Helminths in alternative therapeutics of inflammatory bowel disease

Article information

Intest Res. 2024;.ir.2023.00059
Publication date (electronic) : 2024 January 12
doi : https://doi.org/10.5217/ir.2023.00059
1Redcliffe Labs, Noida, India
2School of Biological Sciences, Nanyang Technological University, Singapore
3Centre for Microbiome Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
4Department of Zoology, Ramjas College, University of Delhi, Delhi, India
Correspondence to Devi Lal, Department of Zoology, Ramjas College, University of Delhi, Delhi 110007, India. Tel: +91-9654723557, E-mail: devilal@ramjas.du.ac.in
Co-correspondence to Sunny H. Wong, Centre for Microbiome Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232. Tel: +65-6592-3927, Fax: +65-6339-2889, E-mail: sunny.wong@ntu.edu.sg
Received 2023 May 25; Revised 2023 October 24; Accepted 2023 November 1.

Abstract

Inflammatory bowel disease (IBD), which includes Crohn’s disease and ulcerative colitis, is a nonspecific chronic inflammation of the gastrointestinal tract. Despite recent advances in therapeutics and newer management strategies, IBD largely remains untreatable. Helminth therapy is a promising alternative therapeutic for IBD that has gained some attention in the last two decades. Helminths have immunomodulatory effects and can alter the gut microbiota. The immunomodulatory effects include a strong Th2 immune response, T-regulatory cell response, and the production of regulatory cytokines. Although concrete evidence regarding the efficacy of helminth therapy in IBD is lacking, clinical studies and studies done in animal models have shown some promise. Most clinical studies have shown that helminth therapy is safe and easily tolerable. Extensive work has been done on the whipworm Trichuris, but other helminths, including Schistosoma, Trichinella, Heligmosomoides, and Ancylostoma, have also been explored for pre-clinical and animal studies. This review article summarizes the potential of helminth therapy as an alternative therapeutic or an adjuvant to the existing therapeutic procedures for IBD treatment.

INTRODUCTION

Inflammatory bowel disease (IBD) is a nonspecific chronic relapsing and remitting inflammation of the gastrointestinal tract [1], which affects about 6.8 million people globally [2]. IBD includes Crohn’s disease (CD) and ulcerative colitis (UC) [3]. The patients with CD and UC show some common clinical symptoms, such as abdominal pain, fatigue, continuous or intermittent diarrhea, and rectal bleeding [4], but show differences in the anatomical distribution of inflammatory lesions [5]. Traditional IBD treatment includes immunosuppressants, corticosteroids, and aminosalicylates. The traditional treatment is now supplemented with biological therapies, which include anti-tumor necrosis factor (TNF) [6,7], anti-integrins [8-10], anti-interleukins (ILs) [11-16], and Janus kinase (JAK) inhibitors [17-20]. Nearly 50% of patients do not respond or respond slowly to traditional and biological therapies [21], which necessitates the continued development of new therapeutic options for IBD treatment.

Researchers have been exploring the potential of helminths in various autoimmune disorders, including IBD [22]. Intestinal helminthic infections are common in developing countries with high population densities and poor sanitation. Coincidentally, these are also the regions of lower IBD prevalence. Autoimmune and inflammatory diseases are common in developed countries, though incidences of such diseases are also rising in developing countries. According to the “hygiene hypothesis,” there exists a relationship between an increase in allergic and autoimmune diseases and a lack of exposure to helminth infections [23]. The hygiene hypothesis is supported by epidemiological and clinical evidence [24-29].

Once considered a rare disease, the frequency of IBD has increased in recent years. According to the “IBD hygiene hypothesis,” modern lifestyle has changed the intestinal microbiota, affecting the development of immune regulatory circuits, predisposing us to IBD [30]. Therefore, the loss of exposure to parasitic worms has been hypothesized to increase the risk of IBD [30,31], suggested by the inverse relationship between intestinal helminthic infections and the incidence of IBD [27-29,31-33], possibly because helminths can modulate host immunity to provide protection against IBD. Helminth infection in childhood can protect against the development of IBD [34]. Kabeerdoss et al. [33] reported that compared to CD patients, control subjects had higher T cell reactivity to the hookworm (Ancylostoma and Necator americanus) antigen, suggesting that hookworm infection may prevent the development of CD. This review describes the potential of using helminth therapy as an alternative therapeutic option or an adjuvant to the existing therapeutic procedures for IBD treatment. It also describes the relationship between helminths and gut microbiota. Furthermore, the review also describes various helminth species that have been explored for pre-clinical and animal studies.

HELMINTHS AND GUT MICROBIOTA

Helminths and microbiota are “old friends” of humans, as humans have coexisted with both for their entire existence. The human gut microbiota includes approximately 40 trillion microorganisms [35]. The composition of the human gut microbiota is affected by a number of factors [35]. Several studies have been conducted to know the effects of helminths on gut microbiota. Although further studies are required to reach a consensus, most of these studies have concluded that helminth infection results in a change in the gut microbiota composition [34,36,37], which can affect anti-inflammatory responses [38]. The therapeutic effects of helminth therapy may be due to a shift in the gut microbiota, which depends on the type of helminth species. Helminths may affect gut microbiota and induce dysbiosis by interacting directly with the gut microbiota through physical contact, by producing chemical products, or by competing for nutrients [39]. Various antimicrobial peptides present in helminth excretory/secretory (ES) products can directly influence gut microbiota [40]. Moreover, helminth-derived extracellular vesicles contain antimicrobial peptides and proteins that can affect the gut microbial composition [41].

Most studies showing the effect of helminths on gut microbiota are carried out in animals. These studies indicate that helminth infection can alter the gut microbiota composition in animal models [39,42-44]. Heligmosomoides polygyrus infection increases Lactobacillaceae/Lactococcus abundance in a susceptible mouse [45,46]. Trichurissuis infection alters the abundance of Proteobacteria and Deferribacteres in porcine colon microbiota [47] and increases the abundance of lactobacilli and decreases the diversity of the murine intestinal microbiota [48]. Moreover, T. suis infection along with dier abundance of Faecalibacterium andetary inulin enhances the modifications of the gut microbiota to promote beneficial bacteria such as Prevotella in the porcine gut [49]. Helminth infection can alter alpha diversity and microbial richness, and fecal short-chain fatty acid content in mice [50]. Trichuris trichiura infection changes gut microbial communities in both mice and humans [51]. Recently, Guiver et al. [52] reported that helminth infection in mice decreased α-diversity along the gradient of infective doses and increased β-diversity as the infective dose increased. Studies have reported that chronic infections with helminths, particularly Trichuris muris or H. polygyrus, reduce the abundance of Bacteroidetes and increase the abundance of Lactobacillaceae [29,45,46,48,53]. Contrary to these studies, Su et al. [54] found that H. polygyrus infection increased the abundance of Bacteroidetes and decreased the abundance of Firmicutes and Lactobacillales. A study carried out by Broadhurst et al. [55] in a primate model of idiopathic chronic diarrhea reported that T. trichiura was associated with an increase in microbial diversity and increased abundance of Bacteroidetes and Tenericutes.

Studies carried out on humans also reported a change in gut microbiota with helminth infection. A study by Ramanan et al. [29] found that the gut microbiota of people living in a region with low helminth infections have a higher abundance of Bacteroides, and those living in a region with high helminth infections have a higher abundance of Faecalibacterium and Prevotella. Another study by Jenkins et al. [56] reported that the intestinal microbiota of people with helminth infection had an increased abundance of Verrucomicrobiaceae and Enterobacteriaceae, and those without helminth infection had an increased abundance of Leuconostocaceae. Gordon et al. [57] reported that helminth infection increased the richness and diversity of the gut microbiota with an increased abundance of Faecalibacterium. A meta-analysis by Kupritz et al. [58] reported that helminth infection can influence the alpha and beta diversity of the human fecal microbiome. Contrary to these studies, Cooper et al. [59] observed that T. trichiura infection did not alter the human fecal microbiota. Other studies carried out in humans also reported that helminth infection could produce little or no changes in gut microbiota [60,61].

Studies have also found a two-way interaction between helminths and microbiota. The eggs of T. muris require direct contact with Escherichia coli to hatch [62]. T. muris also requires the presence of Bacteroides thetaiotaomicron for successful infection [63]. T. muris limits subsequent infections by other parasites of the same species by altering the host gut microbiota [63].

HELMINTHS AND THE HOST IMMUNE SYSTEM

Parasitic helminths have evolved with the human immune system and are capable of immunomodulation to promote their own survival [64]. The immunomodulatory molecules secreted by parasitic helminths to evade the host immune system can down-regulate cellular responses, reducing the excessive proinflammatory responses to some diseases. The helminths inhabiting the human intestine either attach to the intestinal epithelium or live freely in the intestinal lumen. The larvae of some intestinal helminths live within the epithelial layer of the intestine and therefore may interact directly with the host immune system. Helminths or their products can modulate various innate and adaptive immune cells, including dendritic cells (DC) that support the outgrowth of regulatory T cells (Tregs) and macrophages [65-72].

CD4+ T cells are major determinants in the generation of the host’s protective immune response against helminths. CD4+ T cells are divided into different sub-populations, such as Th1 and Th2, based on the type of cytokines produced. Th1 cells produce cytokines that contribute to inflammation and perpetuate autoimmune response, such as IL-2, IL-12, and interferon gamma (IFN-γ), while Th2 cells produce cytokines that have anti-inflammatory response, such as IL-10. The host immune responses to helminths include a strong Th2 immune response, Treg response, and the production of regulatory cytokines, such as IL-10 [73-81]. The specific Th2 response and Th2 cytokines induced by helminths [82-85] can attenuate Th1-driven inflammatory responses in the host [82,85,86]. Helminths are also capable of inducing immune Tregs that express markers such as Foxp3 and CD25 [87-89]. Such Tregs include type 1 Tr (Tr1) cells that secrete high levels of IL-10 [86,90]. The induction of Th2 and Tr1 can explain the beneficial effects of helminth therapy in IBD patients [84]. The Th2 response elicited by helminth infection also induces alternatively activated macrophages [91-94], which can negatively regulate Th1 response [95,96].

HELMINTHS IN IBD THERAPEUTICS

Several helminths, including Trichuris, Schistosoma, Trichinella, Heligmosomoides, and Ancylostoma, have been considered potential candidates for developing alternative or adjuvant therapy for IBD (Fig. 1). Trichuris is a genus of parasitic roundworms in the family Trichuridae. Trichuris is commonly referred to as whipworm. The important species of Trichuris are T. suis, T. muris, and T. trichiura. The natural host of T. suis is the pig, but it can transiently colonize humans without causing disease [97]. T. trichiura is known as the human whipworm because humans are its natural hosts. The natural host of T. muris is the mouse. Schistosoma is a genus of parasitic flatworms, commonly known as blood flukes, belonging to the class Trematoda. Schistosoma mansoni is the leading cause of schistosomiasis in humans. S. mansoni and Schistosoma japonicum are potential candidates for IBD treatment, and considerable research has been carried out on these 2 species. S. japonicum has a wide host range and infects many species of mammals. Trichinella spiralis is a parasitic roundworm that causes trichinosis in humans. It is transmitted to humans by consuming undercooked or raw meat. T. spiralis completes its life cycle in a single host, during which it releases many proteins that help it to evade the host immune response [98-100]. Another species, Trichinella papuae, has also been considered for treating IBD [101]. H. polygyrus is a roundworm commonly found in the intestine of rodents. Ancylostoma is a genus of roundworms commonly referred to as hookworms. Only a few studies have explored the potential of Ancylostoma in IBD treatment [102-105].

Fig. 1.

FigurHelminths in inflammatory bowel disease treatment and their molecular mechanisms. Figure created with BioRender. IL, interleukin; INF-γ, interferon gamma; TNF-α, tumor necrosis factor alpha; TGF-β, transforming growth factor beta; Treg, regulatory T cell.ecaption

Clinical trials have been carried out for only a few, including Trichuris and Schistosoma (Table 1). Most of the studies on the efficacy of helminth therapy in IBD are restricted to animal models. In a recent meta-analysis, Shields and Cooper [106] concluded that it is difficult to reach any conclusion regarding the efficacy of helminth therapy. Nevertheless, helminth therapy is safe and tolerable. Large clinical cohort studies must be carried out to establish the efficacy of helminth therapy in IBD.

Completed Clinical Trials on Helminths in Inflammatory Bowel Disease Therapeutics

1. Human Clinical Trials

Clinical trials have been carried out using Trichuris, protein derived from Schistosoma, and excretory products derived from H. polygyrus. Several studies have been conducted to evaluate the effectiveness of T. suis for IBD treatment [27,28,85,107-110]. T. suis was the organism of choice for early clinical trials in humans. In one of the first small open-label trials, 7 patients were enrolled, 4 with CD, and 3 with UC [111]. Each patient who received a single dose of 2,500 viable ova demonstrated improvement in clinical symptoms [111]. Another study by the same group enrolled 29 patients with CD and found that after 12 weeks of T. suis ova (TSO) therapy, 66% of patients showed remission [112]. A similar study carried out in patients with UC also reported that patients receiving 2,500 viable T. suis eggs showed a significant improvement over placebo [85]. Human clinical trials have revealed that TSO therapy can reduce the disease severity in some patients with UC and CD, is well tolerated and not associated with short- or long-term side effects [85,107,111-113]. In a case study, Broadhurst et al. [114] reported that patients with severe refractory UC achieved clinical remission when infected with T. trichiura. They also showed that Trichuris infection escalated mucosal expression of IL-4 and IL-22.

Despite the potential of TSO therapy, many studies have found contradictory results. In a Cochrane review, Garg et al. [108] concluded that there was insufficient evidence regarding the efficacy and safety of TSO therapy to treat IBD patients. Schölmerich et al. [109] reported that TSO therapy did not show any clinical efficacy against active CD. A meta-analysis by Huang et al. [110] concluded that TSO therapy showed no statistical benefit for IBD patients.

A protein derived from Schistosoma, P28 glutathione-S-transferase (P28GST), combines the activities of antioxidant glutathione S-transferase and prostaglandin D synthase [115,116]. P28GST was first developed as a vaccine against schistosomiasis and demonstrated safety and tolerability, both in adults and children [117,118]. In a pilot Phase 2a study, Capron et al. [119] evaluated the safety of P28GST in patients with mild CD. They found a decrease in baseline Crohn’s Disease Activity Index and blood calprotectin levels, suggesting that P28GST can be used as a safe therapeutic option for CD. Cook et al. [120] reported that a secretory product of H. polygyrus, Hp-TGF-β mimic (Hp-TGM), could induce Foxp3 expression in a more efficient manner than transforming growth factor beta (TGF-β). Moreover, Tregs induced by Hp-TGM had superior suppressive function than Tregs induced by TGF-β. Croese et al. [121] reported that hookworm Necator americanus could improve disease activity in 9 patients with CD after 20 weeks of administration.

2. Studies Carried on Animal Models

While there are only a few studies on humans that suggest potential of helminths in IBD therapeutics, many studies have been carried out in animals, particularly mouse model of IBD. These studies have explored the potential of whole organisms, eggs/ova, antigens, soluble and secreted proteins, metabolites, ES, and recombinant products from different helminths.

1) Whole Organisms

Experiments in animal models have been carried out using both adult worms and larvae from various helminth species. The use of whole organisms has shown some potential in animal models and can be replicated in large-scale clinical trials. In one of the early studies, Reardon et al. [122] showed that the tapeworm Hymenolepis diminuta infection could ameliorate dextran sodium sulfate (DSS)-induced colitis. Studies have found that S. mansoni could attenuate intestinal inflammation in trinitrobenzene sulfonic acid (TNBS)-induced colitis [123], DSS-induced colitis [124,125], and Toxoplasma gondii-induced ileitis [126]. Studies have found that S. japonicum cercariae could attenuate the inflammatory response in a DSS-induced IBD mouse model by promoting IL-10, repressing IFN-γ production and expression, and enhancing the Treg subset population [127,128].

Khan et al. [129] reported that T. spiralis infection could protect mice from dinitrobenzene sulfonic acid (DNBS)-induced colitis. Subsequent studies found that Trichinella can induce a strong Th2/Treg response, induce the production of immunoregulatory cytokines [100,130-133] and recruit Tregs [134]. Studies with T. spiralis have also found that it could ameliorate inflammation in a mouse model of acetic acid-induced colitis [135], DSS-induced colitis [136,137], Citrobacter rodentium-induced colitis [138], and TNBS-induced colitis [139] by recruiting Foxp3-expressing Tregs, immunomodulating Tregs to produce anti-inflammatory cytokines, inhibiting proinflammatory cytokines and PD-1 upregulation and M2 macrophage polarization [135-140].

H. polygyrus improves colitis by affecting mucosal Th1 cytokine production, suppressing IL-17 production from lamina propria mononuclear cells and mesenteric lymph node cells and augmenting IL-4 and IL-10 production [141,142]. H. polygyrus secretes a substance that mimics the mechanism by which TGF-β promotes the expression of Foxp3 in CD4+ T cells [143]. Foxp3+ Tregs maintain immune homeostasis in the intestine, and any defects therein are linked to IBD. This is evident from the fact that mice with IL-10-/- Foxp3-expressing T cells develop spontaneous colitis [144]. H. polygyrus cannot prevent colitis in transgenic mice harboring T cells that do respond to TGF-β [145].

Studies have indicated that H. polygyrus infection can alleviate IBD by reducing the production of IFN-γ and IL-17 [146], inducing Foxp3+ Tregs, mucosal production of IL-10 [147,148], elevating Th2 cytokine expression, increasing mast cell infiltration [149] and by down-modulating Smad7 expression in intestinal CD4+ T cells [150].

H. polygyrus larvae can also reduce inflammation in a DSSinduced colitis model [151]. Blum et al. [152] found that H. polygyrus bakeri modulated intestinal DC function in Rag IBD murine models reconstituted with IL-10⁻/⁻ T cells. Moreover, H. polygyrus exacerbates C. rodentium-induced colitis in murine models, probably due to an influx of alternatively activated macrophages or increased IL-10 production by DC caused by H. polygyrus infection, which impedes host protection against C. rodentium infection [153,154].

2) Eggs/Ova

Leonardi et al. [155] reported that TSO therapy could significantly reduce the disease activity in a rabbit model of DSS-induced colitis. However, TSO exacerbated colitis in immunosuppressed rabbits. Exposure to schistosome eggs protects a mouse model of TNBS-induced [156] and DSS-induced colitis [157] by inhibiting Th1-type inflammation dependent on the IL-4 signaling pathway [156]. S. mansoni egg antigen can induce Foxp3+ Tregs and Th2 cytokines and has modulatory effects on a DSS-induced colitis mouse model [158]. Extracellular vesicles from DC treated with S. japonicum soluble egg antigen can ameliorate DSS-induced colitis in mice [159]. Recently, Hou et al. [160] reported that pre-exposure to S. japonicum eggs could alleviate colitis in the TNBS-induced colitis model, which was related to increased Treg immune response, decreased Th17 immune response, and reconstruction of Treg/Th17 balance. Another study by Zhu et al. [161] reported that Schistosoma soluble egg antigen could decrease colonic inflammation and the disease activity index score in mice with DSS-induced IBD. This study also reported changes in the intestinal microflora with Schistosoma soluble egg antigen intervention.

3) Antigens, and Soluble and Secreted Proteins

T. spiralis antigens can reduce the severity of colitis in a mouse model of DNBS-induced colitis, as evident from a down-regulation of IL-1β production, myeloperoxidase activity, and inducible nitric oxide synthase and an upregulation of IL-13 and TGF-β production [162]. In a meta-analysis, Li et al. [163] concluded that Trichinella and its derived antigens are effective in alleviating IBD in mouse models. S. mansoni soluble proteins can ameliorate TNBS-induced colitis in mice by increasing the expression of regulatory cytokines and suppressing the expression of proinflammatory cytokines [102]. Protein P28GST from schistosomes can reduce intestinal inflammation in experimental colitis by down-regulating the Th1/Th17 inflammatory response [115,164]. Foligné et al. [165] reported that P28GST administration could modulate the diversity and composition of mouse fecal microbiota and significantly reduce colitis in a mouse model. They found that P28GST increased the immunoregulatory response through Th2/Treg cells and M2 macrophages. It also reduces the local expression of proinflammatory mediators, such as TNF-α, and enhances the expression of anti-inflammatory cytokines, such as IL-5, IL-13, and IL-10 [117,164]. Studies have found that a secreted protein of S. japonicum, rSj16, shows protective effects on DSS-induced colitis by inhibiting the peroxisome proliferator-activated receptor (PPAR)-alpha-α signaling pathway [166] and reducing apoptosis [167].

4) Metabolites, ES, and Recombinant Products

Metabolites from H. polygyrus can lower disease activity in a mouse model of DSS-induced colitis by decreasing LPS-induced TNF and increasing IL-10 release by bone marrow DC [168]. The ES products from T. spiralis suppress macrophage-mediated inflammatory responses, reduce proinflammatory cytokine production, stimulate the differentiation of host Treg cells by activating DC, decrease the expression of IFN-γ and nuclear factor (NF), increase the expression of IL-4 and IL-10, change the ratio of M1/M2 phenotypic macrophages in the spleen, reduce TNF-α secretion, inducing IL-10 expression [169-175]. T. spiralis-derived protein paramyosin ameliorates the progression of colitis by inducing Tregs, reducing proinflammatory responses, and enhancing regulatory cytokine production in a mouse model of DSS-induced colitis [176]. Various T. spiralis-derived recombinant products have also been explored for IBD treatment including a recombinant protein from T. spiralis, rTsP53 [177], recombinant serine protease [178], and recombinant serine protease-like protein (rTs-ADSp-7) [179].

H. polygyrus ES products and recombinant protein, TGF mimic (TGM), showed some benefit in DSS-induced and TNBA-induced colitis, with a reduction in inflammatory cytokines [180,181]. Studies have also found anti-inflammatory properties of low-molecular-weight metabolites derived from somatic extracts and ES products of Ancylostoma caninum [104,105] and Ancylostoma ceylanicum [103]. The recombinant macrophage migration inhibitor-like factor from the herring worm Anisakis simplex third-stage larva has shown to ameliorate DSS-induced colitis in a mouse model [182].

RAMIFICATIONS OF HELMINTH THERAPY

The promising results shown by helminth therapy suggest that the therapy can be used as an alternative or adjuvant therapy to treat various autoimmune or allergic diseases [183]. Nevertheless, most helminths have pathogenic potential in humans, which makes their direct use in treatment unethical. Clinical trials have also shown that helminth therapy is not free of side effects (Table 1). Long-term infection by hookworm can result in iron deficiency anemia [184]. Certain worms may increase mucus secretion and gastrointestinal motility, resulting in diarrhea and abdominal cramps [185,186]. Furthermore, colonization by specific helminths may intensify the inflammatory response to an existing enteric bacterial infection [93,153]. Helminths can also suppress host innate or adaptive immune responses to other parasites, predisposing patients to various infections [187-189]. Although many clinical trials in IBD patients have revealed no side effects of T. suis, Kradin et al. [190] reported that TSO therapy for pediatric CD caused infection in one patient. Similarly, acute exposure to N. americanus can cause diarrhea, vomiting, and abdominal pain [186,191]. Several studies have found that the use of the crude extract, purified, or recombinant molecules/proteins derived from helminths can produce similar anti-inflammatory effects as whole organisms [192-195]. Therefore, in place of living helminths, their extracts or recombinant molecules can be used for IBD treatment.

CONCLUSIONS AND FUTURE PERSPECTIVES

Helminths and microbiota are two “old friends” of humans. Helminths play an important immunoregulatory role in mucosal immunity and in maintaining a healthy gut ecosystem. There is sufficient evidence to conclude that helminth infections are immunomodulatory and regulate the host’s immune response. Studies have confirmed the safety and tolerability of helminth therapy. Various parameters need to be standardized to increase the efficacy and success rate of helminth therapy. These parameters include the helminth species to be used, the optimal dose and frequency of administration, and the route and mode of administration. A detailed study of host genetic factors, environment, diet, and the exact mechanism of action will surely help in designing better helminth therapy for IBD. In place of whole live helminths, proteins, and ES products can also be used for IBD treatment. The commercial production of such products can be done by expressing them in a suitable host, such as bacteria or yeast. The use of helminth-derived recombinant anti-inflammatory molecules can rule out the fear and concerns associated with the use of live helminths. Nevertheless, large clinical cohort studies must be carried out to know the safety and antigenicity of such recombinant products.

Notes

Funding Source

The authors would like to acknowledge the Nanyang Technological University (NTU) Start Up Grant (021337–00001), Wang Lee Wah Memorial Fund, and Singapore Ministry of Education (MOE) Tier 1 Academic Research Fund (RG37/22) for support of this work.

Conflict of Interest

No potential conflict of interest relevant to this article was reported.

Data Availability Statement

Not applicable.

Author Contributions

Conceptualization: Pandey H, Wong SH, Lal D. Funding acquisition: Wong SH. Methodology: Pandey H, Tang DWT, Wong SH, Lal D. Supervision: Wong SH, Lal D. Writing - original draft: Pandey H, Tang DWT, Wong SH, Lal D. Writing - review & editing: Pandey H, Tang DWT, Wong SH, Lal D. Approval of final manuscript: all authors.

References

1. Borowitz SM. The epidemiology of inflammatory bowel disease: clues to pathogenesis? Front Pediatr 2023;10:1103713.
2. GBD 2017 Inflammatory Bowel Disease Collaborators. The global, regional, and national burden of inflammatory bowel disease in 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol Hepatol 2020;5:17–30.
3. Pandey H, Jain D, Tang DWT, Wong SH, Lal D. Gut microbiota in pathophysiology, diagnosis, and therapeutics of inflammatory bowel disease. Intest Res 2023;Nov. 8. [Epub]. https://doi.org/10.5217/ir.2023.00080.
4. Maloy KJ, Powrie F. Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature 2011;474:298–306.
5. Kovarik JJ, Tillinger W, Hofer J, et al. Impaired anti-inflammatory efficacy of n-butyrate in patients with IBD. Eur J Clin Invest 2011;41:291–298.
6. Harris MS, Hartman D, Lemos BR, et al. AVX-470, an orally delivered anti-tumour necrosis factor antibody for treatment of active ulcerative colitis: results of a first-in-human trial. J Crohns Colitis 2016;10:631–640.
7. Almon E, Shaaltiel Y, Sbeit W, et al. Novel orally administered recombinant anti-TNF alpha fusion protein for the treatment of ulcerative colitis: results from a phase 2a clinical trial. J Clin Gastroenterol 2021;55:134–140.
8. Feagan BG, Rutgeerts P, Sands BE, et al. Vedolizumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med 2013;369:699–710.
9. Park SC, Jeen YT. Anti-integrin therapy for inflammatory bowel disease. World J Gastroenterol 2018;24:1868–1880.
10. Sandborn WJ, Vermeire S, Tyrrell H, et al. Etrolizumab for the treatment of ulcerative colitis and Crohn’s disease: an overview of the phase 3 clinical program. Adv Ther 2020;37:3417–3431.
11. Feagan BG, Sandborn WJ, D’Haens G, et al. Induction therapy with the selective interleukin-23 inhibitor risankizumab in patients with moderate-to-severe Crohn’s disease: a randomised, double-blind, placebo-controlled phase 2 study. Lancet 2017;389:1699–1709.
12. Sabino J, Verstockt B, Vermeire S, Ferrante M. New biologics and small molecules in inflammatory bowel disease: an update. Therap Adv Gastroenterol 2019;12:1756284819853208.
13. Danese S, Vermeire S, Hellstern P, et al. Randomised trial and open-label extension study of an anti-interleukin-6 antibody in Crohn’s disease (ANDANTE I and II). Gut 2019;68:40–48.
14. Sands BE, Sandborn WJ, Panaccione R, et al. Ustekinumab as induction and maintenance therapy for ulcerative colitis. N Engl J Med 2019;381:1201–1214.
15. Sandborn WJ, Ferrante M, Bhandari BR, et al. Efficacy and safety of mirikizumab in a randomized phase 2 study of patients with ulcerative colitis. Gastroenterology 2020;158:537–549.
16. Al-Bawardy B, Shivashankar R, Proctor DD. Novel and emerging therapies for inflammatory bowel disease. Front Pharmacol 2021;12:651415.
17. Sandborn WJ, Su C, Sands BE, et al. Tofacitinib as induction and maintenance therapy for ulcerative colitis. N Engl J Med 2017;376:1723–1736.
18. Sandborn WJ, Nguyen DD, Beattie DT, et al. Development of gut-selective pan-Janus kinase inhibitor TD-1473 for ulcerative colitis: a translational medicine programme. J Crohns Colitis 2020;14:1202–1213.
19. Salas A, Hernandez-Rocha C, Duijvestein M, et al. JAK-STAT pathway targeting for the treatment of inflammatory bowel disease. Nat Rev Gastroenterol Hepatol 2020;17:323–337.
20. Feagan BG, Danese S, Loftus EV, et al. Filgotinib as induction and maintenance therapy for ulcerative colitis (SELECTION): a phase 2b/3 double-blind, randomised, placebo-controlled trial. Lancet 2021;397:2372–2384.
21. Toskas A, Akbar A. IBD therapeutics: what is in the pipeline? Frontline Gastroenterol 2022;13:e35–e43.
22. Smallwood TB, Giacomin PR, Loukas A, Mulvenna JP, Clark RJ, Miles JJ. Helminth immunomodulation in autoimmune disease. Front Immunol 2017;8:453.
23. Strachan DP. Hay fever, hygiene, and household size. BMJ 1989;299:1259–1260.
24. van Riet E, Hartgers FC, Yazdanbakhsh M. Chronic helminth infections induce immunomodulation: consequences and mechanisms. Immunobiology 2007;212:475–490.
25. Flohr C, Quinnell RJ, Britton J. Do helminth parasites protect against atopy and allergic disease? Clin Exp Allergy 2009;39:20–32.
26. Erb KJ. Can helminths or helminth-derived products be used in humans to prevent or treat allergic diseases? Trends Immunol 2009;30:75–82.
27. Chu KM, Watermeyer G, Shelly L, et al. Childhood helminth exposure is protective against inflammatory bowel disease: a case control study in South Africa. Inflamm Bowel Dis 2013;19:614–620.
28. Wammes LJ, Mpairwe H, Elliott AM, Yazdanbakhsh M. Helminth therapy or elimination: epidemiological, immunological, and clinical considerations. Lancet Infect Dis 2014;14:1150–1162.
29. Ramanan D, Bowcutt R, Lee SC, et al. Helminth infection promotes colonization resistance via type 2 immunity. Science 2016;352:608–612.
30. Elliott DE, Urban JF Jr, Argo CK, Weinstock JV. Does the failure to acquire helminthic parasites predispose to Crohn’s disease? FASEB J 2000;14:1848–1855.
31. Weinstock JV, Summers RW, Elliott DE, Qadir K, Urban JF Jr, Thompson R. The possible link between de-worming and the emergence of immunological disease. J Lab Clin Med 2002;139:334–338.
32. Weinstock JV, Elliott DE. Helminths and the IBD hygiene hypothesis. Inflamm Bowel Dis 2009;15:128–133.
33. Kabeerdoss J, Pugazhendhi S, Subramanian V, Binder HJ, Ramakrishna BS. Exposure to hookworms in patients with Crohn’s disease: a case-control study. Aliment Pharmacol Ther 2011;34:923–930.
34. Schölmerich J. Trichuris suis ova in inflammatory bowel disease. Dig Dis 2013;31:391–395.
35. Pandey H, Tang DWT, Wong SH, Lal D. Gut microbiota in colorectal cancer: biological role and therapeutic opportunities. Cancers (Basel) 2023;15:866.
36. Brosschot TP, Reynolds LA. The impact of a helminth-modified microbiome on host immunity. Mucosal Immunol 2018;11:1039–1046.
37. Rapin A, Chuat A, Lebon L, Zaiss MM, Marsland BJ, Harris NL. Infection with a small intestinal helminth, Heligmosomoides polygyrus bakeri, consistently alters microbial communities throughout the murine small and large intestine. Int J Parasitol 2020;50:35–46.
38. Giacomin P, Croese J, Krause L, Loukas A, Cantacessi C. Suppression of inflammation by helminths: a role for the gut microbiota? Philos Trans R Soc Lond B Biol Sci 2015;370:20140296.
39. Ling F, Steinel N, Weber J, et al. The gut microbiota response to helminth infection depends on host sex and genotype. ISME J 2020;14:1141–1153.
40. Rausch S, Midha A, Kuhring M, et al. Parasitic nematodes exert antimicrobial activity and benefit from microbiota-driven support for host immune regulation. Front Immunol 2018;9:2282.
41. Allen NR, Taylor-Mew AR, Wilkinson TJ, et al. Modulation of rumen microbes through extracellular vesicle released by the rumen fluke Calicophoron daubneyi. Front Cell Infect Microbiol 2021;11:661830.
42. Kreisinger J, Bastien G, Hauffe HC, Marchesi J, Perkins SE. Interactions between multiple helminths and the gut microbiota in wild rodents. Philos Trans R Soc Lond B Biol Sci 2015;370:20140295.
43. Barelli C, Donati C, Albanese D, et al. Interactions between parasitic helminths and gut microbiota in wild tropical primates from intact and fragmented habitats. Sci Rep 2021;11:21569.
44. Myhill LJ, Stolzenbach S, Mejer H, et al. Parasite-probiotic interactions in the gut: Bacillus sp. and Enterococcus faecium regulate type-2 inflammatory responses and modify the gut microbiota of pigs during helminth infection. Front Immunol 2022;12:793260.
45. Walk ST, Blum AM, Ewing SA, Weinstock JV, Young VB. Alteration of the murine gut microbiota during infection with the parasitic helminth Heligmosomoides polygyrus. Inflamm Bowel Dis 2010;16:1841–1849.
46. Reynolds LA, Smith KA, Filbey KJ, et al. Commensal-pathogen interactions in the intestinal tract: lactobacilli promote infection with, and are promoted by, helminth parasites. Gut Microbes 2014;5:522–532.
47. Li RW, Wu S, Li W, et al. Alterations in the porcine colon microbiota induced by the gastrointestinal nematode Trichuris suis. Infect Immun 2012;80:2150–2157.
48. Holm JB, Sorobetea D, Kiilerich P, et al. Chronic Trichuris muris infection decreases diversity of the intestinal microbiota and concomitantly increases the abundance of lactobacilli. PLoS One 2015;10e0125495.
49. Stolzenbach S, Myhill LJ, Andersen LO, et al. Dietary inulin and Trichuris suis infection promote beneficial bacteria throughout the porcine gut. Front Microbiol 2020;11:312.
50. Khudhair Z, Alhallaf R, Eichenberger RM, et al. Gastrointestinal helminth infection improves insulin sensitivity, decreases systemic inflammation, and alters the composition of gut microbiota in distinct mouse models of type 2 diabetes. Front Endocrinol (Lausanne) 2021;11:606530.
51. Rosa BA, Snowden C, Martin J, et al. Whipworm-associated intestinal microbiome members consistent across both human and mouse hosts. Front Cell Infect Microbiol 2021;11:637570.
52. Guiver E, Galan M, Lippens C, Bellenger J, Faivre B, Sorci G. Increasing helminth infection burden depauperates the diversity of the gut microbiota and alters its composition in mice. Curr Res Parasitol Vector Borne Dis 2022;2:100082.
53. Houlden A, Hayes KS, Bancroft AJ, et al. Chronic Trichuris muris infection in C57BL/6 mice causes significant changes in host microbiota and metabolome: effects reversed by pathogen clearance. PLoS One 2015;10e0125945.
54. Su C, Su L, Li Y, et al. Helminth-induced alterations of the gut microbiota exacerbate bacterial colitis. Mucosal Immunol 2018;11:144–157.
55. Broadhurst MJ, Ardeshir A, Kanwar B, et al. Therapeutic helminth infection of macaques with idiopathic chronic diarrhea alters the inflammatory signature and mucosal microbiota of the colon. PLoS Pathog 2012;8e1003000.
56. Jenkins TP, Rathnayaka Y, Perera PK, et al. Infections by human gastrointestinal helminths are associated with changes in faecal microbiota diversity and composition. PLoS One 2017;12e0184719.
57. Gordon CA, Krause L, McManus DP, et al. Helminths, polyparasitism, and the gut microbiome in the Philippines. Int J Parasitol 2020;50:217–225.
58. Kupritz J, Angelova A, Nutman TB, Gazzinelli-Guimaraes PH. Helminth-induced human gastrointestinal dysbiosis: a systematic review and meta-analysis reveals insights into altered taxon diversity and microbial gradient collapse. mBio 2021;12e0289021.
59. Cooper P, Walker AW, Reyes J, et al. Patent human infections with the whipworm, Trichuris trichiura, are not associated with alterations in the faecal microbiota. PLoS One 2013;8e76573.
60. Lee SC, Tang MS, Lim YA, et al. Helminth colonization is associated with increased diversity of the gut microbiota. PLoS Negl Trop Dis 2014;8e2880.
61. Giacomin P, Zakrzewski M, Jenkins TP, et al. Changes in duodenal tissue-associated microbiota following hookworm infection and consecutive gluten challenges in humans with coeliac disease. Sci Rep 2016;6:36797.
62. Hayes KS, Bancroft AJ, Goldrick M, Portsmouth C, Roberts IS, Grencis RK. Exploitation of the intestinal microflora by the parasitic nematode Trichuris muris. Science 2010;328:1391–1394.
63. White EC, Houlden A, Bancroft AJ, et al. Manipulation of host and parasite microbiotas: survival strategies during chronic nematode infection. Sci Adv 2018;4eaap7399.
64. Helmby H. Human helminth therapy to treat inflammatory disorders: where do we stand? BMC Immunol 2015;16:12.
65. Whelan RA, Hartmann S, Rausch S. Nematode modulation of inflammatory bowel disease. Protoplasma 2012;249:871–886.
66. Schnoeller C, Rausch S, Pillai S, et al. A helminth immunomodulator reduces allergic and inflammatory responses by induction of IL-10-producing macrophages. J Immunol 2008;180:4265–4272.
67. Siracusa MC, Reece JJ, Urban JF, Scott AL. Dynamics of lung macrophage activation in response to helminth infection. J Leukoc Biol 2008;84:1422–1433.
68. Klotz C, Ziegler T, Figueiredo AS, et al. A helminth immunomodulator exploits host signaling events to regulate cytokine production in macrophages. PLoS Pathog 2011;7e1001248.
69. Li Z, Liu G, Chen Y, Liu Y, Liu B, Su Z. The phenotype and function of naturally existing regulatory dendritic cells in nematode-infected mice. Int J Parasitol 2011;41:1129–1137.
70. Segura M, Su Z, Piccirillo C, Stevenson MM. Impairment of dendritic cell function by excretory-secretory products: a potential mechanism for nematode-induced immunosuppression. Eur J Immunol 2007;37:1887–1904.
71. Hewitson JP, Grainger JR, Maizels RM. Helminth immunoregulation: the role of parasite secreted proteins in modulating host immunity. Mol Biochem Parasitol 2009;167:1–11.
72. Taylor BC, Zaph C, Troy AE, et al. TSLP regulates intestinal immunity and inflammation in mouse models of helminth infection and colitis. J Exp Med 2009;206:655–667.
73. Finkelman FD, Shea-Donohue T, Morris SC, et al. Interleukin-4- and interleukin-13-mediated host protection against intestinal nematode parasites. Immunol Rev 2004;201:139–155.
74. Harnett W, Harnett MM. Molecular basis of worm-induced immunomodulation. Parasite Immunol 2006;28:535–543.
75. Elliott DE, Weinstock JV. Helminthic therapy: using worms to treat immune-mediated disease. Adv Exp Med Biol 2009;666:157–166.
76. Elliott DE, Weinstock JV. Helminth-host immunological interactions: prevention and control of immune-mediated diseases. Ann N Y Acad Sci 2012;1247:83–96.
77. Weinstock JV. Autoimmunity: the worm returns. Nature 2012;491:183–185.
78. Grencis RK, Humphreys NE, Bancroft AJ. Immunity to gastrointestinal nematodes: mechanisms and myths. Immunol Rev 2014;260:183–205.
79. Anuradha R, Munisankar S, Bhootra Y, et al. Systemic cytokine profiles in Strongyloides stercoralis infection and alterations following treatment. Infect Immun 2016;84:425–431.
80. Anuradha R, Munisankar S, Dolla C, Kumaran P, Nutman TB, Babu S. Parasite antigen-specific regulation of Th1, Th2, and Th17 responses in Strongyloides stercoralis infection. J Immunol 2015;195:2241–2250.
81. Vukman KV, Lalor R, Aldridge A, O’Neill SM. Mast cells: new therapeutic target in helminth immune modulation. Parasite Immunol 2016;38:45–52.
82. Fox JG, Beck P, Dangler CA, et al. Concurrent enteric helminth infection modulates inflammation and gastric immune responses and reduces helicobacter-induced gastric atrophy. Nat Med 2000;6:536–542.
83. Zaccone P, Fehérvári Z, Jones FM, et al. Schistosoma mansoni antigens modulate the activity of the innate immune response and prevent onset of type 1 diabetes. Eur J Immunol 2003;33:1439–1449.
84. Moreels TG, Pelckmans PA. Gastrointestinal parasites: potential therapy for refractory inflammatory bowel diseases. Inflamm Bowel Dis 2005;11:178–184.
85. Summers RW, Elliott DE, Urban JF, Thompson RA, Weinstock JV. Trichuris suis therapy for active ulcerative colitis: a randomized controlled trial. Gastroenterology 2005;128:825–832.
86. Wang LJ, Cao Y, Shi HN. Helminth infections and intestinal inflammation. World J Gastroenterol 2008;14:5125–5132.
87. Takahashi T, Tagami T, Yamazaki S, et al. Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. J Exp Med 2000;192:303–310.
88. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science 2003;299:1057–1061.
89. Belkaid Y, Rouse BT. Natural regulatory T cells in infectious disease. Nat Immunol 2005;6:353–360.
90. Ruyssers NE, De Winter BY, De Man JG, et al. Worms and the treatment of inflammatory bowel disease: are molecules the answer? Clin Dev Immunol 2008;2008:567314.
91. Gordon S. Alternative activation of macrophages. Nat Rev Immunol 2003;3:23–35.
92. Anthony RM, Urban JF, Alem F, et al. Memory T(H)2 cells induce alternatively activated macrophages to mediate protection against nematode parasites. Nat Med 2006;12:955–960.
93. Weng M, Huntley D, Huang IF, et al. Alternatively activated macrophages in intestinal helminth infection: effects on concurrent bacterial colitis. J Immunol 2007;179:4721–4731.
94. Hunter MM, Wang A, Parhar KS, et al. In vitro-derived alternatively activated macrophages reduce colonic inflammation in mice. Gastroenterology 2010;138:1395–1405.
95. Goerdt S, Orfanos CE. Other functions, other genes: alternative activation of antigen-presenting cells. Immunity 1999;10:137–142.
96. Herbert DR, Hölscher C, Mohrs M, et al. Alternative macrophage activation is essential for survival during schistosomiasis and downmodulates T helper 1 responses and immunopathology. Immunity 2004;20:623–635.
97. Beer RJ. The relationship between Trichuris trichiura (Linnaeus 1758) of man and Trichuris suis (Schrank 1788) of the pig. Res Vet Sci 1976;20:47–54.
98. Bruschi F, Chiumiento L. Trichinella inflammatory myopathy: host or parasite strategy? Parasit Vectors 2011;4:42.
99. Sun S, Wang X, Wu X, et al. Toll-like receptor activation by helminths or helminth products to alleviate inflammatory bowel disease. Parasit Vectors 2011;4:186.
100. Bai X, Wu X, Wang X, et al. Regulation of cytokine expression in murine macrophages stimulated by excretory/secretory products from Trichinella spiralis in vitro. Mol Cell Biochem 2012;360:79–88.
101. Adisakwattana P, Nuamtanong S, Kusolsuk T, Chairoj M, Yenchitsomanas PT, Chaisri U. Non-encapsulated Trichinella spp., T. papuae, diminishes severity of DSS-induced colitis in mice. Asian Pac J Allergy Immunol 2013;31:106–114.
102. Ruyssers NE, De Winter BY, De Man JG, et al. Therapeutic potential of helminth soluble proteins in TNBS-induced colitis in mice. Inflamm Bowel Dis 2009;15:491–500.
103. Cançado GG, Fiuza JA, de Paiva NC, et al. Hookworm products ameliorate dextran sodium sulfate-induced colitis in BALB/c mice. Inflamm Bowel Dis 2011;17:2275–2286.
104. Sotillo J, Ferreira I, Potriquet J, et al. Changes in protein expression after treatment with Ancylostoma caninum excretory/secretory products in a mouse model of colitis. Sci Rep 2017;7:41883.
105. Wangchuk P, Shepherd C, Constantinoiu C, et al. Hookwormderived metabolites suppress pathology in a mouse model of colitis and inhibit secretion of key inflammatory cytokines in primary human leukocytes. Infect Immun 2019;87e0085118.
106. Shields VE, Cooper J. Use of helminth therapy for management of ulcerative colitis and Crohn’s disease: a systematic review. Parasitology 2022;149:145–154.
107. Sandborn WJ, Elliott DE, Weinstock J, et al. Randomised clinical trial: the safety and tolerability of Trichuris suis ova in patients with Crohn’s disease. Aliment Pharmacol Ther 2013;38:255–263.
108. Garg SK, Croft AM, Bager P. Helminth therapy (worms) for induction of remission in inflammatory bowel disease. Cochrane Database Syst Rev 2014;:2014–CD009400.
109. Schölmerich J, Fellermann K, Seibold FW, et al. A randomised, double-blind, placebo-controlled trial of Trichuris suis ova in active Crohn’s disease. J Crohns Colitis 2017;11:390–399.
110. Huang X, Zeng LR, Chen FS, Zhu JP, Zhu MH. Trichuris suis ova therapy in inflammatory bowel disease: a meta-analysis. Medicine (Baltimore) 2018;97e12087.
111. Summers RW, Elliott DE, Qadir K, Urban JF, Thompson R, Weinstock JV. Trichuris suis seems to be safe and possibly effective in the treatment of inflammatory bowel disease. Am J Gastroenterol 2003;98:2034–2041.
112. Summers RW, Elliott DE, Urban JF, Thompson R, Weinstock JV. Trichuris suis therapy in Crohn’s disease. Gut 2005;54:87–90.
113. Hoshina T, Sakurai T, Ichimura H, et al. Safety and tolerability of medicinal parasite ova (Trichuris suis) in healthy Japanese volunteers: a randomized, double-blind, placebo-controlled trial. Parasitol Int 2021;85:102441.
114. Broadhurst MJ, Leung JM, Kashyap V, et al. IL-22+ CD4+ T cells are associated with therapeutic Trichuris trichiura infection in an ulcerative colitis patient. Sci Transl Med 2010;2:60–ra88.
115. Sarazin A, Dendooven A, Delbeke M, et al. Treatment with P28GST, a schistosome-derived enzyme, after acute colitis induction in mice: decrease of intestinal inflammation associated with a down regulation of Th1/Th17 responses. PLoS One 2018;13e0209681.
116. Hervé M, Angeli V, Pinzar E, et al. Pivotal roles of the parasite PGD2 synthase and of the host D prostanoid receptor 1 in schistosome immune evasion. Eur J Immunol 2003;33:2764–2772.
117. Riveau G, Deplanque D, Remoué F, et al. Safety and immunogenicity of rSh28GST antigen in humans: phase 1 randomized clinical study of a vaccine candidate against urinary schistosomiasis. PLoS Negl Trop Dis 2012;6e1704.
118. Riveau G, Schacht AM, Dompnier JP, et al. Safety and efficacy of the rSh28GST urinary schistosomiasis vaccine: a phase 3 randomized, controlled trial in Senegalese children. PLoS Negl Trop Dis 2018;12e0006968.
119. Capron M, Béghin L, Leclercq C, et al. Safety of P28GST, a protein derived from a schistosome helminth parasite, in patients with Crohn’s disease: a pilot study (ACROHNEM). J Clin Med 2019;9:41.
120. Cook L, Reid KT, Häkkinen E, et al. Induction of stable human FOXP3+ Tregs by a parasite-derived TGF-β mimic. Immunol Cell Biol 2021;99:833–847.
121. Croese J, O’neil J, Masson J, et al. A proof of concept study establishing Necator americanus in Crohn’s patients and reservoir donors. Gut 2006;55:136–137.
122. Reardon C, Sanchez A, Hogaboam CM, McKay DM. Tapeworm infection reduces epithelial ion transport abnormalities in murine dextran sulfate sodium-induced colitis. Infect Immun 2001;69:4417–4423.
123. Moreels TG, Nieuwendijk RJ, De Man JG, et al. Concurrent infection with Schistosoma mansoni attenuates inflammation induced changes in colonic morphology, cytokine levels, and smooth muscle contractility of trinitrobenzene sulphonic acid induced colitis in rats. Gut 2004;53:99–107.
124. Smith P, Mangan NE, Walsh CM, et al. Infection with a helminth parasite prevents experimental colitis via a macrophagemediated mechanism. J Immunol 2007;178:4557–4566.
125. Floudas A, Aviello G, Schwartz C, Jeffery IB, O’Toole PW, Fallon PG. Schistosoma mansoni worm infection regulates the intestinal microbiota and susceptibility to colitis. Infect Immun 2019;87:e00275–19.
126. Pêgo B, Martinusso CA, Bernardazzi C, et al. Schistosoma mansoni coinfection attenuates murine Toxoplasma gondiiinduced Crohn’s-like ileitis by preserving the epithelial barrier and downregulating the inflammatory response. Front Immunol 2019;10:442.
127. Liu Y, Ye Q, Liu YL, Kang J, Chen Y, Dong WG. Schistosoma japonicum attenuates dextran sodium sulfate-induced colitis in mice via reduction of endoplasmic reticulum stress. World J Gastroenterol 2017;23:5700–5712.
128. Zhou H, Zeng X, Sun D, et al. Monosexual cercariae of Schistosoma japonicum infection protects against DSS-induced colitis by shifting the Th1/Th2 balance and modulating the gut microbiota. Front Microbiol 2021;11:606605.
129. Khan WI, Blennerhasset PA, Varghese AK, et al. Intestinal nematode infection ameliorates experimental colitis in mice. Infect Immun 2002;70:5931–5937.
130. Fabre MV, Beiting DP, Bliss SK, Appleton JA. Immunity to Trichinella spiralis muscle infection. Vet Parasitol 2009;159:245–248.
131. Geiger SM, Fujiwara RT, Freitas PA, et al. Excretory-secretory products from hookworm l(3) and adult worms suppress proinflammatory cytokines in infected individuals. J Parasitol Res 2011;2011:512154.
132. Ilic N, Worthington JJ, Gruden-Movsesijan A, Travis MA, Sofronic-Milosavljevic L, Grencis RK. Trichinella spiralis antigens prime mixed Th1/Th2 response but do not induce de novo generation of Foxp3+ T cells in vitro. Parasite Immunol 2011;33:572–582.
133. Xu J, Yu P, Wu L, Liu M, Lu Y. Effect of Trichinella spiralis intervention on TNBS-induced experimental colitis in mice. Immunobiology 2019;224:147–153.
134. Cho MK, Park MK, Kang SA, Choi SH, Ahn SC, Yu HS. Trichinella spiralis infection suppressed gut inflammation with CD4(+)CD25(+)Foxp3(+) T cell recruitment. Korean J Parasitol 2012;50:385–390.
135. Ashour DS, Othman AA, Shareef MM, Gaballah HH, Mayah WW. Interactions between Trichinella spiralis infection and induced colitis in mice. J Helminthol 2014;88:210–218.
136. Yang X, Yang Y, Wang Y, et al. Excretory/secretory products from Trichinella spiralis adult worms ameliorate DSS-induced colitis in mice. PLoS One 2014;9e96454.
137. Wang Z, Hao C, Zhuang Q, et al. Excretory/secretory products from Trichinella spiralis adult worms attenuated DSSinduced colitis in mice by driving PD-1-mediated M2 macrophage polarization. Front Immunol 2020;11:563784.
138. Xue Y, Xu YF, Zhang B, et al. Trichinella spiralis infection ameliorates the severity of Citrobacter rodentium-induced experimental colitis in mice. Exp Parasitol 2022;238:108264.
139. Yang Y, Liu L, Liu X, et al. Extracellular vesicles derived from Trichinella spiralis muscle larvae ameliorate TNBS-induced colitis in mice. Front Immunol 2020;11:1174.
140. Gao X, Yang Y, Liu X, et al. Extracellular vesicles derived from Trichinella spiralis prevent colitis by inhibiting M1 macrophage polarization. Acta Trop 2021;213:105761.
141. Elliott DE, Setiawan T, Metwali A, Blum A, Urban JF, Weinstock JV. Heligmosomoides polygyrus inhibits established colitis in IL-10-deficient mice. Eur J Immunol 2004;34:2690–2698.
142. Elliott DE, Metwali A, Leung J, et al. Colonization with Heligmosomoides polygyrus suppresses mucosal IL-17 production. J Immunol 2008;181:2414–2419.
143. Grainger JR, Smith KA, Hewitson JP, et al. Helminth secretions induce de novo T cell Foxp3 expression and regulatory function through the TGF-β pathway. J Exp Med 2010;207:2331–2341.
144. Rubtsov YP, Rasmussen JP, Chi EY, et al. Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 2008;28:546–558.
145. Ince MN, Elliott DE, Setiawan T, et al. Role of T cell TGF-beta signaling in intestinal cytokine responses and helminthic immune modulation. Eur J Immunol 2009;39:1870–1878.
146. Hang L, Setiawan T, Blum AM, et al. Heligmosomoides polygyrus infection can inhibit colitis through direct interaction with innate immunity. J Immunol 2010;185:3184–3189.
147. Leung J, Hang L, Blum A, Setiawan T, Stoyanoff K, Weinstock J. Heligmosomoides polygyrus abrogates antigen-specific gut injury in a murine model of inflammatory bowel disease. Inflamm Bowel Dis 2012;18:1447–1455.
148. Setiawan T, Metwali A, Blum AM, et al. Heligmosomoides polygyrus promotes regulatory T-cell cytokine production in the murine normal distal intestine. Infect Immun 2007;75:4655–4663.
149. Sutton TL, Zhao A, Madden KB, et al. Anti-Inflammatory mechanisms of enteric Heligmosomoides polygyrus infection against trinitrobenzene sulfonic acid-induced colitis in a murine model. Infect Immun 2008;76:4772–4782.
150. Hang L, Kumar S, Blum AM, Urban JF Jr, Fantini MC, Weinstock JV. Heligmosomoides polygyrus bakeri infection decreases Smad7 expression in intestinal CD4+ T cells, which allows TGF-β to induce IL-10-producing regulatory T cells that block colitis. J Immunol 2019;202:2473–2481.
151. Donskow-Łysoniewska K, Bien J, Brodaczewska K, Krawczak K, Doligalska M. Colitis promotes adaptation of an intestinal nematode: a Heligmosomoides polygyrus mouse model system. PLoS One 2013;8e78034.
152. Blum AM, Hang L, Setiawan T, et al. Heligmosomoides polygyrus bakeri induces tolerogenic dendritic cells that block colitis and prevent antigen-specific gut T cell responses. J Immunol 2012;189:2512–2520.
153. Chen CC, Louie S, McCormick B, Walker WA, Shi HN. Concurrent infection with an intestinal helminth parasite impairs host resistance to enteric Citrobacter rodentium and enhances Citrobacter-induced colitis in mice. Infect Immun 2005;73:5468–5481.
154. Chen CC, Louie S, McCormick BA, Walker WA, Shi HN. Helminth-primed dendritic cells alter the host response to enteric bacterial infection. J Immunol 2006;176:472–483.
155. Leonardi I, Gerstgrasser A, Schmidt TSB, et al. Preventive Trichuris suis ova (TSO) treatment protects immunocompetent rabbits from DSS colitis but may be detrimental under conditions of immunosuppression. Sci Rep 2017;7:16500.
156. Elliott DE, Li J, Blum A, et al. Exposure to schistosome eggs protects mice from TNBS-induced colitis. Am J Physiol Gastrointest Liver Physiol 2003;:284–G385-G391.
157. Ma ZR, Sun X, Zheng WX, et al. Schistosoma japonicum eggs exerts protective effects in an experimental ulcerative colitis model. Biomed Environ Sci 2022;35:1085–1089.
158. Hasby EA, Hasby Saad MA, Shohieb Z, El Noby K. FoxP3+ T regulatory cells and immunomodulation after Schistosoma mansoni egg antigen immunization in experimental model of inflammatory bowel disease. Cell Immunol 2015;295:67–76.
159. Wang L, Yu Z, Wan S, et al. Exosomes derived from dendritic cells treated with Schistosoma japonicum soluble egg antigen attenuate DSS-induced colitis. Front Pharmacol 2017;8:651.
160. Hou X, Zhu F, Zheng W, et al. Protective effect of Schistosoma japonicum eggs on TNBS-induced colitis is associated with regulating Treg/Th17 balance and reprogramming glycolipid metabolism in mice. Front Cell Infect Microbiol 2022;12:1028899.
161. Zhu T, Xue Q, Liu Y, et al. Analysis of intestinal microflora and metabolites from mice with DSS-induced IBD treated with Schistosoma soluble egg antigen. Front Cell Dev Biol 2021;9:777218.
162. Motomura Y, Wang H, Deng Y, El-Sharkawy RT, Verdu EF, Khan WI. Helminth antigen-based strategy to ameliorate inflammation in an experimental model of colitis. Clin Exp Immunol 2009;155:88–95.
163. Li J, Liu X, Ding J, et al. Effect of Trichinella spp. or derived antigens on chemically induced inflammatory bowel disease (IBD) in mouse models: a systematic review and meta-analysis. Int Immunopharmacol 2020;85:106646.
164. Driss V, El Nady M, Delbeke M, et al. The schistosome glutathione S-transferase P28GST, a unique helminth protein, prevents intestinal inflammation in experimental colitis through a Th2-type response with mucosal eosinophils. Mucosal Immunol 2016;9:322–335.
165. Foligné B, Plé C, Titécat M, et al. Contribution of the gut microbiota in P28GST-mediated anti-inflammatory effects: experimental and clinical insights. Cells 2019;8:577.
166. Wang L, Xie H, Xu L, et al. rSj16 protects against DSS-induced colitis by inhibiting the PPAR-α signaling pathway. Theranostics 2017;7:3446–3460.
167. Zhang LC, Wu XY, Yang RB, et al. Recombinant protein Schistosoma japonicum-derived molecule attenuates dextran sulfate sodium-induced colitis by inhibiting miRNA-217-5p to alleviate apoptosis. World J Gastroenterol 2021;27:7982–7994.
168. Malacco NO, Siciliani EA, Madrigal AG, et al. A4 helminthderived metabolites induce a tolerogenic profile in dendritic cells and alleviate experimental colitis. J Can Assoc Gastroenterol 2022;5(Suppl 1):5–6.
169. Kobpornchai P, Flynn RJ, Reamtong O, et al. A novel cystatin derived from Trichinella spiralis suppresses macrophagemediated inflammatory responses. PLoS Negl Trop Dis 2020;14e0008192.
170. Sun XM, Guo K, Hao CY, Zhan B, Huang JJ, Zhu X. Trichinella spiralis excretory-secretory products stimulate host regulatory T cell differentiation through activating dendritic cells. Cells 2019;8:1404.
171. Xu J, Wu L, Yu P, Liu M, Lu Y. Effect of two recombinant Trichinella spiralis serine protease inhibitors on TNBS-induced experimental colitis of mice. Clin Exp Immunol 2018;194:400–413.
172. Xu J, Wu L, Yu P, Sun Y, Lu Y. Effect of T. spiralis serine protease inhibitors on TNBS-induced experimental colitis mediated by macrophages. Sci Rep 2020;10:3147.
173. Xu N, Bai X, Liu Y, et al. The anti-inflammatory immune response in early Trichinella spiralis intestinal infection depends on serine protease inhibitor-mediated alternative activation of macrophages. J Immunol 2021;206:963–977.
174. Xu J, Liu M, Yu P, Wu L, Lu Y. Effect of recombinant Trichinella spiralis cysteine proteinase inhibitor on TNBS-induced experimental inflammatory bowel disease in mice. Int Immunopharmacol 2019;66:28–40.
175. Long SR, Liu RD, Kumar DV, Wang ZQ, Su CW. Immune protection of a helminth protein in the DSS-induced colitis model in mice. Front Immunol 2021;12:664998.
176. Hao C, Wang W, Zhan B, et al. Trichinella spiralis paramyosin induces colonic regulatory T cells to mitigate inflammatory bowel disease. Front Cell Dev Biol 2021;9:695015.
177. Du L, Tang H, Ma Z, et al. The protective effect of the recombinant 53-kDa protein of Trichinella spiralis on experimental colitis in mice. Dig Dis Sci 2011;56:2810–2817.
178. Qu Z, Jin X, Wang Y, et al. Effect of recombinant serine protease from newborn larval stage of Trichinella spiralis on 2,4,6- trinitrobenzene sulfonic acid-induced experimental colitis in mice. Acta Trop 2020;211:105553.
179. Pang J, Ding J, Zhang L, et al. Effect of recombinant serine protease from adult stage of Trichinella spiralis on TNBS-induced experimental colitis in mice. Int Immunopharmacol 2020;86:106699.
180. Smyth DJ, White MPJ, Johnston CJC, et al. Protection from T cell-dependent colitis by the helminth-derived immunomodulatory mimic of transforming growth factor-β, Hp-TGM. Discov Immunol 2023;:2–kyad001.
181. Smyth DJ, Ren B, White MPJ, et al. Oral delivery of a functional algal-expressed TGF-β mimic halts colitis in a murine DSS model. J Biotechnol 2021;340:1–12.
182. Cho MK, Lee CH, Yu HS. Amelioration of intestinal colitis by macrophage migration inhibitory factor isolated from intestinal parasites through toll-like receptor 2. Parasite Immunol 2011;33:265–275.
183. Navarro S, Ferreira I, Loukas A. The hookworm pharmacopoeia for inflammatory diseases. Int J Parasitol 2013;43:225–231.
184. Hotez PJ, Brooker S, Bethony JM, Bottazzi ME, Loukas A, Xiao S. Hookworm infection. N Engl J Med 2004;351:799–807.
185. Khan WI, Collins SM. Immune-mediated alteration in gut physiology and its role in host defence in nematode infection. Parasite Immunol 2004;26:319–326.
186. Mortimer K, Brown A, Feary J, et al. Dose-ranging study for trials of therapeutic infection with Necator americanus in humans. Am J Trop Med Hyg 2006;75:914–920.
187. Kullberg MC, Pearce EJ, Hieny SE, Sher A, Berzofsky JA. Infection with Schistosoma mansoni alters Th1/Th2 cytokine responses to a non-parasite antigen. J Immunol 1992;148:3264–3270.
188. Sacco R, Hagen M, Sandor M, Weinstock JV, Lynch RG. Established T(H1) granulomatous responses induced by active Mycobacterium avium infection switch to T(H2) following challenge with Schistosoma mansoni. Clin Immunol 2002;104:274–281.
189. Hartmann W, Haben I, Fleischer B, Breloer M. Pathogenic nematodes suppress humoral responses to third-party antigens in vivo by IL-10-mediated interference with Th cell function. J Immunol 2011;187:4088–4099.
190. Kradin RL, Badizadegan K, Auluck P, Korzenik J, Lauwers GY. Iatrogenic Trichuris suis infection in a patient with Crohn disease. Arch Pathol Lab Med 2006;130:718–720.
191. Daveson AJ, Jones DM, Gaze S, et al. Effect of hookworm infection on wheat challenge in celiac disease: a randomised double-blinded placebo controlled trial. PLoS One 2011;6e17366.
192. Osada Y, Kanazawa T. Parasitic helminths: new weapons against immunological disorders. J Biomed Biotechnol 2010;2010:743758.
193. Heylen M, Ruyssers NE, De Man JG, et al. Worm proteins of Schistosoma mansoni reduce the severity of experimental chronic colitis in mice by suppressing colonic proinflammatory immune responses. PLoS One 2014;9e110002.
194. Heylen M, Ruyssers NE, Nullens S, et al. Treatment with egg antigens of Schistosoma mansoni ameliorates experimental colitis in mice through a colonic T-cell-dependent mechanism. Inflamm Bowel Dis 2015;21:48–59.
195. Wu Z, Wang L, Tang Y, Sun X. Parasite-derived proteins for the treatment of allergies and autoimmune diseases. Front Microbiol 2017;8:2164.

Article information Continued

Fig. 1.

FigurHelminths in inflammatory bowel disease treatment and their molecular mechanisms. Figure created with BioRender. IL, interleukin; INF-γ, interferon gamma; TNF-α, tumor necrosis factor alpha; TGF-β, transforming growth factor beta; Treg, regulatory T cell.ecaption

Table 1.

Completed Clinical Trials on Helminths in Inflammatory Bowel Disease Therapeutics

Clinical trial identifier Study title No. of participants Country Side effects
NCT01433471 Mucosal immunity of ulcerative colitis patients undergoing therapy with Trichuris suis ova (MUCUS) 4 USA Excess flatulence, diarrhea, perianal pruritis, nausea/vomiting, abdominal pain, fatigue
NCT03565939 Probiotic treatment of ulcerative colitis with Trichuris suis ova (TSO) (PROCTO) 119 Denmark Not disclosed
NCT01576471 Efficacy and safety of Trichuris suis ova (TSO) as compared to placebo (TRUST-I) 250 USA Small intestinal obstruction, Crohn’s disease
NCT01434693 Safety and tolerability of single doses oral CNDO 201 Trichuris suis ova in patients with Crohn’s disease 36 USA Diarrhea, vomiting, abdominal pain, rectal hemorrhage, Crohn’s disease flare, anovulvar fistula
NCT01279577 Trichuris suis ova (TSO) suspension versus placebo in active Crohn’s Disease (TRUST-2) 254 Germany Not disclosed
NCT03079700 Immune modulation from Trichuris suis 1 Denmark Not disclosed
NCT02281916 Safety study of P28GST treatment in Crohn’s disease patients (ACROHNEM) 10 France Not disclosed