Hostname: page-component-76fb5796d-9pm4c Total loading time: 0 Render date: 2024-04-28T01:23:03.133Z Has data issue: false hasContentIssue false

Human infection with Strongyloides stercoralis and other related Strongyloides species

Published online by Cambridge University Press:  16 May 2016

THOMAS B. NUTMAN*
Affiliation:
Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 4 – Room B1-03, 4 Center Dr., Bethesda, MD 20892-0425, USA
*
*Corresponding author: Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 4 – Room B1-03, 4 Center Dr., Bethesda, MD 20892-0425, USA. Tel: 301-496-5399. Fax: 301-480-3757. E-mail: tnutman@niaid.nih.gov

Summary

The majority of the 30–100 million people infected with Strongyloides stercoralis, a soil transmitted intestinal nematode, have subclinical (or asymptomatic) infections. These infections are commonly chronic and longstanding because of the autoinfective process associated with its unique life cycle. A change in immune status can increase parasite numbers, leading to hyperinfection syndrome, dissemination, and death if unrecognized. Corticosteroid use and HTLV-1 infection are most commonly associated with the hyperinfection syndrome. Strongyloides adult parasites reside in the small intestine and induce immune responses both local and systemic that remain poorly characterized. Definitive diagnosis of S. stercoralis infection is based on stool examinations for larvae, but newer diagnostics – including new immunoassays and molecular tests – will assume primacy in the next few years. Although good treatment options exist for infection and control of this infection might be possible, S. stercoralis remains largely neglected.

Type
Special Issue Review
Copyright
Copyright © Cambridge University Press 2016 

INTRODUCTION

Strongyloidiasis, the disease caused by the infection with Strongyloides stercoralis, and to a lesser extent by Strongyloides fuelleborni fuelleborni and S. fuelleborni kelleyi, is a soil-transmitted helminthiasis with an estimated 30–100 million people infected worldwide (Genta, Reference Genta1989; Schar et al. Reference Schar, Trostdorf, Giardina, Khieu, Muth, Marti, Vounatsou and Odermatt2013). Although the burden of the disease has been felt to be underestimated (Viney and Lok, Reference Viney and Lok2007; Olsen et al. Reference Olsen, van Lieshout, Marti, Polderman, Polman, Steinmann, Stothard, Thybo, Verweij and Magnussen2009; Schar et al. Reference Schar, Giardina, Khieu, Muth, Vounatsou, Marti and Odermatt2015, Reference Schar, Inpankaew, Traub, Khieu, Dalsgaard, Chimnoi, Chhoun, Sok, Marti, Muth and Odermatt2014), S. stercoralis infections in humans range from asymptomatic light infections to chronic symptomatic strongyloidiasis. However, uncontrolled multiplication of the parasite (hyperinfection) and potentially life-threatening dissemination of larvae in immunocompromised patients result in mortality rates of up to 85% (Keiser and Nutman, Reference Keiser and Nutman2004; Mejia and Nutman, Reference Mejia and Nutman2012).

The parasite, occurring naturally in dogs, primates and humans, is endemic to the tropics and subtropics; foci of infection occur in temperate regions as well (Genta, Reference Genta1989; Schar et al. Reference Schar, Trostdorf, Giardina, Khieu, Muth, Marti, Vounatsou and Odermatt2013) where poor sanitation or other factors facilitate the transmission through fecal contamination. In parts of Africa and in Papua New Guinea, human infections caused by S. fuelleborni fuelleborni and S. fuelleborni kelleyi respectively have been reported (Pampiglione and Ricciardi, Reference Pampiglione and Ricciardi1971; Hira and Patel, Reference Hira and Patel1977; Vince et al. Reference Vince, Ashford, Gratten and Bana-Koiri1979; Crouch and Shield, Reference Crouch and Shield1982; Evans et al. Reference Evans, Markus, Joubert and Gunders1991; Freedman, Reference Freedman1991; Ashford et al. Reference Ashford, Barnish and Viney1992). In Africa, S. fuelleborni fuelleborni is primarily a parasite of primates, but in Papua New Guinea no animal host has been demonstrated for S. fuelleborni kelleyi (Ashford et al. Reference Ashford, Barnish and Viney1992; Viney and Lok, Reference Viney and Lok2007).

Strongyloides stercoralis is unique among nematodes infectious for humans in that larvae passing in the feces can give rise to a free-living generation of worms which, in turn, give rise to infective larvae. This so-called heterogonic development process serves as an amplification mechanism that allows for increased numbers of infective larvae in the external environment. The infective larvae are active skin penetrators; infection per os, while possible, is probably of limited importance. Because the parasitic female's eggs hatch often within the gastrointestinal tract, the potential for autoinfection exists when precociously developing larvae attain infectivity while still in the host. When the rate of autoinfection escapes control by the host, massive re-penetration and larval migration may result.

LIFE CYCLE

The S. stercoralis (and S. fuelleborni spp.) life cycle encompasses both free-living and parasitic stages. Adult female worms parasitizing the human small intestine lay eggs in the intestinal mucosa that hatch into rhabditiform larvae, which are shed in the stool. In the environment, under warm moist conditions that often characterize the tropical and subtropical endemic areas, rhabditiform larvae can either moult into infective filariform larvae or develop through succeeding rhabditiform stages into free-living adults. Sexual reproduction occurs exclusively in the free-living stage.

Humans are generally infected transcutaneously, although infection has also been experimentally induced by oral administration of water contaminated with filariform larvae (Grove, Reference Grove1996). After dermal penetration, the filariform larvae, through undefined mechanisms, migrate to the small intestine. The most clinically relevant, though perhaps not the predominant (Mansfield et al. Reference Mansfield, Niamatali, Bhopale, Volk, Smith, Lok, Genta and Schad1996), migration is the classic pulmonary route, in which organisms enter the bloodstream and are carried to the lungs, ascending the tracheobronchial tree to enter the gastrointestinal tract. Only female adults are detectable in humans and subsequent reproduction occurs asexually through parthenogenesis (Neva, Reference Neva1986).

Some rhabditiform larvae transform into invasive filariform larvae before being excreted. As such, they are capable of re-infecting the host by invading the intestinal wall or the perianal skin (Grove, Reference Grove1996). This autoinfective cycle can occur at a low level throughout infection and allows subsequent generations to persist in the host indefinitely (Neva, Reference Neva1986).

In the immunocompetent host, it is felt that cellular immune effector mechanisms and intrinsic parasite biology both serve to regulate the population density of adult worms in the intestine. With an alteration in host immune responsiveness, even one adult female can multiply rapidly by parthenogenesis, leading to accelerated autoinfection and/or dissemination.

EPIDEMIOLOGY

While endemic to the tropics and subtropics, foci of infection occur in temperate regions such as Japan, Italy, Australia and the USA (Genta, Reference Genta1989; Al-Hasan et al. Reference Al-Hasan, McCormick and Ribes2007; Schar et al. Reference Schar, Trostdorf, Giardina, Khieu, Muth, Marti, Vounatsou and Odermatt2013). Immigrants and refugees comprise a significant population at risk for strongyloidiasis in high- and middle-income countries (Posey et al. Reference Posey, Blackburn, Weinberg, Flagg, Ortega, Wilson, Secor, Sanders-Lewis, Won and Maguire2007; Schar et al. Reference Schar, Trostdorf, Giardina, Khieu, Muth, Marti, Vounatsou and Odermatt2013).

Prevalence and global distribution

There is little consensus about prevalence rates and the global distribution of human infections with S. stercoralis. There is, however, a great degree of consensus about the fact that the prevalence of strongyloidiasis has long been underestimated (Olsen et al. Reference Olsen, van Lieshout, Marti, Polderman, Polman, Steinmann, Stothard, Thybo, Verweij and Magnussen2009; Schar et al. Reference Schar, Trostdorf, Giardina, Khieu, Muth, Marti, Vounatsou and Odermatt2013; Khieu et al. Reference Khieu, Schar, Forrer, Hattendorf, Marti, Duong, Vounatsou, Muth and Odermatt2014; Toledo et al. Reference Toledo, Munoz-Antoli and Esteban2015). Although prevalence and global distribution patterns have been recently examined, aggregated detailed distribution maps and country by country data [cf. (Schar et al. Reference Schar, Trostdorf, Giardina, Khieu, Muth, Marti, Vounatsou and Odermatt2013; Toledo et al. Reference Toledo, Munoz-Antoli and Esteban2015)] are beyond the scope of this review

Transmission

While Strongyloides is most commonly acquired transcutaneously, high prevalence rates in institutionalized subjects raise speculation about alternate routes of transmission (Yoeli et al. Reference Yoeli, Most, Hammond and Scheinesson1972; Gatti et al. Reference Gatti, Lopes, Cevini, Ijaoba, Bruno, Bernuzzi, de Lio, Monco and Scaglia2000; Robson et al. Reference Robson, Welch, Beeching and Gill2009). A Japanese study found support to this claim by observing a higher prevalence of Strongyloides infection in patients with Blastocystis hominis, a protozoan acquired by the fecal oral route (Czachor and Jonas, Reference Czachor and Jonas2000). However, standard rather than strict contact precautions appear sufficient for prevention of nosocomial transmission based on case reports of patients with disseminated disease (Sugiyama et al. Reference Sugiyama, Hasegawa, Nagasawa and Hitomi2006). Transmission of Strongyloides infection after transplantation of kidneys, pancreatic allograft or intestines has been suggested by several reports where donors but not recipients had a history of travel to a Strongyloides endemic regions of the world (Ben-Youssef et al. Reference Ben-Youssef, Baron, Edson, Raghavan and Okechukwu2005; Said et al. Reference Said, Nampoory, Nair, Halim, Shetty, Kumar, Mokadas, Elsayed, Johny, Samhan and Al-Mousawi2007; Patel et al. Reference Patel, Arvelakis, Sauter, Gondolesi, Caplivski and Huprikar2008) (see section below).

CLINICAL MANIFESTATIONS

Acute Strongyloidiasis

The clinical manifestations of acute strongyloidiasis are associated with the path of larval migration to the small intestine. Infected individuals may experience irritation at the site of skin penetration by larvae followed occasionally by localized oedema or urticaria. Within a week following infection, a dry cough and/or tracheal irritation may occur. Gastrointestinal symptoms such as diarrhoea, constipation, abdominal pain, or anorexia can occur (Keiser and Nutman, Reference Keiser and Nutman2004) following the establishment of the infection in the small intestine.

