Clinical trials investigating microbiome-based therapies have yielded encouraging results. A randomized, double-blind, placebo-controlled study of SIM01, a synbiotic preparation containing Bifidobacterium species, demonstrated notable symptom improvement in long COVID patients [31]. After six months, the intervention group experienced greater alleviation of fatigue, memory loss, gastrointestinal upset, and concentration difficulties compared to controls [31].


Challenges in Diagnosis and Biomarker Development

Diagnosing long COVID presents considerable obstacles for clinicians, primarily stemming from the absence of definitive laboratory markers and the reliance on subjective symptom reporting. Presently, these diagnostic limitations hinder effective patient management and treatment selection.

Lack of Standardized Diagnostic Tools

The clinical recognition of long COVID remains complicated by inconsistent case definitions across major health organizations. Without a unified international diagnostic standard, healthcare providers encounter substantial variability in identifying affected individuals. Diagnosis currently depends heavily on patient-reported symptoms and physicians’ subjective interpretation, as many cardinal manifestations—including fatigue, brain fog, and memory decline—lack objective assessment parameters [32]. Indeed, this diagnostic uncertainty often results in delayed or missed diagnoses, with many patients being inappropriately labeled with anxiety or depression instead of receiving proper care [33]. Furthermore, long COVID symptoms frequently overlap with those of other chronic conditions, encompassing psychiatric disorders, autoimmune diseases, and various post-infectious syndromes, creating additional diagnostic confusion [33].

Potential Biomarkers: CCL11, Spike Antigen, Cortisol

Recent investigations have identified several promising molecular markers that may facilitate objective diagnosis. Blood transcriptomic analyzes revealed 212 differentially expressed genes in long COVID patients, with antisense ORF1ab RNA and FYN RNA emerging as independent diagnostic biomarkers (93.8% sensitivity, 91.7% specificity) [33]. Persistently elevated proinflammatory cytokines—specifically IL-6, TNF-α, and CCL11—correlate with symptom severity and cognitive dysfunction [34]. Perhaps most compelling, circulating SARS-CoV-2 spike antigen was detected in 60% of long COVID patients up to 12 months after diagnosis compared to 0% of recovered controls, suggesting ongoing viral persistence [35]. Additionally, studies demonstrate consistently low blood cortisol levels in patients even after a year of symptoms, without compensatory increases in ACTH, indicating hypothalamic-pituitary-adrenal axis dysfunction [34].

Role of Corneal Microscopy and Tilt Table Testing

Innovative diagnostic approaches utilizing specialized examinations show promise for objective assessment. Corneal confocal microscopy (CCM), initially developed for evaluating diabetic neuropathy, effectively identifies nerve damage in long COVID patients through non-invasive imaging of corneal nerve fibers [35]. This technique reveals reduced corneal nerve density, shorter nerve length, and increased dendritic cell density persisting over 20 months after infection [35]. Microneuromas, indicative of nerve damage and regeneration, appear in 15% of long COVID patients versus none in control groups [35]. For dysautonomia evaluation, tilt table testing provides valuable diagnostic insights, with studies demonstrating similar haemodynamic and cognitive abnormalities during orthostatic stress between long COVID and ME/CFS patients [30]. These emerging tools may bridge the gap between subjective symptom reporting and objective disease confirmation.


Long Covid Treatment


Conclusion Led   Top Of Page

Long COVID presents physicians with unprecedented challenges due to its multisystemic nature, affecting cardiovascular, neurological, pulmonary, gastrointestinal, and musculoskeletal systems simultaneously. The complex interplay between autoimmunity, microclot formation, endothelial dysfunction, and gut microbiome alterations underpins the persistent symptoms experienced by millions worldwide.

Diagnosis remains primarily clinical, though several promising biomarkers have emerged. Spike protein persistence, altered cytokine profiles, and unique transcriptomic signatures might soon provide objective diagnostic criteria, while specialized testing methods such as corneal confocal microscopy offer new avenues for objective assessment.

