Soothe Inflammation. Ease Pain. All from Home.
Experience the benefits of hypebaric oxygen safely and conveniently at home. Rent or Buy.

Inflammation is a double-edged sword in healing – acute inflammation helps repair tissue, but chronic inflammation can impede recovery. Hyperbaric oxygen therapy (HBOT), breathing 100% oxygen at elevated pressure, may modulate inflammation by flooding tissues with oxygen and affecting immune pathways[1][2]. Higher oxygen availability can reduce hypoxia-driven inflammatory signals and stimulate anti-inflammatory, antioxidant processes[3].
· Ulcerative Colitis: A meta-analysis (~780 patients) suggests adjunct HBOT improves ulcerative colitis outcomes. IL-6 and TNF-α decreased >60%, while anti-inflammatory IL-10 increased ~240% (Chen et al., 2021[4]). Clinical remission rates were higher with HBOT than standard care alone.
· Crohn’s Disease: In a small Crohn’s trial, 10 HBOT sessions (2.0–2.5 ATA) led to a ~59% drop in C-reactive protein (CRP) (from ~81 to 33 mg/L) and lower disease activity scores (Li et al., 2024[5]). HBOT was well-tolerated as an add-on to biologic therapy.
· Chronic Wounds (Diabetic Foot Ulcers): Patients with ischemic diabetic ulcers receiving HBOT (e.g. 30–40 sessions at ~2 ATA) showed reduced inflammation and better healing. At 3 months, erythrocyte sedimentation rate (ESR) fell significantly and CRP trended down, alongside faster ulcer closure (Martins-Mendes et al., 2025[6]).
· Bone Ischemia: In avascular necrosis of the femoral head, 60 HBOT sessions (2.5 ATA, 90 min each) produced significant drops in circulating IL-6 and TNF-α, paralleling reduced bone marrow edema and pain (Bosco et al., 2018[7]). This highlights HBOT’s systemic anti-inflammatory potential in ischemic conditions.
· Exercise Recovery: HBOT has been explored for acute inflammation after intense exercise. In healthy volunteers, a single 2.5 ATA HBOT session post-exercise significantly lowered IL-6 and fibrinogen levels compared to exercise alone, indicating reduced exercise-induced inflammation (Woo et al., 2020[8]).
· Oxygenation & Tissue Repair: HBOT rapidly increases oxygen delivery to hypoxic tissues, aiding cellular energy supply and healing. Improved oxygenation can enhance collagen synthesis, angiogenesis, and microbial killing, while mitigating the hypoxia that drives inflammatory gene activation[1].
· Immune Modulation: Hyperoxia from HBOT triggers mild oxidative stress that activates anti-inflammatory pathways. Studies suggest HBOT suppresses pro-inflammatory cytokines (e.g. TNF-α, IL-1β, IL-6) and boosts antioxidant defenses[2]. This may shift macrophages toward an “M2” reparative phenotype and promote resolution of inflammation.
Most findings come from small or heterogeneous studies. While results are encouraging, HBOT is used as an adjunctive therapy alongside standard treatments[9], not a standalone cure. There is variability in protocols (pressure, duration, number of sessions) and conditions studied. Larger controlled trials are needed to confirm these anti-inflammatory effects, optimize treatment regimens, and understand which patient populations benefit most[10]. Overall, current evidence supports HBOT’s potential to modulate inflammation as a supportive therapy, but more research will clarify its role in clinical practice.
· Chen P et al. Hyperbaric oxygen for ulcerative colitis: a systematic review and meta-analysis. Ther Adv Gastroenterol. 2021. PMID: 34349835. https://pubmed.ncbi.nlm.nih.gov/34349835
· Li Y et al. HBOT ameliorates intestinal inflammation in Crohn’s disease via gut microbiota modulation. J Transl Med. 2024. PMID: 38816750. https://pubmed.ncbi.nlm.nih.gov/38816750
· Bosco G et al. Hyperbaric oxygen therapy ameliorates osteonecrosis by modulating inflammation and oxidative stress. J Enzyme Inhib Med Chem. 2018. PMID: 30274530. https://pubmed.ncbi.nlm.nih.gov/30274530
· Martins-Mendes D et al. HBOT in recalcitrant diabetic foot ulcers: microvascular, biochemical and clinical impacts. Cells. 2025. PMID: 40801628. https://pubmed.ncbi.nlm.nih.gov/40801628
· Woo J et al. Effects of HBOT on inflammation after acute exercise (pilot study). Int J Environ Res Public Health. 2020. PMID: 33050362. https://pubmed.ncbi.nlm.nih.gov/33050362
· Wu X et al. The role of HBOT in inflammatory bowel disease: a narrative review. Med Gas Res. 2021. PMID: 33818446. https://pubmed.ncbi.nlm.nih.gov/33818446
[1] [2] [3] Medical Gas Research
https://journals.lww.com/mgar/fulltext/
[4] [9] Systematic review with meta-analysis: effectiveness of hyperbaric oxygenation therapy for ulcerative colitis - PubMed
https://pubmed.ncbi.nlm.nih.gov/34349835/
[5] Hyperbaric oxygen therapy ameliorates intestinal and systematic inflammation by modulating dysbiosis of the gut microbiota in Crohn's disease - PubMed
https://pubmed.ncbi.nlm.nih.gov/38816750/
[6] [10] Microvascular, Biochemical, and Clinical Impact of Hyperbaric Oxygen Therapy in Recalcitrant Diabetic Foot Ulcers - PubMed
https://pubmed.ncbi.nlm.nih.gov/40801628/
[7] Hyperbaric oxygen therapy ameliorates osteonecrosis in patients by modulating inflammation and oxidative stress - PubMed
https://pubmed.ncbi.nlm.nih.gov/30274530/
[8] Effects of Hyperbaric Oxygen Therapy on Inflammation, Oxidative/Antioxidant Balance, and Muscle Damage after Acute Exercise in Normobaric, Normoxic and Hypobaric, Hypoxic Environments: A Pilot Study - PubMed

Mitochondrial ATP production is fundamental for cellular function, and inefficiencies in mitochondrial energy metabolism are linked to insulin resistance and fatigue[1]. Hyperbaric oxygen therapy (HBOT) involves breathing pure oxygen at elevated pressure, saturating haemoglobin and dissolving up to ~4× more oxygen in blood[2]. This increased oxygen availability may enhance oxidative phosphorylation and tissue ATP generation.
HBOT studies for metabolism are relatively small (often <40 participants) and heterogeneous in protocols (1.5–2.4 ATA; single vs multiple sessions) and endpoints. While short-term improvements in bioenergetic biomarkers are reported, it is unclear how long these benefits persist and whether they translate into meaningful clinical outcomes (e.g. sustained glycaemic control or exercise tolerance). Not all findings are positive – e.g. some trials show no acute benefit on exercise recovery[6] – highlighting the need for larger, longer-term studies. Overall, HBOT may support cellular energy metabolism as an adjunctive intervention, but its clinical utility in metabolic diseases or athletic recovery requires further confirmation. Cautious optimism is warranted, and HBOT should not replace standard therapies.
· Sarabhai et al., 2023 – Hyperbaric oxygen rapidly improves tissue-specific insulin sensitivity and mitochondrial capacity in humans with type 2 diabetes: a randomised placebo-controlled crossover trial (Diabetologia)
· Wilkinson et al., 2012 – Hyperbaric oxygen therapy improves peripheral insulin sensitivity in humans (Diabet. Med.)
· Hadanny et al., 2022 – Effects of Hyperbaric Oxygen Therapy on Mitochondrial Respiration and Physical Performance in Middle-Aged Athletes: A Blinded, Randomized Controlled Trial (Sports Med Open)
· Huang et al., 2021 – Effects of Pre-, Post- and Intra-Exercise Hyperbaric Oxygen Therapy on Performance and Recovery: A Systematic Review and Meta-Analysis (Front. Physiol.)
[1] [2] [3] [7] [9] [10] Hyperbaric oxygen rapidly improves tissue-specific insulin sensitivity and mitochondrial capacity in humans with type 2 diabetes: a randomised placebo-controlled crossover trial | Diabetologia
https://link.springer.com/article/10.1007/s00125-022-05797-0
[4] Hyperbaric oxygen therapy improves peripheral insulin sensitivity in humans - PubMed
https://pubmed.ncbi.nlm.nih.gov/22269009/
[5] Effects of Hyperbaric Oxygen Therapy on Mitochondrial Respiration and Physical Performance in Middle-Aged Athletes: A Blinded, Randomized Controlled Trial | Sports Medicine - Open | Full Text
https://sportsmedicine-open.springeropen.com/articles/10.1186/s40798-021-00403-w
[6] [8] Frontiers | Effects of Pre-, Post- and Intra-Exercise Hyperbaric Oxygen Therapy on Performance and Recovery: A Systematic Review and Meta-Analysis
https://www.frontiersin.org/journals/physiology/articles/10.3389/fphys.2021.791872/full

Injury repair and immune balance rely on mobilising stem/progenitor cells and controlling inflammation. By raising dissolved oxygen levels under pressure, HBOT may influence these processes. Studies suggest HBOT can boost circulating progenitor cells involved in tissue regeneration while modulating immune activity – potentially enhancing anti-inflammatory signals and tempering excessive inflammation in various conditions.
· Stem cell mobilisation: Controlled trial in healthy adults (breathing 100% O₂ at 2.0 ATA for 2 hours) showed circulating CD34⁺ progenitor cells doubled after one HBOT session, and ~8-fold after 20 daily sessions (Thom et al., 2006)[1]. This rise in CD34⁺ cells was linked to nitric oxide-mediated bone marrow release.
· Immune ageing: In a prospective trial of 35 older adults (60 daily HBOT sessions, 2 ATA for 90 min each), HBOT increased telomere length in T‐cells, B‐cells and NK cells by ~20–37%, and reduced the proportion of senescent T-helper cells by ~37% (and senescent cytotoxic T-cells by ~11%) compared to baseline (Hachmo et al., 2020)[2]. These changes suggest a partial reversal of immune cell ageing markers after repeated HBOT.
· Anti-inflammatory shifts in IBD: A 2021 meta-analysis (13 studies, 780 ulcerative colitis patients) found adjunct HBOT improved clinical remission rates and was associated with higher anti-inflammatory IL-10 levels (SMD ≈ +2.40) alongside significantly lower TNF-α and IL-6 levels vs. standard care alone (Chen et al., 2021)[3]. This aligns with clinical reports of reduced inflammation and improved healing in refractory inflammatory bowel disease.
· Acute infection response: Observational data in necrotising soft tissue infections (n=209 HBOT-treated patients) showed each HBOT session (typically ~2.8 ATA, 90 min) acutely lowered excessive inflammatory cytokines. Notably, median plasma IL-6 levels dropped by ~29 pg/mL after the first HBOT session (vs pre-treatment) and G-CSF also decreased (Hedetoft et al., 2021)[4], suggesting HBOT can blunt hyperinflammation in severe infections.
· Innate immunity (NK cells): A small crossover RCT in healthy women exposed to mild HBOT (1.4 ATA, ~35% O₂ for 70 min) reported a significant increase in circulating natural killer (NK) cell counts post-treatment, compared to no change under normoxic conditions. Importantly, the mild HBOT did not elevate IL-6 or oxidative stress markers (Badur Un Nisa et al., 2023)[5]. This indicates even lower-pressure HBOT may enhance innate immune cell activity without triggering systemic inflammation.
· Human: Hyperoxic exposures trigger endothelial nitric oxide (NO) release, which in turn promotes stem cell factor and disrupts the SDF-1/CXCR4 retention of progenitors in bone marrow. HBOT-induced NO surges (↑ ~1 µM in marrow) correlate with the mobilization of CD34⁺ cells, an effect abolished in NO-synthase knockout models[6]. This *NO-mediated signalling is thought to loosen marrow niches, allowing stem/progenitor cells to enter circulation.
· Preclinical: Intermittent high-oxygen sessions followed by normal oxygen can mimic a hypoxia–reoxygenation pattern that modulates immune pathways. In animal studies, HBOT shifts macrophages from an M1 (pro-inflammatory) phenotype to M2 (repair-oriented) phenotype[7], with treated macrophages showing higher arginase-1 and IL-10 levels during clearance of debris[8]. HBOT has also been noted to reduce HIF-1α-driven Th17 T-cell responses and increase regulatory T-cells in models of autoimmunity, aligning with reduced pro-inflammatory cytokine production. These mechanisms suggest HBOT creates a transient oxidative stress that paradoxically activates anti-inflammatory and progenitor-release signals.
Much of the evidence comes from small trials or adjunctive therapy studies with heterogeneous protocols (varying pressure, duration, and patient populations). Many findings rely on surrogate endpoints (cell counts, cytokines) rather than long-term clinical outcomes. It remains unclear how long immune or stem cell changes persist after HBOT, or how they translate into tangible health benefits. Larger controlled trials are needed to confirm HBOT’s immunomodulatory effects and optimal dosing. Overall, HBOT is considered an adjunctive therapy; while studies suggest it may support immune balance and repair, it is not a standalone cure and responses vary. Future research should clarify which patient groups benefit most, and establish whether repeated HBOT can sustainably enhance immune regeneration without adverse effects.
· Thom SR et al. (2006). Stem cell mobilization by hyperbaric oxygen. Am J Physiol Heart Circ Physiol, 290(4):H1378–86. DOI: 10.1152/ajpheart.00888.2005[1][6]
· Hachmo Y et al. (2020). Hyperbaric oxygen therapy increases telomere length and decreases immunosenescence in isolated blood cells: a prospective trial. Aging (Albany NY), 12(22):22445–22456. DOI: 10.18632/aging.202188[9]
· Chen P et al. (2021). Systematic review with meta-analysis: effectiveness of hyperbaric oxygenation therapy for ulcerative colitis. Therap Adv Gastroenterol, 14:17562848211023394. DOI: 10.1177/17562848211023394[3]
· Hedetoft M et al. (2021). Hyperbaric oxygen treatment is associated with a decrease in cytokine levels in patients with necrotizing soft-tissue infection. Physiol Reports, 9(6):e14757. DOI: 10.14814/phy2.14757[4]
· Badur Un Nisa et al. (2023). Mild hyperbaric oxygen exposure enhances peripheral circulatory natural killer cells in healthy young women. Life (Basel), 13(2):408. DOI: 10.3390/life13020408[5]
· Chen L et al. (2024). The new insights of hyperbaric oxygen therapy: focus on inflammatory bowel disease. Precision Clinical Medicine, 7(1):pbae001. DOI: 10.1093/pcmedi/pbae001[7][8]
[1] [6] Stem cell mobilization by hyperbaric oxygen - PubMed
https://pubmed.ncbi.nlm.nih.gov/16299259/
[2] [9] Hyperbaric oxygen therapy increases telomere length and decreases immunosenescence in isolated blood cells: a prospective trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/33206062/
[3] Systematic review with meta-analysis: effectiveness of hyperbaric oxygenation therapy for ulcerative colitis - PubMed
https://pubmed.ncbi.nlm.nih.gov/34349835/
[4] Hyperbaric oxygen treatment is associated with a decrease in cytokine levels in patients with necrotizing soft-tissue infection - PubMed
https://pubmed.ncbi.nlm.nih.gov/33719215/
[5] Mild Hyperbaric Oxygen Exposure Enhances Peripheral Circulatory Natural Killer Cells in Healthy Young Women - PubMed
https://pubmed.ncbi.nlm.nih.gov/36836764/
[7] [8] new insights of hyperbaric oxygen therapy: focus on inflammatory bowel disease | Precision Clinical Medicine | Oxford Academic

Neuroplasticity is the brain’s capacity to adapt and form new neural connections, supporting recovery and cognitive resilience after injury. Hyperbaric Oxygen Therapy (HBOT) involves breathing 100% oxygen at higher-than-normal atmospheric pressure, which markedly increases oxygen delivery to brain tissue[1]. Enhanced oxygen availability can boost cellular metabolism and cerebral blood flow, potentially creating conditions that facilitate neuronal repair and network reorganization.
Evidence for HBOT’s neurocognitive benefits is promising but heterogeneous. Studies span stroke, TBI, and other conditions with varying protocols and outcome measures. Many trials (especially early ones) lacked sham controls, making placebo effects possible; however, recent sham-controlled studies in mTBI have yielded mixed results. The durability of cognitive gains post-HBOT is not yet fully clear, and optimal dosing (pressure, number of sessions) needs clarification. Overall, HBOT is considered an adjunct to standard neurorehabilitation rather than a standalone treatment. Larger, multi-centre trials with standardised protocols are needed to confirm long-term efficacy and to identify which patients are most likely to benefit[11].
[1] [6] [11] Annals of Medicine and Surgery
[2] [3] Hyperbaric oxygen induces late neuroplasticity in post stroke patients--randomized, prospective trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/23335971/
[4] [5] Hyperbaric oxygen therapy can improve post concussion syndrome years after mild traumatic brain injury - randomized prospective trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/24260334/
[7] [8] Cognitive enhancement of healthy older adults using hyperbaric oxygen: a randomized controlled trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/32589613/
[9] [10] Hyperbaric Oxygen Therapy Can Diminish Fibromyalgia Syndrome – Prospective Clinical Trial | PLOS One
https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0127012
[12] [13] Frontiers | Hyperbaric Oxygen Therapy Can Induce Angiogenesis and Regeneration of Nerve Fibers in Traumatic Brain Injury Patients
https://www.frontiersin.org/journals/human-neuroscience/articles/10.3389/fnhum.2017.00508/full

Collagen deposition and tissue perfusion are critical for wound repair; chronic wounds often remain unhealed due to hypoxia impairing fibroblast activity and angiogenesis. By having patients breathe 100% oxygen at elevated pressure, HBOT dramatically increases dissolved oxygen in plasma, which may fuel collagen synthesis and new capillary growth in injured tissues. Under hyperoxia, wounds can receive the oxygen needed for fibroblast proliferation, cross-linking of collagen fibers, and efficient tissue regeneration, potentially accelerating closure and strengthening repairs.
· Diabetic Foot Ulcers (DFU) – A meta-analysis of 14 RCTs in DFU patients (typical HBOT ~2.4 ATA for ~90 min, 30–40 sessions) found adjunctive HBOT roughly doubled short-term wound healing rates (risk ratio ~2.4 at 8–12 weeks) and significantly reduced major amputation risk (RR ~0.31) vs standard care[1] (Faruk/Oley et al., 2024).
· Chronic DFU – 1-Year Outcomes – In a double-blind RCT (n=94, Wagner grade 2–4 DFUs), HBOT at 2.5 ATA for 85 min, 5 days/week for 8 weeks (40 sessions) led to higher 1-year complete ulcer healing (52% vs 29% with sham HBOT; P = 0.03)[2]. Patients receiving HBOT had low adverse event rates and fewer subsequent amputations (Löndahl et al., 2010).
· Radiation-Injured Tissue – A Cochrane review (14 trials) in late radiation wounds (e.g. after head/neck or pelvic radiotherapy) reported that HBOT (2.0–2.5 ATA, ~90 min, ~30 sessions) improved healing outcomes. For example, in mandibular osteoradionecrosis HBOT increased rates of mucosal coverage (RR 1.3, 95% CI 1.1–1.6; NNT ~5)[3], and in chronic radiation proctitis it improved symptom resolution (RR ~1.7; NNT ~5)[4] compared to no HBOT (Bennett et al., 2016).
· Graft/Flap Salvage – Retrospective clinical data indicate HBOT can rescue compromised skin grafts and flaps. In one series, ~75.7% of failing grafts or flaps were salvaged with HBOT (avg. ~30 sessions at 2.0–2.5 ATA)[5]. Similarly, HBOT initiated early post-surgery markedly increased flap survival rates (e.g. one trial noted an ~8.7-fold higher flap success rate, NNT ~4)[6] (Francis & Baynosa, 2017).
· Venous & Mixed Ulcers – Evidence in non-diabetic wounds is limited but suggestive. A small RCT in refractory venous leg ulcers (n=16) showed significantly greater wound area reduction at 6 weeks with HBOT (~33% more area healed than controls)[7]. Another trial in mixed-aetiology leg ulcers found ~62% ulcer area reduction after 30 days of HBOT vs standard care[8] (Cochrane, 2015).
· Preclinical: Elevated oxygen tension in wounds boosts fibroblast function and collagen synthesis while stimulating angiogenesis. Animal and in-vitro studies show HBOT mitigates ischemia–reperfusion injury and triggers growth factor-driven neovascularisation[9]. Hyperoxia-induced reactive oxygen signaling can upregulate VEGF and other factors, leading to new capillary growth and a stronger extracellular matrix.
· Human: In patients, HBOT acutely raises periwound tissue oxygen (tcPO₂), creating an environment for collagen deposition and re-epithelialisation. Studies report that HBOT modulates the wound microenvironment by reducing inflammatory cytokines and increasing angiogenic factors (e.g. vascular endothelial growth factor, epidermal growth factor), which together may accelerate granulation tissue formation and remodeling[10]. Improved microcirculation and oxygen delivery under HBOT support effective collagen cross-linking and bacterial defense, aiding overall healing.
The quality of evidence for HBOT in tissue repair is moderate and varies by wound type. Many RCTs have small sample sizes and heterogeneous methods (different wound etiologies and HBOT protocols), making outcomes difficult to generalise[11]. Robust support exists for diabetic foot ulcers, whereas data for venous, arterial, or pressure ulcers are sparse or inconclusive[11]. Trials often use HBOT as an adjunct to standard care (debridement, offloading, antibiotics, etc.), and it is not a stand-alone cure. Safety profiles in wound care studies are acceptable – no severe adverse events were commonly reported[12][13] – but HBOT requires specialised facilities and patient screening (e.g. to avoid contraindications such as untreated pneumothorax[14]). Further large-scale studies are needed to confirm optimal HBOT dosing, long-term scar quality outcomes (e.g. tensile strength), and cost-effectiveness across various chronic wound populations[15]. Until then, HBOT remains an adjunctive therapy that may support collagen deposition and healing, to be considered on a case-by-case basis alongside standard wound management.
· Oley MH et al. (2024) – Hyperbaric Oxygen Therapy for Diabetic Foot Ulcers Based on Wagner Grading: A Systematic Review and Meta-analysis. Plast Reconstr Surg Glob Open, 12(3): e5692. DOI: 10.1097/GOX.0000000000005692
· Löndahl M et al. (2010) – Hyperbaric oxygen therapy facilitates healing of chronic foot ulcers in patients with diabetes. Diabetes Care, 33(5): 998–1003. DOI: 10.2337/dc09-1754
· Bennett MH et al. (2016) – Hyperbaric oxygen therapy for late radiation tissue injury (Review). Cochrane Database Syst Rev, 2016(4): CD005005. DOI: 10.1002/14651858.CD005005.pub4
· Francis A & Baynosa RC (2017) – Hyperbaric Oxygen Therapy for the Compromised Graft or Flap. Adv Wound Care (New Rochelle), 6(1): 23–32. DOI: 10.1089/wound.2016.0707
· Kranke P et al. (2015) – Hyperbaric oxygen therapy for chronic wounds. Cochrane Database Syst Rev, 2015(6): CD004123. DOI: 10.1002/14651858.CD004123.pub4
· Zhang R et al. (2025) – Effect of hyperbaric oxygen therapy on postoperative muscle damage and inflammation following total knee arthroplasty: a randomized controlled trial. Sci Rep 13: 14273. DOI: 10.1038/s41598-025-06223-2
[1] Hyperbaric Oxygen Therapy for Diabetic Foot Ulcers Based on Wagner Grading: A Systematic Review and Meta-analysis - PMC
https://pmc.ncbi.nlm.nih.gov/articles/PMC10962882/
[2] Hyperbaric oxygen therapy facilitates healing of chronic foot ulcers in patients with diabetes - PubMed
https://pubmed.ncbi.nlm.nih.gov/20427683/
[3] [4] [6] [12] Hyperbaric oxygen therapy for late radiation tissue injury - PubMed
https://pubmed.ncbi.nlm.nih.gov/27123955/
[5] Hyperbaric oxygen for compromised graft
https://www.biobarica.com/en/news/hyperbaric-oxygen-for-compromised-graft-9SDSY6ENFM1phuVom6hm/
[7] [8] [11] [13] Hyperbaric oxygen therapy for treating chronic wounds | Cochrane
https://www.cochrane.org/evidence/CD004123_hyperbaric-oxygen-therapy-treating-chronic-wounds
[9] The effect of hyperbaric oxygen on compromised grafts and flaps
[10] Effect of hyperbaric oxygen therapy on postoperative muscle damage and inflammation following total knee arthroplasty: a randomized controlled trial | Scientific Reports
[14] Hyperbaric Therapy for Skin Grafts and Flaps - StatPearls - NCBI Bookshelf
https://www.ncbi.nlm.nih.gov/books/NBK470219/
[15] Hyperbaric Oxygen Therapy for the Compromised Graft or Flap - PubMed

Neurons have high energy needs, and brain plasticity relies on efficient metabolism and blood flow. Photobiomodulation (PBM) with red to near-infrared light (600–1100 nm) is thought to boost cellular energy by stimulating cytochrome c oxidase in mitochondria, enhancing ATP production[1]. In the brain, transcranial PBM (tPBM) may improve cerebral metabolism and network dynamics by increasing regional cerebral blood flow and triggering anti-inflammatory and neurotrophic pathways[2]. These effects could support synaptic plasticity and cognitive function, although PBM is exploratory and not a standalone treatment.
· Depression: A 2024 meta-analysis of 11 RCTs found PBM significantly alleviated depressive symptoms compared to sham (pooled SMD ≈ –0.55, 95% CI –0.75 to –0.35)[3]. Sleep outcomes did not significantly differ. Subgroup analysis suggested optimal tPBM parameters around 823 nm, 10–100 J/cm² fluence at ≤50 mW/cm², for 30 min sessions <3 times/week over >15 sessions[4] (Ji et al., 2024).
· Healthy Ageing: A single-blind crossover RCT (N=55, healthy older adults) using 1064 nm laser to the prefrontal scalp showed improved working memory: 3-back test accuracy and reaction time significantly better after active tPBM vs sham[5]. fNIRS imaging revealed increased functional connectivity and network efficiency in frontoparietal regions during tPBM, correlating with the cognitive gains[5] (Yang et al., 2025).
· Mild Cognitive Impairment: In a 3-week controlled trial (n=36), older adults with MCI receiving tPBM (versus no treatment) had significant improvements in executive function and memory. The tPBM group improved on the Montreal Cognitive Assessment and Shape Trail Making B (faster by several seconds), with reduced anti-saccade latencies indicating better attention control[6]. Notably, more tPBM-treated participants showed reduced depressive symptoms post-intervention[6] (Lee et al., 2025).
· Traumatic Brain Injury: Preliminary studies in chronic TBI suggest tPBM can augment neuroplasticity. For example, 8 out of 12 patients showed increased regional cerebral blood flow on SPECT scans after multi-session tPBM[7]. Another pilot reported strengthened functional connectivity within the salience and executive control networks and a rise in N-acetylaspartate (a neuronal health metabolite) in the anterior cingulate after 6 weeks of tPBM[8]. These brain changes correlated with improved attention, memory and mood in TBI patients (Naeser et al., 2023).
· Cellular Energy and Blood Flow: Human and animal research indicates tPBM light boosts mitochondrial respiration, increasing ATP and releasing nitric oxide[1]. This can dilate blood vessels and improve cerebral blood flow, while up-regulating growth factors that promote neuron survival and sprouting[2]. PBM also has documented anti-inflammatory and antioxidant effects in neural tissue[2].
· Network and Neurochemical Effects: By energizing cells, tPBM may enhance brain network function. Small studies have observed increased EEG alpha power and functional connectivity after tPBM, suggesting more efficient neural oscillations. In parallel, tPBM has been associated with higher brain levels of metabolites like NAA (linked to neuronal viability), hinting at improved neuronal health[8]. These changes reflect a milieu that may support synaptic plasticity and cognitive processing.
Current evidence comes from relatively small trials with heterogeneous PBM parameters, making it difficult to standardise an optimal dose[9]. Some well-controlled studies have found no significant benefits (e.g. no cognitive or mood improvement in a 30-patient dementia trial)[10], highlighting mixed results. Blinding can be challenging due to noticeable device effects, and the durability of tPBM gains remains uncertain. Larger, multi-centre RCTs – with careful dose finding – are needed to confirm benefits and ideal treatment protocols[9]. At present, tPBM is an adjunctive, investigational approach for brain health, and not an established therapy.
· Salehpour F. et al. (2018). Brain Photobiomodulation Therapy: A Narrative Review. Molecular Neurobiology, 55(8), 6601–6636. DOI: 10.1007/s12035-017-0852-4[11][12]
· Ji Q. et al. (2024). Photobiomodulation Improves Depression Symptoms: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Frontiers in Psychiatry, 14, 1267415. DOI: 10.3389/fpsyt.2023.1267415[3][4]
· Yang Q. et al. (2025). Transcranial Photobiomodulation Improves Functional Brain Networks and Working Memory in Healthy Older Adults: An fNIRS Study. NeuroImage, 316, 121305. DOI: 10.1016/j.neuroimage.2025.121305[5]
· Lee T.L. et al. (2025). Improved Cognitive Function, Efficiency, Saccadic Eye Movement, and Depressive Symptoms in Mild Cognitive Impairment With Transcranial Photobiomodulation. Journal of Alzheimer’s Disease, 107(2), 529–541. DOI: 10.1177/13872877251361033[6]
· Jarrahi S. et al. (2025). The Efficacy of Photobiomodulation Therapy in Improving Cognitive Function and Reducing Depression and Anxiety in Patients With Mild-to-Moderate Dementia: A Double-Blinded Randomized Clinical Trial. Photobiomodulation, Photomedicine, and Laser Surgery, 43(9), 411–416. DOI: 10.1177/25785478251376443[10]
· Zeng J. et al. (2024). Can Transcranial Photobiomodulation Improve Cognitive Function in TBI Patients? A Systematic Review. Frontiers in Psychology, 15, 1378570. DOI: 10.3389/fpsyg.2024.1378570[7][8]
[1] [2] Brain Photobiomodulation Therapy: a Narrative Review-Bohrium
[3] [4] Photobiomodulation improves depression symptoms: a systematic review and meta-analysis of randomized controlled trials - PMC
https://pmc.ncbi.nlm.nih.gov/articles/PMC10866010/
[5] Transcranial photobiomodulation improves functional brain networks and working memory in healthy older adults: An fNIRS study - PubMed
https://pubmed.ncbi.nlm.nih.gov/40482941/
[6] Improved cognitive function, efficiency, saccadic eye movement, and depressive symptoms in mild cognitive impairment with transcranial photobiomodulation
https://www.researchgate.net/publication/
[7] [8] [9] Frontiers | Can transcranial photobiomodulation improve cognitive function in TBI patients? A systematic review
https://www.frontiersin.org/journals/psychology/articles/10
[10] The Efficacy of Photobiomodulation Therapy in Improving Cognitive Function and Reducing Depression and Anxiety in Patients with Mild-to-Moderate Dementia: A Double-Blinded Randomized Clinical Trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/40930542/
[11] [12] Brain Photobiomodulation Therapy: a Narrative Review - PubMed

Cellular energy (ATP) is fundamental for muscle function, brain activity, and tissue recovery. Red and near-infrared photobiomodulation (PBM) is being explored for its ability to enhance mitochondrial ATP production by delivering light absorbed by chromophores like cytochrome c oxidase in the respiratory chain. By modulating mitochondrial activity, PBM may support energy metabolism and exercise capacity in cells and tissues.
Most PBM studies are small and heterogeneous, with varying wavelengths, dosages, and timing protocols, making it challenging to identify optimal treatment parameters. Positive results in tightly controlled trials (often with athletes) may not generalize to all populations, and some studies have found no significant effects on performance or recovery. Meta-analyses note encouraging trends but also potential publication bias and risk of bias in many trials[6]. The durability of PBM’s benefits is unclear (e.g. effects may be acute and diminish with repeated use[11]), and there is no consensus on long-term safety of frequent high-dose use. Overall, PBM is considered an adjunct modality that may support cellular energy and recovery; it is not a substitute for proven training, nutrition, or medical therapies. Larger, well-controlled studies are needed to confirm efficacy, ideal dosing, and the longevity of benefits.
[1] Improvement of Performance and Reduction of Fatigue With Low-Level Laser Therapy in Competitive Cyclists - PubMed
https://pubmed.ncbi.nlm.nih.gov/28422520/
[2] Low-level laser therapy improves the VO2 kinetics in competitive cyclists - PubMed
https://pubmed.ncbi.nlm.nih.gov/29119418/
[3] [11] Effects of Photobiomodulation Therapy on Performance in Successive Time-to-Exhaustion Cycling Tests: A Randomized Double-Blinded Placebo-Controlled Trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/37873903/
[4] Effects of Photobiomodulation Therapy on Performance in Successive Time-to-Exhaustion Cycling Tests: A Randomized Double-Blinded Placebo-Controlled Trial
https://www.mdpi.com/2411-5142/8/4/144
[5] [6] Effects of Low-Level Laser Therapy on Muscular Performance and Soreness Recovery in Athletes: A Meta-analysis of Randomized Controlled Trials - PubMed
https://pubmed.ncbi.nlm.nih.gov/34428975/
[7] Photobiomodulation in human muscle tissue: an advantage in sports performance? - PubMed
https://pubmed.ncbi.nlm.nih.gov/27874264/
[8] Pretreatment of neurons with 670 nm LED once a day for 3 days (A) or 5... | Download Scientific Diagram
https://www.researchgate.net/figure/
[9] [10] Photobiomodulation increases mitochondrial citrate synthase activity in rats submitted to aerobic training - PubMed

Photobiomodulation (PBM) uses red to near-infrared light (600–1100 nm) absorbed by cellular chromophores (e.g. cytochrome‐c oxidase) to modulate immune homeostasis and tissue repair[1]. By improving mitochondrial energy production and redox signaling, PBM may dampen excessive inflammatory responses while supporting pro‐healing pathways. Emerging research suggests PBM can reduce pro-inflammatory cytokines and oxidative stress, potentially promoting a more regulated immune profile conducive to tissue regeneration.
Current evidence is promising but heterogeneous. Small-sample RCTs and varying PBM dosages (wavelengths, fluence, timing) lead to mixed outcomes. While short-term reductions in inflammatory biomarkers are reported, consistency across larger trials is needed. There is scant direct evidence of PBM elevating human anti-inflammatory cytokines (e.g. IL-10) – most human trials show no significant IL-10 rise[2]. Preliminary findings (e.g. stem cell mobilization) require confirmation. Overall, PBM appears safe with adjunctive benefit for inflammation; however, standardized protocols and long-term studies are required to establish optimal parameters and clarify which patient populations may respond best.
[1] [3] Photobiomodulation therapy (PBMT) on acute pain and inflammation in patients who underwent total hip arthroplasty-a randomized, triple-blind, placebo-controlled clinical trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/29909435/
[2] Frontiers | Photobiomodulation Improves Serum Cytokine Response in Mild to Moderate COVID-19: The First Randomized, Double-Blind, Placebo Controlled, Pilot Study
https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2022.929837/full
[4] Efficacy of Photobiomodulation Therapy in the Treatment of Pain and Inflammation: A Literature Review - PMC
https://pmc.ncbi.nlm.nih.gov/articles/PMC10094541/
[5] Photobiomodulation Therapy is Able to Modulate PGE2 Levels in Patients With Chronic Non-Specific Low Back Pain: A Randomized Placebo-Controlled Trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/32330315/
[6] Photobiomodulation Therapy to Autologous Bone Marrow in Humans Significantly Increases the Concentration of Circulating Stem Cells and Macrophages: A Pilot Study - PubMed
https://pubmed.ncbi.nlm.nih.gov/35196142/
[7] [8] Reprogramming Macrophage Phenotypes With Photobiomodulation for Improved Inflammation Control in ENT Organ Tissues - PubMed

Neurons have high energy needs, and brain plasticity relies on efficient metabolism and blood flow. Photobiomodulation (PBM) with red to near-infrared light (600–1100 nm) is thought to boost cellular energy by stimulating cytochrome c oxidase in mitochondria, enhancing ATP production[1]. In the brain, transcranial PBM (tPBM) may improve cerebral metabolism and network dynamics by increasing regional cerebral blood flow and triggering anti-inflammatory and neurotrophic pathways[2]. These effects could support synaptic plasticity and cognitive function, although PBM is exploratory and not a standalone treatment.
· Depression: A 2024 meta-analysis of 11 RCTs found PBM significantly alleviated depressive symptoms compared to sham (pooled SMD ≈ –0.55, 95% CI –0.75 to –0.35)[3]. Sleep outcomes did not significantly differ. Subgroup analysis suggested optimal tPBM parameters around 823 nm, 10–100 J/cm² fluence at ≤50 mW/cm², for 30 min sessions <3 times/week over >15 sessions[4] (Ji et al., 2024).
· Healthy Ageing: A single-blind crossover RCT (N=55, healthy older adults) using 1064 nm laser to the prefrontal scalp showed improved working memory: 3-back test accuracy and reaction time significantly better after active tPBM vs sham[5]. fNIRS imaging revealed increased functional connectivity and network efficiency in frontoparietal regions during tPBM, correlating with the cognitive gains[5] (Yang et al., 2025).
· Mild Cognitive Impairment: In a 3-week controlled trial (n=36), older adults with MCI receiving tPBM (versus no treatment) had significant improvements in executive function and memory. The tPBM group improved on the Montreal Cognitive Assessment and Shape Trail Making B (faster by several seconds), with reduced anti-saccade latencies indicating better attention control[6]. Notably, more tPBM-treated participants showed reduced depressive symptoms post-intervention[6] (Lee et al., 2025).
· Traumatic Brain Injury: Preliminary studies in chronic TBI suggest tPBM can augment neuroplasticity. For example, 8 out of 12 patients showed increased regional cerebral blood flow on SPECT scans after multi-session tPBM[7]. Another pilot reported strengthened functional connectivity within the salience and executive control networks and a rise in N-acetylaspartate (a neuronal health metabolite) in the anterior cingulate after 6 weeks of tPBM[8]. These brain changes correlated with improved attention, memory and mood in TBI patients (Naeser et al., 2023).
· Cellular Energy and Blood Flow: Human and animal research indicates tPBM light boosts mitochondrial respiration, increasing ATP and releasing nitric oxide[1]. This can dilate blood vessels and improve cerebral blood flow, while up-regulating growth factors that promote neuron survival and sprouting[2]. PBM also has documented anti-inflammatory and antioxidant effects in neural tissue[2].
· Network and Neurochemical Effects: By energizing cells, tPBM may enhance brain network function. Small studies have observed increased EEG alpha power and functional connectivity after tPBM, suggesting more efficient neural oscillations. In parallel, tPBM has been associated with higher brain levels of metabolites like NAA (linked to neuronal viability), hinting at improved neuronal health[8]. These changes reflect a milieu that may support synaptic plasticity and cognitive processing.
Current evidence comes from relatively small trials with heterogeneous PBM parameters, making it difficult to standardise an optimal dose[9]. Some well-controlled studies have found no significant benefits (e.g. no cognitive or mood improvement in a 30-patient dementia trial)[10], highlighting mixed results. Blinding can be challenging due to noticeable device effects, and the durability of tPBM gains remains uncertain. Larger, multi-centre RCTs – with careful dose finding – are needed to confirm benefits and ideal treatment protocols[9]. At present, tPBM is an adjunctive, investigational approach for brain health, and not an established therapy.
· Salehpour F. et al. (2018). Brain Photobiomodulation Therapy: A Narrative Review. Molecular Neurobiology, 55(8), 6601–6636. DOI: 10.1007/s12035-017-0852-4[11][12]
· Ji Q. et al. (2024). Photobiomodulation Improves Depression Symptoms: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Frontiers in Psychiatry, 14, 1267415. DOI: 10.3389/fpsyt.2023.1267415[3][4]
· Yang Q. et al. (2025). Transcranial Photobiomodulation Improves Functional Brain Networks and Working Memory in Healthy Older Adults: An fNIRS Study. NeuroImage, 316, 121305. DOI: 10.1016/j.neuroimage.2025.121305[5]
· Lee T.L. et al. (2025). Improved Cognitive Function, Efficiency, Saccadic Eye Movement, and Depressive Symptoms in Mild Cognitive Impairment With Transcranial Photobiomodulation. Journal of Alzheimer’s Disease, 107(2), 529–541. DOI: 10.1177/13872877251361033[6]
· Jarrahi S. et al. (2025). The Efficacy of Photobiomodulation Therapy in Improving Cognitive Function and Reducing Depression and Anxiety in Patients With Mild-to-Moderate Dementia: A Double-Blinded Randomized Clinical Trial. Photobiomodulation, Photomedicine, and Laser Surgery, 43(9), 411–416. DOI: 10.1177/25785478251376443[10]
· Zeng J. et al. (2024). Can Transcranial Photobiomodulation Improve Cognitive Function in TBI Patients? A Systematic Review. Frontiers in Psychology, 15, 1378570. DOI: 10.3389/fpsyg.2024.1378570[7][8]
[1] [2] Brain Photobiomodulation Therapy: a Narrative Review-Bohrium
[3] [4] Photobiomodulation improves depression symptoms: a systematic review and meta-analysis of randomized controlled trials - PMC
https://pmc.ncbi.nlm.nih.gov/articles/PMC10866010/
[5] Transcranial photobiomodulation improves functional brain networks and working memory in healthy older adults: An fNIRS study - PubMed
https://pubmed.ncbi.nlm.nih.gov/40482941/
[6] Improved cognitive function, efficiency, saccadic eye movement, and depressive symptoms in mild cognitive impairment with transcranial photobiomodulation
[7] [8] [9] Frontiers | Can transcranial photobiomodulation improve cognitive function in TBI patients? A systematic review
https://www.frontiersin.org/journals/psychology/articles/10.3389/fpsyg.2024.1378570/
[10] The Efficacy of Photobiomodulation Therapy in Improving Cognitive Function and Reducing Depression and Anxiety in Patients with Mild-to-Moderate Dementia: A Double-Blinded Randomized Clinical Trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/40930542/
[11] [12] Brain Photobiomodulation Therapy: a Narrative Review - PubMed

Collagen is the primary structural protein that supports tissue integrity and plays a central role in wound repair and skin elasticity. Photobiomodulation (PBM) with red to near-infrared light (600–1100 nm) has been explored as a non-invasive way to enhance tissue healing by stimulating cellular processes in the skin and connective tissues. By absorbing photons, cells (particularly fibroblasts) may increase their energy production and activate signaling pathways that promote collagen synthesis and remodeling of the extracellular matrix, potentially improving tissue repair and strength.
Overall, studies suggest PBM may support collagen formation and tissue repair, but evidence quality is moderate and heterogeneous. Clinical trials vary widely in wavelength, dose (energy densities from <5 J/cm² to >100 J/cm²), treatment frequency, and device type (laser vs. LED), making it challenging to identify an optimal protocol. Many studies have small sample sizes or short follow-up, and outcome measures range from subjective scales to objective histology or imaging, with variable consistency. Not all trials show significant benefits, and some meta-analyses rate the evidence as very low to moderate quality[9][6]. The durability of PBM-induced improvements (e.g. whether collagen gains persist long-term) remains unclear. PBM is generally considered an adjunct therapy – for example, used alongside standard wound care or rehabilitation – rather than a standalone treatment. Larger, well-controlled studies are needed to confirm efficacy across different tissues and to refine treatment parameters (dose-response) for reproducible outcomes. As a non-invasive and safe modality, PBM holds promise in accelerating healing and improving scar quality, but claims must be cautious: current evidence suggests benefits without guaranteeing them for every patient or condition.
[1] [8] A prospective, randomized, placebo-controlled, double-blinded, and split-face clinical study on LED phototherapy for skin rejuvenation: clinical, profilometric, histologic, ultrastructural, and biochemical evaluations and comparison of three different treatment settings - PubMed
https://pubmed.ncbi.nlm.nih.gov/17566756/
[2] Clinical trial of a novel non-thermal LED array for reversal of photoaging: clinical, histologic, and surface profilometric results - PubMed
https://pubmed.ncbi.nlm.nih.gov/15654716/
[3] A controlled trial to determine the efficacy of red and near-infrared light treatment in patient satisfaction, reduction of fine lines, wrinkles, skin roughness, and intradermal collagen density increase - PubMed
https://pubmed.ncbi.nlm.nih.gov/24286286/
[4] Photobiomodulation therapy with an 830-nm light-emitting diode for the prevention of thyroidectomy scars: a randomized, double-blind, sham device-controlled clinical trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/36045183/
[5] [6] [9] The effect of low-level red and near-infrared photobiomodulation on pain and function in tendinopathy: a systematic review and meta-analysis of randomized control trials - PMC
https://pmc.ncbi.nlm.nih.gov/articles/PMC8364035/
[7] Efficacy of Light-Emitting Diode-Mediated Photobiomodulation in Tendon Healing in a Murine Model

Osteoarthritis (OA) and rheumatoid arthritis (RA) cause chronic joint pain, stiffness, and functional disability. Adjunctive therapies such as hyperbaric oxygen therapy (HBOT) and red light photobiomodulation (PBM) have been explored for their potential to modulate inflammation and enhance tissue repair via oxygen- and light-mediated pathways. Early studies suggest these modalities may help alleviate pain and inflammatory markers in arthritis when used alongside standard care[1][2], though robust evidence is still emerging.
HBOT:
PBM (Red Light Therapy):
HBOT: By elevating oxygen availability in tissues, HBOT may reverse hypoxia in inflamed joints and disrupt hypoxia-inducible pathways (e.g. down-regulating HIF-1α). Increased oxygen tension can reduce edema and improve microcirculation, fostering tissue repair. Mechanistically, preclinical studies indicate HBOT can attenuate inflammatory signaling cascades like NF-κB, resulting in lower production of pro-inflammatory cytokines (such as TNF-α, IL-1β, IL-6)[17]. Human and animal data show HBOT triggers antioxidative responses (e.g. via Nrf2 activation) and promotes angiogenesis (upregulating VEGF), which together help resolve inflammation and ischemia in injured tissues[18][19]. This multifaceted action (anti-inflammatory, anti-edema, and pro-oxygenation) underpins the reported clinical improvements in arthritis swelling and pain with HBOT.
PBM: Red and near-infrared light in PBM is absorbed by mitochondrial chromophores (notably cytochrome c oxidase), leading to enhanced electron transport and ATP synthesis, along with a transient burst of reactive oxygen species that acts as a cellular signal[20][21]. This photochemical signaling can temper inflammatory pathways – for instance, PBM has been shown to reduce expression of cyclooxygenase-2 (COX-2) and other NF-κB–regulated mediators in treated tissues[22]. It may also increase anti-inflammatory cytokines like IL-10 while reducing IL-1β/IL-6 in joint cells (observed in some preclinical arthritis models). Additionally, PBM’s effects on nerve fibers and microvasculature likely contribute to pain relief and reduced swelling: light therapy can induce vasodilation and lymphatic flow, and modulate neural pain circuits, yielding local analgesic and anti-oedema benefits[23]. Preclinical evidence supports these mechanisms, though translating them to consistent human outcomes requires optimizing PBM dose parameters for each condition. Notably, both HBOT and PBM converge on lowering inflammatory cytokine activity (e.g. IL-6, TNF-α) and improving tissue oxygenation/energy metabolism, which may explain their observed anti-inflammatory effects in joints.
Current evidence for HBOT and PBM in arthritis is limited and heterogeneous. Many studies are small, non-blinded or lack placebo controls, and treatment protocols vary widely (different pressures and durations for HBOT; different wavelengths, energies, and schedules for PBM). This heterogeneity complicates comparisons and may contribute to mixed results. Blinding is a challenge (sham HBOT and sham light devices), which can introduce bias in subjective outcomes like pain. In PBM, efficacy appears highly parameter-dependent – trials that used doses outside the effective window often found no benefit[7]. The modalities also seem more consistently beneficial in osteoarthritis (degenerative and localized inflammation) than in rheumatoid arthritis (systemic autoimmune inflammation), as evidenced by the lack of clear RA benefits in a recent PBM review[13] and the exploratory nature of RA HBOT data. Overall, the quality of evidence is low-to-moderate, and neither therapy is part of standard clinical guidelines for arthritis. Larger, rigorously sham-controlled trials are needed to confirm efficacy, identify which subgroups benefit most, and refine optimal dosing (e.g. standardizing HBOT pressure protocols and PBM wavelength/fluence). Long-term safety and cost-effectiveness also remain to be established. At present, HBOT and PBM should be considered adjunctive options for management of refractory symptoms or post-operative recovery, rather than replacements for proven therapies. Ongoing research will clarify their place, if any, in the comprehensive care of arthritis.
[1] [6] The Effects of Hyperbaric Oxygen on Rheumatoid Arthritis: A Pilot Study - PubMed
https://pubmed.ncbi.nlm.nih.gov/32947434/
[2] [7] [8] [9] Efficacy of low-level laser therapy on pain and disability in knee osteoarthritis: systematic review and meta-analysis of randomised placebo-controlled trials - PubMed
https://pubmed.ncbi.nlm.nih.gov/31662383/
[3] The effects of hyperbaric oxygen on MRI findings in rheumatoid arthritis: A pilot study - PubMed
https://pubmed.ncbi.nlm.nih.gov/36820805/
[4] [5] [17] [18] [19] Effect of hyperbaric oxygen therapy on postoperative muscle damage and inflammation following total knee arthroplasty: a randomized controlled trial | Scientific Reports
[10] [11] [12] Effects of photobiomodulation and a physical exercise program on the expression of inflammatory and cartilage degradation biomarkers and functional capacity in women with knee osteoarthritis: a randomized blinded study | Advances in Rheumatology | Full Text
https://advancesinrheumatology.biomedcentral.com/articles/10.1186/s42358-021-00220-5
[13] [14] Effects of low-level laser therapy in adults with rheumatoid arthritis: A systematic review and meta-analysis of controlled trials | PLOS One
https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0291345
[15] [16] The Impact of Photobiomodulation Therapy on Swelling Reduction and Recovery Enhancement in Total Knee Arthroplasty: A Randomized Clinical Trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/39786308/
[20] [21] [22] [23] NFkB is activated by PBM induced ROS in embryonic fibroblasts. (A)... | Download Scientific Diagram

Infectious and inflammatory complications (e.g. severe wounds, radiation injuries, therapy-induced mucositis) can involve excessive inflammation and impaired healing. Hyperbaric oxygen therapy (HBOT) and photobiomodulation (PBM, red/near-infrared light) are being explored as adjuncts to standard care to modulate immune responses and support tissue repair[1][2]. These therapies may help reduce pro-inflammatory cytokines and improve oxygenation or cellular energy, potentially aiding infection control and recovery, while not replacing antibiotics or surgery.
HBOT (mechanistic): Breathing 100% O₂ under high pressure raises tissue oxygen tension, which can restore hypoxic zones and enhance leukocyte function[13][14]. Hyperoxia directly inhibits anaerobic bacteria and toxin production and reduces oedema via vasoconstriction (while maintaining tissue oxygenation). HBOT may modulate HIF‑1α and NF‑κB pathways, attenuating pro-inflammatory cytokine release. Repeated sessions stimulate angiogenesis and collagen deposition, improving microcirculation in chronic wounds.
PBM (mechanistic): Red and near-infrared light photons (≈630–850 nm) are absorbed by mitochondrial cytochrome-c oxidase, boosting electron transport and ATP synthesis[15]. This triggers transient reactive oxygen and nitric oxide signaling[16] that can alter gene expression. In inflamed or injured tissues, PBM tends to down-regulate excessive inflammation: it can up-regulate antioxidant defenses and shift macrophages to a reparative phenotype[2]. NF‑κB-driven cytokine production is reduced in activated immune cells[17], leading to lower levels of IL-6, TNF-α and other mediators. In oral mucositis, PBM’s promotion of epithelial regeneration and reduced oxidative stress helps maintain the mucosal barrier during cancer therapy.
(Similar IL-6/TNF-α reductions have been observed with both HBOT and PBM in separate human studies[10][6], suggesting convergent anti-inflammatory pathways.)
Current evidence is heterogeneous and often limited by small sample sizes and adjunct use. HBOT and PBM protocols vary widely (doses, timing), complicating comparisons. Blinding is challenging (sham pressure or light), which may introduce bias. Many positive findings come from preliminary or uncontrolled studies; for example, dramatic cytokine drops in PBM-treated COVID-19 need confirmation in larger trials. Thus far, these therapies are adjunctive – standard treatments (antibiotics, surgery, chemo/radiotherapy) were concurrently used in studies[9][1]. Robust, sham-controlled RCTs are needed to determine definitive clinical benefits (e.g. infection resolution, survival, hospital stay, mucositis severity). Consistent dosing guidelines should be established (optimal ATA×minutes or J/cm²) to reproducibly harness the immune-modulating effects. Overall, HBOT and PBM appear generally safe (minor barotrauma with HBOT; no serious PBM effects reported[18]), but further large-scale studies are required to confirm efficacy and refine patient selection criteria.
[1] [13] [14] 09. Necrotizing Soft Tissue Infections - Undersea & Hyperbaric Medical Society
https://www.uhms.org/9-necrotizing-soft-tissue-infections.html
[2] [15] [16] [17] Mechanisms and applications of the anti-inflammatory effects of photobiomodulation
http://www.aimspress.com/article/10.3934/biophy.2017.3.337?ref=aurelienroy
[3] [19] Hyperbaric oxygen therapy facilitates healing of chronic foot ulcers in patients with diabetes - PubMed
https://pubmed.ncbi.nlm.nih.gov/20427683/
[4] [5] [20] Frontiers | The effectiveness of hyperbaric oxygen therapy for managing radiation-induced proctitis – results of a 10-year retrospective cohort study
https://www.frontiersin.org/journals/oncology/articles/10.3389/fonc.2023.1235237/full
[6] Hyperbaric oxygen therapy in preventing mechanical ventilation in COVID-19 patients: a retrospective case series – SerenaGroup® Research Foundation
[7] [8] Five sessions of hyperbaric oxygen for critically ill patients with COVID-19-induced ARDS: A randomised, open label, phase II trial - Respiratory Medicine
https://www.resmedjournal.com/article/S0954-6111(24)00219-1/fulltext
[9] [10] [18] [21] Photobiomodulation Improves Serum Cytokine Response in Mild to Moderate COVID-19: The First Randomized, Double-Blind, Placebo Controlled, Pilot Study - PubMed
https://pubmed.ncbi.nlm.nih.gov/35874678/
[11] [12] Efficacy of low-level laser for treatment of cancer oral mucositis: a systematic review and meta-analysis - PubMed

Diabetic foot ulcers (DFUs) are chronic wounds with a high risk of non-healing, infection, and lower-limb amputation[1]. Standard care (debridement, off-loading, dressings) achieves only ~50% healing[2], so adjunct therapies targeting tissue perfusion, oxygenation, and inflammation are being explored. Hyperbaric oxygen therapy (HBOT) and photobiomodulation (PBM, red/near-infrared light therapy) aim to enhance wound healing via improved oxygen delivery and cellular bioenergetics while modulating inflammation.
· Meta-analysis (20 RCTs, n=1263): Adjunctive HBOT (typically 2.0–2.5 ATA for ~90 min, 20–40 sessions) nearly doubled DFU healing rates (RR ~1.90) and reduced major amputation risk by ~48%[1]. Mean time-to-healing was ~19 days shorter with HBOT (p<0.001)[1]. Larger, well-controlled trials were recommended[3].
· Ischaemic DFUs: In patients with peripheral arterial disease, HBOT significantly lowered major amputation rates (≈11% vs 26%)[4] (NNT ~7) but did not consistently improve complete ulcer healing[5][6]. Selection of hypoxic DFUs (e.g. low TcPO₂) may identify those most likely to benefit[7].
· RCT (double-blind, 94 patients): HBOT at 2.5 ATA for 85 min, 40 sessions over 8 weeks, improved 1-year ulcer closure (52% vs 29% with sham hyperbaric air, p=0.03)[8][9]. A per-protocol subgroup (>35 sessions) showed 61% healing vs 27% in controls[8][10], with few adverse events reported.
· RCT (double-blind, 103 patients): Another trial (30 sessions HBOT at 2.4 ATA vs sham, added to comprehensive care) found no significant difference at 12 weeks in healing (20% HBOT vs 22% sham) or in amputation criteria[11]. This suggests HBOT’s benefits may manifest over longer follow-up or in specific subgroups.
· Inflammation & perfusion: A small prospective study of refractory DFUs noted HBOT led to reduced erythrocyte sedimentation rate and ulcer area by 3 months (p<0.05), trends toward lower C-reactive protein, and increased angiogenic factors (SDF-1α)[12]. HBOT-treated wounds showed higher microvessel density on biopsy[13]. Over 3 years, the HBOT group had fewer amputations and improved wound healing than controls[14][15] (Martins-Mendes, 2025), though the sample was small and non-randomised.
· Meta-analysis (7 RCTs, n=194): Low-level light therapy (red/NIR PBM) significantly improved DFU outcomes when added to standard care. Pooled results showed greater ulcer size reduction (WMD +34% area reduction vs controls) and higher odds of complete closure (OR 6.72, 95% CI 1.99–22.6, p=0.002)[16][17]. PBM also accelerated granulation tissue formation and shortened time to closure, with no reported treatment-related adverse effects[17].
· RCT (23 ulcers, 685 nm laser): PBM (10 J/cm², 50 mW/cm²) given ~6×/week initially then alternate days led to 66.6% complete healing vs 38.4% in the sham group (Wagner grade I–II ulcers)[18][19]. Treated ulcers healed faster, with greater reductions in wound area and duration to closure (Kaviani et al., 2011).
· RCT (68 patients, 15-day course): Daily PBM (multidiode laser, 60 mW, 2–4 J/cm² per spot) for 2 weeks significantly improved wound closure compared to controls[20][21]. Mean ulcer area reduction was ~1043 mm² in PBM vs 322 mm² with standard care alone[20][21], a ~3-fold greater improvement (Kajagar et al., 2012).
· Short-term outcomes: In a 30-patient trial (660 nm LED, 3 J/cm² daily), the PBM group achieved ~37% wound area reduction in 2 weeks versus ~15% in controls[22][23], alongside visibly increased granulation tissue (Mathur et al., 2017). Another study reported PBM (632.8 nm, 4 J/cm² thrice weekly) not only shrank ulcers over 4 weeks but also significantly relieved chronic wound pain[24][25] (Feitosa et al., 2015).
HBOT (human/preclinical): By exposing patients to 100% oxygen at 2–2.5 ATA, HBOT acutely elevates periwound oxygen tension, which can reverse local hypoxia and support normal healing processes. Increased tissue pO₂ boosts fibroblast function and collagen synthesis and promotes angiogenesis via upregulation of VEGF and related pathways[13][14]. Higher oxygen levels also aid leukocyte bactericidal activity and may dampen hypoxia-driven inflammation (e.g. reducing CRP/ESR)[12]. Repeated HBOT sessions induce prolonged microvascular improvements (capillary density) and enhanced oxidative stress defenses, helping chronic wounds to progress from stalled inflammation to granulation.
PBM (human/preclinical): Red and near-infrared light in PBM is absorbed by mitochondrial chromophores (especially cytochrome c oxidase), leading to improved electron transport and ATP synthesis in cells. This photochemical effect triggers redox signalling that can increase growth factor release and cell proliferation. In wounds, PBM has been shown to stimulate fibroblast and keratinocyte activity, accelerating collagen deposition and re-epithelialisation[13]. PBM may also transiently release nitric oxide from cells, causing vasodilation and improved microcirculation around the ulcer. Preclinical models indicate PBM modulates inflammatory cytokines (e.g. lowering TNF-α/IL-6) and oxidative stress, thereby promoting a more regenerative wound-healing environment. (Notably, PBM’s efficacy follows a biphasic dose response – inadequate or excessive fluence may fail to produce these benefits, underscoring the importance of optimal dose windows.)
Many studies are small, single-centre trials with heterogeneous protocols, which limits firm conclusions. For HBOT, blinding is challenging and standard-care co-interventions (off-loading, revascularisation) vary, making it hard to isolate HBOT’s effect. PBM trials likewise differ in wavelength, dose, and application technique, and few are double-blinded due to visible light. Meta-analyses suggest beneficial trends, but also reveal publication bias and generally moderate-quality evidence. There is a need for large, multi-centre RCTs with sham controls for both HBOT and PBM, using standardised outcome measures (e.g. time to complete closure) and optimised dosing regimens. Better patient selection criteria (e.g. based on ulcer perfusion status for HBOT or wound chronicity for PBM) should be defined. Overall, current evidence supports HBOT and PBM as adjuncts to standard care in hard-to-heal diabetic wounds, but neither is a standalone “cure.” Further high-quality research is required to confirm long-term benefits, cost-effectiveness, and safety in broader diabetic populations.
[1] [3] Efficacy of hyperbaric oxygen therapy for diabetic foot ulcers: An updated systematic review and meta-analysis - PubMed
https://pubmed.ncbi.nlm.nih.gov/34376365/
[2] [18] [19] [20] [21] [22] [23] [24] [25] [26] [27] [28] [29] [30] [31] [34] Clinical Effect of Photobiomodulation on Wound Healing of Diabetic Foot Ulcers: Does Skin Color Needs to Be Considered? - PMC
https://pmc.ncbi.nlm.nih.gov/articles/PMC9789897/
[4] [5] [6] A systematic review and meta-analysis of hyperbaric oxygen therapy for diabetic foot ulcers with arterial insufficiency - PubMed
https://pubmed.ncbi.nlm.nih.gov/32040434/
[7] NCA - Hyperbaric Oxygen Therapy for Hypoxic Wounds and Diabetic Wounds of the Lower Extremities (CAG-00060N) - Decision Memo
[8] [9] [10] Hyperbaric oxygen therapy facilitates healing of chronic foot ulcers in patients with diabetes - PubMed
https://pubmed.ncbi.nlm.nih.gov/20427683/
[11] Hyperbaric Oxygen Therapy Does Not Reduce Indications for Amputation in Patients With Diabetes With Nonhealing Ulcers of the Lower Limb: A Prospective, Double-Blind, Randomized Controlled Clinical Trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/26740639/
[12] [13] [14] [15] Microvascular, Biochemical, and Clinical Impact of Hyperbaric Oxygen Therapy in Recalcitrant Diabetic Foot Ulcers - PubMed
https://pubmed.ncbi.nlm.nih.gov/40801628/
[16] [17] Efficacy of low-level light therapy for treatment of diabetic foot ulcer: A systematic review and meta-analysis of randomized controlled trials - PubMed
https://pubmed.ncbi.nlm.nih.gov/30009935/
[32] [33] A randomized clinical trial on the effect of low-level laser therapy on chronic diabetic foot wound healing: a preliminary report - PubMed

Fibromyalgia (FM) is characterized by chronic widespread pain, fatigue, non-restorative sleep, cognitive “fibro-fog,” and markedly reduced quality of life[1][2]. Oxygen- and light-based therapies are being explored as adjuncts to standard care, aiming to improve cellular bioenergetics, tissue perfusion, and neuroimmune balance in FM. Hyperbaric oxygen therapy (HBOT) delivers high-pressure oxygen to increase tissue oxygenation, while photobiomodulation (PBM, red/near-infrared light therapy) targets mitochondrial enzymes to modulate pain and inflammation.
HBOT:
- Meta-analysis (Chen 2023, 9 studies, n=288): HBOT improved pain in FM vs control interventions (pooled standardised mean difference ≈ –1.56, 95% CI –2.18 to –0.93)[3]. Most studies also noted gains in tender-point counts, fatigue, multidimensional function (e.g. Fibromyalgia Impact Questionnaire, FIQ), patient global impression, and sleep quality[4]. Adverse effects (e.g. ear barotrauma) were reported in ~24% of patients, with no serious events; using lower pressure (<2.0 ATA) appeared to reduce side effects[5].
PBM:
- Meta-analysis (Yeh 2019, 9 RCTs, n=325): Low-level laser therapy (LLLT, a form of PBM) applied to multiple tender points (wavelengths 632–904 nm) was superior to sham for improving composite FIQ scores, pain intensity, tender-point counts, fatigue, stiffness, and mood in FM[16]. Effect sizes were moderate to large (e.g. pain and FIQ SMD ~1.1–1.4)[16]. In one RCT, adding PBM to exercise conferred no extra benefit over exercise alone, but using a combined laser+LED device did improve pain, tender points and fatigue beyond exercise alone[17][18]. Overall, LLLT/PBM was well tolerated with no serious adverse events reported[19][20].
HBOT (human/preclinical): Breathing 100% oxygen at high pressure super-oxygenates the blood and tissues, which may enhance mitochondrial ATP production and tissue repair in oxygen-starved muscle[28]. The transient oxidative burst from hyperoxia can paradoxically induce an anti-inflammatory effect: HBOT has been shown to reduce pro-inflammatory cytokines (e.g. IL-6, TNF-α) and oxidative stress markers while upregulating growth factors for healing[29]. In FM, HBOT’s neuroplastic effects include “resetting” aberrant pain processing in the central nervous system – SPECT scans confirm previously hyperactive pain-related brain regions regain normal perfusion after HBOT alongside symptom relief[6][13]. This suggests HBOT may break the cycle of central sensitisation by improving regional brain blood flow and reducing neuroinflammation.
PBM (human/preclinical): Red and near-infrared light (λ ~600–900 nm) penetrates tissues and is absorbed by mitochondrial chromophores like cytochrome-c oxidase, boosting electron transport, ATP synthesis and modulating cellular redox signaling[30]. By restoring efficient mitochondrial function, PBM can lessen chronic pain pathways: studies show PBM reduces oxidative stress and alters expression of genes linked to nociception and inflammation[30]. At the tissue level, PBM of sufficient dose can raise pressure-pain thresholds and decrease allodynia in FM, indicating a reduction in central sensitisation of pain[31][21]. Transcranial PBM, applying light to scalp regions, has shown enhanced cerebral blood flow and cognitive improvements in brain injury trials[32]. This raises the prospect that PBM could alleviate “fibro-fog” cognitive symptoms by modulating neural activity and connectivity in frontal and limbic networks – though direct evidence in fibromyalgia patients is still preliminary.
Current evidence is limited by small sample sizes and heterogeneity in treatment protocols and outcomes. Many FM studies are unblinded or use suboptimal shams (e.g. inactive light that may still produce warmth, or no chamber pressurisation), which can bias subjective endpoints. Follow-up durations are relatively short, and the long-term durability of HBOT/PBM benefits needs confirmation beyond 6 months. Additionally, patient selection varies (e.g. FM with specific triggers like trauma), making it hard to generalise results. Standardised outcome measures (e.g. updated ACR criteria, FIQR, pain thresholds) and rigorous sham-controlled trial designs are required to validate these modalities. Importantly, HBOT and PBM should be viewed as adjunctive therapies – while studies suggest meaningful improvements in pain and function, neither is a proven standalone “cure” for fibromyalgia. Larger, multi-center trials integrating mechanistic biomarkers (imaging, autonomic measures, inflammatory markers) will help establish the true efficacy and optimal dosing parameters for HBOT and PBM in fibromyalgia management.
[1] [2] [6] [7] [8] Hyperbaric Oxygen Therapy Can Diminish Fibromyalgia Syndrome – Prospective Clinical Trial | PLOS One
https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0127012
[3] [4] [5] Efficacy and safety of hyperbaric oxygen therapy for fibromyalgia: a systematic review and meta-analysis - PubMed
https://pubmed.ncbi.nlm.nih.gov/36690401/
[9] [10] [11] [12] [13] [14] Hyperbaric oxygen therapy compared to pharmacological intervention in fibromyalgia patients following traumatic brain injury: A randomized, controlled trial | PLOS One
https://journals.plos.org/plosone/article?id=10.1371/journal.pone.0282406
[15] [26] [27] Frontiers | Outcomes of whole-body photobiomodulation on pain, quality of life, leisure physical activity, pain catastrophizing, kinesiophobia, and self-efficacy: a prospective randomized triple-blinded clinical trial with 6 months of follow-up
https://www.frontiersin.org/journals/neuroscience/articles/10.3389/fnins.2024.1264821/full
[16] Low-Level Laser Therapy for Fibromyalgia: A Systematic Review and Meta-Analysis - PubMed
https://pubmed.ncbi.nlm.nih.gov/31151332/
[17] [18] [19] [20] painphysicianjournal.com
[21] [24] [25] [30] [31] [32] Effectiveness of Photobiomodulation Therapy in the Management of Fibromyalgia Syndrome: A Systematic Review
https://www.mdpi.com/2076-3417/15/8/4161
[22] [23] Photobiomodulation therapy combined with static magnetic field is better than placebo in patients with fibromyalgia: a randomized placebo-controlled trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/37847246/
[28] Frontiers | Hyperbaric Oxygen Therapy Can Induce Neuroplasticity and Significant Clinical Improvement in Patients Suffering From Fibromyalgia With a History of Childhood Sexual Abuse—Randomized Controlled Trial
https://www.frontiersin.org/journals/psychology/articles/10.3389/fpsyg.2018.02495/full
[29] The Effects of Hyperbaric Oxygenation on Oxidative Stress, Inflammation and Angiogenesis

MS is a chronic neuroinflammatory disease causing disability, with common symptoms including fatigue, mobility limitations, and cognitive impairment. Hyperbaric oxygen therapy (HBOT) and photobiomodulation (PBM, red to near-infrared light therapy) are being explored as adjunct interventions aiming to enhance tissue oxygenation, cellular energy (mitochondrial ATP production), and neuroimmune modulation[1][2]. While neither approach is proven to alter MS disease progression, they have been investigated for potential short-term relief of MS-related symptoms and functional improvement in combination with standard care.
HBOT
- Systematic reviews/meta-analyses: A meta-analysis of 12 randomized trials (mostly relapsing MS, 1980s) using ~20 HBOT sessions at 1.75–2.5 ATA for 60–120 min/day found no consistent benefit in disability outcomes (no significant EDSS improvement)[3]. No plausible disease-modifying effect was identified; any subgroup benefit is considered unlikely to be clinically significant[4]. Routine HBOT for MS is not recommended based on available evidence (Bennett & Heard, 2010)[5].
- Clinical course (long-term): An uncontrolled multicenter series (n = 312 MS patients) receiving an initial 20 daily HBOT sessions (~2.0 ATA, 1 hour) plus monthly boosters for 2 years showed no slowing of disability progression: mean EDSS worsened by ~0.9 point over 2 years[6][7]. Approximately 20% reported transient symptom relief (e.g. improved bladder function) during HBOT, but only ~5% maintained any improvement at 2-year follow-up[8][9].
- Guideline stance: National guidelines advise against HBOT as a therapy for MS. For example, NICE (2022) explicitly recommends not offering HBOT for MS-related fatigue due to insufficient evidence of benefit[10]. No significant reductions in relapse rate or MRI lesion activity have been demonstrated in trials to support HBOT as a disease-modifying therapy in MS[11].
- Safety/tolerability: When administered according to standard protocols, HBOT is generally well tolerated. The most common issue is middle ear/sinus barotrauma from pressure changes[12]. Serious adverse events are rare (e.g. oxygen toxicity seizures are infrequent at MS treatment pressures).
PBM (Red Light / Low-Level Laser Therapy)
- Fatigue and cognition: A small controlled trial in relapsing–remitting MS (RRMS, n = 32) examined pulsed low-level laser therapy (~810 nm GaAlAs laser) applied to the cervical spine 3×/week for 4 weeks[13][14]. The PBM-treated group showed significant improvements in fatigue (Fatigue Severity Scale scores) and cognitive performance (reaction time accuracy) compared to baseline, alongside modest EDSS disability score improvements[14][15]. (Adding adjunct UV-B light in a comparison group provided no additional benefit, suggesting the LLLT itself drove the effect[16][17].)
- Functional status: A randomized trial of 120 MS patients (mixed types) receiving 21 days of rehab including low-level laser therapy to spinal nerve roots (with or without pulsed magnetotherapy) reported improved neurological function. All treatment arms showed statistically significant gains in EDSS (lower disability) and Barthel Index (activities of daily living) after the 3-week course[18][19]. The greatest improvement occurred in the combined laser+magnet stimulation group[19]. Notably, functional benefits persisted at least 1 month post-therapy, indicating a short-term carryover effect (Kubsik et al., 2016).
- Mixed results on fatigue: Not all PBM studies have been positive. A sham-controlled pilot RCT in RRMS (Tamiris Silva et al., 2022) delivered 808 nm laser PBM (36 J per 6 min session) either sublingually or over the radial artery 3×/week for 4 weeks. It found no significant difference in fatigue outcomes (Modified Fatigue Impact Scale scores) between active PBM and placebo-treatment groups[20][21], underscoring the need to identify optimal dosing and targets for fatigue relief.
- Muscle performance: Transcranial PBM aimed at peripheral neuromuscular function has shown promise. In a double-blind crossover trial (n = 17, mild-moderate MS), near-infrared PBM (wavelength 600–1100 nm, up to 120 J) was applied to a leg muscle (tibialis anterior). After a personalized 2-week PBM course, patients had significantly greater muscle strength gains versus sham (mean +22.9 N vs –4.1 N, p = 0.02)[22][23]. However, there was no significant change in measured muscle fatigue or endurance, nor any subjective fatigue improvement in this short-term study (Rouhani et al., 2024)[22][24].
- Immune and safety profile: Preliminary biomarker evidence suggests PBM may exert anti-inflammatory effects. In a 14-person RRMS trial, 12 weeks of PBM (808 nm, 100 mW, 6 min to sublingual or radial artery sites, 24 sessions) led to a ~3–4 fold increase in serum IL-10 (an anti-inflammatory cytokine) from baseline[25][26], whereas pro-oxidative nitrite levels were unchanged. No adverse effects were reported. In fact, across published PBM studies in MS, no serious treatment-related adverse events have been noted[27]. PBM parameters and delivery sites have varied widely (wavelengths ~630–1064 nm; dosimetry from ~36 J up to 120 J per site; applied to scalp, neck, or peripheral blood targets), and all studies to date have been small.
HBOT: By delivering 100% oxygen at elevated pressure, HBOT can markedly increase oxygen saturation in blood and tissues. The rationale in MS is that this might counteract lesion hypoxia and support energy metabolism in chronically inflamed CNS areas. Preclinical evidence (EAE models) shows that correcting CNS hypoxia can rapidly improve neurological deficits and reduce demyelination despite ongoing inflammation[28][29]. HBOT may also transiently reduce edema and modulate the immune response (e.g. reducing certain inflammatory mediators), thereby lowering oxidative stress in lesions. Improved microvascular perfusion and oxygen delivery to impaired nerves could underlie reported short-term relief in some MS symptoms. However, these effects appear temporary, and there is no confirmed impact on the autoimmune attack on myelin.
PBM: Red/NIR light in the 600–1100 nm range can penetrate tissue and is absorbed by mitochondrial chromophores (notably cytochrome c oxidase). This absorption is thought to enhance mitochondrial respiration and ATP synthesis, while also triggering secondary cellular signaling that promotes neuroprotection. In MS and related neuroinflammatory conditions, PBM has been observed to shift cytokine profiles towards anti-inflammatory states (increasing IL-10 and IL-4, with relative suppression of Th1/Th17 pro-inflammatory cytokines in animal models)[30][31]. PBM may additionally improve neuronal function via increased regional cerebral blood flow and altered cortical activity. For example, transcranial PBM at 1064 nm has been associated with improved cognitive performance and normalized EEG rhythms in human studies[2][32]. These mechanisms – boosting cellular energy availability and dampening inflammatory oxidative damage – align with the observed preliminary benefits in MS fatigue, cognition, and muscle function. Importantly, PBM’s effects are dose-dependent and biphasic (insufficient or excessive dose may fail to produce benefit), which highlights the need for optimized treatment parameters in MS.
Overall, the evidence for HBOT or PBM in MS is limited and heterogeneous. The HBOT trials are mostly older, with variable quality and often negative primary outcomes[33]. No high-level evidence shows that HBOT can alter the course of MS (relapses or MRI lesions), and expert consensus is that it should not be used as a disease-modifying therapy[5]. PBM studies in MS, while more recent, have involved small sample sizes and sometimes lack proper blinding or sham controls. There is significant variability in PBM protocols (wavelength, dosage, treatment location), making it difficult to compare results across studies[34]. Both modalities require more rigorous research: well-powered, placebo-controlled trials in defined MS populations (e.g. patients with refractory fatigue or gait impairment) using standardized outcome measures (fatigue scales, cognitive tests, EDSS, etc.). Long-term safety and durability of any benefits also remain to be established – especially for PBM, where optimal dosing and scheduling are still being determined[35]. In summary, current evidence does not support HBOT or PBM as standalone disease-modifying treatments in MS, but they may have a role as adjunctive, symptom-focused therapies. Well‑designed future studies are needed to confirm the preliminary functional improvements and to identify which MS patients, if any, are most likely to benefit from these energy-based interventions.
· Bennett MH, Heard R. Hyperbaric oxygen therapy for multiple sclerosis. CNS Neurosci Ther. 2010;16(2):115-124. DOI: 10.1111/j.1755-5949.2009.00129.x[3][4].
· Kindwall EP et al. Treatment of multiple sclerosis with hyperbaric oxygen: results of a national registry. Arch Neurol. 1991;48(2):195–199. DOI: 10.1001/archneur.1991.00530140091021[6][8].
· National Institute for Health and Care Excellence (NICE). Multiple sclerosis in adults: management. NICE Guideline NG220, Oct 2022[10].
· Kubsik A et al. Application of laser radiation and magnetostimulation in therapy of patients with multiple sclerosis. NeuroRehabilitation. 2016;38(2):183-190. DOI: 10.3233/NRE-161309[18][19].
· Rouhani M et al. Effects of photobiomodulation therapy on muscle function in individuals with multiple sclerosis. Mult Scler Relat Disord. 2024;86:105598. DOI: 10.1016/j.msard.2024.105598[22][23].
· Silva T et al. Effect of photobiomodulation on fatigue in individuals with relapsing–remitting multiple sclerosis: a pilot study. Lasers Med Sci. 2022;37(8):3107-3113. DOI: 10.1007/s10103-022-03567-3[20][21].
· Silva T et al. Effects of photobiomodulation on interleukin-10 and nitrites in individuals with relapsing-remitting MS – a randomized clinical trial. PLoS One. 2020;15(4):e0230551. DOI: 10.1371/journal.pone.0230551[25][26].
· Andrade Filho VO et al. Systematic review of photobiomodulation for multiple sclerosis. Front Neurol. 2024;15:1465621. DOI: 10.3389/fneur.2024.1465621[36][34].
[1] [28] [29] Oxygen treatment reduces neurological deficits and demyelination in two animal models of multiple sclerosis - PMC
https://pmc.ncbi.nlm.nih.gov/articles/PMC10107096/
[2] [27] [30] [31] [32] [34] [35] [36] Frontiers | Systematic review of photobiomodulation for multiple sclerosis
https://www.frontiersin.org/journals/neurology/articles/10.3389/fneur.2024.1465621/full
[3] [4] [5] Hyperbaric oxygen therapy for multiple sclerosis - PubMed
https://pubmed.ncbi.nlm.nih.gov/20415839/
[6] [7] [8] [9] Treatment of multiple sclerosis with hyperbaric oxygen. Results of a national registry - PubMed
https://pubmed.ncbi.nlm.nih.gov/1993011/
[10] Recommendations | Multiple sclerosis in adults: management | Guidance | NICE
https://www.nice.org.uk/guidance/ng220/chapter/Recommendations
[11] [12] Multiple Sclerosis and Complementary Health Approaches: What the Science Says | NCCIH
[13] [14] [15] [16] [17] kamj.journals.ekb.eg
https://kamj.journals.ekb.eg/article_359634_0d61317c59e4e1e26e4738584f22ebc6.pdf
[18] [19] Application of laser radiation and magnetostimulation in therapy of patients with multiple sclerosis - PubMed
https://pubmed.ncbi.nlm.nih.gov/26923357/
[20] [21] Effect of photobiomodulation on fatigue in individuals with relapsing-remitting multiple sclerosis: a pilot study - PubMed
https://pubmed.ncbi.nlm.nih.gov/35499744/
[22] [23] [24] Effects of photobiomodulation therapy on muscle function in individuals with multiple sclerosis - PubMed
https://pubmed.ncbi.nlm.nih.gov/38614054/
[25] [26] Effects of photobiomodulation on interleukin-10 and nitrites in individuals with relapsing-remitting multiple sclerosis - Randomized clinical trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/32255785/
[33] Microsoft Word - Treatment_of_Multiple_Sclerosis.doc
https://www.uhms.org/images/Position-Statements/treatment_of_multiple_sclero.pdf

Acute surgical wounds and chronic ulcers (diabetic, venous, pressure) pose a major clinical burden, often complicated by hypoxia and impaired healing[1]. Adjunctive therapies like hyperbaric oxygen (HBOT) and photobiomodulation (red light, PBM) aim to enhance tissue oxygenation and cellular repair processes, respectively, to support standard wound care.
HBOT (Hyperbaric Oxygen Therapy) – Administering 100% O₂ at >1 atmosphere absolute (ATA) pressure:
PBM (Photobiomodulation – Low-Level Red/Infrared Light Therapy):
HBOT: By elevating plasma O₂ levels, HBOT raises tissue oxygen tension (TcPO₂) in hypoxic wound areas, which may jump-start oxygen-dependent healing processes. Increased O₂ availability supports collagen synthesis and cross-linking and promotes angiogenesis (e.g. up-regulating VEGF and fibroblast growth factors) to spur new capillary growth. Hyperoxia also directly enhances leukocyte oxidative killing of bacteria and inhibits toxin-producing anaerobes, helping to control infection[14]. Preclinical studies and human wound fluid analyses show HBOT can stimulate fibroblast proliferation and neovascularisation while reducing tissue edema[15][14]. By improving microcirculation and oxygen delivery, HBOT may dampen chronic inflammation driven by tissue hypoxia (e.g. down-regulating hypoxia-inducible cytokines).
PBM: Red and near-infrared light can penetrate skin and is absorbed by mitochondrial chromophores (notably cytochrome c oxidase). This absorption boosts cellular respiration and ATP production, and induces mild oxidative signaling that triggers growth factor release[16]. In wounds, PBM has been shown to activate key repair cells: increasing fibroblast proliferation and migration, accelerating keratinocyte re-epithelialisation, and enhancing deposition of collagen I/III in the extracellular matrix[17]. PBM also modulates the inflammatory response – studies report reductions in IL-6 and TNF-α levels in laser-treated wounds (preclinical) alongside improved local blood flow, partly due to nitric oxide release causing vasodilation in the peri-wound microvasculature. Collectively, these photobiological effects (sometimes termed “laser bio-stimulation”) can create a more favorable environment for wound closure, especially in chronic ulcers with stalled healing. Similar perfusion and anti-inflammatory trends have been noted in diabetic wound models receiving PBM, complementing the oxygenation gains seen with HBOT.
Current evidence is limited by heterogeneity and study size. Many trials are small single-centre studies with varying HBOT pressures (2.0–2.5 ATA protocols) or diverse PBM parameters (wavelengths 600–900 nm, dose 1–10 J/cm²), making comparisons difficult. Co-interventions like off-loading (for DFUs) or compression (for venous ulcers) were not uniform, and true sham controls are challenging (e.g. visible light in PBM). Meta-analyses flag a very low certainty of evidence for PBM outcomes[18] and risk of bias in HBOT studies[19]. Blinding issues and publication bias further temper confidence. Both modalities are generally well-tolerated (HBOT’s main risks are barotrauma or mild oxygen toxicity, while PBM has no significant adverse effects reported[20]), but robust data on long-term safety are lacking. Overall, larger high-quality RCTs are needed – ideally multi-centre, sham-controlled trials with standardized dosing and clinically important endpoints (complete healing, time to closure, amputation rates). Future research should also clarify optimal patient selection (e.g. wounds with critical ischemia might benefit more from HBOT, whereas superficial non-ischaemic wounds might respond well to PBM). Until then, HBOT and red light therapy should be viewed as adjunctive therapies to standard wound management rather than standalone cures, with further confirmation required to define their precise roles in improving wound healing outcomes.
[1] [2] [19] Hyperbaric oxygen therapy for chronic wounds - PubMed
https://pubmed.ncbi.nlm.nih.gov/26106870/
[3] Hyperbaric oxygen therapy facilitates healing of chronic foot ulcers in patients with diabetes - PubMed
https://pubmed.ncbi.nlm.nih.gov/20427683/
[4] Hyperbaric Oxygen Therapy Does Not Reduce Indications for Amputation in Patients With Diabetes With Nonhealing Ulcers of the Lower Limb: A Prospective, Double-Blind, Randomized Controlled Clinical Trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/26740639/
[5] [6] [7] Hyperbaric oxygen therapy for late radiation tissue injury - PubMed
https://pubmed.ncbi.nlm.nih.gov/27123955/
[8] [10] [20] Efficacy of low-level light therapy for treatment of diabetic foot ulcer: A systematic review and meta-analysis of randomized controlled trials - PubMed
https://pubmed.ncbi.nlm.nih.gov/30009935/
[9] [18] The effect of low-level laser therapy on diabetic foot ulcers: A meta-analysis of randomised controlled trials - PubMed
https://pubmed.ncbi.nlm.nih.gov/33751853/
[11] [12] [16] [17] cureus.com
[13] A Systematic Review and Meta-Analysis of the Effects of Low-Level Laser Therapy in the Treatment of Diabetic Foot Ulcers - PubMed
https://pubmed.ncbi.nlm.nih.gov/32394760/
[14] [15] Hyperbaric Therapy for Skin Grafts and Flaps - StatPearls - NCBI Bookshelf

Chronic pain is a pervasive cause of disability, impairing daily function and quality of life while often necessitating long-term analgesics. Hyperbaric oxygen therapy (HBOT) and red light photobiomodulation (PBM) are being explored as adjunctive treatments to modulate inflammation, tissue metabolism, and nociceptive processing. Both approaches aim to improve pain and function by altering the cellular and physiological environment (e.g. oxygenation or mitochondrial activity) in ways conventional therapies do not.
· Complex Regional Pain Syndrome (CRPS) – A double-blind RCT (n=71) using HBOT (≈2.0 ATA, 60 min, 15 sessions) showed greater reductions in limb pain and edema vs sham (normal air)[1]. A larger case series (n=83, CRPS I/II) with ~22 sessions at 2.0–2.4 ATA for 80–90 min (with air break) found rest-pain VAS halved (mean 3.2→1.6) and activity-pain improved (6.1→3.7, p<0.001)[2], alongside functional gains and 86% symptom relief; no serious HBOT-related adverse events were reported[3][4]. (Kiralp 2004; Hájek 2024)
· Fibromyalgia (widespread pain) – A meta-analysis of 5 RCTs (n≈180) reported HBOT (typically 2.0 ATA, 60–90 min for 15–40 sessions) improved multiple outcomes. Tender point counts dropped by ~6 points and pain-pressure thresholds rose (SMD +0.57)[5], with modest pain score reductions (VAS difference ~5 points)[5] and better sleep and quality-of-life. These findings suggest potential benefit but are based on preliminary trials (Kulshreshtha 2024).
· Post-concussive Headache – Limited evidence suggests HBOT may relieve some headache syndromes. A Cochrane review pooled 3 small trials of acute migraine and found HBOT increased the chance of pain relief vs sham (relative risk ~1.5)[6], though evidence quality was low and no benefit was seen for preventing attacks. HBOT for chronic headaches remains experimental (Bennett 2015).
· Neck and Back Pain – A systematic review of 16 trials (820 patients) found that low-level laser PBM (red/infrared) can significantly reduce chronic neck pain. Pooled results showed an ≈20 mm greater VAS pain improvement (0–100 mm scale) vs placebo[7], and pain relief persisted up to 22 weeks post-therapy[8]. Only mild, transient side effects (e.g. skin warmth) were noted (Chow 2009). Evidence in low back pain is mixed; some trials report benefit while others using suboptimal dosages found no significant pain or disability change[9].
· Knee Osteoarthritis – PBM has shown analgesic effects in degenerative joint pain. A network meta-analysis of 13 RCTs (n=673) reported that PBM significantly reduced knee OA pain compared to sham (pooled SMD ~0.96)[10], although improvements in WOMAC function or stiffness were not consistently significant[11]. Notably, only specific light parameters were effective – wavelengths around 904 nm or 785–850 nm yielded the greatest pain reduction[12] – underscoring dose-dependence (Fan 2024).
· Neuropathic Pain – Preliminary studies indicate PBM may alleviate peripheral neuropathic pain. In diabetic peripheral neuropathy, trials have observed reduced neuropathic pain scores and even improved nerve conduction velocities with PBM[13]. A small pilot in post-herpetic neuralgia (laser 650 nm, 16 sessions) reported striking pain relief: 11 of 15 patients had complete resolution of pain (VAS 0) and the remainder improved to mild pain[14]. While promising, these outcomes in neuropathy need confirmation in larger controlled studies (Korada 2023; Mukhtar 2020).
· Comparative Note: Both HBOT and PBM have been associated with pain reductions in select chronic pain populations, but their roles appear adjunctive. PBM outcomes are highly contingent on correct dose parameters (appropriate wavelength, irradiance, and sufficient treatment course)[15]. HBOT, meanwhile, shows potential benefit in certain refractory pain syndromes (e.g. CRPS, fibromyalgia) but evidence comes from small or exploratory trials. Neither modality is a stand-alone “cure,” and results vary by condition.
HBOT: By raising oxygen availability in blood and tissues, HBOT may counteract hypoxia-driven inflammation and support tissue healing. Increased plasma O₂ tension improves microcirculation and can reduce edema and ischemia in injured tissues[16]. Both animal and human studies suggest HBOT attenuates pain partly via anti-inflammatory effects – for example, down-regulating overactive glial cells and inflammatory cytokines involved in chronic pain sensitisation[17] (preclinical). The net result may be a reset of aberrant nociceptive signaling pathways contributing to chronic pain.
PBM: Red and near-infrared light can penetrate tissues and is absorbed by mitochondrial chromophores (especially cytochrome-c-oxidase)[18]. This photochemical activation enhances cellular respiration (ATP production) and triggers redox signalling that modulates inflammation and promotes tissue repair. PBM has been shown to reduce the release of pro-inflammatory mediators and can increase local blood flow, which together may lower peripheral nerve sensitivity and muscle spasm (preclinical). In central pain, transcranial PBM (tPBM) is being studied for its neuromodulatory effects – early studies indicate it might influence cortical activity and cerebral blood flow, potentially reducing central sensitisation and headache intensity. Similar analgesic trends are reported in PBM trials for arthritis pain (see Arthritis card).
Current evidence is limited by heterogeneity and small sample sizes. Studies vary widely in HBOT/PBM protocols (pressure or wavelength/dosage, session length, total sessions) and outcome measures, making comparisons difficult. Blinding remains a challenge: patients can sometimes sense pressure changes (HBOT) or warmth/tingling (PBM), which may bias placebo-controlled trials. Many positive findings come from exploratory or unblinded studies, so true efficacy is uncertain. The durability of pain relief also varies – some benefits waned after treatment, underscoring the need to assess long-term outcomes. PBM in particular shows a narrow therapeutic window: inappropriate dose (too low or high power, or insufficient sessions) can yield null results[9]. Meanwhile, high-quality HBOT trials for chronic pain are few – evidence is promising (e.g. large pain drops in fibromyalgia and CRPS) but of low certainty[19]. Both interventions are best considered adjuncts to standard care. More robust, sham-controlled trials are needed, with standardized pain and function endpoints and tracking of medication usage, to validate these modalities’ roles in chronic pain management.
[1] [16] [20] Effectiveness of hyperbaric oxygen therapy in the treatment of complex regional pain syndrome - PubMed
https://pubmed.ncbi.nlm.nih.gov/15174218/
[2] [3] [4] [17] [19] The Effectiveness of Hyperbaric Oxygen Treatment in Patients with Complex Regional Pain Syndrome: A Retrospective Case series
https://www.medsci.org/v21p2021.htm
[5] Journal of Medical Evidence
https://journals.lww.com/jome/fulltext/2024/05010/assessment_of_the_efficacy_and_safety_of.8.aspx
[6] Normobaric and hyperbaric oxygen therapy for the treatment and ...
https://www.cochranelibrary.com/cdsr/doi/10.1002/14651858.CD005219.pub3/full
[7] [8] [21] Efficacy of low-level laser therapy in the management of neck pain: a systematic review and meta-analysis of randomised placebo or active-treatment controlled trials - PubMed
https://pubmed.ncbi.nlm.nih.gov/19913903/
[9] Photobiomodulation therapy does not decrease pain and disability ...
https://pubmed.ncbi.nlm.nih.gov/32680739/
[10] [11] [12] [15] A systematic review and network meta-analysis on the optimal wavelength of low-level light therapy (LLLT) in treating knee osteoarthritis symptoms | Aging Clinical and Experimental Research
https://link.springer.com/article/10.1007/s40520-024-02853-0
[13] Effectiveness of Photobiomodulation Therapy on Neuropathic Pain, Nerve Conduction and Plantar Pressure Distribution in Diabetic Peripheral Neuropathy - A Systematic Review - PubMed
https://pubmed.ncbi.nlm.nih.gov/37622461/
[14] Role of low-level laser therapy in post-herpetic neuralgia: a pilot study - PubMed
https://pubmed.ncbi.nlm.nih.gov/32006261/
[18] Photobiomodulation presents an anti-inflammatory effect, reflecting ...
https://www.sciencedirect.com/science/article/abs/pii/S1011134425001083

Post-viral fatigue syndromes (such as ME/CFS and Long COVID) are characterised by prolonged exhaustion, “brain fog” (cognitive impairment), autonomic dysfunction, and diminished quality of life[1][2]. Hyperbaric oxygen therapy (HBOT) and photobiomodulation (PBM, red to near-infrared light) are being explored as adjunctive interventions to support cellular bioenergetics and modulate inflammation. HBOT increases tissue oxygen availability and may trigger regenerative anti-inflammatory pathways[2], while PBM is thought to stimulate mitochondrial function and microcirculation, potentially alleviating fatigue and cognitive symptoms[3].
Current evidence is preliminary and heterogeneous. Most studies are small and vary in HBOT/PBM dosages, timing, and endpoints, making comparisons difficult. Blinding and placebo effects are challenges – e.g. a sham HBOT group showed notable improvements alongside treatment[6], and an uncontrolled registry saw some patients worsen after HBOT[8]. Safety profiles so far appear acceptable (no serious adverse events reported for either therapy[7][16]), but data on long-term outcomes are limited (though one study noted sustained benefits 1 year post-HBOT[23][24]). Both HBOT and PBM are being investigated as adjuncts rather than standalone cures. Larger, well-powered sham-controlled trials in well-defined Long COVID populations are needed to confirm efficacy on fatigue, cognitive (“brain fog”), exercise tolerance and to determine optimal treatment parameters and durability of responses.
[1] [2] [8] [9] Hyperbaric oxygen therapy for long COVID: a prospective registry | Scientific Reports
[3] [11] Frontiers | Photobiomodulation Improves Serum Cytokine Response in Mild to Moderate COVID-19: The First Randomized, Double-Blind, Placebo Controlled, Pilot Study
https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2022.929837/full
[4] [5] Hyperbaric oxygen therapy improves neurocognitive functions and symptoms of post-COVID condition: randomized controlled trial | Scientific Reports
[6] [7] Ten sessions of hyperbaric oxygen versus sham treatment in patients with long covid (HOT-LoCO): a randomised, placebo-controlled, double-blind, phase II trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/40228859/
[10] [25] Systematic review with meta-analysis: effectiveness of hyperbaric oxygenation therapy for ulcerative colitis - PubMed
https://pubmed.ncbi.nlm.nih.gov/34349835/
[12] [13] Use of either transcranial or whole-body photobiomodulation treatments improves COVID-19 brain fog - PubMed
https://pubmed.ncbi.nlm.nih.gov/37018063/
[14] [15] Can transcranial photobiomodulation improve cognitive function? A systematic review of human studies - PubMed
https://pubmed.ncbi.nlm.nih.gov/36371017/
[16] [22] Transcranial Photobiomodulation for the Treatment of Major Depressive Disorder. The ELATED-2 Pilot Trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/30346890/
[17] [18] Stem cell mobilization by hyperbaric oxygen - PubMed
https://pubmed.ncbi.nlm.nih.gov/16299259/
[19] [20] Hyperbaric oxygen therapy increases telomere length and decreases immunosenescence in isolated blood cells: a prospective trial - PubMed
https://pubmed.ncbi.nlm.nih.gov/33206062/
[21] What Lies at the Heart of Photobiomodulation: Light, Cytochrome C Oxidase, and Nitric Oxide-Review of the Evidence - PubMed
https://pubmed.ncbi.nlm.nih.gov/32716711/
[23] [24] Long term outcomes of hyperbaric oxygen therapy in post covid condition: longitudinal follow-up of a randomized controlled trial | Scientific Reports

Cancer treatments such as chemotherapy, radiotherapy, and surgery can cause severe side effects (e.g. oral mucositis, chronic radiation injuries, wound-healing problems). Hyperbaric oxygen therapy (HBOT) and photobiomodulation (PBM, low-level red/infrared light) are being explored as supportive care interventions to improve healing and symptom relief, helping patients tolerate and complete their cancer therapy[1]. These modalities are not anticancer treatments but aim to reduce therapy-related complications and improve quality of life.
HBOT:
- Late radiation injuries: A 2023 Cochrane review of 18 RCTs (n=1071) found that HBOT (typical dose ~100% O₂ at 2.0–2.5 ATA for ~90 min, 30–40 sessions) improved resolution of late radiation tissue injuries in head-neck, bladder, and rectal tissues (complete healing/improvement in 39% more patients vs controls, RR ~1.4)[2][3]. HBOT also markedly reduced wound breakdown in irradiated head-neck surgical sites (RR 0.24, 95% CI 0.06–0.94)[4].
- Radiation proctitis: In a double-blind RCT for chronic refractory radiation proctitis (n=120), HBOT at 2.0 ATA (90 min for 30 sessions) led to 89% of patients achieving clinical improvement vs 63% with sham (p=0.0009), yielding a 32% absolute risk reduction (NNT ~3) in moderate-to-severe symptoms[5][6]. Treated patients had discontinuation of other interventions and improved bowel-specific quality of life.
- Osteoradionecrosis (ORN): Cohort studies suggest adjunctive HBOT can promote bone and mucosal healing in ORN of the jaw (e.g. improved mucosal coverage and pain)[7][8]. However, evidence is mixed: a multicenter trial reported no added benefit of HBOT in early-stage (Grade I) ORN when compared to standard care[9]. HBOT is often used prophylactically (e.g. 20–30 dives at 2.4 ATA pre- and 10 dives post-dental extraction) in irradiated patients to reduce ORN risk[10].
- Wound healing in irradiated tissue: HBOT has been associated with improved healing of surgical flaps and chronic wounds in previously irradiated areas. For example, pooled RCT data show significantly lower postoperative wound dehiscence in irradiated head-neck cancer patients receiving HBOT (roughly 76% relative risk reduction)[4]. Clinically, HBOT is usually combined with standard wound care (and surgery if needed) in these settings.
PBM (Red Light Therapy):
- Oral mucositis prevention: Evidence-based guidelines (MASCC/ISOO 2019) recommend PBM therapy to prevent severe oral mucositis (OM) in patients undergoing head & neck radiotherapy (with or without chemotherapy) and in stem cell transplant conditioning[1]. Effective PBM protocols use red or near-infrared light (commonly 630–660 nm) delivered intraorally at specific dosimetry (e.g. ~6 J/cm² per point, with power ~20–100 mW/cm²) prior to and during cancer treatment[11]. Trials adhering to these parameters showed significant reductions in OM incidence and severity.
- Clinical outcomes in mucositis: Multiple RCTs and meta-analyses demonstrate that PBM can markedly reduce the severity of therapy-induced mucositis and associated pain. A 2023 systematic review (10 trials, n=759 head-neck cancer patients) reported that prophylactic PBM (lasers/LEDs 632–850 nm) halved the occurrence of grade 3–4 OM and shortened the duration of mucositis, with treated patients experiencing lower peak pain scores and better oral functional quality of life[12]. PBM has also been linked to reduced need for opioid analgesics, fewer feeding tube placements, and fewer unplanned treatment breaks in comparison to controls in these studies[13][12].
- Other side-effect applications: Emerging studies suggest PBM may benefit other cancer therapy side effects. For example, small trials in breast cancer found PBM (e.g. red light-emitting diodes, ~3–6 J/cm²) can lessen acute radiodermatitis severity[14][15]. Pilot studies have also explored PBM for lymphedema and neuropathy, but data remain limited.
- Safety: PBM as used in supportive care has not shown any promotion of tumor growth or recurrence to date[16]. No significant adverse events have been reported in human trials for mucositis; PBM is non-invasive and painless. Professional guidelines emphasize using recommended dose parameters to ensure safety and efficacy[16].
Comparison: HBOT is primarily applied for chronic, late-arising radiation injuries (often months to years after therapy), whereas PBM is typically used during active treatment to mitigate acute toxicities. In practice, the two adjuncts address different phases of supportive care – HBOT for repairing established damage in hypoxic irradiated tissues, and PBM for preventing or reducing immediate mucosal and skin reactions[17][1]. Both modalities aim to improve patients’ tolerance of cancer treatments and maintain treatment continuity by reducing side-effect burden.
HBOT: Breathing 100% oxygen under high pressure increases tissue oxygenation and has been shown (in both preclinical models and patient observations) to stimulate new blood vessel growth and collagen formation in damaged tissue[18][19]. This enhanced perfusion can counteract radiation-induced hypoxia and fibrosis, supporting wound healing in irradiated areas. HBOT may also down-regulate chronic inflammation mediated by hypoxia-inducible factors in late radiation injury.
PBM: Low-level red/infrared light is absorbed by mitochondrial cytochrome-c oxidase in cells, boosting ATP production and modulating reactive oxygen species and NF-κB signaling[20]. Preclinical studies indicate that PBM can reduce the release of pro-inflammatory cytokines (e.g. TNF-α, IL-1, IL-6) and promote epithelial regeneration and angiogenesis in mucosal tissues[21][20]. Clinically, this mechanism translates into faster healing of oral mucosa and reduction in pain. Both HBOT and PBM ultimately support the body’s repair processes, with the goal of mitigating treatment-related injury while not interfering with the anticancer effects of therapy.
Current evidence is promising but has limitations. The clinical studies to date are heterogeneous in protocols (varying oxygen pressures, light wavelengths/doses) and endpoints, making direct comparisons difficult. Many HBOT findings come from small trials or observational series; benefits seem context-dependent (e.g. HBOT appears effective in established ORN or soft-tissue necrosis, but one trial showed no benefit for early mild ORN[9]). PBM evidence for oral mucositis is stronger, with consistent positive outcomes, but trials differ in laser parameters and timing. Both modalities are intended as adjunctive therapies, and neither replaces standard cancer treatment. Larger, sham-controlled trials are needed to confirm optimal dosing schedules and to define which patient subgroups benefit most. Long-term oncologic safety data are also important – so far no increase in tumor recurrence has been seen with supportive PBM[16], and HBOT has not shown any negative effect on survival[22][23], but continued vigilance is warranted. Overall, a cautious interpretation of the existing data is advised[24], and further research (including standardized protocols and cost-effectiveness analyses) is encouraged to solidify these therapies’ roles in cancer supportive care.
· Lin ZC et al. (2023). Hyperbaric oxygen therapy for late radiation tissue injury. Cochrane Database Syst Rev 2023(8):CD005005. DOI: 10.1002/14651858.CD005005.pub5[2][3].
· Clarke RE et al. (2008). Hyperbaric oxygen treatment of chronic refractory radiation proctitis: a randomized, double-blind, crossover trial. Int J Radiat Oncol Biol Phys 72(1):134-143. DOI: 10.1016/j.ijrobp.2007.12.048[5][6].
· Bennett MH et al. (2016). Hyperbaric oxygen therapy for late radiation tissue injury (Review). Cochrane Database Syst Rev 2016(4):CD005005. PMID: 27093034[25][9].
· Zadik Y et al. (2019). Systematic review of photobiomodulation for the management of oral mucositis in cancer patients and clinical practice guidelines. Support Care Cancer 27(10):3969-3983. DOI: 10.1007/s00520-019-04890-2[1][11].
· Sánchez-Martos R et al. (2023). Therapeutic outcomes of photobiomodulation in treatment-induced oral mucositis: a systematic review. Med Oral Patol Oral Cir Bucal 28(6):e170- e178. PMID: 37799759[12][13].
· MASCC/ISOO Mucositis Guidelines (2019 update). Photobiomodulation for the prevention of oral mucositis. MASCC/ISOO Clinical Practice Guidelines[1][26].
· Chung H et al. (2020). Safety of photobiomodulation in oncology: no evidence of tumor stimulation. Medical Lasers 9(2):93-103[16].
· Undersea & Hyperbaric Medical Society (UHMS). Delayed radiation injury (soft tissue and bony necrosis) – HBO2 therapy mechanism and outcomes. UHMS Guidance (2016)[18][19].
· Gautam AP et al. (2015). Low-level laser therapy for preventing radiotherapy-induced oral mucositis in head & neck cancer: a randomized study. Support Care Cancer 23(12):3915-3924. DOI: 10.1007/s00520-015-2810-1[12][20].
· Ferreira B et al. (2021). Photobiomodulation therapy for acute radiation dermatitis in breast cancer: a pilot randomized trial. Support Care Cancer 29(12):7209-7218. DOI: 10.1007/s00520-021-06187-8[14][15].
[1] Systematic review of photobiomodulation for the management of oral mucositis in cancer patients and clinical practice guidelines - PubMed
https://pubmed.ncbi.nlm.nih.gov/31286228/
[2] [3] [4] [8] [17] [22] [23] [24] Hyperbaric oxygen therapy for the treatment of the late effects of radiotherapy | Cochrane
[5] [6] Hyperbaric oxygen treatment of chronic refractory radiation proctitis: a randomized and controlled double-blind crossover trial with long-term follow-up - PubMed
https://pubmed.ncbi.nlm.nih.gov/18342453/
[7] Hyperbaric oxygen therapy for late radiation tissue injury - PubMed
https://pubmed.ncbi.nlm.nih.gov/27123955/
[9] [18] [19] 11. Delayed Radiation Injury (Soft Tissue and Bony Necrosis)
https://www.uhms.org/11-delayed-radiation-injury-soft-tissue-and-bony-necrosis.html
[10] The role of hyperbaric oxygen in osteoradionecrosis—a prophylactic ...
https://onlinelibrary.wiley.com/doi/10.1111/adj.12963
[11] Management of Cancer Therapy-Induced Oral Mucositis Using ...
https://www.liebertpub.com/doi/pdf/10.1089/photob.2023.0091?download=true
[12] [13] Therapeutic Outcomes of Photobiomodulation in Cancer Treatment-induced Oral Mucositis: A Systematic Review - PMC
https://pmc.ncbi.nlm.nih.gov/articles/PMC10550082/
[14] Photobiomodulation therapy for the prevention of acute radiation ...
https://www.thegreenjournal.com/article/S0167-8140(21)06117-X/fulltext
[15] Effects of photobiomodulation therapy for acute radiation dermatitis ...
https://www.sciencedirect.com/science/article/abs/pii/S0167814024035679
[16] Unraveling the role of photobiomodulation in tumor biology and ...
https://www.jkslms.or.kr/journal/view.html?uid=378&vmd=Full&
[20] The potential effect of photobiomodulation on oral mucositis induced ...
https://www.sciencedirect.com/science/article/pii/S1991790225001862
[21] Mechanisms of PhotoBioModulation (PBM) focused on oral ...
https://pmc.ncbi.nlm.nih.gov/articles/PMC8086292/
[25] [PDF] Hyperbaric oxygen therapy for late radiation tissue injury (Review)
http://neilhampson.com/uploads/3/4/7/0/34704948/2016cochrane_review_rad_nec.pdf
[26] Update of photobiomodulation in oral mucositis: a systematic review
https://www.jomos.org/articles/mbcb/pdf/2022/04/mbcb210178.pdf
Put your long text here…
Put your long text here…
Put your long text here…
Put your long text here…
Put your long text here…
Put your long text here…
Put your long text here…
Put your long text here…

Swedish international and Premier League forward Anthony Elanga is known for his pace, power, and relentless work ethic on the pitch. Playing at the highest level demands more than training — it requires recovery strategies that keep him sharp, resilient, and ready for every challenge.
With Hyperbaric Oxygen Therapy (HBOT) and Red Light Therapy, Anthony supports faster recovery, reduced muscle fatigue, and sustained performance throughout the season. These tools give him the edge to perform at his best week in, week out.
“Recovery is just as important as training — HBOT helps me go again at full speed.”t here…

Premier League and Nigerian international defender Ola Aina knows the demands of competing at the highest level. To stay sharp and resilient throughout the season, recovery is key.
With Hyperbaric Oxygen Therapy (HBOT), Ola accelerates recovery, reduces fatigue, and maintains the energy needed to perform week after week.
Known for her pace, sharp finishing, and relentless drive, Sam Kerr is one of the most dynamic forwards in the women’s game. Competing at the top level demands not only world-class training, but also recovery strategies that keep her performing at her best.
By incorporating Hyperbaric Oxygen Therapy (HBOT), Sam supports faster muscle recovery, reduced fatigue, and sustained energy throughout intense training and competition schedules.ext here…
Tom and Luke Stoltman, known as the World’s Strongest Brothers, have dominated the sport of strongman with titles and records that showcase their unrivalled power and endurance. Competing at the very highest level demands more than just training — it requires advanced recovery strategies to keep their bodies performing under extreme pressure.
Through Hyperbaric Oxygen Therapy (HBOT), the Stoltmans support faster muscle recovery, reduce fatigue, and prepare their bodies to handle the toughest challenges in strength sport.
Put your long Former WBO Heavyweight Champion Joseph Parker knows the demands of competing at the top of world boxing. Every fight requires not just power and skill, but recovery that keeps him sharp, resilient, and ready for the ring.
With Hyperbaric Oxygen Therapy (HBOT), Joseph supports faster healing, reduced fatigue, and the energy needed to perform at his best.text here…
Pushing the limits of endurance means recovery has to be as strong as the training itself. Philly Bowden, elite marathon runner, relies on Hyperbaric Oxygen Therapy (HBOT) to speed up recovery, boost energy, and prepare her body for the next challenge on the road.
Known as the “Flying Man,” Harry Aikines has built his career on speed, power, and consistency at the top of athletics. Competing at that level demands recovery tools that keep him sharp and resilient.
With Hyperbaric Oxygen Therapy (HBOT), Harry unlocks faster recovery, improved energy, and the edge to keep pushing beyond his limits.

Olympic silver medallist and rising star Ben Whittaker is known for his flair, skill, and dedication inside the ring. Competing at the elite level means recovery is just as important as training.
By using Hyperbaric Oxygen Therapy (HBOT) and Red Light Therapy, Ben accelerates recovery, reduces inflammation, and maintains the sharpness needed for peak performance.ere…
Both treatments work at a cellular level — HBOT increases oxygen saturation in your blood to accelerate recovery and healing, while red light stimulates mitochondria to boost energy production and reduce inflammation. You simply relax and let the technology do the work. We will provide you with protocol that will be tailored according to your needs.
Yes. Both are non-invasive, well-studied, and widely used in clinics and elite sports facilities worldwide. Our equipment is safety-certified and installed by trained professionals, with clear protocols for safe home use.
A few conditions require caution or medical clearance (e.g., untreated pneumothorax for HBOT, or light sensitivity disorders for red light). We’ll run through a quick checklist and liaise with your healthcare provider if needed.
Some people notice benefits after the first session — better sleep, sharper focus, or reduced soreness. For lasting improvements, most follow a personalised plan of 2–5 sessions per week over several weeks.See our page with what to expect after certain number of sessions
We handle everything — delivery, setup, and demonstration. HBOT chambers are about the size of a small sofa; red light panels are 180 cm long placed on top of the massage beds.
Yes — you can come to our Clinique in New market to try the equipment before you rent or buy.
"Our mission is to make the healing benefits of hyperbaric oxygen therapy accessible and affordable for everyone in the UK, ensuring that cost is never a barrier to health and well-being."
"My mission is simple: to cut through the noise and deliver practical, science-backed strategies that fit real life."
"I teach individuals and clinics how to integrate advanced tools like HBOT, Red Light Therapy, and PEMF to restore energy, sharpen focus, and support long-term health."
"Born with a passion for greatness. Tom Joyce founded Built Not Born with a sole mission; to unearth the inner champion within all of us."
Operated by its founders from Cambridge, Oxydise combines genuine care, scientific understanding, and hands-on experience at every stage of your journey. From initial consultation to installation and ongoing use, you work directly with the people who built the business and understand the technology inside out.
Our founder-led model means faster decision-making, more flexible solutions, and a level of personal accountability that simply doesn’t exist in large, faceless corporations. You’re supported by real experts — not call centres — ensuring your system performs exactly as it should and your results speak for themselves.
We believe in our systems so much that we make it easy to try them without risk. Whether you prefer to rent, buy, or rent-then-buy, you have the freedom to find what fits best for you.
Our goal is simple — to make advanced wellness technology accessible. You can spread payments over 12 months through our trusted finance partner, or choose our 3-month in-house payment option directly with Oxydise.
Every plan is fully transparent — no hidden fees, no long applications — just straightforward options that reflect our confidence in the equipment and our commitment to your results.
Every Oxydise system — from red-light panels to HBOT chambers — meets CE and UKCA standards and complies with ISO 9001, 14001, and 45001 certifications. Each unit is independently pressure-tested, electrically inspected, and safety-verified before leaving our facility.
Our 1.5 ATA chambers are pressure-tested to 1.75 ATA, and our 2.0 ATA hard-shell units are tested to 3.0 ATA — well above their normal operating range — to ensure exceptional structural integrity and long-term reliability.
Our expertise is anchored by real credentials — Alan Trim, Oxydise Co-Founder, is certified by the International Board of Undersea Medicine (IBUM), bringing clinical insight and hands-on experience to every client protocol. Our Clinical Board, chaired by Oli Patrick, oversees best-practice standards across all Oxydise programmes, ensuring every protocol aligns with the latest evidence in recovery and wellness. Dr Masha, a certified HBOT specialist, leads our HBOT Safety Training and Certification Course, giving every client and practitioner complete confidence in correct, safe operation.
We take contraindications seriously — from screening and consultation to ongoing use, safety always comes first. When we deliver your system, our team assembles it, performs full functional checks, and provides hands-on training so you know exactly how to use it from day one.
At Oxydise, our relationship doesn’t end when your system arrives — it begins there. Every client receives personalised guidance and structured training to ensure their chamber or red-light system is used safely, confidently, and effectively from day one.
Your induction is delivered by trained Oxydise technicians, who assemble the system on-site, perform full safety checks, and walk you through correct operation and maintenance. Beyond setup, you’ll have access to ongoing clinical and technical support, so you always know how to get the most from your system.
Our expertise is anchored by real credentials — Alan Trim, Oxydise Co-Founder, is certified by the International Board of Undersea Medicine (IBUM), bringing clinical insight and hands-on experience to every client protocol. Our Clinical Board, chaired by Oli Patrick, and our HBOT safety lead, Dr Masha, who delivers the Oxydise HBOT Safety and Certification Course, together ensure every programme reflects best practice and up-to-date science.
We take contraindications and safety seriously — every recommendation is designed to optimise results while protecting client wellbeing. And when you need help, we’re just a call away — direct access to real experts who know the equipment and the science behind it.
All Oxydise systems are stocked, assembled, and serviced here in the UK — so your experience is fast, seamless, and stress-free. There are no customs delays, import fees, or long lead times to worry about.
For our 1.5 ATA chambers, typical delivery and setup take around seven days, with installation arranged at your convenience. Hard-shell 2.0 ATA systems may take slightly longer due to logistics and final testing, but our team will always give you a clear, realistic delivery window.
When your system arrives, our trained technicians deliver, assemble, and test everything on-site, then walk you through safe operation and usage protocols before handing it over. If you have specific timing needs, you can always call us directly — we’ll do everything possible to accommodate your schedule and ensure a smooth start to your wellness journey.
Every Oxydise system comes with a 2-year comprehensive warranty as standard — reflecting the confidence we have in our build quality and testing. If an issue ever arises, our process is straightforward and fast.
Once a fault is reported, our team will assess the problem the same day and decide whether a repair or full replacement is the best course of action. In most cases, issues are resolved within three days — minimising disruption to your recovery or wellness routine.
We have a dedicated HBOT engineer on our team who can diagnose and fix most issues quickly. If on-site repair isn’t possible, we’ll arrange a rapid unit replacement to get you back up and running without delay.
With Oxydise, you’re never left waiting — just direct access to the people who built, tested, and stand behind every system we deliver.