Lactiplantibacillus plantarum IS-10506 is a novel probiotic strain with a substantial and growing body of scientific evidence supporting its therapeutic potential across a range of clinical conditions. Isolated from 'dadih', a traditional Indonesian fermented buffalo milk, this strain has demonstrated a robust safety profile and significant efficacy rooted in its potent immunomodulatory and epithelial barrier-enhancing properties.[1] Its primary mechanisms of action involve the systemic rebalancing of T-helper cell responses, characterized by the suppression of pro-inflammatory Th2 and Th17 pathways and the promotion of Th1 and regulatory T-cell (Treg) functions.[3] This is coupled with the fortification of mucosal barriers, evidenced by increased expression of tight junction proteins in both the intestinal and respiratory tracts and enhanced production of secretory IgA (sIgA).[5]
Clinically, L. plantarum IS-10506 has shown significant efficacy in randomized controlled trials for inflammatory skin diseases, markedly improving clinical scores in both atopic dermatitis and psoriasis vulgaris.[4] In the gastrointestinal tract, it accelerates the healing of gastric mucosal injury and alleviates functional constipation by modulating gut microbiota and increasing the production of beneficial short-chain fatty acids (SCFAs).[8] The therapeutic influence of this probiotic extends beyond the gut, with compelling preclinical evidence demonstrating its ability to positively modulate the gut-brain axis by increasing levels of neurotrophic factors like BDNF, and the gut-kidney axis by promoting renal tubular regeneration following injury.[10] Furthermore, it has been shown to act as a pharmacokinetic modulator, significantly enhancing the absorption of the antihypertensive drug amlodipine and the micronutrient zinc, opening novel avenues for its use as an adjuvant therapy.[2] Supported by a strong safety record, including in pediatric and immunocompromised populations, and confirmed by genomic analysis,
L. plantarum IS-10506 stands out as a promising, multi-faceted therapeutic probiotic.
The subject of this monograph, IS-10506, is a specific, well-characterized strain of the lactic acid bacterium Lactiplantibacillus plantarum, a species formerly classified within the Lactobacillus genus.[3] Throughout the scientific literature, it is consistently referred to as
Lactobacillus plantarum (LP) IS-10506 or, following recent taxonomic updates, Lactiplantibacillus plantarum IS-10506.[3]
This is a novel probiotic strain with a unique origin, having been isolated from 'dadih', a traditional fermented buffalo milk indigenous to West Sumatra, Indonesia.[1] Its isolation from a traditional food source that has been part of the regional diet for generations suggests a long history of safe human consumption. This co-evolution with a human diet may contribute to its notable compatibility with the human gastrointestinal environment, potentially enhancing its viability, colonization, and therapeutic efficacy.
The strain's genetic identity is definitively established and publicly available through its GenBank accession number, DQ860148.[6] The availability of its complete genome sequence has enabled detailed
in silico analyses, which have confirmed its safety profile and provided insights into the genetic basis of its probiotic functions.[11] The development and extensive investigation of this strain have been spearheaded by a consortium of academic institutions in Indonesia, including the Fakultas Kedokteran at Universitas Indonesia, Universitas Airlangga, and Gadjah Mada University, indicating a focused and sustained regional research effort to validate a promising indigenous biological asset.[1]
A point of critical importance for safety and accurate scientific discourse is the need to distinguish IS-10506 from a similarly designated chemical compound. One document identifies "NIH 10506" as a synonym for β-hydroxy Fentanyl, an analytical reference standard that is structurally categorized as an opioid.[36] This designation refers to a chemical entity within the U.S. National Institutes of Health (NIH) compound registry.
It must be unequivocally stated that IS-10506, the probiotic Lactiplantibacillus plantarum strain, is an entirely separate and distinct entity from NIH 10506, the opioid compound. The numerical similarity is a coincidence arising from independent nomenclature systems—one for designating a microbial strain isolate and the other for cataloging a chemical compound. Failure to recognize this distinction could lead to severe and hazardous confusion. All research and discussion pertaining to IS-10506 in this monograph refer exclusively to the probiotic bacterium.
For any probiotic to exert a therapeutic effect, it must survive transit through the hostile environment of the upper gastrointestinal (GI) tract and successfully interact with the host's intestinal mucosa. L. plantarum IS-10506 possesses intrinsic characteristics that facilitate this process and can be further enhanced through advanced formulation technology.
Acid and Bile Resistance: The strain has been demonstrated to possess inherent resistance to the harsh acidic conditions of the stomach and the detergent-like action of bile salts in the small intestine.[4] This is a fundamental prerequisite for its classification as a probiotic, ensuring that a sufficient number of viable cells can reach the lower intestine to colonize and exert their biological functions.
Microencapsulation and Viability: While the strain has natural resistance, its survival is dramatically improved by microencapsulation technology. An advanced in vitro study utilizing the TNO in vitro model of the stomach and small intestine (TIM-1), a dynamic, computer-controlled system that closely mimics human physiological conditions, provided a stark comparison. The survival rate of free, unencapsulated L. plantarum IS-10506 cells after transit through the complete upper GI model was found to be approximately 18.5%. In contrast, the survival rate of microencapsulated cells was 84.5%.[28] This greater than four-fold increase in viability highlights the critical importance of formulation. The study further isolated the effect of gastric stress, revealing that free cells were particularly sensitive to gastric acid, with only 32% survival in the gastric compartment alone as the pH dropped to 2.0.[29] This underscores that microencapsulation provides a crucial protective shield, particularly against stomach acidity.
This finding has profound implications for both clinical research and commercial product development. The efficacy of a probiotic is dose-dependent, requiring the delivery of "adequate amounts" of live microorganisms. A significant loss of viability during GI transit is functionally equivalent to a substantial reduction in the administered dose. Therefore, the colony-forming unit (CFU) count stated on a product label may not accurately reflect the bioactive dose delivered to the colon. A product formulated with 2 × 10^10 CFU of free cells might only deliver approximately 0.37 × 10^10 viable cells to the target site, whereas the same dose in a microencapsulated formulation could deliver around 1.69 × 10^10 cells. This discrepancy means that the delivery technology is a pivotal variable that can determine therapeutic success and must be considered when comparing clinical trial outcomes or evaluating the potential efficacy of different commercial products.
Adhesion Capabilities: Beyond survival, effective probiotic action requires interaction with the host. L. plantarum IS-10506 demonstrates strong adhesion to the intestinal mucosa, a process mediated by surface molecules on its cell wall, specifically lipoteichoic acid and peptidoglycan.[2] This physical attachment is a critical first step that prevents the bacteria from being quickly washed out of the GI tract, facilitating transient colonization and allowing for sustained interaction with intestinal epithelial cells. This adhesion is not merely passive; it actively triggers signaling cascades between the probiotic and nearby host immune cells, such as plasma cells, initiating the immunomodulatory effects for which the strain is known.[2] In one comparative analysis, IS-10506 was identified as the most adhesive
L. plantarum strain tested, with a 9.8% adhesion rate, and demonstrated a superior ability to competitively exclude and inhibit the adhesion of known pathogens.[37]
The therapeutic efficacy of L. plantarum IS-10506 is largely attributable to its profound and multi-faceted immunomodulatory capabilities. By influencing key cytokine profiles and T-cell populations, it can restore immune homeostasis, making it particularly effective in managing inflammatory conditions of the skin and other epithelial surfaces.
A central theme in the mechanism of action of L. plantarum IS-10506 is the rebalancing of the adaptive immune system, particularly the interplay between different subsets of T-helper (Th) cells.
Modulation of the Th1/Th2/Th17 Axis: Many allergic and inflammatory diseases, most notably atopic dermatitis, are characterized by a pathological dominance of the Th2 immune response.[19]
L. plantarum IS-10506 directly counteracts this imbalance. Clinical studies have consistently shown that supplementation leads to a significant reduction in key Th2- and Th17-associated cytokines, including Interleukin-4 (IL-4), the primary driver of allergic inflammation and IgE production, and Interleukin-17 (IL-17), a potent pro-inflammatory cytokine involved in autoimmune conditions like psoriasis.[3]
Simultaneously, the probiotic promotes a shift towards Th1 and regulatory responses. This is evidenced by a significant increase in Interferon-gamma (IFN-γ), a hallmark Th1 cytokine that can inhibit Th2 cell proliferation and function.[3] Perhaps most importantly, it significantly elevates levels of Forkhead box P3 (Foxp3+), a transcription factor that is the master regulator for the development and function of regulatory T-cells (Tregs).[3] Tregs are crucial for maintaining immune tolerance and actively suppressing excessive inflammatory responses. By boosting the Treg population,
L. plantarum IS-10506 helps to re-establish control over a dysregulated immune system.
Enhancement of Mucosal Immunity: The probiotic's influence extends to the first line of immune defense at mucosal surfaces. Supplementation has been shown to significantly increase fecal concentrations of secretory Immunoglobulin A (sIgA) in young children.[1] The mechanism for this appears to be the stimulation of Transforming Growth Factor-β1 (TGF-β1), a cytokine that promotes B-cell switching to produce IgA.[1] sIgA plays a critical role in immune exclusion by binding to pathogens and allergens in the gut lumen, preventing them from adhering to and breaching the epithelial barrier.
The immunomodulatory effects of L. plantarum IS-10506 translate directly into clinical benefits for patients with atopic dermatitis, as demonstrated in rigorous clinical trials across different age groups.
Adult Population: A randomized, double-blind, placebo-controlled trial was conducted on 30 adults with mild-to-moderate AD. The intervention group received microencapsulated L. plantarum IS-10506 at a dose of 2×1010 CFU/day for eight weeks. The primary clinical outcome was a significant reduction in the Scoring Atopic Dermatitis Index (SCORAD) in the probiotic group compared to the placebo group at the end of the study period.[3] This improvement in clinical symptoms was directly correlated with the expected immunological shifts: a significant decrease in serum IL-4 and IL-17 levels, and a significant increase in IFN-γ and Foxp3+ levels.[3] An interesting and consistent finding was that while cellular immunity was profoundly affected, total serum Immunoglobulin E (IgE) levels remained unchanged.[1] This suggests that the primary mechanism of action is the modulation of T-cell-mediated inflammatory pathways rather than a direct impact on the systemic production of IgE antibodies.
Pediatric Population: The efficacy of the probiotic was confirmed in children in a separate randomized, double-blind, placebo-controlled trial involving 22 children with mild-to-moderate AD. In this study, a dose of 1010 CFU/day for 12 weeks also resulted in a significant reduction in SCORAD scores.[5] The underlying immunological changes mirrored those seen in adults, with significant decreases in IL-4, IFN-γ, and IL-17, and a significant increase in the ratio of Foxp3+ to IL-10, indicating a strong shift towards an anti-inflammatory, regulatory immune profile.[22]
The application of L. plantarum IS-10506 has been successfully extended to another chronic inflammatory skin disease, psoriasis. In a randomized, double-blind, placebo-controlled trial involving 49 patients with mild-to-moderate psoriasis vulgaris, supplementation with 2×1010 CFU/day for 12 weeks yielded significant clinical and immunological benefits.[7]
Clinically, the probiotic group experienced a significant reduction in the Psoriasis Area and Severity Index (PASI) score, with a statistically significant difference from placebo emerging as early as week 6 and sustained through week 12. Patients also reported a better quality of life, reflected in lower Dermatology Life Quality Index (DLQI) scores.[7] The immunological mechanism was consistent with its known actions, showing a significant decrease in the key pro-inflammatory cytokine IL-17, which is a major therapeutic target in psoriasis, alongside a significant increase in the anti-inflammatory cytokine IL-10 and the Treg marker Foxp3+.[7]
A particularly compelling finding from this study was the observation of a lasting therapeutic effect. Six months after the cessation of treatment, subjects who had received the probiotic had a significantly lower probability of experiencing a disease flare (52.2%) compared to those who had received the placebo (79.2%).[7] This suggests an effect that goes beyond simple, transient symptom suppression. Standard treatments for chronic inflammatory diseases often lose their effect upon discontinuation. The durable reduction in flare rates implies that the 12-week course of
L. plantarum IS-10506 may have induced a more profound and lasting re-regulation of the patient's immune system. This is likely mediated by the observed enhancement of the regulatory T-cell population, which is fundamental to establishing long-term immune tolerance. This elevates the probiotic's potential from that of a simple adjuvant therapy to a genuine disease-modifying agent capable of "retraining" the immune system to be less auto-reactive.
The principle that L. plantarum IS-10506 restores epithelial barrier integrity and calms inflammation is not limited to the gut-skin axis. Preclinical research demonstrates its potential in the respiratory tract, supporting the concept of a "gut-lung axis." In a mouse model of allergic airway inflammation induced by the dust mite allergen Dermatophagoides pteryonissinus, administration of the probiotic was shown to significantly enhance the structural integrity of the bronchial epithelium.[5] This was demonstrated by a significant increase in the expression of three critical tight junction proteins: claudin-18, occludin, and zonula occludens-1 (ZO-1). By strengthening the physical barrier of the airway, the probiotic can reduce the penetration of inhaled allergens, thereby mitigating the initial trigger for the allergic inflammatory cascade.
In a separate model of acute lung injury caused by smoke inhalation, treatment with L. plantarum IS-10506 was associated with lower median scores for histopathological lung inflammation and reduced neutrophil counts in bronchoalveolar lavage fluid, although these trends did not reach statistical significance in that particular study design.[18] Together, these studies provide a mechanistic basis for the potential use of this probiotic in managing allergic and inflammatory respiratory conditions.
As an orally administered probiotic, the primary site of action for L. plantarum IS-10506 is the gastrointestinal tract. Within this complex ecosystem, it exerts a range of beneficial effects, from protecting the gastric lining and improving gut function to restoring cellular health and barrier integrity at a molecular level.
The stomach presents a formidable challenge to ingested microbes, but evidence suggests L. plantarum IS-10506 not only survives this environment but can also confer protective benefits to the gastric mucosa itself. Preclinical studies using rat models have demonstrated its ability to accelerate the healing of gastric injury induced by nonsteroidal anti-inflammatory drugs (NSAIDs) such as ketorolac and ibuprofen, which are common causes of gastric ulcers.[8]
The mechanism of this protective effect is twofold. First, the probiotic enhances the stomach's natural defense systems by promoting the expression of key protective molecules:
Second, it actively mitigates the inflammatory damage caused by NSAIDs. It achieves this by inhibiting the expression of the pro-inflammatory enzyme Cyclooxygenase-2 (COX-2), thereby reducing the local inflammatory response that can exacerbate tissue damage.[45] At a cellular level, the probiotic was also shown to directly enhance the regeneration of the gastric epithelium and modulate the activity of fibroblast cells to promote organized and accelerated healing.[8]
L. plantarum IS-10506 has also been investigated for its ability to improve gut function, specifically in the context of functional constipation. A randomized, double-blind, placebo-controlled trial (NCT03885973) in women with this condition found that supplementation for 21 days with fermented milk containing the probiotic significantly influenced the metabolic output of the gut microbiota.[9]
The primary finding was a change in the fecal concentration of Short-Chain Fatty Acids (SCFAs), which are beneficial metabolites produced by bacterial fermentation of dietary fiber. The probiotic group exhibited a statistically significant increase in acetate levels (p=0.032) and showed marginally significant increases in propionate (p=0.063) and butyrate (p=0.068).[9] SCFAs play a crucial role in colonic health; they serve as the primary energy source for colonocytes (especially butyrate), help maintain an acidic luminal pH that discourages pathogens, and can directly influence gut motility. The proposed mechanisms by which
L. plantarum IS-10506 alleviates constipation are therefore thought to include the modification of the gut microbiota composition to favor SCFA production, the direct effect of these metabolites on gut sensation and motility, and the overall regulation of the intraluminal environment.[9] The clinical trial was designed to measure these metabolic changes alongside clinical symptom improvement using validated questionnaires such as the Patient Assessment of Constipation Symptoms (PAC-Sym) and Quality of Life (PAC-QoL).[47]
At a more fundamental level, L. plantarum IS-10506 promotes the health and integrity of the intestinal lining itself. In preclinical models of intestinal injury induced by bacterial lipopolysaccharide (LPS), a potent inflammatory trigger, the probiotic has been shown to stimulate key regenerative processes.
Cellular Regeneration: The probiotic enhances the regeneration of Paneth cells, specialized epithelial cells located at the base of the intestinal crypts that secrete antimicrobial peptides, thereby shaping the microbiota and protecting the crucial intestinal stem cell niche.[16] This effect was confirmed by measuring increased expression of MATH-1, a transcription factor essential for secretory cell differentiation, and DEFA-6, a Paneth cell-specific defensin.[16] Furthermore, the probiotic was shown to directly activate intestinal stem cells that express the markers Lgr5 and Bmi1, driving the proliferation and renewal of the intestinal epithelium to repair damage.[32]
Barrier and Signaling Modulation: The structural integrity of the gut barrier is reinforced by the probiotic's ability to increase the expression of essential brush border and tight junction proteins, including Galectin-4, Myosin-1a, Occludin, and ZO-1.[5] This action effectively "tightens" the junctions between epithelial cells, reducing intestinal permeability or "leaky gut."
The probiotic also intervenes in the complex signaling environment of the gut. In response to inflammatory challenge, it was found to modulate the expression of serotonin (5-hydroxytryptamine, 5-HT) and the serotonin transporter (SERT) in the ileum.[13] This is a particularly insightful finding, as over 90% of the body's serotonin is produced in the gut, where it is a critical regulator of motility, secretion, and sensation. By influencing this local serotonergic system, the probiotic can directly impact gut function, such as constipation. This local action on the enteric nervous system also serves as the mechanistic foundation for its more systemic effects on the gut-brain axis, as these gut-derived signals are communicated to the central nervous system.
The influence of L. plantarum IS-10506 extends far beyond the confines of the gastrointestinal tract. By modulating the gut microbiome and its metabolic output, this probiotic initiates signals that can impact distant organs, including the brain and kidneys, and can alter the way the body absorbs nutrients and pharmaceutical drugs. This highlights its role as a systemic biological response modifier.
Compelling preclinical evidence demonstrates that L. plantarum IS-10506 actively stimulates the gut-brain axis, the bidirectional communication network linking the enteric and central nervous systems. A study in Wistar rats showed that oral supplementation led to significant and positive changes in the brain's neurochemical environment.[10]
The mechanism of this effect was elucidated through the measurement of key neuro-active molecules. The probiotic-treated group showed significant upregulation of expression in the brain for:
Crucially, these changes in the brain were accompanied by a significant increase in serotonin (5-HT)-producing cells within the ileum, confirming that the gut is the likely origin of this signaling cascade.[10] These findings strongly imply that
L. plantarum IS-10506 has the potential to be used in promoting brain health and function, and may warrant investigation for conditions related to mood, memory, and neurodevelopment.
The systemic benefits of modulating the gut environment with L. plantarum IS-10506 have also been observed in the kidneys, supporting the concept of a "gut-kidney axis." In a rat model of pyelonephritis—a severe kidney infection that causes inflammation and tissue damage—oral administration of the probiotic demonstrated a significant ability to improve renal injury and promote healing.[1]
The mechanism of action involves the activation of the kidney's own regenerative processes. The probiotic was found to stimulate endogenous renal tubular stem cells to proliferate and differentiate into mature, functional renal tubular epithelial cells, effectively repairing the damaged tissue. This was evidenced by immunohistochemical analysis of key molecular markers within the kidney:
One of the most significant findings with broad clinical implications is the ability of L. plantarum IS-10506 to alter the absorption of other ingested substances. This positions the probiotic not just as a therapeutic agent in its own right, but as a potential pharmacokinetic modulator.
Drug Absorption: In a study using a rabbit model, pre-treatment with the probiotic for 14 days was found to significantly increase the absorption of amlodipine, a widely used calcium channel blocker for treating hypertension. Plasma concentrations of amlodipine were substantially higher in the probiotic group at all time points measured after 30 minutes. At 120 minutes, the mean concentration was 12.48 ng/ml in the probiotic group versus 7.17 ng/ml in the control group, representing a nearly 75% increase in plasma levels.[12]
Nutrient Absorption: This effect is not limited to pharmaceuticals. A clinical trial in Indonesian pre-school children demonstrated that co-supplementation of L. plantarum IS-10506 with zinc led to a significantly greater improvement in the children's zinc status compared to supplementation with zinc alone or the probiotic alone.[2]
These findings carry considerable weight. On one hand, they open up a novel therapeutic paradigm where the probiotic could be used as an adjuvant to enhance the efficacy of certain drugs, potentially allowing for lower doses and fewer side effects, or to improve nutritional status by boosting the absorption of essential micronutrients. On the other hand, this capability necessitates a higher level of clinical caution. If the probiotic has a general effect on intestinal absorption—perhaps by improving gut barrier health or modulating intestinal transporter proteins—its co-administration with drugs that have a narrow therapeutic index (e.g., warfarin, digoxin) must be carefully studied to avoid the risk of inadvertently increasing their plasma levels to toxic concentrations. This elevates the probiotic from a simple "supplement" to a bioactive agent with the potential for significant drug interactions, a critical area for future research and regulatory consideration.
The translation of Lactiplantibacillus plantarum IS-10506 from a novel isolate to a potential therapeutic agent is supported by a growing portfolio of human clinical trials and a robust safety profile. This body of evidence provides a strong foundation for its current applications and guides future research into its full therapeutic potential.
L. plantarum IS-10506 has been rigorously evaluated in multiple randomized, double-blind, placebo-controlled trials, which represent the gold standard for establishing clinical evidence. These studies have spanned various indications and patient populations, from infants to adults.
Key Indications Studied: The primary areas of clinical investigation have been inflammatory skin conditions (atopic dermatitis and psoriasis vulgaris), gastrointestinal function (functional constipation), and the modulation of the immune system in both healthy and specialized populations.[3]
Clinical Trial Registry Data: Several trials have been registered, providing transparency into the scope of research:
Use in Specialized Populations: A particularly important study was conducted in HIV-infected children aged 2-18 years who were receiving antiretroviral therapy. In this immunocompromised population, supplementation with L. plantarum IS-10506 was not only safe but also effective at reducing blood lipopolysaccharide (LPS) levels.[55] High LPS is a marker of microbial translocation from a "leaky gut," which contributes to chronic inflammation in HIV. The ability to reduce LPS without causing adverse events demonstrates both the strain's barrier-restoring function and its safety in a vulnerable patient group.
To synthesize the extensive clinical data, the following table summarizes the key human trials conducted with L. plantarum IS-10506. This provides a clear, at-a-glance overview of the evidence base, allowing for direct comparison of study designs, dosages, and outcomes across different therapeutic areas. The consistent positive results across multiple, well-designed trials underscore the strain's significant clinical potential.
Indication | Trial ID / Reference | Patient Population | Study Design | Intervention (Dose & Duration) | Primary Outcomes & Key Results |
---|---|---|---|---|---|
Atopic Dermatitis | 3 | 30 Adults (mild-moderate) | RCT, DB, PC | 2×1010 CFU/day for 8 weeks | ↓ SCORAD score; ↓ IL-4, IL-17; ↑ IFN-γ, Foxp3+ |
Atopic Dermatitis | 22 | 22 Children (mild-moderate) | RCT, DB, PC | 1010 CFU/day for 12 weeks | ↓ SCORAD score; ↓ IL-4, IFN-γ, IL-17; ↑ Foxp3+/IL-10 ratio |
Psoriasis Vulgaris | 7 | 49 Adults (mild-moderate) | RCT, DB, PC | 2×1010 CFU/day for 12 weeks | ↓ PASI score; ↓ DLQI; ↓ IL-17; ↑ IL-10, Foxp3+; Lower flare rate at 6 months |
Functional Constipation | NCT03885973, 9 | 37 Women | RCT, DB, PC | 108 CFU/day in fermented milk for 3 weeks | ↑ Fecal SCFAs (Acetate, Propionate, Butyrate); Improved PAC-Sym/QoL scores |
Immune Response | NCT03924076, 33 | 79 Elementary School Children | RCT, DB, PC | 5×1010 CFU/day (paraprobiotic) for 90 days | ↑ Humoral immune response (faecal sIgA, serum IgG/IgA/IgM) |
Immune Response | 30 | Indonesian pre-school children | RCT, DB, PC | 1010 CFU/day for 90 days | ↑ Fecal sIgA; Improved zinc status when co-administered |
HIV Enteropathy | 56 | 21 HIV-infected children (2-18 yrs) | RCT, DB, PC | 2.86×1010 CFU/day for 6 days | ↓ Blood LPS level; No change in CD4/CD8 counts; No serious adverse events |
RCT: Randomized Controlled Trial; DB: Double-Blind; PC: Placebo-Controlled; SCORAD: Scoring Atopic Dermatitis Index; PASI: Psoriasis Area and Severity Index; DLQI: Dermatology Life Quality Index; SCFA: Short-Chain Fatty Acid; LPS: Lipopolysaccharide.
The safety of L. plantarum IS-10506 is well-established through its traditional use, extensive clinical trials, and genomic analysis.
General Safety: As a species, Lactobacillus plantarum has been granted "generally recognized as safe" (GRAS) status by regulatory bodies like the U.S. Food and Drug Administration. It is a common inhabitant of the human gut and is widely present in fermented foods consumed globally.[15]
Strain-Specific Safety in Clinical Trials: Across all reported clinical trials, L. plantarum IS-10506 has demonstrated an excellent safety and tolerability profile. Crucially, no serious adverse events have been reported, even in vulnerable populations such as young children and immunocompromised pediatric patients with HIV.[55]
Reported Adverse Events: The side effects that have been noted are consistently mild, transient, and gastrointestinal in nature. These include minor changes in defecation frequency or mild nausea.[7] Importantly, these effects were observed at similar frequencies in both the probiotic and placebo groups, suggesting they may not be directly attributable to the probiotic itself. The general profile for the species includes the possibility of mild gas or bloating, which is typically well-tolerated.[60]
Genomic Safety Assessment: The safety profile is further reinforced by a full genomic analysis of the IS-10506 strain. This in silico investigation confirmed the absence of significant transferable antimicrobial resistance genes or known virulence factors, providing genetic evidence that supports its long history of safe use.[11]
Precautions: While the strain has a strong safety record, standard precautions for probiotic use should be observed. Caution is generally advised for individuals who are critically ill, severely immunocompromised, or have specific medical conditions such as damaged heart valves, where extremely rare cases of systemic infection have been documented for other probiotic strains.[60]
Conclusion: Lactiplantibacillus plantarum IS-10506 is a well-characterized probiotic strain with a compelling body of evidence supporting its role as a potent immunomodulatory and epithelial barrier-fortifying agent. Its indigenous origin, robust safety profile, and proven efficacy in gold-standard clinical trials establish it as a significant therapeutic candidate. The strongest evidence supports its use in managing inflammatory skin conditions like atopic dermatitis and psoriasis, and for alleviating functional constipation. The emerging preclinical data on its systemic effects on the gut-brain axis, the gut-kidney axis, and its ability to modulate pharmacokinetics are particularly promising, suggesting a therapeutic potential that extends far beyond the gut. The critical role of microencapsulation in ensuring its viability highlights that formulation technology is key to unlocking its full clinical benefit.
Recommendations for Future Research:
Published at: September 10, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.