Treatment approaches must address both symptom management and underlying pathophysiology. Low-dose naltrexone shows promise for cognitive dysfunction, whereas careful pacing strategies help mitigate post-exertional malaise. Beta-blockers effectively manage tachycardia and POTS symptoms, while pulmonary rehabilitation programs benefit respiratory manifestations when appropriately tailored to avoid symptom exacerbation.

Emerging therapies target specific pathophysiological mechanisms. Leronlimab may normalize immune dysregulation, montelukast potentially reduces respiratory inflammation, and hyperbaric oxygen therapy demonstrates cognitive benefits through enhanced tissue oxygenation. Additionally, probiotic interventions with Bifidobacterium species appear to ameliorate symptoms through microbiome modulation.

The healthcare community must recognize Long COVID as a distinct clinical entity requiring multidisciplinary care. Patients need validation of their symptoms alongside targeted interventions based on their unique symptom clusters. Future research should focus on defining subgroups of patients who might benefit from specific therapeutic approaches.

Medical education must evolve rapidly to incorporate Long COVID management into clinical training programs. Physicians require tools to distinguish Long COVID from other post-infectious syndromes and to implement evidence-based treatment protocols as they emerge. Multidisciplinary collaboration between immunologists, cardiologists, neurologists, pulmonologists, and rehabilitation specialists will prove essential for addressing this complex condition effectively.

Though Long COVID presents formidable challenges, scientific understanding continues to advance rapidly. The convergence of clinical observation with sophisticated immunological and microbiological research offers hope for patients suffering from this debilitating condition. Therefore, physicians must remain vigilant about emerging evidence while providing compassionate, personalized care to those affected by the long-term sequelae of SARS-CoV-2 infection.

Key Takeaways

Long COVID affects 10-30% of infected individuals with multisystem symptoms requiring targeted, evidence-based treatment approaches across cardiovascular, neurological, respiratory, and gastrointestinal manifestations.

• Autoimmune mechanisms drive persistence: Autoantibodies against ACE2 and β2-adrenergic receptors, plus microclots and endothelial inflammation, create ongoing dysfunction requiring immunomodulatory interventions.

• Symptom-specific treatments show promise: Low-dose naltrexone for brain fog, pacing strategies for fatigue, beta-blockers for cardiac symptoms, and pulmonary rehabilitation for respiratory issues.

• Gut microbiome restoration is crucial: Reduced butyrate-producing bacteria and fungal translocation contribute to symptoms; probiotic interventions with Bifidobacterium species demonstrate clinical improvement.

• Emerging therapies target root causes: Leronlimab for immune modulation, hyperbaric oxygen for cognitive function, and montelukast for respiratory inflammation show encouraging clinical trial results.

• Objective biomarkers are developing: Persistent spike protein, elevated CCL11, and corneal nerve damage provide diagnostic tools beyond subjective symptom reporting for clinical validation.

The convergence of pathophysiological understanding with targeted therapeutics offers hope for the millions suffering from this complex post-viral syndrome. Healthcare providers must adopt multidisciplinary approaches while staying current with rapidly evolving treatment evidence.

 

Long Covid Treatment

Frequently Asked Questions:    Top Of Page

FAQs

Q1. What are the latest breakthroughs in long COVID treatment? Recent research has identified several promising approaches, including low-dose naltrexone for cognitive symptoms, montelukast for respiratory issues, and hyperbaric oxygen therapy for fatigue. Additionally, probiotic interventions targeting gut microbiome restoration have shown encouraging results in clinical trials.

Q2. How is long COVID diagnosed? Diagnosis remains primarily clinical, based on persistent symptoms following COVID-19 infection. However, emerging biomarkers like circulating spike protein, altered cytokine profiles, and corneal nerve damage detected through confocal microscopy are providing more objective diagnostic tools.

Q3. What are the most common symptoms of long COVID? The most frequently reported symptoms include fatigue, cognitive dysfunction (“brain fog”), shortness of breath, and cardiovascular issues like tachycardia. Many patients also experience gastrointestinal problems, musculoskeletal pain, and neurological symptoms.

Q4. How long do long COVID symptoms typically last? The duration of long COVID symptoms varies greatly between individuals. While some patients recover within a few months, others continue to experience symptoms for a year or more. Ongoing research aims to better understand the factors influencing symptom persistence and recovery.

Q5. Are there any effective treatments for long COVID fatigue? Several approaches show promise for managing long COVID-related fatigue. These include carefully structured pacing strategies to avoid post-exertional malaise, low-dose naltrexone, and in some cases, hyperbaric oxygen therapy. Additionally, addressing underlying issues like sleep disturbances and potential nutritional deficiencies may help improve energy levels.

 


References:   Top Of Page

[1] – https://www.who.int/publications/i/item/WHO-2019-nCoV-Post_COVID-19_condition-Clinical_case_definition-2021.1
[2] – https://pmc.ncbi.nlm.nih.gov/articles/PMC8691845/
[3] – https://pmc.ncbi.nlm.nih.gov/articles/PMC9877908/
[4] – https://pmc.ncbi.nlm.nih.gov/articles/PMC10036151/
[5] – https://pmc.ncbi.nlm.nih.gov/articles/PMC10113134/
[6] – https://pmc.ncbi.nlm.nih.gov/articles/PMC9250701/
[7] – https://www.mdpi.com/1422-0067/23/13/7209
[8] – https://pubmed.ncbi.nlm.nih.gov/35951203/
[9] – https://www.nature.com/articles/s41401-022-00998-0
[10] – https://pmc.ncbi.nlm.nih.gov/articles/PMC12595092/
[11] – https://www.nature.com/articles/s41584-023-00964-y
[12] – https://www.bmj.com/content/387/bmj-2024-081318
[13] – https://jamanetwork.com/journals/jamaneurology/fullarticle/2841155
[14] – https://www.cdc.gov/me-cfs/hcp/clinical-care/treating-the-most-disruptive-symptoms-first-and-preventing-worsening-of-symptoms.html
[15] – https://workwellfoundation.org/pacing-with-a-heart-rate-monitor-to-minimize-post-exertional-malaise-pem-in-me-cfs-and-long-covid/
[16] – https://patientresearchcovid19.com/clinicians-pacing-and-management-guide-for-me-cfs-and-long-covid/
[17] – https://www.ahajournals.org/doi/10.1161/CIR.0000000000001290
[18] – https://pmc.ncbi.nlm.nih.gov/articles/PMC11345335/
[19] – https://pmc.ncbi.nlm.nih.gov/articles/PMC10967081/
[20] – https://pubmed.ncbi.nlm.nih.gov/34991703/
[21] – https://pmc.ncbi.nlm.nih.gov/articles/PMC9383814/
[22] – https://www.eatg.org/hiv-news/cytodyn-inc-announces-positive-preliminary-results-of-unblinded-data-from-its-covid-19-long-haulers-clinical-trial/
[23] – https://pubmed.ncbi.nlm.nih.gov/39545965/
[24] – https://pubmed.ncbi.nlm.nih.gov/38672710/
[25] – https://www.massgeneralbrigham.org/en/about/newsroom/articles/trial-of-vitamin-b3-derivative-in-people-with-long-covid
[26] – https://www.nature.com/articles/s41380-023-02161-5
[27] – https://www.nature.com/articles/s41575-022-00698-4
[28] – https://pmc.ncbi.nlm.nih.gov/articles/PMC11756069/
[29] – https://insight.jci.org/articles/view/164813
[30] – https://www.nature.com/articles/s41579-022-00846-2
[31] – https://www.thelancet.com/journals/laninf/article/PIIS1473-3099(23)00735-1/fulltext
[32] – https://www.cdc.gov/long-covid/php/scientific-approach/index.html
[33] – https://pmc.ncbi.nlm.nih.gov/articles/PMC12408318/
[34] – https://www.frontiersin.org/journals/medicine/articles/10.3389/
fmed.2023.1085988/full
[35] – https://pmc.ncbi.nlm.nih.gov/articles/PMC10605628/


[Internal Medicine -Home]

 

Video Section


 

Recent Articles

Cardiology

   


 

 

About Author

Similar Articles

Leave a Reply


thpxl