Chronic Strongyloidiasis

Chronic infection with S. stercoralis is most often clinically asymptomatic (Grove, Reference Grove1989). Since up to 75% of persons may have peripheral eosinophilia or elevated IgE levels (Rossi et al. Reference Rossi, Takahashi, Partel, Teodoro and da Silva1993), Strongyloides should be considered in the differential diagnosis of high grade and/or persistent eosinophilia in travellers or expatriates from endemic areas (O'Connell and Nutman, Reference O'Connell and Nutman2015).

Symptomatic individuals may complain of diarrhoea, constipation, intermittent vomiting or borborygmus. Dermatologic manifestations such as recurrent urticaria can occur (Leighton and MacSween, Reference Leighton and MacSween1990) as can larva currens (pruritic linear streaks located along the lower trunk, thighs and buttocks) as a result of migrating larvae (Pelletier, Reference Pelletier1984; Pelletier and Gabre-Kidan, Reference Pelletier and Gabre-Kidan1985; Grove, Reference Grove1996). Unusual manifestations of chronic strongyloidiasis include arthritis (Richter et al. Reference Richter, Muller-Stover, Strothmeyer, Gobels, Schmitt and Haussinger2006); nephrotic syndrome (Hsieh et al. Reference Hsieh, Wen and Chen2006), chronic malabsorption (Atul et al. Reference Atul, Ajay, Ritambhara, Harsh and Ashish2005), duodenal obstruction (Harish et al. Reference Harish, Sunilkumar, Varghese and Feroze2005; Suvarna et al. Reference Suvarna, Mehta, Sadasivan, Raj and Balakrishnan2005), focal hepatic lesions (Gulbas et al. Reference Gulbas, Kebapci, Pasaoglu and Vardareli2004) and recurrent asthma (Tullis, Reference Tullis1970; Dunlap et al. Reference Dunlap, Shin, Polt and Ho1984).

Hyperinfection syndrome/disseminated infections

Hyperinfection describes the syndrome of accelerated autoinfection, generally – although not always (Husni et al. Reference Husni, Gordon, Longworth and Adal1996; Tiwari et al. Reference Tiwari, Rautaraya and Tripathy2012; Dogan et al. Reference Dogan, Gayaf, Ozsoz, Sahin, Aksel, Karasu, Aydogdu and Turgay2014) – the result of an alteration in immune status. The distinction between autoinfection and hyperinfection is quantitative and not strictly defined. Therefore, hyperinfection syndrome implies the presence of signs and symptoms attributable to increased larval migration. Development or exacerbation of gastrointestinal and pulmonary symptoms is seen, and the detection of increased numbers of larvae in stool and/or sputum is the hallmark of hyperinfection. Larvae in non-disseminated hyperinfection are increased in numbers but confined to the organs normally involved in the pulmonary autoinfective cycle (i.e. gastrointestinal tract, peritoneum and lungs), although enteric bacteria, that can be carried by the filariform larvae or gain systemic access through intestinal ulcers, may affect any organ system.

The term disseminated infection is often used to refer to migration of larvae to organs beyond the range of the pulmonary autoinfective cycle. This does not necessarily imply a greater severity of disease. Extra-pulmonary migration of larvae has been shown to occur routinely during the course of chronic S. stercoralis infections in experimental dogs (Schad et al. Reference Schad, Aikens and Smith1989) and has been reported to cause symptoms in humans without other manifestations of hyperinfection syndrome (Lai et al. Reference Lai, Hsu, Wang and Lin2002). Similarly, many cases of hyperinfection are fatal without larvae being detected outside the pulmonary autoinfective route.

General features

The clinical manifestations of S. stercoralis hyperinfection vary widely. The onset may be acute (Thomas and Costello, Reference Thomas and Costello1998) or insidious (Wurtz et al. Reference Wurtz, Mirot, Fronda, Peters and Kocka1994). Fever and chills are not uniformly present and should prompt a search for an associated bacterial infection. Other constitutional symptoms include fatigue (Liepman, Reference Liepman1975), weakness (Chu et al. Reference Chu, Whitlock and Dietrich1990) and total body pain (Chaudhuri et al. Reference Chaudhuri, Nanos, Soco and McGrew1980). Blood counts performed during hyperinfection may show eosinophilia but more often show a suppressed eosinophil count (Grove, Reference Grove1996). Patients who have increased peripheral eosinophilia during hyperinfection appear to have a better prognosis (Jamil and Hilton, Reference Jamil and Hilton1992).

Gastrointestinal manifestations

Gastrointestinal symptoms are common but are non-specific. Some case reports do not mention any gastrointestinal symptoms (Liepman, Reference Liepman1975). Abdominal pain (Celedon et al. Reference Celedon, Mathur-Wagh, Fox, Garcia and Wiest1994), often described as crampy or bloating in nature, watery diarrhoea, constipation anorexia, weight loss (Scowden et al. Reference Scowden, Schaffner and Stone1978), difficulty swallowing (Yee et al. Reference Yee, Boylen, Noguchi, Klatt and Sharma1987), sore throat, nausea (Liepman, Reference Liepman1975), vomiting and gastrointestinal bleeding, and small bowel obstruction (Newton et al. Reference Newton, Limpuangthip, Greenberg, Gam and Neva1992; Thomas and Costello, Reference Thomas and Costello1998) may result, with diffuse abdominal tenderness and hypoactive bowel sounds. Protein-losing enteropathy may give rise to acute or worsening hypoalbuminaemia with peripheral oedema (Ho et al. Reference Ho, Luk, Chan and Yuen1997; Yoshida et al. Reference Yoshida, Endo, Tanaka, Ishikawa, Kondo and Nakamura2006) or ascites (Liepman, Reference Liepman1975). Hypokalaemia (Jain et al. Reference Jain, Agarwal and el-Sadr1994) or other electrolyte abnormalities may reflect these gastrointestinal disturbances. Direct stool exam usually shows numerous rhabditiform and filariform larvae. Occasionally, adult worms (Ho et al. Reference Ho, Luk, Chan and Yuen1997) and eggs (Armignacco et al. Reference Armignacco, Capecchi, De Mori and Grillo1989; Cahill and Shevchuk, Reference Cahill and Shevchuk1996) are also seen. Occult or gross blood is a common finding. Esophagitis and gastritis are reported, in addition to duodenitis, jejunitis, ileitis, colitis, including pseudomembranous colitis and proctitis. Mucosal ulceration is most common in the small intestine, but can occur at any level from the oesophagus (Levi et al. Reference Levi, Kallas and Ramos Moreira Leite1997) and stomach (Wurtz et al. Reference Wurtz, Mirot, Fronda, Peters and Kocka1994) to the rectum. Larvae may be seen in these ulcers on biopsy (Gompels et al. Reference Gompels, Todd, Peters, Main and Pinching1991; Wurtz et al. Reference Wurtz, Mirot, Fronda, Peters and Kocka1994; Ho et al. Reference Ho, Luk, Chan and Yuen1997). Crypts are often distorted by the numerous larvae (Wurtz et al. Reference Wurtz, Mirot, Fronda, Peters and Kocka1994) Inflammatory infiltrates (Mori et al. Reference Mori, Konishi, Matsuoka, Deguchi, Ohta, Mizuno, Ueno, Okinaka, Nishimura, Ito and Nakano1998) or areas of necrosis (Neefe et al. Reference Neefe, Pinilla, Garagusi and Bauer1973; Yee et al. Reference Yee, Boylen, Noguchi, Klatt and Sharma1987) in involved intestinal mucosa may be present (Newton et al. Reference Newton, Limpuangthip, Greenberg, Gam and Neva1992). The appendix may also be invaded by larvae (Scowden et al. Reference Scowden, Schaffner and Stone1978; Kramer et al. Reference Kramer, Gregg, Goldstein, Llamas and Krieger1990). Abdominal imaging may show small bowel distension with air-fluid levels (Newton et al. Reference Newton, Limpuangthip, Greenberg, Gam and Neva1992; Celedon et al. Reference Celedon, Mathur-Wagh, Fox, Garcia and Wiest1994). Mucosal oedema (Neefe et al. Reference Neefe, Pinilla, Garagusi and Bauer1973; Mori et al. Reference Mori, Konishi, Matsuoka, Deguchi, Ohta, Mizuno, Ueno, Okinaka, Nishimura, Ito and Nakano1998) and findings consistent with protein-losing enteropathy may also be demonstrated radiographically. Computed tomography scans can occasionally reveal intra-abdominal lymphadenopathy (Thomas and Costello, Reference Thomas and Costello1998).

Cardiopulmonary manifestations

Cardiopulmonary manifestations range from none at all to cough (Nomura and Rekrut, Reference Nomura and Rekrut1996), wheezing (Kramer et al. Reference Kramer, Gregg, Goldstein, Llamas and Krieger1990), (Yee et al. Reference Yee, Boylen, Noguchi, Klatt and Sharma1987), a choking sensation (Cahill  and Shevchuk, Reference Cahill and Shevchuk1996), hoarseness (Yee et al. Reference Yee, Boylen, Noguchi, Klatt and Sharma1987), chest pain (Chaudhuri et al. Reference Chaudhuri, Nanos, Soco and McGrew1980; Cahill  and Shevchuk, Reference Cahill and Shevchuk1996), haemoptysis, palpitations, atrial fibrillation (Gordon et al. Reference Gordon, Gal, Solomon and Bryan1994), dyspnoea (Nomura  and Rekrut, Reference Nomura and Rekrut1996), and, rarely, respiratory collapse. Respiratory alkalosis is common (Thompson  and Berger, Reference Thompson and Berger1991). Pneumothorax is rarely seen (McNeely et al. Reference McNeely, Inouye, Tam and Ripley1980). Sputum may demonstrate filariform or rhabditiform larvae and even, occasionally, eggs (Kennedy et al. Reference Kennedy, Campbell, Lawrence, Nichol and Rao1989). These findings suggest that filariform larvae develop into adults in the lungs, and a new generation of rhabditiform larvae is produced locally (Cirioni et al. Reference Cirioni, Giacometti, Burzacchini, Balducci and Scalise1996). This hypothesis is supported by reports of adult parasites being expectorated post treatment (McLarnon  and Ma, Reference McLarnon and Ma1981) and autopsy studies showing adult worms in lung tissue (Cahill  and Shevchuk, Reference Cahill and Shevchuk1996). Chest imaging most frequently show bilateral or focal interstitial infiltrates. Lung tissues may show alveolar haemorrhage. Petechial haemorrhage or hyperaemia of the bronchial, tracheal and laryngeal mucosa has also been reported (Yee et al. Reference Yee, Boylen, Noguchi, Klatt and Sharma1987; Cahill  and Shevchuk, Reference Cahill and Shevchuk1996).

Dermatologic manifestations

Pruritic linear streaks of the lower trunk, thighs and buttocks (larva currens) frequently accompany hyperinfection (Ho et al. Reference Ho, Luk, Chan and Yuen1997). Petechial and purpuric rashes of these same areas, in which larvae have been demonstrated on skin biopsy is common (Ronan et al. Reference Ronan, Reddy, Manaligod, Alexander and Fu1989; Stewart et al. Reference Stewart, Ramanathan, Mahanty, Fedorko, Janik and Morris2011). Skin manifestations of vasculitis (Harcourt-Webster et al. Reference Harcourt-Webster, Scaravilli and Darwish1991) or of disseminated intravascular coagulation seen associated with

Gram-negative sepsis (Neefe et al. Reference Neefe, Pinilla, Garagusi and Bauer1973) may, of course, also present during hyperinfection.

Central nervous system (CNS) manifestations

Meningeal signs and symptoms (Kramer et al. Reference Kramer, Gregg, Goldstein, Llamas and Krieger1990) are the most common manifestation of CNS involvement in hyperinfection syndrome. Hyponatremia may accompany meningitis (Harcourt-Webster et al. Reference Harcourt-Webster, Scaravilli and Darwish1991; Jain et al. Reference Jain, Agarwal and el-Sadr1994). In patients with meningitis, spinal fluid may show parameters of aseptic meningitis [i.e. pleocytosis, elevated protein, normal glucose, negative bacterial cultures (Scowden et al. Reference Scowden, Schaffner and Stone1978; Vishwanath et al. Reference Vishwanath, Baker and Mansheim1982)] or demonstrate characteristics of a Gram-negative bacterial infection. Larvae have been found in spinal fluid (Dutcher et al. Reference Dutcher, Marcus, Tanowitz, Wittner, Fuks and Wiernik1990), meningeal vessels (Cahill  and Shevchuk, Reference Cahill and Shevchuk1996), dura, epidural, subdural and subarachnoid spaces (Neefe et al. Reference Neefe, Pinilla, Garagusi and Bauer1973). Eosinophilic meningitis has not been reported.

Sepsis

Hyperinfection syndrome/disseminated are often complicated, and rarely preceded by infections caused by gut flora that gain access to extraintestinal sites, presumably through ulcers induced by the filariform larvae or by virtue of being carried on the surface or in the intestinal tract of larvae themselves. Organisms that have been reported to cause sepsis in such patients include Group D Streptococci, Candida Streptococcus bovis, Escherichia coli, Klebsiella pneumoniae, Proteus mirabilis, Pseudomonas, Enterococcus faecalis, coagulase negative staphylococci and Streptococcus pneumoniae. Polymicrobial infections can also occur (Link  and Orenstein, Reference Link and Orenstein1999).

Disseminated infections

Organs to which larvae have disseminated include skin, mesenteric lymph nodes, gallbladder, liver, diaphragm, heart, pancreas, skeletal muscle, kidneys, ovaries and brain (Keiser  and Nutman, Reference Keiser and Nutman2004) based largely on autopsy studies. Chronic inflammation or necrosis frequently surrounds the larvae, but tissue reactions are also frequently absent (Neefe et al. Reference Neefe, Pinilla, Garagusi and Bauer1973; Takayanagui et al. Reference Takayanagui, Lofrano, Araugo and Chimelli1995).

Conditions associated with hyperinfection syndrome and dissemination (Table 1)

Corticosteroids and other agents

Corticosteroids (most commonly prednisone and methyl-prednisilone) have a particularly strong and specific association with the development of hyperinfection syndrome and dissemination. Beyond their known (and broad) effects on the host immune system, it has been postulated that corticosteroids have a direct effect on the S. stercoralis parasite (Genta, Reference Genta1992; Ramanathan et al. Reference Ramanathan, Varma, Ribeiro, Myers, Nolan, Abraham, Lok and Nutman2011) though this has not been shown definitively. Other immunosuppressive therapies and underlying conditions (Table 1) may also predispose to dissemination. However, the concomitant administration of corticosteroids in most of these other conditions makes it difficult to assign a direct causal association. Hyperinfection syndrome has been described regardless of dose, duration or route of administration of corticosteroids. Even short courses (6–17 days) of corticosteroids in immunocompetent patients without underlying immunosuppressive conditions have even been associated with hyperinfection syndrome and death (Ghosh  and Ghosh, Reference Ghosh and Ghosh2007).

Table 1. Conditions associated with hyperinfection syndrome

HTLV-1 Infection

Human T-cell lymphotropic virus type 1 (HTLV-1) represents a significant risk factor for the development of hyperinfection syndrome or disseminated strongyloidiasis (Carvalho  and Da Fonseca Porto, Reference Carvalho and Da Fonseca Porto2004) that may be related to HTLV-I driven alterations in IgE or associated Type-2 responses (Neva et al. Reference Neva, Filho, Gam, Thompson, Freitas, Melo and Carvalho1998; Porto et al. Reference Porto, Neva, Bittencourt, Lisboa, Thompson, Alcantara and Carvalho2001; Mitre et al. Reference Mitre, Thompson, Carvalho, Nutman and Neva2003; Santos et al. Reference Santos, Porto, Muniz, Jesus and Carvalho2004). A growing body of evidence points to the synergistic relationship between HTLV-1 and S. stercoralis. Higher rates of S. stercoralis infection have been found in HTLV-1 patients (Carvalho  and Da Fonseca Porto, Reference Carvalho and Da Fonseca Porto2004). Strongyloides stercoralis infection has been shown to influence the natural history of HTLV-1 infection (Marcos et al. Reference Marcos, Terashima, Canales and Gotuzzo2011) and has been considered a co-factor in the development of HTLV-1-associated diseases (Gotuzzo et al. Reference Gotuzzo, Arango, de Queiroz-Campos and Isturiz2000).

HIV

Strongyloidiasis was once considered an AIDS defining illness (Keiser  and Nutman, Reference Keiser and Nutman2004) yet there is no evidence that a low CD4 count will increase the risk of dissemination or decrease the chance of clearing an infection (Walson et al. Reference Walson, Stewart, Sangare, Mbogo, Otieno, Piper, Richardson and John-Stewart2010). Severe infection with Strongyloides has not been observed frequently with HIV-infected patients (Celedon et al. Reference Celedon, Mathur-Wagh, Fox, Garcia and Wiest1994). Hyperinfection syndrome is associated with the use of corticosteroids in the treatment of immune reconstitution inflammatory syndrome (IRIS) (Brown et al. Reference Brown, Cartledge and Miller2006; Mascarello et al. Reference Mascarello, Gobbi, Angheben, Gobbo, Gaiera, Pegoraro, Lanzafame, Buonfrate, Concia and Bisoffi2011). Whether IRIS occurs after the initiation of antiretroviral therapy in patients with single infections with S. stercoralis remains unclear.

Strongyloides infection in the transplanted patient

Solid organ transplants (Stone and Schaffner, Reference Stone and Schaffner1990; Lichtenberger et al. Reference Lichtenberger, Rosa-Cunha, Morris, Nishida, Akpinar, Gaitan, Tzakis and Doblecki-Lewis2009; Mokaddas et al. Reference Mokaddas, Shati, Abdulla, Nampoori, Iqbal, Nair, Said, Abdulhalim and Hira2009) haematopoietic stem cell transplants (HSCT) and their pre-conditioning regimens and subsequent immunosuppression have been linked to dissemination of S. stercoralis. Among the different types of transplants, HSCT has the highest incidence of fatal dissemination with a higher mortality than in other types of transplants (Wirk  and Wingard, Reference Wirk and Wingard2009). A unique complication of transplants is the development of graft vs host disease (GVHD). In HSCT the risk of GVHD is greater than for other types of transplants because of the use of allogeneic stem cells (non-ablative conditioning). Because the main therapy for acute GVHD is corticosteroids, it is at the time that steroids are given in the setting of chronic strongyloidiasis that the risk for dissemination is high (Choi  and Reddy, Reference Choi and Reddy2014).

The geographical proximity to either North America or Europe by immigrants from Central and South America and Africa that are being transplanted are a sizeable ‘at risk’ population for dissemination of S. stercoralis (Wolfe et al. Reference Wolfe, Roys and Merion2010; Guermani et al. Reference Guermani, Potenza, Isnardi, Peluso, Bosco and Donadio2013). Organ donors have also been shown to transmit Strongyloides infection with cases of solid organ transplant-associated S. stercoralis infections having been reported (Weiser et al. Reference Weiser, Scully, Bulman, Husain and Grossman2011).

Other

Several case reports have supported an association between S. stercoralis infection hypogammaglobulinaemia associated with multiple myeloma and nephrotic syndrome (Seet et al. Reference Seet, Lau and Tambyah2005; Hsieh et al. Reference Hsieh, Wen and Chen2006; Yassin et al. Reference Yassin, El Omri, Al-Hijji, Taha, Hassan, Aboudi and El-Ayoubi2010).

HUMAN IMMUNE RESPONSES AND PROTECTIVE IMMUNITY

The human immune response to S. stercoralis has not been studied in great detail. Most of our knowledge about immune responsiveness and protective immunity has come from animal studies [reviewed in this issue (Breloer  and Abraham, Reference Breloer and Abraham2016)]. These studies, that have as an added benefit the knowledge of exactly when the infection was initiated, have suggested a role for antibodies (of many isotypes) as well as for innate and adaptive immune responses in mediating resistance to infection (Fig. 1).

Fig. 1. Immune responses in Strongyloides stercoralis infection as a function of time after infection initiation. The infective L3 larval parasites initiate infection at skin sites and activate a variety of different cell types such as innate lymphoid cells (ILCs), macrophages (MAC), dendritic cells (DCs), natural killer cells (NK), eosinophils (Eos) and basophils/mast cells (Baso/MC). At this relatively early phase of infection (or by the time the adult worms are established in the small intestine) the parasite induces the differentiation of a small number of effector Th1/Th17 and a relatively larger number of Th2 cells which together with IgE antibody, may lead to attrition of some of the parasites. At the time of patency (when larval production occurs) there is an expansion of Th2/Th9 CD4+ cells, a further contraction of Th1/Th17 cells and the induction of alternatively activated macrophages (AAM). With the evolution of chronic longstanding infection, there is an associated expansion of IL-10- and/or TGFβ-producing regulatory T cells (Tregs) and a small contraction of Th2/Th9 cells.

In humans, it has been shown, however, that Th2 response are essential to protect against hyperinfection (Porto et al. Reference Porto, Neva, Bittencourt, Lisboa, Thompson, Alcantara and Carvalho2001; Iriemenam et al. Reference Iriemenam, Sanyaolu, Oyibo and Fagbenro-Beyioku2010) and that individuals with strongyloidiasis develop S. stercoralis-specific antibodies of the IgM, IgG, IgA and IgE isotypes (McRury et al. Reference McRury, De Messias, Walzer, Huitger and Genta1986; Genta  and Lillibridge, Reference Genta and Lillibridge1989; Atkins et al. Reference Atkins, Lindo, Lee, Conway, Bailey, Robinson and Bundy1997; Rodrigues et al. Reference Rodrigues, de Oliveira, Sopelete, Silva, Campos, Taketomi and Costa-Cruz2007).

The evolution of the antibody response in S. stercoralis infection has been difficult to discern given that only cross-sectional studies of infected people have been performed. Nevertheless, these studies of S. stercoralis infection have suggested that there is rapid induction of parasite-specific IgE, IgG1, IgG2 and IgG3 antibodies directed against crude S. stercoralis soluble extracts that is followed (often weeks to months later) with a rise in parasite-specific IgG4. In that the IgE and IgG4 antibodies often are directed at a similar, but restricted, set of antigens (Genta  and Lillibridge, Reference Genta and Lillibridge1989), it is the IgG4 antibodies that allow for the blocking of IgE-mediated effector responses (Genta et al. Reference Genta, Ottesen, Poindexter, Gam, Neva, Tanowitz and Wittner1983; Barrett et al. Reference Barrett, Neva, Gam, Cicmanec, London, Phillips and Metcalfe1988; Genta  and Lillibridge, Reference Genta and Lillibridge1989) thereby modulating some of the Type-2-mediated inflammation.

Recent work has suggested that once S. stercoralis establishes patency [usually within 6–7 weeks following infection (Freedman, Reference Freedman1991)] that the infection drives a systemic cytokine response that is dominated by Th2-associated and anti-inflammatory cytokines (Anuradha et al. Reference Anuradha, Munisankar, Bhootra, Jagannathan, Dolla, Kumaran, Shen, Nutman and Babu2015a ) (Fig. 1). This systemic response appears to reflect an expansion of antigen-specific Th2/Th9 cells with a concomitant contraction of Th1 and Th17 cells, the latter being dependent on IL-10 (George et al. Reference George, Anuradha, Kumar, Sridhar, Banurekha, Nutman and Babu2014; Anuradha et al. Reference Anuradha, Munisankar, Bhootra, Jagannathan, Dolla, Kumaran, Nutman and Babu2016, Reference Anuradha, Munisankar, Dolla, Kumaran, Nutman and Babu2015b ). With appropriate anthelmintic therapy leading to cure of the S. stercoralis infection, many of these cytokine levels and T-cell perturbations return to their homeostatic state (Anuradha et al. Reference Anuradha, Munisankar, Bhootra, Jagannathan, Dolla, Kumaran, Shen, Nutman and Babu2015a , Reference Anuradha, Munisankar, Dolla, Kumaran, Nutman and Babu b ).

Like many other systemic helminth infections (e.g. Schistosoma mansoni, Wuchereria bancrofti), S. stercoralis also, given its capacity for chronic longstanding infection, can modulate responses to bystander antigens particularly in the context of infection with other pathogens such as Mycobacterium tuberculosis (George et al. Reference George, Pavan Kumar, Jaganathan, Dolla, Kumaran, Nair, Banurekha, Shen, Nutman and Babu2015) and HTLV-1 (Mitre et al. Reference Mitre, Thompson, Carvalho, Nutman and Neva2003; Montes et al. Reference Montes, Sanchez, Verdonck, Lake, Gonzalez, Lopez, Terashima, Nolan, Lewis, Gotuzzo and White2009; Salles et al. Reference Salles, Bacellar, Amorim, Orge, Sundberg, Lima, Santos, Porto and Carvalho2013).

DIAGNOSIS

For the chronically infected, asymptomatic individual, diagnosis of strongyloidiasis can be challenging (Levenhagen  and Costa-Cruz, Reference Levenhagen and Costa-Cruz2014; Buonfrate et al. Reference Buonfrate, Formenti, Perandin and Bisoffi2015; Toledo et al. Reference Toledo, Munoz-Antoli and Esteban2015). Diagnosis of hyperinfection syndrome/disseminated S. stercoralis infection is much less difficult given the large numbers of larvae often seen in the stool or other bodily fluids including CSF, pleural fluid, bronchoalveolar lavage fluid.

Parasitological methods

Definitive diagnosis relies on detection of larvae in the stool. However, intermittent and scanty excretion of larvae limits the utility of standard stool studies. Various investigators have attempted to improve the diagnostic yield of stool examination using techniques such as direct smear of feces in saline/Lugol's iodine stain, Baermann concentration, Harada-Mori filter paper culture, quantitative formalin ethyl acetate concentration technique and nutrient agar plate cultures [see (Sato et al. Reference Sato, Kobayashi, Toma and Shiroma1995)]. Sensitivity improved to 100% when seven stool samples were studied (Siddiqui  and Berk, Reference Siddiqui and Berk2001). Duodenal aspiration, while more sensitive than stool examination, is an invasive procedure that makes it a less favourable option. Duodenal biopsy, when performed, can demonstrate parasites nested in the gastric crypts or duodenal glands, as well as eosinophil infiltration of the lamina propria (Rivasi et al. Reference Rivasi, Pampiglione, Boldorini and Cardinale2006).

Immunological methods

Antibody detection

A number of immunoassays, most notably enzyme-linked immunosorbent assays (ELISAs), have been increasingly used in conjunction with stool studies to increase diagnostic sensitivity. The high negative predictive value of these immunoassays can be particularly useful in excluding S. stercoralis infection as part of the differential diagnosis. Despite their utility, antibody-based immunoassays have several limitations including: (1) cross-reactivity in patients with active filarial infections; (2) lower sensitivity in patients with haematologic malignancies or HTLV-1 infection; and (3) the inability to distinguish between current and past infection. Moreover, the current available immunoassays [see (Levenhagen  and Costa-Cruz, Reference Levenhagen and Costa-Cruz2014; Buonfrate et al. Reference Buonfrate, Formenti, Perandin and Bisoffi2015; Toledo et al. Reference Toledo, Munoz-Antoli and Esteban2015) for a comprehensive discussion] relies on the preparation of S. stercoralis larval antigen from stool samples of heavily infected humans or experimentally infected animals or from related (but not S. stercoralis) Strongyloides species (e.g. S. ratti).

To overcome some of these drawbacks, S. stercoralis-specific recombinant antigens, such as NIE (Ravi et al. Reference Ravi, Ramachandran, Thompson, Andersen and Neva2002) and SsIR (Ramachandran et al. Reference Ramachandran, Thompson, Gam and Neva1998), were proposed as alternatives to the crude antigen-based immunoassays currently in use. Using a number of formats including ELISA [(Krolewiecki et al. Reference Krolewiecki, Ramanathan, Fink, McAuliffe, Cajal, Won, Juarez, Di Paolo, Tapia, Acosta, Lee, Lammie, Abraham and Nutman2010), luciferase immunoprecipitation systems (Ramanathan et al. Reference Ramanathan, Burbelo, Groot, Iadarola, Neva and Nutman2008; Krolewiecki et al. Reference Krolewiecki, Ramanathan, Fink, McAuliffe, Cajal, Won, Juarez, Di Paolo, Tapia, Acosta, Lee, Lammie, Abraham and Nutman2010; Bisoffi et al. Reference Bisoffi, Buonfrate, Sequi, Mejia, Cimino, Krolewiecki, Albonico, Gobbo, Bonafini, Angheben, Requena-Mendez, Munoz and Nutman2014)] and diffraction-based biosensors (Pak et al. Reference Pak, Vasquez-Camargo, Kalinichenko, Chiodini, Nutman, Tanowitz, McAuliffe, Wilkins, Smith, Ward, Libman and Ndao2014) the use of recombinant NIE and/or SsIR has improved greatly the diagnostic accuracy and utility of these antibody-based assays (Bisoffi et al. Reference Bisoffi, Buonfrate, Sequi, Mejia, Cimino, Krolewiecki, Albonico, Gobbo, Bonafini, Angheben, Requena-Mendez, Munoz and Nutman2014; Levenhagen  and Costa-Cruz, Reference Levenhagen and Costa-Cruz2014; Buonfrate et al. Reference Buonfrate, Formenti, Perandin and Bisoffi2015; Toledo et al. Reference Toledo, Munoz-Antoli and Esteban2015).

Antigen detection

Coproantigen detection assays have the ability to overcome some of the limitations seen in immunoassays that measure antibody (see above). There have been several capture ELISA assays developed for S. stercoralis coproantigen detection (El-Badry, Reference El-Badry2009; Sykes  and McCarthy, Reference Sykes and McCarthy2011), and both of these assays have been performed on relatively few samples and are only available in a research setting.

Molecular diagnosis

Molecular diagnostics – using standard (and/or nested-) PCR, qPCR or loop-mediated isothermal amplification assays – have been increasingly gaining traction for use stool-based assays given their high degree of specificity and sensitivity (ten Hove et al. Reference ten Hove, van Esbroeck, Vervoort, van den Ende, van Lieshout and Verweij2009; Verweij et al. Reference Verweij, Canales, Polman, Ziem, Brienen, Polderman and van Lieshout2009; Taniuchi et al. Reference Taniuchi, Verweij, Noor, Sobuz, Lieshout, Petri, Haque and Houpt2011; Mejia et al. Reference Mejia, Vicuna, Broncano, Sandoval, Vaca, Chico, Cooper and Nutman2013; Sultana et al. Reference Sultana, Jeoffreys, Watts, Gilbert and Lee2013; Watts et al. Reference Watts, James, Sultana, Ginn, Outhred, Kong, Verweij, Iredell, Chen and Lee2014; Easton et al. Reference Easton, Oliveira, O'Connell, Kepha, Mwandawiro, Njenga, Kihara, Mwatele, Odiere, Brooker, Webster, Anderson and Nutman2016; Llewellyn et al. Reference Llewellyn, Inpankaew, Nery, Gray, Verweij, Clements, Gomes, Traub and McCarthy2016). Indeed, the improved specificity relies on the specific DNA targets used [18S rRNA, IST1, cytochrome c oxidase subunit 1 or the highly repetitive interspersed repeat sequence (Moore et al. Reference Moore, Ramachandran, Gam, Neva, Lu, Saunders, Williams and Nutman1996)] and the improved sensitivity has resulted from better methods for DNA extraction in stool (ten Hove et al. Reference ten Hove, van Esbroeck, Vervoort, van den Ende, van Lieshout and Verweij2009; Taniuchi et al. Reference Taniuchi, Verweij, Noor, Sobuz, Lieshout, Petri, Haque and Houpt2011; Liu et al. Reference Liu, Gratz, Amour, Kibiki, Becker, Janaki, Verweij, Taniuchi, Sobuz, Haque, Haverstick and Houpt2013; Mejia et al. Reference Mejia, Vicuna, Broncano, Sandoval, Vaca, Chico, Cooper and Nutman2013; Sultana et al. Reference Sultana, Jeoffreys, Watts, Gilbert and Lee2013; Easton et al. Reference Easton, Oliveira, O'Connell, Kepha, Mwandawiro, Njenga, Kihara, Mwatele, Odiere, Brooker, Webster, Anderson and Nutman2016). These molecular diagnostic techniques likely identify active S. stercoralis infection as positivity has been shown to be lost following definitive treatment.

TREATMENT

The goals for therapy for S. stercoralis infection are to: (1) clear the organism completely thereby eliminating the possibility of autoinfection: (2) treat symptomatic infection; and (3) prevent complications associated with asymptomatic infection. Oral ivermectin (200 µg kg−1 for 2 days) remains the treatment of choice for uncomplicated S. stercoralis infections (Keiser  and Nutman, Reference Keiser and Nutman2004; Suputtamongkol et al. Reference Suputtamongkol, Premasathian, Bhumimuang, Waywa, Nilganuwong, Karuphong, Anekthananon, Wanachiwanawin and Silpasakorn2011; Mejia  and Nutman, Reference Mejia and Nutman2012; Toledo et al. Reference Toledo, Munoz-Antoli and Esteban2015; Henriquez-Camacho et al. Reference Henriquez-Camacho, Gotuzzo, Echevarria, White, Terashima, Samalvides, Perez-Molina and Plana2016) as it targets both adults and larvae. Albendazole at 400 mg twice a day for 3–7 days has been shown to be slightly less effective than ivermectin for the treatment of uncomplicated S. stercoralis (Suputtamongkol et al. Reference Suputtamongkol, Premasathian, Bhumimuang, Waywa, Nilganuwong, Karuphong, Anekthananon, Wanachiwanawin and Silpasakorn2011; Henriquez-Camacho et al. Reference Henriquez-Camacho, Gotuzzo, Echevarria, White, Terashima, Samalvides, Perez-Molina and Plana2016) and should be considered an alternative therapy. This is likely because albendazole primarily targets only the adult parasites. Thiabendazole (25 mg kg−1 day−1) for three days can also be used, but because of gastrointestinal side effects, its use has been supplanted by ivermectin.

Hyperinfection syndrome should be considered a potential medical emergency. Thus, treatment should be started immediately if this diagnosis is being considered. Although no controlled trials have been performed in hyperinfection syndrome, daily ivermectin has been the de facto treatment with the length of treatment being for a minimum of 2 weeks (and often until there has been evidence of two full weeks of negative stool examination). Reduction of immunosuppressive therapy should also be an important part of treatment, but obviously needs to be weighed against long-term outcomes of the underlying condition. There have been case reports of the improved efficacy of combination treatment with ivermectin and albendazole (Pornsuriyasak et al. Reference Pornsuriyasak, Niticharoenpong and Sakapibunnan2004) but no randomized trials have been done.

Other methods of ivermectin administration may have to be used, particularly when patients are unable to take oral medication (even through a nasogastric tube) because of severe systemic illness or paralytic ileus. These include per rectal and parenteral formulations (Grein et al. Reference Grein, Mathisen, Donovan and Fleckenstein2010). The parenteral formulation is a veterinary formulation of ivermectin and should be reserved for extreme situations with no other options for clearing the Strongyloides infection (Marty et al. Reference Marty, Lowry, Rodriguez, Milner, Pieciak, Sinha, Fleckenstein and Baden2005; Salluh et al. Reference Salluh, Feres, Velasco, Holanda, Toscano and Soares2005; Turner et al. Reference Turner, Maclean, Fleckenstein and Greenaway2005; Suputtamongkol et al. Reference Suputtamongkol, Kungpanichkul, Silpasakorn and Beeching2008; Lichtenberger et al. Reference Lichtenberger, Rosa-Cunha, Morris, Nishida, Akpinar, Gaitan, Tzakis and Doblecki-Lewis2009; Marcos et al. Reference Marcos, Terashima, Canales and Gotuzzo2011; Moura et al. Reference Moura, Maia Mde, Ghazi, Amorim and Pinhati2012; Donadello et al. Reference Donadello, Cristallini, Taccone, Lorent, Vincent, de Backer and Jacobs2013; Barrett et al. Reference Barrett, Broderick, Soulsby, Wade and Newsholme2016).

In conclusion, the gaps in our understanding of human strongyloidiasis, among the most neglected of the neglected tropical diseases (NTDs) (Olsen et al. Reference Olsen, van Lieshout, Marti, Polderman, Polman, Steinmann, Stothard, Thybo, Verweij and Magnussen2009) are extraordinary given the rapid pace of scientific and clinical advances seen in related areas of parasitic and other tropical infectious diseases. Given its increasing importance as a significant public health problem (in high-, middle- and low-income countries) and the lack of a public health response (Krolewiecki et al. Reference Krolewiecki, Lammie, Jacobson, Gabrielli, Levecke, Socias, Arias, Sosa, Abraham, Cimino, Echazu, Crudo, Vercruysse and Albonico2013), harnessing the insights made, to date, in our understanding of the basic biology and genetic makeup of Strongyloides (Hunt et al. Reference Hunt, Tsai, Coghlan, Reid, Holroyd, Foth, Tracey, Cotton, Stanley, Beasley, Bennett, Brooks, Harsha, Kajitani, Kulkarni, Harbecke, Nagayasu, Nichol, Ogura, Quail, Randle, Xia, Brattig, Soblik, Ribeiro, Sanchez-Flores, Hayashi, Itoh, Denver, Grant, Stoltzfus, Lok, Murayama, Wastling, Streit, Kikuchi, Viney and Berriman2016), the host response to this long-lived parasite (Breloer  and Abraham, Reference Breloer and Abraham2016), and molecularly based approaches to diagnosis and intervention must be made an imperative if we are to consider a world free of soil transmitted helminths (of which S. stercoralis is one of the most important).

FINANCIAL SUPPORT

This work was supported by the Division of Intramural Research (DIR) of the National Institute of Allergy and Infectious Diseases.

References

REFERENCES

Al-Hasan, M. N., McCormick, M. and Ribes, J. A. (2007). Invasive enteric infections in hospitalized patients with underlying strongyloidiasis. American Journal of Clinical Pathology 128, 622627.Google Scholar
Anuradha, R., Munisankar, S., Bhootra, Y., Jagannathan, J., Dolla, C., Kumaran, P., Nutman, T. B. and Babu, S. (2016). IL-10- and TGFbeta-mediated Th9 Responses in a Human Helminth Infection. PLoS Neglected Tropical Diseases 10, e0004317.Google Scholar
Anuradha, R., Munisankar, S., Bhootra, Y., Jagannathan, J., Dolla, C., Kumaran, P., Shen, K., Nutman, T. B. and Babu, S. (2015 a). Systemic Cytokine profiles in Strongyloides stercoralis infection and alterations following treatment. Infection and Immunity 84, 425431.CrossRefGoogle ScholarPubMed
Anuradha, R., Munisankar, S., Dolla, C., Kumaran, P., Nutman, T. B. and Babu, S. (2015 b). Parasite antigen-specific regulation of Th1, Th2, and Th17 responses in Strongyloides stercoralis infection. Journal of Immunology 195, 22412250.Google Scholar
Armignacco, O., Capecchi, A., De Mori, P. and Grillo, L. R. (1989). Strongyloides stercoralis hyperinfection and the acquired immunodeficiency syndrome. American Journal of Medicine 86, 258.Google Scholar
Ashford, R. W., Barnish, G. and Viney, M. E. (1992). Strongyloides fuelleborni kellyi: infection and disease in Papua New Guinea. Parasitology Today 8, 314318.Google Scholar
Atkins, N. S., Lindo, J. F., Lee, M. G., Conway, D. J., Bailey, J. W., Robinson, R. D. and Bundy, D. A. (1997). Humoral responses in human strongyloidiasis: correlations with infection chronicity. Transactions of the Royal Society of Tropical Medicine and Hygiene 91, 609613.Google Scholar
Atul, S., Ajay, D., Ritambhara, N., Harsh, M. and Ashish, B. (2005). An unusual cause of malabsorption in an immunocompetent host. Journal of Ayub Medical College 17, 8586.Google Scholar
Barrett, K. E., Neva, F. A., Gam, A. A., Cicmanec, J., London, W. T., Phillips, J. M. and Metcalfe, D. D. (1988). The immune response to nematode parasites: modulation of mast cell numbers and function during Strongyloides stercoralis infections in nonhuman primates. American Journal of Tropical Medicine and Hygiene 38, 574581.Google Scholar
Barrett, J., Broderick, C., Soulsby, H., Wade, P. and Newsholme, W. (2016). Subcutaneous ivermectin use in the treatment of severe Strongyloides stercoralis infection: two case reports and a discussion of the literature. Journal of Antimicrobial Chemotherapy 71, 220225.Google Scholar
Ben-Youssef, R., Baron, P., Edson, F., Raghavan, R. and Okechukwu, O. (2005). Stronglyoides stercoralis infection from pancreas allograft: case report. Transplantation 80, 997998.Google Scholar
Bisoffi, Z., Buonfrate, D., Sequi, M., Mejia, R., Cimino, R. O., Krolewiecki, A. J., Albonico, M., Gobbo, M., Bonafini, S., Angheben, A., Requena-Mendez, A., Munoz, J. and Nutman, T. B. (2014). Diagnostic accuracy of five serologic tests for Strongyloides stercoralis infection. PLoS Neglected Tropical Diseases 8, e2640.Google Scholar
Breloer, M. and Abraham, D. (2016). Strongyloides infection in rodents: immune response and immune regulation. Parasitology 121 (in press).Google Scholar
Brown, M., Cartledge, J. D. and Miller, R. F. (2006). Dissemination of Strongyloides stercoralis as an immune restoration phenomenon in an HIV-1-infected man on antiretroviral therapy. International Journal of STD and AIDS 17, 560561.CrossRefGoogle Scholar
Buonfrate, D., Formenti, F., Perandin, F. and Bisoffi, Z. (2015). Novel approaches to the diagnosis of Strongyloides stercoralis infection. Clinical Microbiology and Infection 21, 543552.CrossRefGoogle Scholar
Cahill, K. M. and Shevchuk, M. (1996). Fulminant, systemic strongyloidiasis in AIDS. Annals of Tropical Medicine and Parasitology 90, 313318.Google Scholar
Carvalho, E. M. and Da Fonseca Porto, A. (2004). Epidemiological and clinical interaction between HTLV-1 and Strongyloides stercoralis . Parasite Immunology 26, 487497.Google Scholar
Celedon, J. C., Mathur-Wagh, U., Fox, J., Garcia, R. and Wiest, P. M. (1994). Systemic strongyloidiasis in patients infected with the human immunodeficiency virus. A report of 3 cases and review of the literature. Medicine (Baltimore) 73, 256263.CrossRefGoogle ScholarPubMed
Chaudhuri, B., Nanos, S., Soco, J. N. and McGrew, E. A. (1980). Disseminated Strongyloides stercoralis infestation detected by sputum cytology. Acta Cytologica 24, 360362.Google Scholar
Choi, S. W. and Reddy, P. (2014). Current and emerging strategies for the prevention of graft-versus-host disease. Nature Reviews: Clinical Oncology 11, 536547.Google Scholar
Chu, E., Whitlock, W. L. and Dietrich, R. A. (1990). Pulmonary hyperinfection syndrome with Strongyloides stercoralis . Chest 97, 14751477.Google Scholar
Cirioni, O., Giacometti, A., Burzacchini, F., Balducci, M. and Scalise, G. (1996). Strongyloides stercoralis first-stage larvae in the lungs of a patient with AIDS: primary localization or a noninvasive form of dissemination? Clinical Infectious Diseases 22, 737.CrossRefGoogle ScholarPubMed
Crouch, P. R. and Shield, J. M. (1982). Strongyloides fulleborni-like infections in Anga children. Papua New Guinea Medical Journal 25, 164165.Google ScholarPubMed
Czachor, J. S. and Jonas, A. P. (2000). Transmission of Strongyloides steracolis person to person. Journal of Travel Medicine 7, 211212.Google Scholar
Dogan, C., Gayaf, M., Ozsoz, A., Sahin, B., Aksel, N., Karasu, I., Aydogdu, Z. and Turgay, N. (2014). Pulmonary Strongyloides stercoralis infection. Respiratory Medicines Case Reports 11, 1215.Google Scholar
Donadello, K., Cristallini, S., Taccone, F. S., Lorent, S., Vincent, J. L., de Backer, D. and Jacobs, F. (2013). Strongyloides disseminated infection successfully treated with parenteral ivermectin: case report with drug concentration measurements and review of the literature. International Journal of Antimicrobial Agents 42, 580583.Google Scholar
Dunlap, N. E., Shin, M. S., Polt, S. S. and Ho, K. J. (1984). Strongyloidiasis manifested as asthma. Southern Medical Journal 77, 7778.Google Scholar
Dutcher, J. P., Marcus, S. L., Tanowitz, H. B., Wittner, M., Fuks, J. Z. and Wiernik, P. H. (1990). Disseminated strongyloidiasis with central nervous system involvement diagnosed antemortem in a patient with acquired immunodeficiency syndrome and Burkitts lymphoma. Cancer 66, 24172420.3.0.CO;2-G>CrossRefGoogle Scholar
Easton, A. V., Oliveira, R. G., O'Connell, E. M., Kepha, S., Mwandawiro, C. S., Njenga, S. M., Kihara, J. H., Mwatele, C., Odiere, M. R., Brooker, S. J., Webster, J. P., Anderson, R. M. and Nutman, T. B. (2016). Multi-parallel qPCR provides increased sensitivity and diagnostic breadth for gastrointestinal parasites of humans: field-based inferences on the impact of mass deworming. Parasites & Vectors 9, 38.Google Scholar
El-Badry, A. A. (2009). ELISA-based coproantigen in human strongyloidiaisis: a diagnostic method correlating with worm burden. Journal of the Egyptian Society of Parasitology 39, 757768.Google Scholar
Evans, A. C., Markus, M. B., Joubert, J. J. and Gunders, A. E. (1991). Bushman children infected with the nematode Strongyloides fulleborni . South African Medical Journal 80, 410411.Google Scholar
Freedman, D. O. (1991). Experimental infection of human subject with Strongyloides species. Reviews of Infectious Diseases 13, 12211226.Google Scholar
Gatti, S., Lopes, R., Cevini, C., Ijaoba, B., Bruno, A., Bernuzzi, A. M., de Lio, P., Monco, A. and Scaglia, M. (2000). Intestinal parasitic infections in an institution for the mentally retarded. Annals of Tropical Medicine and Parasitology 94, 453460.Google Scholar
Genta, R. M. (1989). Global prevalence of strongyloidiasis: critical review with epidemiologic insights into the prevention of disseminated disease. Reviews of Infectious Diseases 11, 755767.Google Scholar
Genta, R. M. (1992). Dysregulation of strongyloidiasis: a new hypothesis. Clinical Microbiology Reviews 5, 345355.Google Scholar
Genta, R. M. and Lillibridge, J. P. (1989). Prominence of IgG4 antibodies in the human responses to Strongyloides stercoralis infection. Journal of Infectious Diseases 160, 692699.Google Scholar
Genta, R. M., Ottesen, E. A., Poindexter, R., Gam, A. A., Neva, F. A., Tanowitz, H. B. and Wittner, M. (1983). Specific allergic sensitization to Strongyloides antigens in human strongyloidiasis. Laboratory Investigation 48, 633638.Google Scholar
George, P. J., Anuradha, R., Kumar, N. P., Sridhar, R., Banurekha, V. V., Nutman, T. B. and Babu, S. (2014). Helminth infections coincident with active pulmonary tuberculosis inhibit mono- and multifunctional CD4+ and CD8+ T cell responses in a process dependent on IL-10. PLoS Pathogens 10, e1004375.Google Scholar
George, P. J., Pavan Kumar, N., Jaganathan, J., Dolla, C., Kumaran, P., Nair, D., Banurekha, V. V., Shen, K., Nutman, T. B. and Babu, S. (2015). Modulation of pro- and anti-inflammatory cytokines in active and latent tuberculosis by coexistent Strongyloides stercoralis infection. Tuberculosis (Edinb) 95, 822828.CrossRefGoogle ScholarPubMed
Ghosh, K. and Ghosh, K. (2007). Strongyloides stercoralis septicaemia following steroid therapy for eosinophilia: report of three cases. Transactions of the Royal Society of Tropical Medicine and Hygiene 101, 11631165.CrossRefGoogle ScholarPubMed
Gompels, M. M., Todd, J., Peters, B. S., Main, J. and Pinching, A. J. (1991). Disseminated strongyloidiasis in AIDS: uncommon but important. AIDS 5, 329332.Google Scholar
Gordon, S. M., Gal, A. A., Solomon, A. R. and Bryan, J. A. (1994). Disseminated strongyloidiasis with cutaneous manifestations in an immunocompromised host. Journal of the American Academy of Dermatology 31, 255259.CrossRefGoogle Scholar
Gotuzzo, E., Arango, C., de Queiroz-Campos, A. and Isturiz, R. E. (2000). Human T-cell lymphotropic virus-I in Latin America. Infectious Disease Clinics of North America 14, 211239, x-xi.Google Scholar
Grein, J. D., Mathisen, G. E., Donovan, S. and Fleckenstein, L. (2010). Serum ivermectin levels after enteral and subcutaneous administration for Strongyloides hyperinfection: a case report. Scandinavian Journal of Infectious Diseases 42, 234236.Google Scholar
Grove, D. I. (1989). Strongyloidiasis: A Major Roundworm Infection of Man. Taylor & Francis, Philadelphia, PA.Google Scholar
Grove, D. I. (1996). Human strongyloidiasis. Advances in Parasitology 38, 251309.Google Scholar
Guermani, A., Potenza, R., Isnardi, D., Peluso, M., Bosco, R. and Donadio, P. (2013). Organ donation and transplantation in migrants: Piedmont reality from 2004 to 2011. Transplantation Proceedings 45, 25912593.Google Scholar
Gulbas, Z., Kebapci, M., Pasaoglu, O. and Vardareli, E. (2004). Successful ivermectin treatment of hepatic strongyloidiasis presenting with severe eosinophilia. Southern Medical Journal 97, 907910.Google Scholar
Harcourt-Webster, J. N., Scaravilli, F. and Darwish, A. H. (1991). Strongyloides stercoralis hyperinfection in an HIV positive patient. Journal of Clinical Pathology 44, 346348.Google Scholar
Harish, K., Sunilkumar, R., Varghese, T. and Feroze, M. (2005). Strongyloidiasis presenting as duodenal obstruction. Tropical Gastroenterology 26, 201202.Google ScholarPubMed
Henriquez-Camacho, C., Gotuzzo, E., Echevarria, J., White, A. C. Jr., Terashima, A., Samalvides, F., Perez-Molina, J. A. and Plana, M. N. (2016). Ivermectin versus albendazole or thiabendazole for Strongyloides stercoralis infection. Cochrane Database of Systematic Reviews 1, CD007745.Google Scholar
Hira, P. R. and Patel, B. G. (1977). Strongyloides fulleborni infections in man in Zambia. American Journal of Tropical Medicine and Hygiene 26, 640643.Google Scholar
Ho, P. L., Luk, W. K., Chan, A. C. and Yuen, K. Y. (1997). Two cases of fatal strongyloidiasis in Hong Kong. Pathology 29, 324326.Google Scholar
Hsieh, Y. P., Wen, Y. K. and Chen, M. L. (2006). Minimal change nephrotic syndrome in association with strongyloidiasis. Clinical Nephrology 66, 459463.Google Scholar
Hunt, V. L., Tsai, I. J., Coghlan, A., Reid, A. J., Holroyd, N., Foth, B. J., Tracey, A., Cotton, J. A., Stanley, E. J., Beasley, H., Bennett, H. M., Brooks, K., Harsha, B., Kajitani, R., Kulkarni, A., Harbecke, D., Nagayasu, E., Nichol, S., Ogura, Y., Quail, M. A., Randle, N., Xia, D., Brattig, N. W., Soblik, H., Ribeiro, D. M., Sanchez-Flores, A., Hayashi, T., Itoh, T., Denver, D. R., Grant, W., Stoltzfus, J. D., Lok, J. B., Murayama, H., Wastling, J., Streit, A., Kikuchi, T., Viney, M. and Berriman, M. (2016). The genomic basis of parasitism in the Strongyloides clade of nematodes. Nature Genetics 48, 299307.Google Scholar
Husni, R. N., Gordon, S. M., Longworth, D. L. and Adal, K. A. (1996). Disseminated Strongyloides stercoralis infection in an immunocompetent patient. Clinical Infectious Diseases 23, 663.Google Scholar
Iriemenam, N. C., Sanyaolu, A. O., Oyibo, W. A. and Fagbenro-Beyioku, A. F. (2010). Strongyloides stercoralis and the immune response. Parasitology International 59, 914.Google Scholar
Jain, A. K., Agarwal, S. K. and el-Sadr, W. (1994). Streptococcus bovis bacteremia and meningitis associated with Strongyloides stercoralis colitis in a patient infected with human immunodeficiency virus. Clinical Infectious Diseases 18, 253254.CrossRefGoogle Scholar
Jamil, S. A. and Hilton, E. (1992). The Strongyloides hyperinfection syndrome. New York State Journal of Medicine 92, 6768.Google ScholarPubMed
Keiser, P. B. and Nutman, T. B. (2004). Strongyloides stercoralis in the immunocompromised population. Clinical Microbiology Reviews 17, 208217.Google Scholar
Kennedy, S., Campbell, R. M., Lawrence, J. E., Nichol, G. M. and Rao, D. M. (1989). A case of severe Strongyloides stercoralis infection with jejunal perforation in an Australian ex-prisoner-of-war. Medical Journal of Australia 150, 9293.Google Scholar
Khieu, V., Schar, F., Forrer, A., Hattendorf, J., Marti, H., Duong, S., Vounatsou, P., Muth, S. and Odermatt, P. (2014). High prevalence and spatial distribution of Strongyloides stercoralis in rural Cambodia. PLoS Neglected Tropical Diseases 8, e2854.Google Scholar
Kramer, M. R., Gregg, P. A., Goldstein, M., Llamas, R. and Krieger, B. P. (1990). Disseminated strongyloidiasis in AIDS and non-AIDS immunocompromised hosts: diagnosis by sputum and bronchoalveolar lavage. Southern Medical Journal 83, 12261229.Google Scholar
Krolewiecki, A. J., Ramanathan, R., Fink, V., McAuliffe, I., Cajal, S. P., Won, K., Juarez, M., Di Paolo, A., Tapia, L., Acosta, N., Lee, R., Lammie, P., Abraham, D. and Nutman, T. B. (2010). Improved diagnosis of Strongyloides stercoralis using recombinant antigen-based serologies in a community-wide study in northern Argentina. Clinical and Vaccine Immunology 17, 16241630.Google Scholar
Krolewiecki, A. J., Lammie, P., Jacobson, J., Gabrielli, A. F., Levecke, B., Socias, E., Arias, L. M., Sosa, N., Abraham, D., Cimino, R., Echazu, A., Crudo, F., Vercruysse, J. and Albonico, M. (2013). A public health response against Strongyloides stercoralis: time to look at soil-transmitted helminthiasis in full. PLoS Neglected Tropical Diseases 7, e2165.Google Scholar
Lai, C. P., Hsu, Y. H., Wang, J. H. and Lin, C. M. (2002). Strongyloides stercoralis infection with bloody pericardial effusion in a non-immunosuppressed patient. Circulation Journal 66, 613614.CrossRefGoogle Scholar
Leighton, P. M. and MacSween, H. M. (1990). Strongyloides stercoralis. The cause of an urticarial-like eruption of 65 years’ duration. Archives of Internal Medicine 150, 17471748.Google Scholar
Levenhagen, M. A. and Costa-Cruz, J. M. (2014). Update on immunologic and molecular diagnosis of human strongyloidiasis. Acta Tropica 135, 3343.Google Scholar
Levi, G. C., Kallas, E. G. and Ramos Moreira Leite, K. (1997). Disseminated Strongyloides stercoralis infection in an AIDS patient: the role of suppressive therapy. Brazilian Journal of Infectious Diseases 1, 4851.Google Scholar
Lichtenberger, P., Rosa-Cunha, I., Morris, M., Nishida, S., Akpinar, E., Gaitan, J., Tzakis, A. and Doblecki-Lewis, S. (2009). Hyperinfection strongyloidiasis in a liver transplant recipient treated with parenteral ivermectin. Transplant Infectious Disease 11, 137142.Google Scholar
Liepman, M. (1975). Disseminated Strongyloides stercoralis. A complication of immunosuppression. JAMA 231, 387388.Google Scholar
Link, K. and Orenstein, R. (1999). Bacterial complications of strongyloidiasis: Streptococcus bovis meningitis. Southern Medical Journal 92, 728731.Google Scholar
Liu, J., Gratz, J., Amour, C., Kibiki, G., Becker, S., Janaki, L., Verweij, J. J., Taniuchi, M., Sobuz, S. U., Haque, R., Haverstick, D. M. and Houpt, E. R. (2013). A laboratory-developed TaqMan Array Card for simultaneous detection of 19 enteropathogens. Journal of Clinical Microbiology 51, 472480.Google Scholar
Llewellyn, S., Inpankaew, T., Nery, S. V., Gray, D. J., Verweij, J. J., Clements, A. C., Gomes, S. J., Traub, R. and McCarthy, J. S. (2016). Application of a multiplex quantitative PCR to assess prevalence and intensity of intestinal parasite infections in a controlled clinical trial. PLoS Neglected Tropical Diseases 10, e0004380.Google Scholar
Mansfield, L. S., Niamatali, S., Bhopale, V., Volk, S., Smith, G., Lok, J. B., Genta, R. M. and Schad, G. A. (1996). Strongyloides stercoralis: maintenance of exceedingly chronic infections. American Journal of Tropical Medicine and Hygiene 55, 617624.Google Scholar
Marcos, L. A., Terashima, A., Canales, M. and Gotuzzo, E. (2011). Update on strongyloidiasis in the immunocompromised host. Current Infectious Disease Reports 13, 3546.Google Scholar
Marty, F. M., Lowry, C. M., Rodriguez, M., Milner, D. A., Pieciak, W. S., Sinha, A., Fleckenstein, L. and Baden, L. R. (2005). Treatment of human disseminated strongyloidiasis with a parenteral veterinary formulation of ivermectin. Clinical Infectious Diseases 41, e5e8.Google Scholar
Mascarello, M., Gobbi, F., Angheben, A., Gobbo, M., Gaiera, G., Pegoraro, M., Lanzafame, M., Buonfrate, D., Concia, E. and Bisoffi, Z. (2011). Prevalence of Strongyloides stercoralis infection among HIV-positive immigrants attending two Italian hospitals, from 2000 to 2009. Annals of Tropical Medicine and Parasitology 105, 617623.Google Scholar
McLarnon, M. and Ma, P. (1981). Brain stem glioma complicated by Strongyloides stercoralis . Annals of Clinical and Laboratory Science 11, 546549.Google Scholar
McNeely, D. J., Inouye, T., Tam, P. Y. and Ripley, S. D. (1980). Acute respiratory failure due to strongyloidiasis in polymyositis. Journal of Rheumatology 7, 745750.Google Scholar
McRury, J., De Messias, I. T., Walzer, P. D., Huitger, T. and Genta, R. M. (1986). Specific IgE responses in human strongyloidiasis. Clinical and Experimental Immunology 65, 631638.Google Scholar
Mejia, R. and Nutman, T. B. (2012). Screening, prevention, and treatment for hyperinfection syndrome and disseminated infections caused by Strongyloides stercoralis . Current Opinion in Infectious Diseases 25, 458463.Google Scholar
Mejia, R., Vicuna, Y., Broncano, N., Sandoval, C., Vaca, M., Chico, M., Cooper, P. J. and Nutman, T. B. (2013). A novel, multi-parallel, real-time polymerase chain reaction approach for eight gastrointestinal parasites provides improved diagnostic capabilities to resource-limited at-risk populations. American Journal of Tropical Medicine and Hygiene 88, 10411047.Google Scholar
Mitre, E., Thompson, R. W., Carvalho, E. M., Nutman, T. B. and Neva, F. A. (2003). Majority of interferon-gamma-producing CD4+ cells in patients infected with human T cell lymphotrophic virus do not express tax protein. Journal of Infectious Diseases 188, 428432.Google Scholar
Mokaddas, E. M., Shati, S., Abdulla, A., Nampoori, N. R., Iqbal, J., Nair, P. M., Said, T., Abdulhalim, M. and Hira, P. R. (2009). Fatal strongyloidiasis in three kidney recipients in Kuwait. Medical Principles and Practice 18, 414417.Google Scholar
Montes, M., Sanchez, C., Verdonck, K., Lake, J. E., Gonzalez, E., Lopez, G., Terashima, A., Nolan, T., Lewis, D. E., Gotuzzo, E. and White, A. C. Jr. (2009). Regulatory T cell expansion in HTLV-1 and strongyloidiasis co-infection is associated with reduced IL-5 responses to Strongyloides stercoralis antigen. PLoS Neglected Tropical Diseases 3, e456.Google Scholar
Moore, T. A., Ramachandran, S., Gam, A. A., Neva, F. A., Lu, W., Saunders, L., Williams, S. A. and Nutman, T. B. (1996). Identification of novel sequences and codon usage in Strongyloides stercoralis . Molecular and Biochemical Parasitology 79, 243248.Google Scholar
Mori, S., Konishi, T., Matsuoka, K., Deguchi, M., Ohta, M., Mizuno, O., Ueno, T., Okinaka, T., Nishimura, Y., Ito, N. and Nakano, T. (1998). Strongyloidiasis associated with nephrotic syndrome. Internal Medicine 37, 606610.Google Scholar
Moura, E. B., Maia Mde, O., Ghazi, M., Amorim, F. F. and Pinhati, H. M. (2012). Salvage treatment of disseminated strongyloidiasis in an immunocompromised patient: therapy success with subcutaneous ivermectin. Brazilian Journal of Infectious Diseases 16, 479481.CrossRefGoogle Scholar
Neefe, L. I., Pinilla, O., Garagusi, V. F. and Bauer, H. (1973). Disseminated strongyloidiasis with cerebral involvement. A complication of corticosteroid therapy. American Journal of Medicine 55, 832838.Google Scholar
Neva, F. A. (1986). Biology and immunology of human strongyloidiasis. Journal of Infectious Diseases 153, 397406.Google Scholar
Neva, F. A., Filho, J. O., Gam, A. A., Thompson, R., Freitas, V., Melo, A. and Carvalho, E. M. (1998). Interferon-gamma and interleukin-4 responses in relation to serum IgE levels in persons infected with human T lymphotropic virus type I and Strongyloides stercoralis . Journal of Infectious Diseases 178, 18561859.Google Scholar
Newton, R. C., Limpuangthip, P., Greenberg, S., Gam, A. and Neva, F. A. (1992). Strongyloides stercoralis hyperinfection in a carrier of HTLV-I virus with evidence of selective immunosuppression. American Journal of Medicine 92, 202208.CrossRefGoogle Scholar
Nomura, J. and Rekrut, K. (1996). Strongyloides stercoralis hyperinfection syndrome in a patient with AIDS: diagnosis by fluorescent microscopy. Clinical Infectious Diseases 22, 736.Google Scholar
O'Connell, E. M. and Nutman, T. B. (2015). Eosinophilia in infectious diseases. Immunology and Allergy Clinics of North America 35, 493522.Google Scholar
Olsen, A., van Lieshout, L., Marti, H., Polderman, T., Polman, K., Steinmann, P., Stothard, R., Thybo, S., Verweij, J. J. and Magnussen, P. (2009). Strongyloidiasis – the most neglected of the neglected tropical diseases? Transactions of the Royal Society of Tropical Medicine and Hygiene 103, 967972.Google Scholar
Pak, B. J., Vasquez-Camargo, F., Kalinichenko, E., Chiodini, P. L., Nutman, T. B., Tanowitz, H. B., McAuliffe, I., Wilkins, P., Smith, P. T., Ward, B. J., Libman, M. D. and Ndao, M. (2014). Development of a rapid serological assay for the diagnosis of strongyloidiasis using a novel diffraction-based biosensor technology. PLoS Neglected Tropical Diseases 8, e3002.Google Scholar
Pampiglione, S. and Ricciardi, M. L. (1971). The presence of Strongyloides fulleborni von Linstow, 1905, in man in Central and East Africa. Parassitologia 13, 257269.Google Scholar
Patel, G., Arvelakis, A., Sauter, B. V., Gondolesi, G. E., Caplivski, D. and Huprikar, S. (2008). Strongyloides hyperinfection syndrome after intestinal transplantation. Transplant Infectious Disease 10, 137141.CrossRefGoogle ScholarPubMed
Pelletier, L. L. Jr. (1984). Chronic strongyloidiasis in World War II Far East ex-prisoners of war. American Journal of Tropical Medicine and Hygiene 33, 5561.Google Scholar
Pelletier, L. L. Jr. and Gabre-Kidan, T. (1985). Chronic strongyloidiasis in Vietnam veterans. American Journal of Medicine 78, 139140.Google Scholar
Pornsuriyasak, P., Niticharoenpong, K. and Sakapibunnan, A. (2004). Disseminated strongyloidiasis successfully treated with extended duration ivermectin combined with albendazole: a case report of intractable strongyloidiasis. Southeast Asian Journal of Tropical Medicine and Public Health 35, 531534.Google Scholar
Porto, A. F., Neva, F. A., Bittencourt, H., Lisboa, W., Thompson, R., Alcantara, L. and Carvalho, E. M. (2001). HTLV-1 decreases Th2 type of immune response in patients with strongyloidiasis. Parasite Immunology 23, 503507.CrossRefGoogle ScholarPubMed
Posey, D. L., Blackburn, B. G., Weinberg, M., Flagg, E. W., Ortega, L., Wilson, M., Secor, W. E., Sanders-Lewis, K., Won, K. and Maguire, J. H. (2007). High prevalence and presumptive treatment of schistosomiasis and strongyloidiasis among African refugees. Clinical Infectious Diseases 45, 13101315.CrossRefGoogle ScholarPubMed
Ramachandran, S., Thompson, R. W., Gam, A. A. and Neva, F. A. (1998). Recombinant cDNA clones for immunodiagnosis of strongyloidiasis. Journal of Infectious Diseases 177, 196203.Google Scholar
Ramanathan, R., Burbelo, P. D., Groot, S., Iadarola, M. J., Neva, F. A. and Nutman, T. B. (2008). A luciferase immunoprecipitation systems assay enhances the sensitivity and specificity of diagnosis of Strongyloides stercoralis infection. Journal of Infectious Diseases 198, 444451.Google Scholar
Ramanathan, R., Varma, S., Ribeiro, J. M., Myers, T. G., Nolan, T. J., Abraham, D., Lok, J. B. and Nutman, T. B. (2011). Microarray-based analysis of differential gene expression between infective and noninfective larvae of Strongyloides stercoralis . PLoS Neglected Tropical Diseases 5, e1039.Google Scholar
Ravi, V., Ramachandran, S., Thompson, R. W., Andersen, J. F. and Neva, F. A. (2002). Characterization of a recombinant immunodiagnostic antigen (NIE) from Strongyloides stercoralis L3-stage larvae. Molecular and Biochemical Parasitology 125, 7381.Google Scholar
Richter, J., Muller-Stover, I., Strothmeyer, H., Gobels, K., Schmitt, M. and Haussinger, D. (2006). Arthritis associated with Strongyloides stercoralis infection in HLA B-27-positive African. Parasitology Research 99, 706707.Google Scholar
Rivasi, F., Pampiglione, S., Boldorini, R. and Cardinale, L. (2006). Histopathology of gastric and duodenal Strongyloides stercoralis locations in fifteen immunocompromised subjects. Archives of Pathology and Laboratory Medicine 130, 17921798.Google Scholar
Robson, D., Welch, E., Beeching, N. J. and Gill, G. V. (2009). Consequences of captivity: health effects of far East imprisonment in World War II. QJM 102, 8796.CrossRefGoogle ScholarPubMed
Rodrigues, R. M., de Oliveira, M. C., Sopelete, M. C., Silva, D. A., Campos, D. M., Taketomi, E. A. and Costa-Cruz, J. M. (2007). IgG1, IgG4, and IgE antibody responses in human strongyloidiasis by ELISA using Strongyloides ratti saline extract as heterologous antigen. Parasitology Research 101, 12091214.Google Scholar
Ronan, S. G., Reddy, R. L., Manaligod, J. R., Alexander, J. and Fu, T. (1989). Disseminated strongyloidiasis presenting as purpura. Journal of the American Academy of Dermatology 21, 11231125.Google Scholar
Rossi, C. L., Takahashi, E. E., Partel, C. D., Teodoro, L. G. and da Silva, L. J. (1993). Total serum IgE and parasite-specific IgG and IgA antibodies in human strongyloidiasis. Revista do Instituto de Medicina Tropical de São Paulo 35, 361365.Google Scholar
Said, T., Nampoory, M. R., Nair, M. P., Halim, M. A., Shetty, S. A., Kumar, A. V., Mokadas, E., Elsayed, A., Johny, K. V., Samhan, M. and Al-Mousawi, M. (2007). Hyperinfection strongyloidiasis: an anticipated outbreak in kidney transplant recipients in Kuwait. Transplantation Proceedings 39, 10141015.CrossRefGoogle ScholarPubMed
Salles, F., Bacellar, A., Amorim, M., Orge, G., Sundberg, M., Lima, M., Santos, S., Porto, A. and Carvalho, E. (2013). Treatment of strongyloidiasis in HTLV-1 and Strongyloides stercoralis coinfected patients is associated with increased TNFalpha and decreased soluble IL2 receptor levels. Transactions of the Royal Society of Tropical Medicine and Hygiene 107, 526529.Google Scholar
Salluh, J. I., Feres, G. A., Velasco, E., Holanda, G. S., Toscano, L. and Soares, M. (2005). Successful use of parenteral ivermectin in an immunosuppressed patient with disseminated strongyloidiasis and septic shock. Intensive Care Medicine 31, 1292.Google Scholar
Santos, S. B., Porto, A. F., Muniz, A. L., Jesus, A. R. and Carvalho, E. M. (2004). Clinical and immunological consequences of human T cell leukemia virus type-I and Schistosoma mansoni co-infection. Memorias do Instituto Oswaldo Cruz 99, 121126.Google Scholar
Sato, Y., Kobayashi, J., Toma, H. and Shiroma, Y. (1995). Efficacy of stool examination for detection of Strongyloides infection. American Journal of Tropical Medicine and Hygiene 53, 248250.Google Scholar
Schad, G. A., Aikens, L. M. and Smith, G. (1989). Strongyloides stercoralis: is there a canonical migratory route through the host? Journal of Parasitology 75, 740749.Google Scholar
Schar, F., Trostdorf, U., Giardina, F., Khieu, V., Muth, S., Marti, H., