Skip to main content
Clinical Microbiology Reviews logoLink to Clinical Microbiology Reviews
. 2023 Nov 8;36(4):e00033-23. doi: 10.1128/cmr.00033-23

Human strongyloidiasis: complexities and pathways forward

Dora Buonfrate 1,✉,#, Richard S Bradbury 2,#, Matthew R Watts 3,#, Zeno Bisoffi 1,#
Editor: Louisa A Messenger4
PMCID: PMC10732074  PMID: 37937980

SUMMARY

Strongyloidiasis is a World Health Organization neglected tropical disease usually caused by Strongyloides stercoralis, a parasitic worm with a complex life cycle. Globally, 300–600 million people are infected through contact with fecally contaminated soil. An autoinfective component of the life cycle can lead to chronic infection that may be asymptomatic or cause long-term symptoms, including malnourishment in children. Low larval output can limit the sensitivity of detection in stool, with serology being effective but less sensitive in immunocompromise. Host immunosuppression can trigger catastrophic, fatal hyperinfection/dissemination, where large numbers of larvae pierce the bowel wall and disseminate throughout the organs. Stable disease is effectively treated by single-dose ivermectin, with disease in immunocompromised patients treated with multiple doses. Strategies for management include raising awareness, clarifying zoonotic potential, the development and use of effective diagnostic tests for epidemiological studies and individual diagnosis, and the implementation of treatment programs with research into therapeutic alternatives and medication safety.

KEYWORDS: Strongyloides, strongyloidiasis, soil-transmitted helminth, neglected tropical diseases

INTRODUCTION

Strongyloidiasis is a human parasitic infection that is most commonly caused by the nematode Strongyloides stercoralis, with other species (Strongyloides fuelleborni fuelleborni and Strongyloides fuelleborni kellyi) being less frequent pathogens (1). Exposure to environments contaminated with feces is the main risk factor for acquisition, and the autoinfective life cycle can result in decades-long chronic infection that, if untreated, can persist in populations long after improvements in sanitation (2). While chronic infections may be asymptomatic, about 50% of cases have long-term symptoms (3). Malnourishment in children is a significant morbidity, and immunosuppressive treatments can lead to a life-threatening flare in disease severity, with the need for screening based on risk factors (48). In terms of prevalence, it is estimated that globally 300 to 600 million people are infected, with most living in disadvantaged areas (9, 10). The World Health Organization (WHO) has consequently recognized strongyloidiasis as a neglected tropical disease (NTD) with defined management targets (11).

In this review, we aim to navigate the complexities of this infection and highlight strategies developed to improve disease management on a population and individual level. We will discuss the unusual biology and host-parasite interaction of S. stercoralis and how this has impacted diagnostics, available epidemiological data, and management of the disease. Box 1 outlines the search strategy and reference selection criteria, and Box 2 lists the key points.

Box 1. Search strategy and selection criteria.

References for this review were identified through searches of PubMed and Embase for articles published up to October 2022, by use of the following key terms: ("Strongyloides stercorali*" OR Strongyloidiasis) OR ("Strongyloides stercorali*" OR Strongyloidiasis AND treatment*). Articles resulting from these searches and relevant references cited in those articles were reviewed to verify their relevance for this work.

Box 2. Strongyloidiasis—Key Points.

  1. The autoinfective cycle leads to chronic infection, unlike the other soil-transmitted helminthiases. If not treated, the infection can persist for life.

  2. Chronic infection may be asymptomatic in about half of cases, and larval output in the stool may be low, reducing detection with stool microscopy.

  3. Immunosuppression can lead to an increase in larval production and result in potentially fatal hyperinfection/dissemination.

  4. Treatment recommendations vary according to disease severity, parasite burden, and host immunity.

  5. Based on current evidence, a single dose of 200 µg/kg ivermectin could be used as an effective treatment for strongyloidiasis in population-based soil-transmitted helminthiases control programs.

BIOLOGY AND PATHOGENESIS

The three life cycles of Strongyloides stercoralis

Strongyloides stercoralis has the most complex life cycle of any human helminth, with the capacity for environmental (homogonic and heterogonic) cycles and an autoinfective cycle (Fig. 1) that include a number of developmental stages (12). Parasitic adults are female and are embedded in the mucosa of the duodenal crypts, sometimes the antrum, and rarely the fundus, of the stomach (13). From animal studies, these parthenogenetic females have between 10 and 12 eggs in utero and are estimated to pass a maximum of 15 eggs per day (14). Eggs in the intestine develop and hatch as L1r (rhabditiform) larvae in the mucosal epithelium or intestinal crypts of Lieberkűhn (2). Most larvae passed in the feces are L1r, with some molting to other stages within the gut, including L3a (autoinfective larvae) or L3i (filariform) larvae (14, 15). If passed onto soil, L1r larvae have been observed to molt to the larger L2r (rhabditiform) stage within 10–15 hours, and some or all transform to infective L3i (filariform) larvae within 30–40 hours of passage (16). Under favorable environmental conditions, these L3i larvae will survive for 3 to 4 weeks until they must find a host or die (17, 18). This is known as the direct (homogonic) life cycle (Fig. 1).

FIG 1.

FIG 1

Life cycles of Strongyloides stercoralis. (a) Homogonic life cycle. The L1r larvae are passed per rectum in feces ① and then mature in the soil to longer L2r ②, followed by molting further to become L3 filariform infective (L3i) larvae ③. These L3i larvae must find a host and infect it via transdermal penetration. These larvae randomly migrate (light blue arrows) or via the cardio-pulmonary-esophagus route (red arrows) to reach the small intestine. Once in the small intestine, they further mature to become new parasitic females. (b) Heterogonic life cycle. The L1r larvae are passed per rectum in feces ① and then mature in the soil to L2r larvae ②, followed by further molting again into morphologically identical but larger L3r larvae ③, where sexual differentiation begins, and then to the final L4r stage ④. The L4r larvae further mature and differentiate into free-living adult male ⑤ and female ⑥ worms, which mate and produce eggs ⑦. These eggs hatch in the soil into a new generation of L1r larvae ⑧, which molt into L2r larvae ⑨ and are then all transformed into L3i larvae ⑩, which must find a host to infect via transdermal penetration or perish. Once within the host, these larvae migrate and develop into new parasitic females as per the homogonic life cycle (blue and red arrows). (c) Autoinfective life cycle. The parasitic female S. stercoralis ① passes eggs ②, which typically develop in the intestinal crypts of Lieberkűhn ③ before hatching into L1 rhabditiform (L1r) larvae ④. These molt within the intestine to L2 rhabditiform (L2r) larvae ⑤ and further molt into L3 filariform autoinfective (L3a) larvae ⑥. These larvae randomly migrate (light blue arrows) or via the cardio-pulmonary-esophagus route (red arrows) to reach the small intestine. Once in the small intestine, they further mature to become new parasitic females.

Depending on environmental conditions and possibly genotype, all or some of the L2r larvae in the environment may choose a different developmental pathway, transforming into an L3r (rhabditiform) life stage and then differentiating into male and female free-living adult worms (2, 16). Free-living adults undergo sexual reproduction (19). Only one generation of the free-living adult stage has been observed, as all offspring must transform into infective L3i larvae and find a host or perish (20). This entire life cycle takes approximately 6 days at 25°C–28°C and is known as the indirect (heterogonic) life cycle (Fig. 1) (21). Environmental L3i larvae from the homogonic or heterogonic cycles usually infect humans via transdermal penetration and actively target hosts, being attracted to heat, sweat, and odorants found in human skin, sweat, and skin microbiota (22). Oral ingestion of larvae may be another potential source of infection, but the significance is uncertain (23). While larvae of S. fuelleborni have been demonstrated in the breastmilk of nursing mothers in Africa, transmammary transmission of S. stercoralis in humans has not been proven (24).

In the third, autoinfective life cycle of S. stercoralis, L3a larvae enter the host tissue through the wall of the lower gut or via the skin, usually in the perianal region (Fig. 1) (14). In the latter case, external subdermal migration may cause the pathognomonic larva currens (see Clinical Features) (25). Untreated strongyloidiasis can be a chronic, lifelong condition, with infection documented 75 years post-exposure (26). It is unclear if spontaneous cure may occur in some patients (27).

Once larvae enter the host, they migrate to the stomach and small intestine via two possible routes. Some will undertake this journey via the blood or lymphatic circulation to be transported into the alveoli of the lungs, thereafter migrating up the respiratory tree to be swallowed and then enter the gut (14). Others may randomly migrate through the organs to reach the small intestine (28). At this site, they will molt twice, develop into parasitic adult females, burrow into the intestinal wall, and produce eggs.

Parasite morphology

Morphological appearance is the classical reference standard for identification and is used to differentiate the various life stages of S. stercoralis (Fig. 2). The adult parasitic female (Fig. 3) has a transparent and thread-like appearance and is large enough to be seen with the naked eye (Table 1). The posterior region is wider than the anterior region, tapering to a cone-shaped, bluntly rounded tail (29). The reflected ovary is straight, and the stoma is hexagonal in shape (19, 29, 30). S. fuelleborni can be differentiated from S. stercoralis as it is longer, with an x-shaped stoma and spiral ovaries (29).

FIG 2.

FIG 2

Major life stages of Strongyloides stercoralis—light micrographs. (a) Rhabditiform larva; (b) autoinfective filariform larva; (c) infective filariform larva [reproduced from reference (12)]; (d) parasitic female; (e) mating free-living female and male.

FIG 3.

FIG 3

Anatomy of Strongyloides stercoralis adult life stages. (A) Parasitic adult; (B) free-living adult female; (C) free-living adult male. An, anus; Eg, eggs; Es, esophagus; Gu, gubernaculum; In, intestine; Nr, nerve ring; Rt, reproductive tubule; Sp, spicule; Te, testis; Ut, uterus; Vo, vaginal opening. (Image licensed under the terms of the Creative Commons Attribution 4.0 International license.)

TABLE 1.

Measurements of the major life stages of Strongyloides stercoralis (19, 3032)

Life stage Anatomical feature (length) Number measured (n) Mean lengthb (range) (µm)
Parasitic female Total body 25 2,415 (2,000–2,800)
Widtha 25 37 (37–40)
Esophagus 25 575 (480–670)
Mouth to vulva 25 1,670 (1,400–1,900)
Anus to tail 25 54 (40–70)
Free-living female Total body 31 1,140 (920–1,700)
Widtha 31 62 (52–85)
Esophagus 31 144 (125–150)
Mouth to vulva 31 580 (470–820)
Anus to tail 31 110 (80–170)
Free-living male Total body 21 900 (810–1,000)
Widtha 21 43 (40–50)
Esophagus 21 118 (110–125)
Anus to tail 21 74 (55–95)
Spicule 21 37 (35–40)
Rhabditiform (L1) larvae Total body ns 210 (180–240)
Widtha ns 15 (ns)
Esophagus ns ns (33–38)
Infective filariform (L3i) larvae Total body 31 563 (490–630)
Widtha 31 15.8 (15–16)
Esophagus 31 242 (220–270)
Anus to tail 31 69 (60–80)
Autoinfective filariform (L3a) larvae Total body ns 269 (234–317)
Widtha ns 11 (10–13)
Esophagus ns ns (39–73)
Eggs Diameter ns ns (74–104)
a

At widest point.

b

ns, number not specified.

S. stercoralis rhabditiform larvae have a rhabditoid esophagus with a single posterior bulb (Fig. 4) (19). The four stages are all slug-like in appearance and morphologically similar, differentiated primarily by an increase in size following each molt (Fig. 5), while other subtle morphological changes do occur (19). The rhabditiform-stage larvae of all Strongyloides spp. are essentially indistinguishable, but in human feces, care must be taken to differentiate them from other superficially similar larvae such as those of the hookworms, Oesophagostomum spp., Rhabditis spp., Ternidens deminutus, and Trichostrongylus spp. (Fig. 6) (33). Distinguishing features of Strongyloides larvae are the very short buccal cavity, straight gut, and lateral rhomboid genital primordium halfway down the larval body (33).

FIG 4.

FIG 4

Anatomy of Strongyloides stercoralis larval life stages. (A) Larvated egg; (B) L1 rhabditiform larva; (C) L3 autoinfective filariform larvae; (D) L3 infective filariform larvae. An, anus; Bc, buccal cavity; Eg, egg; Es/In, esophageal-intestinal junction; Gp, genital primordium; Nr, nerve ring. (Image licensed under the terms of the Creative Commons Attribution 4.0 International license.)

FIG 5.

FIG 5

Development of the four stages of rhabditiform larvae of Strongyloides stercoralis in the heterogonic (free-living) life cycle. (Adapted from original drawings provided by Emeritus Professor Richard “Rick” Speare.)

FIG 6.

FIG 6

Morphological comparison of rhabditiform larvae that may be recovered from human fecal samples. (Reproduced from reference 31.)

Two filariform stages of Strongyloides have been reported. The more commonly observed L3i stage is found in both external life cycles. These larvae are quite distinct from other filariform larvae that may be found in human stool (Fig. 7). Differentiating features are the smaller size of Strongyloides filariform larvae and their far more slender and serpentine appearance. The intestine-to-esophagus ratio is 1:1, whereas it is 1:2 in other human-infecting filariform larvae (Fig. 7). The intestine is straight, and the tail tapers to a point with a distinct notch. The L3a stage is very similar in appearance to L3i larvae; it is shorter and has a more bluntly rounded anterior than L3i (31, 34).

FIG 7.

FIG 7

Morphological comparison of filariform larvae that may be cultured from human fecal samples. (Reproduced from reference 31.)

The free-living female stage (Fig. 3) has a rhabditoid esophagus, a cylindrical procorpus, a narrow isthmus, and a large posterior bulb (19). These stages are relatively large (Table 1), allowing detection with the naked eye on agar plate cultures (Fig. 7). The females are spindle-shaped and have a tapering anterior and posterior, with marked central widening to allow for the egg-filled uterus, which takes up most of the body. The tail gently tapers to a point (30).

The anterior of the free-living male is essentially the same as the female (Fig. 3), but males are much shorter and smaller (Table 1). The posterior curls more than 90° and rapidly tapers to a point. The reproductive system is a straight tube; at the anterior, there is a single testis, which transforms without clear demarcation into the vas deferens and seminal vesicle. There are two small, sickle-shaped spicules and a single gubernaculum (19, 30, 32).

Unlike S. fuelleborni, the eggs of S. stercoralis (Fig. 4) are not passed in feces, except in cases of hyperinfection or systemic disease (19). Eggs are produced by both adult parasitic females and free-living females, and the latter may be observed on agar plate cultures if they are incubated for sufficient time (~5 days). Strongyloides stercoralis eggs are thin-shelled, round to oval, and contain partially developed L1r larvae on passage (19).

Innate and adaptive immune responses

Most information about the pathogenesis of Strongyloides infection comes from studies carried out in gerbils (infected with S. stercoralis) or rats (infected with either Strongyloides ratti or Strongyloides venezuelensis), which do not develop autoinfection unless immunosuppression is induced (35).

In line with other helminth infections, eosinophils play a prominent role in the innate immune response to S. stercoralis. This explains why eosinophilia is frequently observed in people with chronic infection, while eosinophil depletion is common in individuals with hyperinfection and associated immunocompromise (36). As a first line of defense against the parasite, eosinophils, along with neutrophils and complement activation, entrap and kill larvae intercepted in the skin through the release of toxic molecules (innate immunity) (35). In addition, eosinophils elicit the adaptive immune response by acting as antigen-presenting cells, thus inducing a T-helper 2 response with the consequent production of cytokines and specific antibodies (both IgM and IgG) against the worm (35). The cytokines interleukin (IL)-4 and IL-5 are relevant for modulating the cellular and humoral responses, respectively. IL-4 induces the development of alternatively activated macrophages, which is associated with larval killing, whereas IL-5 is required to stimulate B-cells to produce IgM (35).

Host-parasite balance, the autoinfective cycle, and hyperinfection/dissemination

Helminths have the capacity to dampen the immune response, and in immunocompetent individuals, the interaction between Strongyloides and the host reaches a steady state that leads to the persistence of the infection, with sometimes nil, relatively mild, or transient clinical manifestations, where eosinophilia may be absent or the only diagnostic clue (1, 37, 38). Animal studies have indicated that in chronic infections, many adult females enter a life stage where they cease producing eggs (39). This would explain the low larval output seen in chronic infections, which does not necessarily reflect low numbers of parasitic adults in the intestine. The number of parasitic females in the host and their fertility are controlled by poorly understood factors involving interactions with host immunity and possibly interactions between crowded adult parasites via chemical signaling (14, 39, 40). If host immunity is suppressed, the proportion of egg-producing females and L3a larvae can increase, resulting in an autoinfective burst (39). This accelerated autoinfection leads to an amplification of the parasite burden that can result in the potentially fatal consequence of hyperinfection (7). This severe illness is characterized as disseminated strongyloidiasis since large numbers of larvae can be readily located throughout the internal organs of the body, with adult females and ova occasionally found in the lung (7, 41).

Hyperinfection/disseminated disease is associated with specific types of immunosuppression, most commonly corticosteroid exposure, transplantation with the associated immunosuppression, chemotherapy, malignancy, including some leukemias and lymphomas, human T-lymphotropic virus type 1 (HTLV-1) infection, diabetes mellitus, liver cirrhosis, alcoholism, and biological and small molecule immune-modulating therapies, e.g., tumor necrosis factor-α inhibitors (68, 4245). Importantly, corticosteroids act directly upon the L1 larvae in vivo, accelerating ecdysis (molting) and transformation into L3a, increasing autoinfection (4650). There is some debate about the impact of HIV infection on strongyloidiasis; while initially disseminated strongyloidiasis was considered an AIDS-defining illness, it was then removed from the list (51). The immunological impairment caused by HIV does not cause a depletion of T-helper 2 lymphocytes, which induce the activation of eosinophils and the production of IgE and cytokines (e.g., IL-4, IL-5), essential for immunity to helminth infections (52). Conversely, human T-lymphotropic virus type 1 (HTLV-1) decreases the type 2 response and reduces immunity to helminth infections (53). This virus is clearly associated with a high risk of hyperinfection/disseminated strongyloidiasis [odds ratio = 59.9, 95% confidence interval (CI) 18.1–199] and a poor treatment response (53). Moreover, Strongyloides and HTLV-1 co-infection seem to worsen the viral infection (53). In tuberculosis, S. stercoralis co-infection leads to significantly lower levels of type 1 cytokines and higher levels of type 2 cytokines (54, 55). This immune modulation was associated with higher bacillary numbers in cases of tuberculous lymphadenitis (55). Interestingly, in tuberculous meningitis, the reduction in pro-inflammatory cytokines (interferon-γ, IL-2, tumor necrosis factor-α) associated with strongyloidiasis led to improved clinical outcomes (56). Finally, co-infection with strongyloidiasis and severe acute respiratory syndrome coronavirus 2 was relevant due to the widespread use of corticosteroids and other immune modulatory therapies to manage coronavirus disease 2019 (5759). This highlighted the importance of systematic screening for strongyloidiasis in patients with epidemiological risk factors and immunocompromise.

Is Strongyloides stercoralis a zoonosis?

S. stercoralis can infect other mammals, including dogs, cats, and nonhuman primates (6063). The cox1 lineage A genotype has been reported in all hosts, while the lineage B genotype of S. stercoralis appears restricted to dogs (6063). It remains unclear at this time if S. stercoralis may be acquired by humans as a zoonosis from animals or by companion animals as a reverse zoonosis from humans. While similarities in genotypes indicate cross-species transmission (64, 65), the sparsity of reports of human cases acquired from dogs (66), the absence of reported cross-infections between dogs and owners, and the difficulty often encountered in establishing lasting cross-species infection under experimental conditions without immunosuppression indicate otherwise (16, 18, 6774). Further investigation using long-read sequencing of dog and human isolates taken from the same community may clarify the question of zoonotic transmission (64).

Omics and bioassays

Advanced genomic, transcriptomic, and proteomic approaches, as well as bioassays, often previously trialed in the similar nematode Caenorhabditis elegans, have been applied to S. stercoralis, S. ratti, and other Strongyloides spp. (75, 76). This knowledge about the fundamental biology of Strongyloides may be translated into improved diagnostics, control strategies, and therapeutics.

The S. stercoralis genome is 42.6 MB in length and AT-rich, with a GC content of 22% (77). Multiple whole genome sequences of isolates from dogs and humans from various geographical locations are now available. A comparison of isolates from humans in Myanmar and Japan with the reference strain from a dog in the USA demonstrated significant population variation, especially in samples from Myanmar (78). The publication of high-quality genomes of several Strongyloides species, along with the genomes of related parasitic and free-living nematode genera, has allowed investigation into the genetic basis of parasitism (76). This work included the determination of the transcriptomes of parasitic and free-living females and the L3i stages of S. stercoralis (76). Notably, excretory/secretory (E/S) proteins, which are upregulated in the parasitic female stages of S. ratti, are likely essential to parasitism and may present effective targets for drug therapies and vaccines (76, 77). The proteomes of multiple L3i stage larvae have been published by several authors, providing insights into biology and novel targets for both serological markers and stool antigen detection assays (7983).

Bioassays have shown that in free-living S. stercoralis, the nuclear hormone receptor DAF-12 is required for the developmental arrest that results in L3i larvae rather than adult worms (75, 84). This was also dependent on a co-activator, DAF-12-interacting protein 1 (84). Such insights might be used to develop environmental control strategies or treatments that slow or arrest human infection (75). Investigations into the effects of temperature, metalloproteases, and other interventions on the skin penetration rate of S. ratti may translate into improved post-exposure prophylaxis protocols for laboratory staff working with S. stercoralis L3i (75). Finally, data from bioassays evaluating Strongyloides infective processes and circadian rhythms may lead to improvements in the timing and dosage of anthelmintic therapies (75).

EPIDEMIOLOGY

In the past, many field studies with the aim of assessing the prevalence of soil-transmitted helminthiases (STH) were based on Kato-Katz and/or other diagnostic methods that have poor sensitivity for the detection of S. stercoralis (see Diagnostics). This impacted attempts to assess the global prevalence, which was previously largely underestimated, further contributing to the neglect of the infection (85). Recent studies have revised the estimates of prevalence. They either compared the distribution of S. stercoralis with that of hookworm, or used a statistical modeling approach that, based on the characteristics of the settings and populations where the infection was found, enabled the extension of estimates to areas with unknown data (9, 10). According to these novel estimates, the global prevalence should be between 386 (95% CI 324–449) and 613.9 (95% CI 313.1–910.1) million people, significantly higher than the previous estimate of 30–100 million individuals at risk of infection (9, 10, 86). Given the transmission route, the increased risk of infection in rural areas is not surprising, where there is often a lack of access to proper latrines and where barefoot walking is common (9, 8792). Moreover, prevalence tends to be higher in adults than in children, reflecting the chronic nature of infection and increased opportunities for exposure over time (9, 88, 9092). Other risk factors for acquisition include institutionalization, household contact, laboratory exposure, and solid organ donor-derived infection, e.g., following kidney or kidney-pancreas transplantation (1, 93, 94).

Although the large majority of infections occur in the poorest areas of Southeast Asia, Africa, and the Western Pacific Region, localized foci of low-level transmission have been identified in high-income countries and areas of temperate climate (9). In the United States of America (USA), Strongyloides transmission was documented mainly in southern states and in rural Appalachia (95). Infection might be endemic in 10 additional states of the USA, as indicated by a study based on a species distribution model (95). In nonendemic areas, infection was found in migrants from areas of endemicity (96). In any case, strongyloidiasis tends to be more frequently diagnosed in people living in disadvantaged conditions, as pointed out by an analysis of hospitalizations throughout the USA, which found an increased risk of infection associated with low socioeconomic status, along with nonwhite ethnicity, male sex, and older age (96).

In Europe, cases of recent local transmission are occasionally reported, mostly in countries along the Mediterranean basin and in Eastern states (97, 98). In fact, strongyloidiasis is generally diagnosed in immigrants from endemic areas and, in some southern countries (epidemiological studies were mostly carried out in Italy and Spain), in elderly people who never traveled abroad, frequently had a history of agricultural work, and who presumably acquired the infection in their youth (99103). Similarly, in Japan, the infection is diagnosed mostly in older people, and local transmission is probably very rare or absent as a result of improved sanitation and the ban on using human feces as fertilizer (104, 105). In Northern and Central Australia, ongoing local transmission is still an issue in remote Aboriginal communities, with up to 59% of people tested with serology found positive in some parts of the Northern Territory (106108). Like in the USA and Europe, cases in metropolitan areas are mostly due to immigration and travel to endemic areas (108).

Traveling abroad does not seem to represent a major risk of infection, particularly when trips are of short duration. Extended stays lead to an increased cumulative risk of infection (109, 110). Conversely, migrants arriving in areas of low endemicity from highly endemic areas have a greater risk of infection, which should be considered for screening policies (101, 111, 112). In a systematic review with meta-analysis, the pooled prevalence of strongyloidiasis in migrants was estimated at 12.2% (95% CI 9.0–15.9) with serology and 1.8% (95% CI 1.2–2.6%; 98%) with stool tests (112). The seroprevalence was higher in people originating from East Asia and the Pacific (17.3%, 95% CI 4.1–37.0), followed by sub-Saharan Africa (14.6%, 95% CI 7.1–24.2), then Latin America and the Caribbean (11.4%, 95% CI 7.8–15.7).

CLINICAL FEATURES

Acute infection has been reported rarely in travelers to endemic areas (113). Skin rash may be seen at the site of skin penetration by the infective larvae, and urticaria, pruritus, and cough (Loëffler syndrome) may appear as early as 7 to 10 days after skin penetration of larvae, while gastrointestinal symptoms (diarrhea, abdominal pain) occur after 3 to 4 weeks, when larvae start to be shed in feces (114, 115). Eosinophilia can be a diagnostic clue in the earlier phases, though it is frequently present in other parasitic infections (114).

In chronic strongyloidiasis, according to a systematic review with meta-analysis, about half of the populations surveyed were asymptomatic, and about half were symptomatic, with 50.4% (95% CI 47.6–53.1) reporting at least one symptom (3). In those infected, the rate of eosinophilia was 70% (3). The remaining 30% with no symptoms and no indicative eosinophilia are reliant on the completeness of the medical history and clinician awareness to ensure screening prior to immunosuppressive therapies (116).

Where symptoms are present, they most frequently involve the skin, intestine, and respiratory tract and can range from mild to severe (3). Some studies have also reported systemic manifestations, including malnourishment and stunting in children (4, 5). Larva currens is a pathognomonic, subcutaneous larval migration of 5–10 cm/hour that is visible as urticarial serpiginous tracks on the skin of the buttocks, thighs, or abdomen (25, 117). Other symptoms are nonspecific, with features in common with other intestinal infections (e.g., diarrhea, abdominal pain, pruritus) or chronic respiratory conditions (e.g., asthma-like features, cough) (118). In a systematic review with meta-analysis, urticaria showed a clear association with strongyloidiasis, being reported by 28% of infected individuals, while diarrhea, abdominal pain, or respiratory symptoms showed a weaker association (118). In general, data from studies adequately designed with proper diagnostic tests are scarce, and this has limited the evaluation of the burden of strongyloidiasis (118).

As discussed in the Biology and Pathogenesis, immunocompromise can trigger the development of life-threatening strongyloidiasis with a dramatic increase in larval load (7). In this circumstance, hyperinfection and dissemination are probably better identified as different descriptions of the same clinical condition, characterized by severe symptoms (e.g., paralytic ileus, respiratory impairment, altered state of consciousness), sometimes associated with sepsis and/or meningitis by intestinal bacteria. Of note, in a systematic review of the literature describing cases of Streptococcus gallolyticus meningitis, a large proportion of cases retrieved (41%) were associated with S. stercoralis infection (119).

DIAGNOSTICS

Body fluid microscopy and histopathology

While stool and serum are the usual diagnostic specimens, Strongyloides larvae may be identified on microscopy of respiratory secretions, ascitic fluid, cerebrospinal fluid (CSF), and rarely the urine, and are typically associated with immunosuppression and dissemination/hyperinfection (7, 41, 120, 121). There are case reports of larvae being identified in cervical smears and a single report of male infertility associated with larvae in the semen (41, 122, 123). Where there is a higher burden of disease, parasites may also be visualized in histopathological specimens from tissue biopsies or at autopsy (41).

In the small intestine (Fig. 8), disease may range from congestion and mild inflammation to severe ulcerative enteritis, where ulcers may extend to the muscularis layer, the epithelium is atrophic, and large numbers of parasites are seen, including invasive filariform larvae (41). Granulation tissue and fibrosis may be present, and there may be damage to the myenteric plexus, increasing transit time and contributing to ileus (41). In the large intestine, there may be significant edema of the lamina propria, associated with penetration of the larvae, patchy ulceration, and granuloma formation. Eosinophils may not be prominent unless there are dead larvae, and fibrosis may develop from recurrent inflammation (41). Strongyloides may be present in the stomach and lower esophagus, with a range from an absent or mild reaction to widespread damage (41). In the liver, there may be granulomatous hepatitis (124).

FIG 8.

FIG 8

Various life stages of Strongyloides stercoralis in the histological section of the duodenum. Active chronic duodenitis. (a) eggs (Eg); (b) L1 rhabditiform larvae (Rh) developing within eggs; (c) parasitic adult female, sectioned at the midline and demonstrating the two branches of the reflected uterus (Ut) and the single intestinal tube (In); all 400× magnification; (d) larvated eggs (white darts) and parasitic females in transverse (black darts) and longitudinal (arrows) section; 100× magnification. (Photographs courtesy of K. Hart, reproduced with permission.)

Although strongyloidiasis is frequently associated with hemorrhagic bronchopneumonia in the lungs, relatively few larvae are present on histopathology (41, 125). Larvae may be found in the mesenteric arteries and the portal system and can invade the myocardium (41, 126). Bacterial meningitis may indicate larval migration to the central nervous system, and while infiltration of the brain is described, it is less common (41, 127). When larva currens is present, larvae can be identified on skin biopsy, and in hyperinfection, larvae may also be found in purpuric lesions (41, 128, 129).

Conventional stool-based tests

The diagnosis of S. stercoralis infection in the stool can be complicated by variable and low larval output in chronic infections (130, 131). This affects the sensitivity of conventional stool-based tests, where morphological identification is used as a reference standard for molecular and serological diagnostics (33). An understanding of the limitations of these reference tests leads to a clearer interpretation of individual patient results, assay validations, epidemiological studies, and drug trials (33, 131, 132).

Routine direct-smear fecal microscopy and Kato-Katz microscopy have a low sensitivity of 0%–18% (Fig. 9) (33). While there are only limited data on the efficacy of mini-FLOTAC for the detection of S. stercoralis larvae, it suggests a sensitivity equal to, or below, direct smear microscopy (133136). Filtration-based stool concentration can also have a low sensitivity, presumably due to larvae retained by the filter (33). Concentration by formalin-ether sedimentation or subsequent modifications of the technique have published sensitivities ranging from 6% to 60% when compared to more sensitive fecal diagnostic methods (Fig. 9) (33). In the hands of an experienced morphologist, microscopy is highly specific, but poorly trained operators may confuse other rhabditiform larvae found infecting human feces (see Parasite Morphology) (33).

FIG 9.

FIG 9

Sensitivity ranges of Strongyloides diagnostic tests. Limited studies have indicated that mini-FLOTAC and filtration stool concentration methods have sensitivities similar to direct smear. Formalin-ether concentration includes modified methods. Baermann sedimentation and agar plate culture require live larvae. PCR sensitivity is dependent on the amount of stool extracted and the effectiveness of the stool extraction method. Serology false-negative results may occur in immunosuppression/hyperinfection.

Methods that utilize the motility of live S. stercoralis larvae to separate them from larger volumes of feces are among the most sensitive approaches for the diagnosis of strongyloidiasis. Baermann sedimentation and agar plate culture (APC) have reported sensitivities of between 40%–80% and 60%–98%, respectively, when compared to composite reference standards (Fig. 9) (33). Like microscopy, these methods are highly specific in the hands of well-trained operators. Several variations of each technique have been proposed to increase larval yield (137143). The analysis of multiple fecal collections further increases sensitivity (144). Ideally, three stool collections are recommended, and these need to be transported at ambient temperature as refrigeration lowers the percentage of viable larvae (33, 145, 146). Larvae should be identified morphologically in these assays to confirm their identity as S. stercoralis.

As these approaches yield highly motile, infective L3i, they present a significant laboratory infection risk (Video S1). All operators should wear two sets of gloves and cover exposed arms and legs when performing these assays (147). With the absence of data on the larvicidal effects of disinfectants on S. stercoralis L3i, it is recommended that larvae at least be treated with 70% ethanol or formalin prior to microscopy (147). Should an operator be exposed to L3i, the affected area should be flooded with 1% povidine iodine (147). Contaminated equipment should be cleaned with 70% ethanol and allowed to dry (147).

Molecular testing

Molecular approaches have a significant advantage over tests based on larval separation (e.g., APC and Baermann sedimentation) since they can be performed on preserved fecal samples without the need for viable larvae (33). This allows transportation and long-term storage of samples where required (33). Both PCR and loop-mediated isothermal amplification (LAMP) assays for the detection of S. stercoralis have been described (33). The most widely used and well-validated assay, a TaqMan real-time PCR developed by Verweij and colleagues, has a sensitivity of 30.9%–100% and a specificity of 65.4%–98.9% when compared to composite reference standards, including larval separation techniques (Fig. 9) (33, 148). This assay is the only S. stercoralis real-time PCR to have undergone thorough clinical validation in an endemic population setting (33, 149). The reported analytical sensitivity and specificity claimed for other assays may not reflect their efficacy when applied in an endemic context. Furthermore, variations in preanalytical variables, such as extraction methods, will affect the sensitivity of any chosen molecular test (33). In addition, almost all molecular assays for S. stercoralis will amplify other members of the genus, including other Strongyloides species infecting humans (viz. S. fuelleborni) and animals (61, 150). To provide ongoing quality assurance for nucleic acid tests, an external quality assessment scheme for S. stercoralis and other human helminths has recently been established in the Netherlands, with another program planned for introduction in Australia (151153).

Next-generation sequencing and metagenomic analysis

Next-generation sequencing (NGS) with metagenomic analysis has been used to investigate the effects of strongyloidiasis on the gut microbiome and as a diagnostic tool (154, 155). Currently, the routine diagnosis of strongyloidiasis from stool based on NGS is limited by cost, laboratory infrastructure, the availability of sequencing instrumentation, and the need to manipulate large bioinformatic data sets, including DNA sequences from the host and microbiota (156, 157). Where the technology is available, metagenomic analysis may be useful for specimens from sterile sites, e.g., CSF, where Strongyloides meningitis may not be suspected or made with microscopy, culture, or routine molecular tests (155).

Serological testing

Serological assays have the advantage of being fast, relatively reproducible, and able to be performed in batches; using widely available technology platforms; and being able to be performed on samples such as serum or finger prick blood (158). The time to seroconversion in humans is unknown, but in animal studies, IgG antibodies were detectable within 2 weeks of infection and reached their highest titer 6 weeks post-infection (74). Sero-reversion following treatment (IgG antibodies dropping to a negative level titer) occurs within 6 months to 2 years after treatment (159), and while results may vary between assays, it does indicate that seropositivity represents a current or relatively recent infection. Whether residual seropositivity indicates treatment failure or residual antibody titers remains unclear.

Various commercial crude larval and recombinant antigen assays are available, with reported sensitivities of between 75.4% and 94.7% and specificities of between 42.1% and 95.4% when compared to parasitological tests (Fig. 9) (33, 160164). A bead-based serological assay using the NIE recombinant antigen had a sensitivity of 93% and a specificity of 95% compared to a microscopy-based reference standard, and this technology has been applied to a multiplex assay to detect soil-transmitted helminths and Plasmodium falciparum (165, 166). Various rapid immunochromatographic serological assays have been developed using crude larval extracts or recombinant antigens (NIE and the S. stercoralis immunoreactive antigen, SsIR) (167169). Only one of these has undergone clinical evaluation using frozen sera in a retrospective study (sensitivity 82.4%, specificity 73.8%, with a fecal test reference standard) and whole blood in a prospective study (sensitivity 79.4%, specificity 93.6%, with a Bayesian latent class analysis) (167, 170). An immunoblot and an enzyme-linked immunosorbent assay(ELISA) combining the recombinant antigens NIE and SsIR have recently been described, and diagnostic evaluations in a clinical setting have been performed for the latter only (161, 170, 171). A prospective evaluation of the NIE/SsIR ELISA assay performed in the field using dried blood spots was found to have a sensitivity of 83.5% (95% CI 73.8–91.8) and a specificity of 91.7% (95% CI 89.6–93.8) (170).

A coproantigen ELISA employing antibodies targeting the S. ratti E/S antigen has been developed but has not reached commercialization (172).

Diagnostic approach

The choice of diagnostic test will depend on the cohort to be tested (population-based versus individual), sensitivity and specificity, and laboratory capacity (Table 2) (33). Due to its convenient methodology and high sensitivity, serology is suited to population screening for strongyloidiasis, with a subgroup having stool testing for quality assurance purposes (33, 158). In routine laboratory use, serology also has a high diagnostic yield compared to stool-based tests, likely due to preanalytical issues affecting the latter, e.g., sampling error from low parasite numbers, larval mortality, and limitations of nucleic acid extraction methods (33, 173). However, not all people infected with S. stercoralis will have positive serology. Immunocompromised patients, at risk of severe disease, may be seronegative while fecal tests are positive (162, 174176). For screening and diagnosis in asymptomatic individuals with no current or planned immunocompromise, serology testing alone would generally be adequate (33). If a patient is symptomatic or is being screened due to immunosuppression, and to maximize the opportunity for diagnosis, serological testing and testing of multiple stool samples would be recommended for diagnosis and monitoring of treatment efficacy (33, 132, 177). A high-sensitivity conventional method for stool testing, such as APC or Baermann sedimentation, can be performed where molecular testing is not available (33). In laboratories where molecular assays are used, separation tests may still play a role, as they can be positive when molecular tests are negative, possibly due to larger sample volumes (33).

TABLE 2.

Population-based and individual disease control

Disease control Population-based Individual
Immune status Immunocompetent Immunocompetent Immunocompromiseda
Diagnostic tests Serology + subset for stool testingb and follow-up testing to evaluate program efficacy Serology + stool testing if symptoms are present Serology + stool testing repeat for test of cure
Treatment Ivermectin single dose Ivermectin single dose or albendazolec Multiple doses of ivermectin duration based on disease severity/larval burden, consider adding albendazole if there is severe infection
a

Or imminent immunosuppression.

b

Quality assurance for serology and test of cure.

c

If ivermectin contraindicated.

TREATMENT

Treatment for strongyloidiasis can be instituted on the basis of mass drug administration, for control in a population assumed to be immunocompetent, or in the context of individual therapy, where host immunity and illness severity are considered (Table 2) (132). In a healthy population with uncomplicated disease, first-line treatment is ivermectin, and randomized controlled trials have demonstrated that a single dose of 200 µg/kg is as effective as multiple doses (178180). The efficacy of a single dose ranged from 76% to 100%, depending on the study setting and methods for assessing cure (178, 179). Although based on expert opinion, in cases of immunocompromise and particularly hyperinfection/dissemination, repeated doses of ivermectin are recommended where the length of treatment is guided by parasitological clearance (1, 132). Subcutaneous administration of parenteral ivermectin has been reported in severe cases with major obstacles to oral intake, e.g., in cases of unconsciousness or paralytic ileus (181). While parenteral formulations of the drug are only approved for veterinary usage, they have been effective, on occasion, for the management of severe disease (132, 181).

Ivermectin targets the glutamate-gated chloride channels of the helminths, causing their paralysis and consequent death (182). In humans, it induces the release of gamma-aminobutyric acid from neurons, which are protected from the drug by the blood-brain barrier (182). Hence, ivermectin has a high tolerability profile and can be administered during breastfeeding (the WHO recommends excluding women during the first week of lactation only) and in children over 5 years old/15 kg (183). In the second and third trimesters of pregnancy, the drug is presumably safe, but due to the lack of evidence of high quality, it cannot be recommended (184). A thorough risk-benefit evaluation should be done for single cases when the drug is not approved.

Albendazole has lower efficacy than ivermectin, so usage would usually be limited to treatment where ivermectin is not available or not currently recommended, e.g., in the second or third trimesters of pregnancy and in children >1 year old, with post-treatment follow-up to confirm cure (132, 180). The recommended duration of albendazole therapy for uncomplicated strongyloidiasis is 3 or 7 days, while single dosing as part of a helminth control strategy demonstrated no significant reduction in infection (132, 146). Longer courses of albendazole have been used in combination with ivermectin to manage hyperinfection, where benefits may include the treatment of larvae disseminated to the central nervous system (132, 185, 186). Thiabendazole has demonstrated high efficacy for the treatment of strongyloidiasis, similar to ivermectin, but it is generally not recommended due to its low tolerability (180).

A possible alternative treatment in the future may be moxidectin, a macrocyclic lactone that acts on glutamate-gated chloride channels like ivermectin (187). The main advantage of moxidectin is its fixed-dose (rather than weight-based) administration, which would be useful for control programs in endemic areas based on preventive chemotherapy (187). Another benefit is that the resistance patterns of the two drugs are different; hence, moxidectin might be used in the event of the emergence of resistance to ivermectin (188). However, approval for use in individuals under 12 years old is still needed, and the drug still needs to demonstrate at least noninferiority to ivermectin for the treatment of strongyloidiasis (187).

CONCLUSIONS

In this review, we illustrate the main characteristics of this unique parasite and summarize management strategies, including opportunities for future research (Table 3). Some of the main consequences for individual and public health are due to the peculiar and complex nature of the life cycle. In the absence of adequate treatment, the continuous process of autoinfection guarantees a very long presence in the individual host and at the population level.

TABLE 3.

Characteristics of strongyloidiasis and strategies to improve disease management

Characteristic Strategy
Potential zoonotic transmission Studies that characterize subpopulations of Strongyloides stercoralis in humans and animal hosts
Extent of disease distribution and population infected Epidemiological studies and mapping that take into consideration the limitations of diagnostic tests—overestimates vs underestimates
Development of disease registries
Limited awareness about chronic asymptomatic infections or nonspecific symptoms Health care worker education regarding clinical features, risk factors for acquisition, and the autoinfective cycle
Lack of a single, high-sensitivity reference standard diagnostic test Use of composite reference standards and/or latent class analysis for test validation and strategic use of the most effective diagnostics, with awareness of test limitations
Optimal diagnostics for use in mass drug administration programs versus individual diagnosis Use of simple testing strategies (e.g., serology) for mass screening and more intensive strategies (serology and stool-based testing) for test-of-cure and individual diagnostics
Treatment of a spectrum of disease severity Effective regimens for mass drug administration: chronic asymptomatic infection in immunocompetent persons versus individualized therapy based on disease burden and host immune status
Treatment of special groups Further research into safe and effective regimens for pregnant/breastfeeding women and young children

While the prevalence of the other STH (Ascaris lumbricoides, Trichuris trichiura, and hookworm) is declining in several endemic countries due to community treatment and possibly improvements in hygiene and sanitation, for Strongyloides, the current community treatment programs (largely based on regimens of albendazole that are ineffective for this parasite) are inadequate. Although improved hygiene and sanitation would potentially impact transmission, it would not prevent the long-term persistence of the parasite in the infected population, potentially mirroring what happened in temperate, high-income countries such as Italy, Spain, or Portugal (99, 101103, 142). The fact that this parasite has now been formally included in the WHO control targets is good news, and the inclusion of ivermectin in STH control programs will have an impact on global prevalence (11, 189). The first WHO guidelines for the control of strongyloidiasis are currently under development, and they will include specific recommendations about the target population (school-age children or the community), diagnostic tools to estimate local prevalence, and thresholds of prevalence above which intervention is indicated.

The real clinical burden of strongyloidiasis is still poorly understood because most reported case series did not have a control group, although this gap has been partially filled by recent studies (5, 118). In particular, the true burden of severe strongyloidiasis and consequent mortality is still a gray area. Published cases are the tip of the iceberg of those diagnosed, and the latter are a tiny fraction of the real ones, as the severe clinical presentations are far from specific and the underlying infection is unlikely to be diagnosed if not specifically suspected (42).

Moreover, although the risk factors for severe complications are well known, the extent of this risk, that is, the likelihood that a patient with an undiagnosed chronic S. stercoralis infection will complicate following treatment with corticosteroids or other immunosuppressive treatments, is unknown. A case registry would be an important step toward a better understanding of this serious syndrome.

Indeed, the lack of a high-sensitivity diagnostic gold standard has played a major role in the neglect of this parasite, causing underestimation of its global prevalence and misdiagnoses at the individual level. The relative absence of appropriate and comparable validation data for all tests in the currently available literature makes it difficult to select and develop efficient diagnostic protocols, both in the context of population screening and individual diagnostics. In recent years, more attention has been given to the proper evaluation of novel diagnostic tests, i.e., using adequate composite reference standards and/or latent class analysis, which are recommended methods in the absence of a single gold standard. However, choosing the appropriate test can still be challenging. It is imperative that clinicians ordering, performing, and interpreting diagnostic tests for strongyloidiasis are aware of the limitations and advantages of each type of test. A “one size fits all” approach will not yield the best diagnostic outcomes and may cause fatal errors.

Another gray area concerns the zoonotic potential of this parasite. Two genetically distinct populations of S. stercoralis exist in dogs, one of which is shared with humans (60). However, the epidemiological relevance of this finding for human or cross-species transmission remains unknown.

Knowledge and research into strongyloidiasis continue to grow as awareness of its importance in public health and as an individual disease increases. Despite many remaining gaps, our current understanding reinforces the gravity of this disease, which until very recently was so neglected that it was not even formally recognized as an NTD.

ACKNOWLEDGMENTS

The authors would like to gratefully acknowledge the late Emeritus Professor Richard “Rick” Speare for his images of the growth stages of the rhabditiform larvae of Strongyloides, which were adapted for use in this paper; from the Institute of Clinical Pathology and Medical Research-New South Wales Health Pathology, A/Prof. Rogan Lee for his insights into Strongyloides diagnostics; from the Royal North Shore Hospital, Sydney, Australia, A/Prof. Bernie Hudson for his contribution to the histopathology figure and video; and Dr. Matthew Greenwood, Dr. Kim Hart, and Dr. Daniel Lennon for their contribution to the histopathology figure.

This work received funds from the Italian Ministry of Health: “Ricerca Corrente”, Linea 2.

All authors declare no conflict of interest.

Biographies

graphic file with name cmr.00033-23.f010.gif

Dora Buonfrate, MD, DTM&H, FFTM RCPS (Glasg), PhD, is an infectious diseases specialist, leading the clinical research in tropical diseases at the research hospital IRCCS Sacro Cuore Don Calabria in Negrar, Verona, Italy. She is co-heading the WHO collaborating center on strongyloidiasis and other neglected tropical diseases since 2014, and currently is a member of the WHO Guideline Development Group (GDG) on “Public health measures for the control of strongyloidiasis”. Her main research interest is strongyloidiasis, including control in endemic areas, diagnosis and treatment. Her research over the last 10 years has also focused on other neglected tropical diseases (mainly schistosomiasis and Chagas disease), and migrant’s health.

graphic file with name cmr.00033-23.f011.gif

Richard Bradbury is a Senior Lecturer in Microbiology at Federation University in Australia. He began his career as a Medical Laboratory Scientist in Australia, often seeing Strongyloides stercoralis cases from Australian born and immigrant patients. His career has involved both academic and clinical laboratory positions in Australia, West Africa, Slovakia, and the United States of America. His areas of research include diagnostics and epidemiology in medical parasitology, with a particular interest in strongyloidiasis and in maintaining knowledge for diagnostic morphological parasitology. He is the former Team Lead of the Parasitic Diseases Diagnostic Reference Laboratory at the CDC in the United States and a member of the World Health Organisation (WHO) Diagnostic Technical Advisory sub-groups for surveillance of neglected tropical diseases and strongyloidiasis. He was the 2020 Australian Society for Microbiology Lyn Gilbert award winner for major contributions in any area of diagnostic laboratory microbiology nationally or internationally.

graphic file with name cmr.00033-23.f012.gif

Matthew R. Watts has a PhD in parasitology and is a Clinical Microbiologist and Infectious Diseases Physician, with fellowships of the Royal College of Pathologists of Australasia and the Royal Australasian College of Physicians. He is an Associate Professor at the Western Clinical School, University of Sydney. He is a senior staff specialist based at the Centre for Infectious Diseases and Microbiology, Institute for Clinical Pathology and Medical Research – New South Wales Health Pathology (NSWHP), Westmead Hospital, Sydney. This is a government reference laboratory providing services to the state and more locally to Western Sydney and regional NSW, which includes clients from first nations communities and culturally and linguistically diverse backgrounds. More broadly, Dr. Watts is an Associate Director of Clinical Operations for NSWHP and a Local Pathology Director for Southern New South Wales. He has a strong commitment to research and education, improving diagnostic capacity and high-quality service provision.

graphic file with name cmr.00033-23.f013.gif

Zeno Bisoffi, MD, DTM&H, PhD in Clinical Sciences, is former Director of the Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria, Negrar di Valpolicella, Italy, currently Scientific Director of the IRCCS (Clinical and Research Institute), a major research institute of the Ministry of Health on Infectious and Tropical Diseases in Italy. Former co-director of the WHO Collaborating Center on Strongyloidiasis and Other Neglected Tropical Diseases, his main research interests are in tropical and parasitic diseases, particularly NTDs, with a focus on strongyloidiasis.

Contributor Information

Dora Buonfrate, Email: dora.buonfrate@sacrocuore.it.

Louisa A. Messenger, University of Nevada Las Vegas, Las Vegas, Nevada, USA

SUPPLEMENTAL MATERIAL

The following material is available online at https://doi.org/10.1128/cmr.00033-23.

Legend of video. cmr.00033-23-s0001.docx.

Description of Strongyloides larvae video.

cmr.00033-23-s0001.docx (12.2KB, docx)
DOI: 10.1128/cmr.00033-23.SuF1
Video: Strongyloides larvae. cmr.00033-23-s0002.avi.

Highly motile Strongyloides stercoralis larvae on the surface of an agar plate culture.

Download video file (4.1MB, avi)
DOI: 10.1128/cmr.00033-23.SuF2

ASM does not own the copyrights to Supplemental Material that may be linked to, or accessed through, an article. The authors have granted ASM a non-exclusive, world-wide license to publish the Supplemental Material files. Please contact the corresponding author directly for reuse.

REFERENCES

  • 1. Nutman TB. 2017. Human infection with Strongyloides stercoralis and other related Strongyloides species. Parasitology 144:263–273. doi: 10.1017/S0031182016000834 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Schad GA. 1989. Morphology and life history of Strongyloides stercoralis, p 85–104. In Grove DI (ed), Strongyloidiasis: a major roundworm infection of man. Taylor & Francis Ltd, London. [Google Scholar]
  • 3. Buonfrate D, Fittipaldo A, Vlieghe E, Bottieau E. 2021. Clinical and laboratory features of Strongyloides stercoralis infection at diagnosis and after treatment: a systematic review and meta-analysis. Clin Microbiol Infect 27:1621–1628. doi: 10.1016/j.cmi.2021.07.016 [DOI] [PubMed] [Google Scholar]
  • 4. Forrer A, Khieu V, Schär F, Hattendorf J, Marti H, Neumayr A, Char MC, Hatz C, Muth S, Odermatt P. 2017. Strongyloides stercoralis is associated with significant morbidity in rural Cambodia, including stunting in children. PLoS Negl Trop Dis 11:e0005685. doi: 10.1371/journal.pntd.0005685 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Jember TH, Amor A, Nibret E, Munshea A, Flores-Chavez M, Ta-Tang T-H, Saugar JM, Benito A, Anegagrie M. 2022. Prevalence of Strongyloides stercoralis infection and associated clinical symptoms among schoolchildren living in different altitudes of Amhara National Regional State, northwest Ethiopia. PLoS Negl Trop Dis 16:e0010299. doi: 10.1371/journal.pntd.0010299 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Davis JS, Ferreira D, Paige E, Gedye C, Boyle M. 2020. Infectious complications of biological and small molecule targeted immunomodulatory therapies. Clin Microbiol Rev 33:e00035-19. doi: 10.1128/CMR.00035-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Keiser PB, Nutman TB. 2004. Strongyloides stercoralis in the immunocompromised population. Clin Microbiol Rev 17:208–217. doi: 10.1128/CMR.17.1.208-217.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Mejia R, Nutman TB. 2012. Screening, prevention, and treatment for hyperinfection syndrome and disseminated infections caused by Strongyloides stercoralis. Curr Opin Infect Dis 25:458–463. doi: 10.1097/QCO.0b013e3283551dbd [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Buonfrate D, Bisanzio D, Giorli G, Odermatt P, Fürst T, Greenaway C, French M, Reithinger R, Gobbi F, Montresor A, Bisoffi Z. 2020. The global prevalence of Strongyloides stercoralis infection. Pathogens 9:468. doi: 10.3390/pathogens9060468 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Fleitas PE, Travacio M, Martí-Soler H, Socías ME, Lopez WR, Krolewiecki AJ. 2020. The Strongyloides stercoralis-hookworms association as a path to the estimation of the global burden of strongyloidiasis: a systematic review. PLoS Negl Trop Dis 14:e0008184. doi: 10.1371/journal.pntd.0008184 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. 2030 targets for soil-transmitted Helminthiases control programmes. 2019. Licence: CC BY-NC-SA 3.0 IGO. Geneva: World Health Organization. [Google Scholar]
  • 12. Page W, Judd JA, Bradbury RS. 2018. The unique life cycle of Strongyloides stercoralis and implications for public health action. Trop Med Infect Dis 3:53. doi: 10.3390/tropicalmed3020053 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Rivasi F, Pampiglione S, Boldorini R, Cardinale L. 2006. istopathology of gastric and duodenal Strongyloides stercoralis locations in fifteen immunocompromised subjects. Arch Pathol Lab Med 130:1792–1798. doi: 10.5858/2006-130-1792-HOGADS [DOI] [PubMed] [Google Scholar]
  • 14. Genta RM. 1992. Dysregulation of strongyloidiasis: a new hypothesis. Clin Microbiol Rev 5:345–355. doi: 10.1128/CMR.5.4.345 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Grove DI. 1989. Diagnosis, p 175–197. In Grove DI (ed), Strongyloidiasis: a major roundworm infectionn of man. Taylor & Francis Ltd, London. [Google Scholar]
  • 16. Kadhim JK. 1968. Studies on experimental and natural infections of Strongyloides stercoralis (Bavay, 1876) Grassi, 1879, in dogs Ph.D, Oklahoma State University, Stillwater, OK [Google Scholar]
  • 17. Galliard H. 1939. Recherches sur La strongyloïdose au Tonkin. Rôle des animaux domestiques dans l’étiologie de l’infestation humaine. Ann Parasitol Hum Comp 17:533–541. doi: 10.1051/parasite/1939-1940176533 [DOI] [Google Scholar]
  • 18. Galliard H. 1951. Recherches sur l'infestation expérimentale a strongyloides au Tonkin XII. action des facteurs physiques et chimiques sur le développement exogéne et endogéne A. role de la temérature dans le dévelopment exogéne. Ann Parasitol Hum Comp 25:441–473. [PubMed] [Google Scholar]
  • 19. Grove DI. 1996. Human strongyloidiasis. Adv Parasitol 38:251–309. doi: 10.1016/s0065-308x(08)60036-6 [DOI] [PubMed] [Google Scholar]
  • 20. Yamada M, Matsuda S, Nakazawa M, Arizono N. 1991. Species-specific differences in heterogonic development of serially transferred free-living generations of Strongyloidies planiceps and Strongyloides stercoralis. J Parasitol 77:592–594. [PubMed] [Google Scholar]
  • 21. Zhou S, Harbecke D, Streit A. 2019. From the feces to the genome: a guideline for the isolation and preservation of Strongyloides stercoralis in the field for genetic and genomic analysis of individual worms. Parasit Vectors 12:496. doi: 10.1186/s13071-019-3748-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Castelletto ML, Gang SS, Okubo RP, Tselikova AA, Nolan TJ, Platzer EG, Lok JB, Hallem EA. 2014. Diverse host-seeking behaviors of skin-penetrating nematodes. PLoS Pathog 10:e1004305. doi: 10.1371/journal.ppat.1004305 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Yoshida S. 1920. A new course for migrating Ancylostoma and Strongyloides larvae after oral ingestion. J Parasitol Res 7:46. doi: 10.2307/3271157 [DOI] [Google Scholar]
  • 24. Brown RC, Girardeau HF. 1977. Transmammary passage of Strongyloides sp. larvae in the human host. Am J Trop Med Hyg 26:215–219. doi: 10.4269/ajtmh.1977.26.215 [DOI] [PubMed] [Google Scholar]
  • 25. Puerta-Peña M, Calleja Algarra A. 2022. Larva currens in Strongyloides hyperinfection syndrome. N Engl J Med 386:1559. doi: 10.1056/NEJMicm2115708 [DOI] [PubMed] [Google Scholar]
  • 26. Prendki V, Fenaux P, Durand R, Thellier M, Bouchaud O. 2011. Strongyloidiasis in man 75 years after initial exposure. Emerg Infect Dis 17:931–932. doi: 10.3201/eid1705.100490 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Satoh M, Kokaze A. 2004. Treatment strategies in controlling strongyloidiasis. Expert Opin Pharmacother 5:2293–2301. doi: 10.1517/14656566.5.11.2293 [DOI] [PubMed] [Google Scholar]
  • 28. Schad GA, Aikens LM, Smith G. 1989. Strongyloides stercoralis: Is there a canonical migratory route through the host J Parasitol 75:740–749. [PubMed] [Google Scholar]
  • 29. Speare R. 1989. Identification of species of Strongyloides, p 11–84. In Grove DI (ed), Stongyloidiasis: A major Roundworm infection of man. Taylor & Francis Ltd, London. [Google Scholar]
  • 30. Little MD. 1966. Comparative morphology of six species of Strongyloides (Nematoda) and redefinition of the genus. J Parasitol 52:69–84. [PubMed] [Google Scholar]
  • 31. Kim J, Joo H-S, Ko H-M, Na M-S, Hwang S-H, Im J-C. 2005. A case of fatal hyperinfective strongyloidiasis with discorvery of autoinfective filariform larvae in the sputum. Korean J Parasitol 43:51–55. doi: 10.3347/kjp.2005.43.2.51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Little MD. 1966. Seven new species of Strongyloides (Nematoda) from Louisiana. J Parasitol 52:85–97. [PubMed] [Google Scholar]
  • 33. Buonfrate D, Tamarozzi F, Paradies P, Watts MR, Bradbury RS, Bisoffi Z, Cringoli G. 2022. The diagnosis of human and companion animal Strongyloides stercoralis infection: Challenges and solutions. A Scoping review. Edited by Rinaldi L.. Advances in parasitology 118:1–84. doi: 10.1016/bs.apar.2022.07.001 [DOI] [PubMed] [Google Scholar]
  • 34. Schad GA, Smith G, Megyeri Z, Bhopale VM, Niamatali S, Maze R. 1993. Strongyloides stercoralis: an intial autoinfective burst amplifies primary infection. Am J Trop Med Hyg 48:716–725. doi: 10.4269/ajtmh.1993.48.716 [DOI] [PubMed] [Google Scholar]
  • 35. Breloer M, Abraham D. 2017. Strongyloides infection in rodents: immune response and immune regulation. Parasitology 144:295–315. doi: 10.1017/S0031182016000111 [DOI] [PubMed] [Google Scholar]
  • 36. Carvalho EM, Andrade TM, Andrade JA, Rocha H. 1983. Immunological features in different clinical forms of strongyloidiasis. Trans R Soc Trop Med Hyg 77:346–349. doi: 10.1016/0035-9203(83)90162-1 [DOI] [PubMed] [Google Scholar]
  • 37. McSorley HJ, Maizels RM. 2012. Helminth infections and host immune regulation. Clin Microbiol Rev 25:585–608. doi: 10.1128/CMR.05040-11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Higashiarakawa M, Hirata T, Tanaka T, Parrott G, Kinjo T, Naka H, Hokama A, Fujita J. 2017. Normal serum IGE levels and eosinophil counts exhibited during Strongyloides stercoralis infection. Parasitol Int 66:807–812. doi: 10.1016/j.parint.2016.10.004 [DOI] [PubMed] [Google Scholar]
  • 39. Schad GA, Thompson F, Talham G, Holt D, Nolan TJ, Ashton FT, Lange AM, Bhopale VM. 1997. Barren female Strongyloides stercoralis from occult chronic infections are rejuvenated by transfer to parasite-naive recipient hosts and give rise to an autoinfective burst. J Parasitol 83:785–791. doi: 10.2307/3284268 [DOI] [PubMed] [Google Scholar]
  • 40. Mansfield LS, Niamatali S, Bhopale V, Volk S, Smith G, Lok JB, Genta RM, Schad GA. 1996. Strongyloides stercoralis: maintenance of exceedingly chronic infections. Am J Trop Med Hyg 55:617–624. doi: 10.4269/ajtmh.1996.55.617 [DOI] [PubMed] [Google Scholar]
  • 41. Genta RM, Caymmi Gomes M. 1989. Pathology, p 105–132. In Grove DI (ed), Strongyloidiasis: A major Roundworm infection of man. Taylor & Francis Ltd, London. [Google Scholar]
  • 42. Buonfrate D, Requena-Mendez A, Angheben A, Muñoz J, Gobbi F, Van Den Ende J, Bisoffi Z. 2013. Severe strongyloidiasis: a systematic review of case reports. BMC Infect Dis 13:78. doi: 10.1186/1471-2334-13-78 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Mukaigawara M, Narita M, Shiiki S, Takayama Y, Takakura S, Kishaba T. 2020. Clinical characteristics of disseminated strongyloidiasis, Japan, 1975-2017. Emerg Infect Dis 26:401–408. doi: 10.3201/eid2603.190571 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Teixeira MCA, Pacheco FTF, Souza JN, Silva MLS, Inês EJ, Soares NM. 2016. Strongyloides stercoralis infection in alcoholic patients. Biomed Res Int 2016:4872473. doi: 10.1155/2016/4872473 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Dykie A, Wijesinghe T, Rabson AB, Madugula K, Farinas C, Wilson S, Abraham D, Jain P. 2020. Human t-cell leukemia virus type 1 and Strongyloides stercoralis: partners in pathogenesis. Pathogens 9:904. doi: 10.3390/pathogens9110904 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Albarqi MMY, Stoltzfus JD, Pilgrim AA, Nolan TJ, Wang Z, Kliewer SA, Mangelsdorf DJ, Lok JB. 2016. Regulation of life cycle checkpoints and developmental activation of infective larvae in Strongyloides stercoralis by dafachronic acid. PLoS Pathog 12:e1005358. doi: 10.1371/journal.ppat.1005358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Siddiqui AA, Stanley CS, Berk SL. 2000. A cDNA encoding the highly immunodominant antigen of Strongyloides stercoralis: γ-subunit of isocitrate dehydrogenase (NAD+). Parasitol Res 86:279–283. doi: 10.1007/s004360050043 [DOI] [PubMed] [Google Scholar]
  • 48. Patton JB, Bonne-Année S, Deckman J, Hess JA, Torigian A, Nolan TJ, Wang Z, Kliewer SA, Durham AC, Lee JJ, Eberhard ML, Mangelsdorf DJ, Lok JB, Abraham D. 2018. Methylprednisolone acetate induces, and Δ7-dafachronic acid suppresses, Strongyloides stercoralis hyperinfection in NSG mice. Proc Natl Acad Sci U S A 115:204–209. doi: 10.1073/pnas.1712235114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Debussche X, Toublanc M, Camillieri JP, Assan R. 1988. Overwhelming strongyloidiasis in a diabetic patient following ACTH treatment and keto-acidosis. Diabete Metab 14:294–298. [PubMed] [Google Scholar]
  • 50. Krishnamurthy R, Dincer HE, Whittemore D. 2007. Strongyloides stercoralis hyperinfection in a patient with rheumatoid arthritis after anti-TNF-α therapy. J Clin Rheumatol 13:150–152. doi: 10.1097/RHU.0b013e3180690933 [DOI] [PubMed] [Google Scholar]
  • 51. Vazquez Guillamet LJ, Saul Z, Miljkovich G, Vilchez GA, Mendonca N, Gourineni V, Lillo N, Pinto M, Baig A, Gangcuangco LM. 2017. Strongyloides stercoralis infection among human immunodeficiency virus (HIV)-infected patients in the United States of America: a case report and review of literature. Am J Case Rep 18:339–346. doi: 10.12659/ajcr.902626 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Weatherhead JE, Mejia R. 2014. Immune response to infection with Strongyloides stercoralis in patients with infection and hyperinfection. Curr Trop Med Rep 1:229–233. doi: 10.1007/s40475-014-0032-9 [DOI] [Google Scholar]
  • 53. Ye L, Taylor GP, Rosadas C. 2022. Human T-cell Lymphotropic virus type 1 and Strongyloides stercoralis Co-infection: a systematic review and meta-analysis. Front Med (Lausanne) 9:832430. doi: 10.3389/fmed.2022.832430 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. George PJ, Pavan Kumar N, Jaganathan J, Dolla C, Kumaran P, Nair D, Banurekha VV, Shen K, Nutman TB, Babu S. 2015. Modulation of pro- and anti-inflammatory cytokines in active and latent tuberculosis by coexistent Strongyloides stercoralis infection. Tuberculosis (Edinb) 95:822–828. doi: 10.1016/j.tube.2015.09.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Kathamuthu GR, Munisankar S, Sridhar R, Baskaran D, Babu S. 2019. Helminth mediated modulation of the systemic and mycobacterial antigen - stimulated cytokine profiles in extra-pulmonary tuberculosis. PLoS Negl Trop Dis 13:e0007265. doi: 10.1371/journal.pntd.0007265 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Donovan J, Tram TTB, Phu NH, Hiep NTT, Van VTT, Mui DTH, Ny NTH, Nghia HDT, Hanh NHH, Tan LV, Thuong NTT, Thwaites GE. 2022. The influence of Strongyloides stercoralis co-infection on the presentation, pathogenesis and outcome of tuberculous meningitis. J Infect Dis 225:1653–1662. doi: 10.1093/infdis/jiaa672 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Lier AJ, Tuan JJ, Davis MW, Paulson N, McManus D, Campbell S, Peaper DR, Topal JE. 2020. Case report: disseminated strongyloidiasis in a patient with COVID-19. Am J Trop Med Hyg 103:1590–1592. doi: 10.4269/ajtmh.20-0699 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Marchese V, Crosato V, Gulletta M, Castelnuovo F, Cristini G, Matteelli A, Castelli F. 2021. Strongyloides infection manifested during immunosuppressive therapy for SARS-CoV-2 pneumonia. Infection 49:539–542. doi: 10.1007/s15010-020-01522-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Jenks NP, Driscoll B, Locke T. 2022. Strongyloidiasis hyperinfection syndrome in COVID-19 positive migrants treated with corticosteroids. J Immigr Minor Health 24:1431–1434. doi: 10.1007/s10903-022-01386-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Jaleta TG, Zhou S, Bemm FM, Schär F, Khieu V, Muth S, Odermatt P, Lok JB, Streit A. 2017. Different but overlapping populations of Strongyloides stercoralis in dogs and humans-dogs as a possible source for zoonotic strongyloidiasis. PLoS Negl Trop Dis 11:e0005752. doi: 10.1371/journal.pntd.0005752 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Barratt JLN, Lane M, Talundzic E, Richins T, Robertson G, Formenti F, Pritt B, Verocai G, Nascimento de Souza J, Mato Soares N, Traub R, Buonfrate D, Bradbury RS. 2019. A global genotyping survey of Strongyloides stercoralis and Strongyloides fuelleborni using deep amplicon sequencing. PLoS Negl Trop Dis 13:e0007609. doi: 10.1371/journal.pntd.0007609 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Wulcan JM, Dennis MM, Ketzis JK, Bevelock TJ, Verocai GG. 2019. Strongyloides spp. in cats: a review of the literature and the first report of zoonotic Strongyloides stercoralis in colonic epithelial nodular hyperplasia in cats. Parasit Vectors 12:349. doi: 10.1186/s13071-019-3592-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Nagayasu E, Aung M, Hortiwakul T, Hino A, Tanaka T, Higashiarakawa M, Olia A, Taniguchi T, Win SMT, Ohashi I, Odongo-Aginya EI, Aye KM, Mon M, Win KK, Ota K, Torisu Y, Panthuwong S, Kimura E, Palacpac NMQ, Kikuchi T, Hirata T, Torisu S, Hisaeda H, Horii T, Fujita J, Htike WW, Maruyama H. 2017. A possible origin population of pathogenic intestinal nematodes, Strongyloides stercoralis, unveiled by molecular phylogeny. Sci Rep 7:4844. doi: 10.1038/s41598-017-05049-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Bradbury RS, Pafčo B, Nosková E, Hasegawa H. 2021. Strongyloides genotyping: a review of methods and application in public health and population genetics. Int J Parasitol 51:1153–1166. doi: 10.1016/j.ijpara.2021.10.001 [DOI] [PubMed] [Google Scholar]
  • 65. Aupalee K, Wijit A, Singphai K, Rödelsperger C, Zhou S, Saeung A, Streit A. 2020. Genomic studies on Strongyloides stercoralis in northern and western Thailand. Parasit Vectors 13:250. doi: 10.1186/s13071-020-04115-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Georgi JR, Sprinkle CL. 1974. A case of human strongyloidosis apparently contracted from asymptomatic colony dogs. Am J Trop Med Hyg 23:899–901. doi: 10.4269/ajtmh.1974.23.899 [DOI] [PubMed] [Google Scholar]
  • 67. Sandground JH. 1926. Biological studies on the life-cycle in the genus Strongyloides Grassi,1879. Am J Epidemiol 6:337–388. doi: 10.1093/oxfordjournals.aje.a120018 [DOI] [Google Scholar]
  • 68. Fuelleborn F. 1927. Über das Verhalten der Larven von Strongyloides stercoralis, Hakenwürmern und Ascaris lumbricoides Im Körper des Wirtes. Arch Schiffs-u Tropenhyg 31:1–56. [Google Scholar]
  • 69. Augustine DL, Davey DG. 1939. Observations on a natural infection with Strongyloides in the dog. J Parasitol 25:117. doi: 10.2307/3272350 [DOI] [Google Scholar]
  • 70. Sandosham AA. 1952. An investigation into the association of creeping eruption with Strongyloides infection contracted in the far East. J Helminthol 26:1–24. doi: 10.1017/S0022149X00032569 [DOI] [Google Scholar]
  • 71. Galliard H. 1950. Recherches sur l’Infestation expérimentale a Strongyloides stercoralis au Tonkin (1Re NOTE). Annales de Parasitologie Humaine et Comparée 25:441–473. [PubMed] [Google Scholar]
  • 72. Sandground JH. 1928. Some studies on susceptibility, resistance, and acquired immunity to infection with Strongyloides stercoralis (Nematoda) in dogs and cats. Am J Hyg 8:507–538. doi: 10.1093/oxfordjournals.aje.a121015 [DOI] [Google Scholar]
  • 73. Sandground JH. 1925. Speciation and specificity in the nematode genus Strongyloides. J Parasitol 12:59. doi: 10.2307/3270768 [DOI] [Google Scholar]
  • 74. Grove DI, Northern C. 1982. Infection and immunity in dogs infected with a human strain of Strongyloides stercoralis. Trans R Soc Trop Med Hyg 76:833–838. doi: 10.1016/0035-9203(82)90120-1 [DOI] [PubMed] [Google Scholar]
  • 75. Cadd LC, Crooks B, Marks NJ, Maule AG, Mousley A, Atkinson LE. 2022. The Strongyloides bioassay toolbox: a unique opportunity to accelerate functional biology for nematode parasites. Mol Biochem Parasitol 252:111526. doi: 10.1016/j.molbiopara.2022.111526 [DOI] [PubMed] [Google Scholar]
  • 76. Hunt VL, Tsai IJ, Coghlan A, Reid AJ, Holroyd N, Foth BJ, Tracey A, Cotton JA, Stanley EJ, Beasley H, et al. 2016. The Genomic basis of Parasitism in the Strongyloides clade of nematodes. Nat Genet 48:299–307. doi: 10.1038/ng.3495 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Jaleta TG, Lok JB. 2019. Advances in the molecular and cellular biology of Strongyloides spp. Curr Trop Med Rep 6:161–178. doi: 10.1007/s40475-019-00186-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Kikuchi T, Hino A, Tanaka T, Aung M, Afrin T, Nagayasu E, Tanaka R, Higashiarakawa M, Win KK, Hirata T, Htike WW, Fujita J, Maruyama H. 2016. Genome-wide analyses of individual Strongyloides stercoralis (Nematoda: Rhabditoidea) provide insights into population structure and reproductive life cycles. PLoS Negl Trop Dis 10:e0005253. doi: 10.1371/journal.pntd.0005253 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Soblik H, Younis AE, Mitreva M, Renard BY, Kirchner M, Geisinger F, Steen H, Brattig NW. 2011. Life cycle stage-resolved proteomic analysis of the excretome/secretome from Strongyloides ratti--identification of stage-specific proteases. Mol Cell Proteomics 10:M111. doi: 10.1074/mcp.M111.010157 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Dishnica K, Piubelli C, Manfredi M, Kondaveeti RT, Longoni SS, Degani M, Buonfrate D, Giorgetti A, Tiberti N. 2023. Novel insights into the somatic proteome of Strongyloides stercoralis infective third-stage larvae. Parasit Vectors 16:45. doi: 10.1186/s13071-023-05675-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Corral MA, Paula FM, Meisel DMCL, Castilho VLP, Gonçalves EMN, Levy D, Bydlowski SP, Chieffi PP, Castro-Borges W, Gryschek RCB. 2017. Potential immunological markers for diagnosis of human strongyloidiasis using heterologous antigens. Parasitology 144:124–130. doi: 10.1017/S0031182016001645 [DOI] [PubMed] [Google Scholar]
  • 82. Marcilla A, Sotillo J, Pérez-Garcia A, Igual-Adell R, Valero ML, Sánchez-Pino MM, Bernal D, Muñoz-Antolí C, Trelis M, Toledo R, Esteban JG. 2010. Proteomic analysis of Strongyloides stercoralis L3 larvae. Parasitology 137:1577–1583. doi: 10.1017/S0031182010000314 [DOI] [PubMed] [Google Scholar]
  • 83. Rodpai R, Intapan PM, Thanchomnang T, Sanpool O, Janwan P, Laummaunwai P, Wongkham C, Insawang T, Maleewong W. 2017. Identification of antigenic proteins in Strongyloides stercoralis by proteomic analysis. Parasitol Res 116:1687–1693. doi: 10.1007/s00436-017-5443-9 [DOI] [PubMed] [Google Scholar]
  • 84. Cheong MC, Wang Z, Jaleta TG, Li X, Lok JB, Kliewer SA, Mangelsdorf DJ. 2021. Identification of a nuclear receptor/coactivator developmental signaling pathway in the nematode parasite Strongyloides stercoralis. Proc Natl Acad Sci U S A 118:e2021864118. doi: 10.1073/pnas.2021864118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Olsen A, van Lieshout L, Marti H, Polderman T, Polman K, Steinmann P, Stothard R, Thybo S, Verweij JJ, Magnussen P. 2009. Strongyloidiasis--the most neglected of the neglected tropical diseases?. Trans R Soc Trop Med Hyg 103:967–972. doi: 10.1016/j.trstmh.2009.02.013 [DOI] [PubMed] [Google Scholar]
  • 86. Bethony J, Brooker S, Albonico M, Geiger SM, Loukas A, Diemert D, Hotez PJ. 2006. Soil-transmitted helminth infections: ascariasis, trichuriasis, and hookworm. Lancet 367:1521–1532. doi: 10.1016/S0140-6736(06)68653-4 [DOI] [PubMed] [Google Scholar]
  • 87. Forrer A, Khieu V, Schär F, Vounatsou P, Chammartin F, Marti H, Muth S, Odermatt P. 2018. Strongyloides stercoralis and hookworm co-infection: spatial distribution and determinants in Preah Vihear Province, Cambodia. Parasit Vectors 11:33. doi: 10.1186/s13071-017-2604-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Muadica AS, Balasegaram S, Beebeejaun K, Köster PC, Bailo B, Hernández-de-Mingo M, Dashti A, Dacal E, Saugar JM, Fuentes I, Carmena D. 2021. Risk associations for intestinal parasites in symptomatic and asymptomatic schoolchildren in central Mozambique. Clin Microbiol Infect 27:624–629. doi: 10.1016/j.cmi.2020.05.031 [DOI] [PubMed] [Google Scholar]
  • 89. Senephansiri P, Laummaunwai P, Laymanivong S, Boonmar T. 2017. Status and risk factors of Strongyloides stercoralis infection in rural communities of Xayaburi province, Lao PDR. Korean J Parasitol 55:569–573. doi: 10.3347/kjp.2017.55.5.569 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Al-Mekhlafi HM, Nasr NA, Lim YAL, Elyana FN, Sady H, Atroosh WM, Dawaki S, Anuar TS, Noordin R, Mahmud R. 2019. Prevalence and risk factors of Strongyloides stercoralis infection among Orang Asli schoolchildren: new insights into the epidemiology, transmission and diagnosis of strongyloidiasis in Malaysia. Parasitology 146:1602–1614. doi: 10.1017/S0031182019000945 [DOI] [PubMed] [Google Scholar]
  • 91. Aung M, Hino A, Oo KM, Win KK, Maruyama H, Htike WW, Nagayasu E. 2018. Prevalence and associated risk factors of Strongyloides stercoralis infection in Lower Myanmar. Trop Med Health 46:43. doi: 10.1186/s41182-018-0126-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Morales ML, Lopez M, Ly P, Anjum S, Fernandez-Baca MV, Valdivia-Rodriguez AM, Mamani-Licona FM, Baca-Turpo B, Farfan-Gonzales N, Chaman-Illanes Y, Cabada MM. 2019. Strongyloides stercoralis infection at different altitudes of the Cusco region in Peru. Am J Trop Med Hyg 101:422–427. doi: 10.4269/ajtmh.18-0568 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Lindo JF, Robinson RD, Terry SI, Vogel P, Gam AA, Neva FA, Bundy DA. 1995. Age-prevalence and household clustering of Strongyloides stercoralis infection in Jamaica. Parasitology 110:97–102. doi: 10.1017/s0031182000081099 [DOI] [PubMed] [Google Scholar]
  • 94. Herwaldt BL. 2001. Laboratory-acquired parasitic infections from accidental exposures. Clin Microbiol Rev 14:659–688. doi: 10.1128/CMR.14.3.659-688.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Singer R, Sarkar S. 2020. Modeling strongyloidiasis risk in the United States. Int J Infect Dis 100:366–372. doi: 10.1016/j.ijid.2020.09.002 [DOI] [PubMed] [Google Scholar]
  • 96. Inagaki K, Bradbury RS, Hobbs CV. 2022. Hospitalizations associated with strongyloidiasis in the United States, 2003-2018. Clin Infect Dis 75:1548–1555. doi: 10.1093/cid/ciac220 [DOI] [PubMed] [Google Scholar]
  • 97. Štrkolcová G, Goldová M, Bocková E, Mojžišová J. 2017. The roundworm Strongyloides stercoralis in children, dogs, and soil inside and outside a segregated settlement in Eastern Slovakia: frequent but hardly detectable parasite. Parasitol Res 116:891–900. doi: 10.1007/s00436-016-5362-1 [DOI] [PubMed] [Google Scholar]
  • 98. Duvignaud A, Pistone T, Malvy D. 2016. Strongyloidiasis in a young French woman raises concern about possible ongoing autochthonous transmission in Spain. Int J Infect Dis 42:43–44. doi: 10.1016/j.ijid.2015.11.015 [DOI] [PubMed] [Google Scholar]
  • 99. Barroso M, Salvador F, Sánchez-Montalvá A, Bosch-Nicolau P, Molina I. 2019. Strongyloides stercoralis infection: a systematic review of endemic cases in Spain. PLoS Negl Trop Dis 13:e0007230. doi: 10.1371/journal.pntd.0007230 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Ottino L, Buonfrate D, Paradies P, Bisoffi Z, Antonelli A, Rossolini GM, Gabrielli S, Bartoloni A, Zammarchi L. 2020. Autochthonous human and canine Strongyloides stercoralis infection in Europe: report of a human case in an Italian teen and systematic review of the literature. Pathogens 9:439. doi: 10.3390/pathogens9060439 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Buonfrate D, Baldissera M, Abrescia F, Bassetti M, Caramaschi G, Giobbia M, Mascarello M, Rodari P, Scattolo N, Napoletano G, Bisoffi Z, CCM Strongyloides Study Group . 2016. Epidemiology of Strongyloides stercoralis in northern Italy: results of a multicentre case-control study, February 2013 to July 2014. Euro Surveill 21:30310. doi: 10.2807/1560-7917.ES.2016.21.31.30310 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Pinto J, Almeida P, Meireles D, Araújo A. 2021. Strongyloidiasis: a diagnosis to consider in previously endemic regions in Portugal. Acta Med Port 34:552–556. doi: 10.20344/amp.12960 [DOI] [PubMed] [Google Scholar]
  • 103. Román-Sánchez P, Pastor-Guzmán A, Moreno-Guillén S, Igual-Adell R, Suñer-Generoso S, Tornero-Estébanez C. 2003. High prevalence of Strongyloides stercoralis among farm workers on the mediterranean coast of Spain: analysis of the predictive factors of infection in developed countries. Am J Trop Med Hyg 69:336–340. doi: 10.4269/ajtmh.2003.69.336 [DOI] [PubMed] [Google Scholar]
  • 104. Ikuno H, Ishikawa T, Norose K. 2020. Status of strongyloidiasis in Japan, 2000-2017. Am J Trop Med Hyg 103:727–734. doi: 10.4269/ajtmh.19-0969 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Kobayashi JUN, Sato Y, Toma H, Shimabukuro I, Tasaki T, Takara M, Shiroma Y. 2000. Epidemiological features of Strongyloides infection in Okinawa, Japan: comparative study with other endemic areas. Jap J Trop Med Hyg 28:9–14. doi: 10.2149/tmh1973.28.9 [DOI] [Google Scholar]
  • 106. Flannery G, White N. 1993. Immunological parameters in northeast Arnhem land aborigines: Consequences of changing settlement patterns and lifestyles, p 202–220. Urban ecology and health in the third world Cambridge University Press, Cambridge. [Google Scholar]
  • 107. Holt DC, Shield J, Harris TM, Mounsey KE, Aland K, McCarthy JS, Currie BJ, Kearns TM. 2017. Soil-transmitted helminths in children in a remote aboriginal community in the Northern territory: hookworm is rare but Strongyloides stercoralis and trichuris trichiura persist. Trop Med Infect Dis 2:51. doi: 10.3390/tropicalmed2040051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Shield J, Braat S, Watts M, Robertson G, Beaman M, McLeod J, Baird RW, Hart J, Robson J, Lee R, McKessar S, Nicholson S, Mayer-Coverdale J, Biggs B-A, Babu S. 2021. Seropositivity and geographical distribution of Strongyloides stercoralis in Australia: a study of pathology laboratory data from 2012-2016. PLoS Negl Trop Dis 15:e0009160. doi: 10.1371/journal.pntd.0009160 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Baaten GG, Sonder GJ, van Gool T, Kint JA, van den Hoek A. 2011. Travel-related schistosomiasis, strongyloidiasis, filariasis, and toxocariasis: the risk of infection and the diagnostic relevance of blood eosinophilia. BMC Infect Dis 11:84. doi: 10.1186/1471-2334-11-84 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Libman MD, MacLean JD, Gyorkos TW. 1993. Screening for schistosomiasis, filariasis, and strongyloidiasis among expatriates returning from the tropics. Clin Infect Dis 17:353–359. doi: 10.1093/clinids/17.3.353 [DOI] [PubMed] [Google Scholar]
  • 111. Agbata EN, Morton RL, Bisoffi Z, Bottieau E, Greenaway C, Biggs B-A, Montero N, Tran A, Rowbotham N, Arevalo-Rodriguez I, Myran DT, Noori T, Alonso-Coello P, Pottie K, Requena-Méndez A. 2018. Effectiveness of screening and treatment approaches for schistosomiasis and strongyloidiasis in newly-arrived migrants from endemic countries in the EU/EEA: a systematic review. Int J Environ Res Public Health 16:11. doi: 10.3390/ijerph16010011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Asundi A, Beliavsky A, Liu XJ, Akaberi A, Schwarzer G, Bisoffi Z, Requena-Méndez A, Shrier I, Greenaway C. 2019. Prevalence of strongyloidiasis and schistosomiasis among migrants: a systematic review and meta-analysis. Lancet Glob Health 7:e236–e248. doi: 10.1016/S2214-109X(18)30490-X [DOI] [PubMed] [Google Scholar]
  • 113. Caumes E, Keystone JS. 2011. Acute strongyloidiasis: a rarity. Chronic strongyloidiasis: a time bomb!. J Travel Med 18:71–72. doi: 10.1111/j.1708-8305.2010.00492.x [DOI] [PubMed] [Google Scholar]
  • 114. Angheben A, Gobbi F, Rossanese A, Anselmi M, Marocco S, Monteiro G, Mistretta M, Sepe A, Lacovazzi T, Bisoffi Z. 2009. Acute strongyloidiasis: clinical features and diagnostic problems in three returning travellers seen at the centre for tropical disease, Negrar, Verona. Trop Med Int Health 14:235. [Google Scholar]
  • 115. Alabi A, Boggild AK, Bitnun A. 2017. Acute strongyloidiasis in a child recently returned from vacation in Cuba. CMAJ 189:E1416–E1420. doi: 10.1503/cmaj.170464 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Marcos LA, Terashima A, Dupont HL, Gotuzzo E. 2008. Strongyloides hyperinfection syndrome: an emerging global infectious disease. Trans R Soc Trop Med Hyg 102:314–318. doi: 10.1016/j.trstmh.2008.01.020 [DOI] [PubMed] [Google Scholar]
  • 117. Grove DI. 1989. Strongyloidiasis: a major roundworm infection of man, p 155–173. In Grove DI (ed), Clinical manifestations. Taylor & Francis Ltd, London. [Google Scholar]
  • 118. Tamarozzi F, Martello E, Giorli G, Fittipaldo A, Staffolani S, Montresor A, Bisoffi Z, Buonfrate D. 2019. Morbidity associated with chronic Strongyloides stercoralis infection: a systematic review and meta-analysis. Am J Trop Med Hyg 100:1305–1311. doi: 10.4269/ajtmh.18-0895 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. van Samkar A, Brouwer MC, Pannekoek Y, van der Ende A, van de Beek D. 2015. Streptococcus gallolyticus meningitis in adults: report of five cases and review of the literature. Clin Microbiol Infect 21:1077–1083. doi: 10.1016/j.cmi.2015.08.003 [DOI] [PubMed] [Google Scholar]
  • 120. Keloth T, Rajkumari N, Gochhait D, Gudivada V, Toi PC, Siddaraju N. 2019. Microfilaria and Strongyloides larva diagnosed in cerebrospinal fluid and ascitic fluid, respectively: approach to their morphology on cytology. Diagn Cytopathol 47:1055–1058. doi: 10.1002/dc.24257 [DOI] [PubMed] [Google Scholar]
  • 121. Baker ML, Brewster UC. 2022. Strongyloides stercoralis infection identified on urine microscopy. BMJ Case Rep 15:e250078. doi: 10.1136/bcr-2022-250078 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Sundar PM, Sivanandam S, Chitra TV. 2022. A usual parasite in an unusual location- Incidental detection of Strongyloides stercoralis in liquid-based cytology. J Postgrad Med 68:60–61. doi: 10.4103/jpgm.jpgm_614_21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Agbo K, Deniau M. 1987. Anguillospermia resistant to treatment. Apropos of a case diagnosed in Togo. Bull Soc Pathol Exot Filiales 80:271–273. [PubMed] [Google Scholar]
  • 124. Poltera AA, Katsimbura N. 1974. Granulomatous hepatitis due to Strongyloides stercoralis. J Pathol 113:241–246. doi: 10.1002/path.1711130408 [DOI] [PubMed] [Google Scholar]
  • 125. Byard RW. 2019. Lethal Strongyloidiasis - diagnostic and forensic issues. J Forensic Leg Med 62:103–106. doi: 10.1016/j.jflm.2019.01.014 [DOI] [PubMed] [Google Scholar]
  • 126. Dimitroglou Y, Alexopoulos T, Aggeli C, Kalantzi M, Nouli A, Dourakis SP, Tsioufis K. 2021. Eosinophilic myocarditis in a patient with Strongyloides stercoralis infection. JACC Case Rep 3:954–959. doi: 10.1016/j.jaccas.2021.04.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Hutabarat SN, Domazetovska A, Ziochos H, Hassett G, Foo H. 2023. Streptococcus gallolyticus meningitis and Strongyloides stercoralis hyperinfection in a patient with systemic lupus erythematosus. Eur J Clin Microbiol Infect Dis 42:665–667. doi: 10.1007/s10096-023-04589-7 [DOI] [PubMed] [Google Scholar]
  • 128. Corte LD, da Silva MVS, Souza PRM. 2013. Simultaneous larva migrans and larva currens caused by Strongyloides stercoralis: a case report. Case Rep Dermatol Med 2013:381583. doi: 10.1155/2013/381583 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Purvis RS, Beightler EL, Diven DG, Sanchez RL, Tyring SK. 1992. Strongyloides hyperinfection presenting with petechiae and purpura. Int J Dermatol 31:169–171. doi: 10.1111/j.1365-4362.1992.tb03925.x [DOI] [PubMed] [Google Scholar]
  • 130. Schär F, Hattendorf J, Khieu V, Muth S, Char MC, Marti HP, Odermatt P. 2014. Strongyloides stercoralis larvae excretion patterns before and after treatment. Parasitology 141:892–897. doi: 10.1017/S0031182013002345 [DOI] [PubMed] [Google Scholar]
  • 131. Dreyer G, Fernandes-Silva E, Alves S, Rocha A, Albuquerque R, Addiss D. 1996. Patterns of detection of Strongyloides stercoralis in stool specimens: implications for diagnosis and clinical trials. J Clin Microbiol 34:2569–2571. doi: 10.1128/jcm.34.10.2569-2571.1996 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Buonfrate D, Rodari P, Barda B, Page W, Einsiedel L, Watts MR. 2022. Current pharmacotherapeutic strategies for strongyloidiasis and the complications in its treatment. Expert Opin Pharmacother 23:1617–1628. doi: 10.1080/14656566.2022.2114829 [DOI] [PubMed] [Google Scholar]
  • 133. Knopp S, Salim N, Schindler T, Karagiannis Voules DA, Rothen J, Lweno O, Mohammed AS, Singo R, Benninghoff M, Nsojo AA, Genton B, Daubenberger C. 2014. Diagnostic accuracy of Kato-Katz, FLOTAC, Baermann, and PCR methods for the detection of light-intensity hookworm and Strongyloides stercoralis infections in Tanzania. Am J Trop Med Hyg 90:535–545. doi: 10.4269/ajtmh.13-0268 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Barda BD, Rinaldi L, Ianniello D, Zepherine H, Salvo F, Sadutshang T, Cringoli G, Clementi M, Albonico M. 2013. Mini-FLOTAC, an innovative direct diagnostic technique for intestinal parasitic infections: experience from the field. PLoS Negl Trop Dis 7:e2344. doi: 10.1371/journal.pntd.0002344 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Barda B, Cajal P, Villagran E, Cimino R, Juarez M, Krolewiecki A, Rinaldi L, Cringoli G, Burioni R, Albonico M. 2014. Mini-FLOTAC, Kato-Katz and McMaster: three methods, one goal; highlights from north Argentina. Parasit Vectors 7:271. doi: 10.1186/1756-3305-7-271 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Glinz D, Silué KD, Knopp S, Lohourignon LK, Yao KP, Steinmann P, Rinaldi L, Cringoli G, N’Goran EK, Utzinger J. 2010. Comparing diagnostic accuracy of Kato-Katz, Koga agar plate, ether-concentration, and FLOTAC for Schistosoma mansoni and soil-transmitted helminths. PLoS Negl Trop Dis 4:e754. doi: 10.1371/journal.pntd.0000754 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Arakaki T, Hasegawa H, Asato R, Ikeshiro T, Kinjo F, Saito A, Iwanaga M. 1988. A new method to detect Strongyloides stercoralis from human stool. Jap J Trop Med Hyg 16:11–17. doi: 10.2149/tmh1973.16.11 [DOI] [Google Scholar]
  • 138. Kaewrat W, Sengthong C, Yingklang M, Intuyod K, Haonon O, Onsurathum S, Dangtakot R, Saisud P, Sangka A, Anutrakulchai S, Pinlaor S, Cha’on U, Pinlaor P. 2020. Improved agar plate culture conditions for diagnosis of Strongyloides stercoralis. Acta Trop 203:105291. doi: 10.1016/j.actatropica.2019.105291 [DOI] [PubMed] [Google Scholar]
  • 139. Pocaterra LA, Ferrara G, Peñaranda R, Rojas E, Pérez-Chacón G, Hernán A, Certad G, Goldstein C, Núñez L. 2017. Improved detection of Strongyloides stercoralis in modified agar plate cultures. Am J Trop Med Hyg 96:863–865. doi: 10.4269/ajtmh.16-0414 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Khanna V, Tilak K, Prakash PY, Mukhopadhyay C. 2015. Modified agar plate culture method for culture of Strongyloides stercoralis. Trop Parasitol 5:136–138. doi: 10.4103/2229-5070.162535 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Koga K, Kasuya S, Khamboonruang C, Sukhavat K, Ieda M, Takatsuka N, Kita K, Ohtomo H. 1991. A modified agar plate method for detection of Strongyloides stercoralis. Am J Trop Med Hyg 45:518–521. doi: 10.4269/ajtmh.1991.45.518 [DOI] [PubMed] [Google Scholar]
  • 142. World Health Organization (WHO), The United Nations Children’s Fund (UNICEF) . 2021. Progress on household drinking water, sanitation and hygiene 2000-2020: Five years into the Sdgs, licence: CC BY-NC-SA 3.0 IGO. Geneva [Google Scholar]
  • 143. Gelaye W, Williams NA, Kepha S, Junior AM, Fleitas PE, Marti-Soler H, Damtie D, Menkir S, Krolewiecki AJ, van Lieshout L, Enbiale W, Stopping Transmission of Intestinal Parasites (STOP) project consortium . 2021. Performance evaluation of Baermann techniques: the quest for developing a microscopy reference standard for the diagnosis of Strongyloides stercoralis. PLoS Negl Trop Dis 15:e0009076. doi: 10.1371/journal.pntd.0009076 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Nielsen PB, Mojon M. 1987. Improved diagnosis of Strongyloides stercoralis by seven consecutive stool specimens. Zentralbl Bakteriol Mikrobiol Hyg A 263:616–618. doi: 10.1016/s0176-6724(87)80207-9 [DOI] [PubMed] [Google Scholar]
  • 145. Hirata T, Nakamura H, Kinjo N, Hokama A, Kinjo F, Yamane N, Fujita J. 2007. Short report: increased detection rate of Strongyloides stercoralis by repeated stool examinations using the agar plate culture method. Am J Trop Med Hyg 77:683–684. doi: 10.4269/ajtmh.2007.77.683 [DOI] [PubMed] [Google Scholar]
  • 146. Steinmann P, Zhou X-N, Du Z-W, Jiang J-Y, Wang L-B, Wang X-Z, Li L-H, Marti H, Utzinger J. 2007. Occurrence of Strongyloides stercoralis in Yunnan Province, China, and comparison of diagnostic methods. PLoS Negl Trop Dis 1:e75. doi: 10.1371/journal.pntd.0000075 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Bradbury RS, Herwaldt BL.. 2020. Section VIII-C: parasitic agents, p 223-238. In Meechan PJ, Potts J (ed), Biosafety in microbiological and biomedical laboratories, 6th ed. U.S. Department of Health and Human Services, Washington D.C. [Google Scholar]
  • 148. Verweij Jaco J, Canales M, Polman K, Ziem J, Brienen EAT, Polderman AM, van Lieshout L. 2009. Molecular diagnosis of Strongyloides stercoralis in faecal samples using real-time PCR. Trans R Soc Trop Med Hyg 103:342–346. doi: 10.1016/j.trstmh.2008.12.001 [DOI] [PubMed] [Google Scholar]
  • 149. Verweij JJ, Claas ECJ, Van M, Van L. 2009. Molecular diagnosis of Strongyloides stercoralis infection in faecal samples of patients living in a nonendemic region. ANTON LEEUW INT J G 95:51–52. [Google Scholar]
  • 150. Beknazarova M, Barratt JLN, Bradbury RS, Lane M, Whiley H, Ross K. 2019. Detection of classic and cryptic Strongyloides genotypes by deep amplicon sequencing: a preliminary survey of dog and human specimens collected from remote Australian communities. PLoS Negl Trop Dis 13:e0007241. doi: 10.1371/journal.pntd.0007241 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. SKML (Dutch foundation for quality assessment in Laboratories) Parasitology section. 2022. Available from: https://www.skml.nl/en/home/sections/parasitology
  • 152. Verweij JJ. 2022. Validation and maintaining laboratory developed molecular tests compliant with ISO15189 for diagnosis of intestinal parasitic infections. Expert Rev Mol Diagn 22:595–601. doi: 10.1080/14737159.2021.1971974 [DOI] [PubMed] [Google Scholar]
  • 153. 2023. RCPAQAP (the royal college of pathologists of Australasia quality assurance programs). Available from: https://rcpaqap.com.au [DOI] [PubMed]
  • 154. Mejia R, Damania A, Jeun R, Bryan PE, Vargas P, Juarez M, Cajal PS, Nasser J, Krolewiecki A, Lefoulon E, Long C, Drake E, Cimino RO, Slatko B. 2020. Impact of intestinal parasites on microbiota and cobalamin gene sequences: a pilot study. Parasit Vectors 13:200. doi: 10.1186/s13071-020-04073-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Qu J, Zong Z. 2022. Strongyloidiasis in a patient diagnosed by metagenomic next-generation sequencing: a case report. Front Med (Lausanne) 9:835252. doi: 10.3389/fmed.2022.835252 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Chiu CY, Miller SA. 2019. Clinical metagenomics. Nat Rev Genet 20:341–355. doi: 10.1038/s41576-019-0113-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Gu W, Miller S, Chiu CY. 2019. Clinical metagenomic next-generation sequencing for pathogen detection. Annu Rev Pathol 14:319–338. doi: 10.1146/annurev-pathmechdis-012418-012751 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158. Diagnostic methods for the control of strongyloidiasis, virtual meeting, 29 September 2020. 2020. Geneva: World Health Organization. Licence: CC BY-NC-SA 3.0 IGO. Available from: https://creativecommons.org/licenses/by-nc-sa/3.0/igo [Google Scholar]
  • 159. Page WA, Dempsey K, McCarthy JS. 2006. Utility of serological follow-up of chronic strongyloidiasis after anthelminthic chemotherapy. Trans R Soc Trop Med Hyg 100:1056–1062. doi: 10.1016/j.trstmh.2005.12.006 [DOI] [PubMed] [Google Scholar]
  • 160. Sears WJ, Nutman TB. 2022. Strongy detect: preliminary validation of a prototype recombinant SS-NIE/SS-IR based ELISA to detect Strongyloides stercoralis infection. PLoS Negl Trop Dis 16:e0010126. doi: 10.1371/journal.pntd.0010126 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Tamarozzi F, Longoni SS, Mazzi C, Pettene S, Montresor A, Mahanty S, Bisoffi Z, Buonfrate D. 2021. Diagnostic accuracy of a novel enzyme-linked immunoassay for the detection of IgG and IgG4 against Strongyloides stercoralis based on the recombinant antigens NIE/SsIR. Parasit Vectors 14:412. doi: 10.1186/s13071-021-04916-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Rodriguez EA, Abraham T, Williams FK. 2015. Severe strongyloidiasis with negative serology after corticosteroid treatment. Am J Case Rep 16:95–98. doi: 10.12659/AJCR.892759 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Osiro S, Hamula C, Glaser A, Rana M, Dunn D. 2017. A case of Strongyloides hyperinfection syndrome in the setting of persistent eosinophilia but negative serology. Diagn Microbiol Infect Dis 88:168–170. doi: 10.1016/j.diagmicrobio.2017.02.016 [DOI] [PubMed] [Google Scholar]
  • 164. Bisoffi Z, Buonfrate D, Sequi M, Mejia R, Cimino RO, Krolewiecki AJ, Albonico M, Gobbo M, Bonafini S, Angheben A, Requena-Mendez A, Muñoz J, Nutman TB, Siddiqui AA. 2014. Diagnostic accuracy of five serologic tests for Strongyloides stercoralis infection. PLoS Negl Trop Dis 8:e2640. doi: 10.1371/journal.pntd.0002640 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Rascoe LN, Price C, Shin SH, McAuliffe I, Priest JW, Handali S, Taylan Ozkan A. 2015. Development of Ss-NIE-1 recombinant antigen based assays for immunodiagnosis of strongyloidiasis. PLoS Negl Trop Dis 9:e0003694. doi: 10.1371/journal.pntd.0003694 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Santano R, Rubio R, Grau-Pujol B, Escola V, Muchisse O, Cuamba I, Vidal M, Ruiz-Olalla G, Aguilar R, Gandasegui J, et al. 2022. Evaluation of antibody serology to determine current helminth and Plasmodium falciparum infections in a co-endemic area in Southern Mozambique. PLoS Negl Trop Dis 16:e0010138. doi: 10.1371/journal.pntd.0010138 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167. Tamarozzi F, Longoni SS, Mazzi C, Rizzi E, Noordin R, Buonfrate D. 2022. The accuracy of a recombinant antigen immunochromatographic test for the detection of Strongyloides stercoralis infection in migrants from sub-Saharan Africa. Parasit Vectors 15:142. doi: 10.1186/s13071-022-05249-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Noordin R, Anuar NS, Juri NM, Wongphutorn P, Ruantip S, Kopolrat KY, Worasith C, Sithithaworn J, Sithithaworn P. 2021. Evaluation of a rapid IgG4 lateral flow dipstick test to detect Strongyloides stercoralis infection in northeast Thailand. Am J Trop Med Hyg 105:688–691. doi: 10.4269/ajtmh.21-0317 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Noordin R, Osman E, Kalantari N, Anuar NS, Gorgani-Firouzjaee T, Sithithaworn P, Juri NM, Rahumatullah A. 2022. A point-of-care cassette test for detection of Strongyloides stercoralis. Acta Trop 226:106251. doi: 10.1016/j.actatropica.2021.106251 [DOI] [PubMed] [Google Scholar]
  • 170. Tamarozzi F, Guevara ÁG, Anselmi M, Vicuña Y, Prandi R, Marquez M, Vivero S, Robinzón Huerlo F, Racines M, Mazzi C, Denwood M, Buonfrate D. 2023. Accuracy, acceptability, and feasibility of diagnostic tests for the screening of Strongyloides stercoralis in the field (ESTRELLA): a cross-sectional study in Ecuador. Lancet Glob Health 11:e740–e748. doi: 10.1016/S2214-109X(23)00108-0 [DOI] [PubMed] [Google Scholar]
  • 171. De Souza JN, Langford I, Wang Y, Soares NM, Handali S. 2021. Development of rSs-NIE-1 and rSs-IR recombinant antigen-based immunoblot for detection of antibody to Strongyloides stercoralis. Am J Trop Med Hyg 104:2038–2041. doi: 10.4269/ajtmh.20-1539 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Sykes AM, McCarthy JS. 2011. A coproantigen diagnostic test for Strongyloides infection. PLoS Negl Trop Dis 5:e955. doi: 10.1371/journal.pntd.0000955 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Buonfrate D, Perandin F, Formenti F, Bisoffi Z. 2017. A retrospective study comparing agar plate culture, indirect immunofluorescence and real-time PCR for the diagnosis of Strongyloides stercoralis infection. Parasitology 144:812–816. doi: 10.1017/S0031182016002559 [DOI] [PubMed] [Google Scholar]
  • 174. Schaffel R, Nucci M, Carvalho E, Braga M, Almeida L, Portugal R, Pulcheri W. 2001. The value of an immunoenzymatic test (enzyme-linked immunosorbent assay) for the diagnosis of strongyloidiasis in patients immunosuppressed by hematologic malignancies. Am J Trop Med Hyg 65:346–350. doi: 10.4269/ajtmh.2001.65.346 [DOI] [PubMed] [Google Scholar]
  • 175. Luvira V, Trakulhun K, Mungthin M, Naaglor T, Chantawat N, Pakdee W, Phiboonbanakit D, Dekumyoy P. 2016. Comparative diagnosis of strongyloidiasis in immunocompromised patients. Am J Trop Med Hyg 95:401–404. doi: 10.4269/ajtmh.16-0068 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176. Roxby AC, Gottlieb GS, Limaye AP. 2009. Strongyloidiasis in transplant patients. Clin Infect Dis 49:1411–1423. doi: 10.1086/630201 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Marathe A, Date V. 2008. Strongyloides stercoralis hyperinfection in an immunocompetent patient with extreme eosinophilia. J Parasitol 94:759–760. doi: 10.1645/GE-1392.1 [DOI] [PubMed] [Google Scholar]
  • 178. Buonfrate D, Salas-Coronas J, Muñoz J, Maruri BT, Rodari P, Castelli F, Zammarchi L, Bianchi L, Gobbi F, Cabezas-Fernández T, Requena-Mendez A, Godbole G, Silva R, Romero M, Chiodini PL, Bisoffi Z. 2019. Multiple-dose versus single-dose ivermectin for Strongyloides stercoralis infection (Strong Treat 1 to 4): a multicentre, open-label, phase 3, randomised controlled superiority trial. Lancet Infect Dis 19:1181–1190. doi: 10.1016/S1473-3099(19)30289-0 [DOI] [PubMed] [Google Scholar]
  • 179. Suputtamongkol Y, Premasathian N, Bhumimuang K, Waywa D, Nilganuwong S, Karuphong E, Anekthananon T, Wanachiwanawin D, Silpasakorn S, Siddiqui AA. 2011. Efficacy and safety of single and double doses of ivermectin versus 7-day high dose albendazole for chronic strongyloidiasis. PLoS Negl Trop Dis 5:e1044. doi: 10.1371/journal.pntd.0001044 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Henriquez-Camacho C, Gotuzzo E, Echevarria J, White AC, Terashima A, Samalvides F, Pérez-Molina JA, Plana MN. 2016. Ivermectin versus albendazole or thiabendazole for Strongyloides stercoralis infection. Cochrane Database Syst Rev 2016:CD007745. doi: 10.1002/14651858.CD007745.pub3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Barrett J, Newsholme W. 2016. Subcutaneous ivermectin use in the treatment of severe Strongyloides stercoralis infection: two case reports and a discussion of the literature. J Antimicrob Chemother 71:1131. doi: 10.1093/jac/dkw007 [DOI] [PubMed] [Google Scholar]
  • 182. Omura S. 2008. Ivermectin: 25 years and still going strong. Int J Antimicrob Agents 31:91–98. doi: 10.1016/j.ijantimicag.2007.08.023 [DOI] [PubMed] [Google Scholar]
  • 183. World Health Organization . 2021. Safety in administering medicines for neglected tropical diseases. licence: CC BY-NC-SA 3.0 IGO. Geneva [Google Scholar]
  • 184. Nicolas P, Maia MF, Bassat Q, Kobylinski KC, Monteiro W, Rabinovich NR, Menéndez C, Bardají A, Chaccour C. 2020. Safety of oral ivermectin during pregnancy: a systematic review and meta-analysis. Lancet Glob Health 8:e92–e100. doi: 10.1016/S2214-109X(19)30453-X [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Boggild AK, Libman M, Greenaway C, McCarthy AE, Committee to Advise on Tropical Medicine, Travel (CATMAT) . 2016. CATMAT statement on disseminated strongyloidiasis: prevention, assessment and management guidelines. Can Commun Dis Rep 42:12–19. doi: 10.14745/ccdr.v42i01a03 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Takayanagui OM, Bonato PS, Dreossi SAC, Lanchote VL. 2002. Enantioselective distribution of albendazole metabolites in cerebrospinal fluid of patients with neurocysticercosis. Br J Clin Pharmacol 54:125–130. doi: 10.1046/j.1365-2125.2002.01634.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Hofmann D, Smit C, Sayasone S, Pfister M, Keiser J. 2022. Optimizing moxidectin dosing for Strongyloides stercoralis infections: insights from pharmacometric modeling. Clin Transl Sci 15:700–708. doi: 10.1111/cts.13189 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Pankavich JA, Berger H, Simkins KL. 1992. Efficacy of moxidectin, nemadectin and ivermectin against an ivermectin-resistant strain of Haemonchus contortus in sheep. Vet Rec 130:241–242. doi: 10.1136/vr.130.12.241 [DOI] [PubMed] [Google Scholar]
  • 189. Gandasegui J, Onwuchekwa C, Krolewiecki AJ, Doyle SR, Pullan RL, Enbiale W, Kepha S, Hatherell HA, van Lieshout L, Cambra-Pellejà M, Escola V, Muñoz J. 2022. Ivermectin and albendazole coadministration: opportunities for strongyloidiasis control. Lancet Infect Dis 22:e341–e347. doi: 10.1016/S1473-3099(22)00369-3 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Legend of video. cmr.00033-23-s0001.docx.

Description of Strongyloides larvae video.

cmr.00033-23-s0001.docx (12.2KB, docx)
DOI: 10.1128/cmr.00033-23.SuF1
Video: Strongyloides larvae. cmr.00033-23-s0002.avi.

Highly motile Strongyloides stercoralis larvae on the surface of an agar plate culture.

Download video file (4.1MB, avi)
DOI: 10.1128/cmr.00033-23.SuF2

Articles from Clinical Microbiology Reviews are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES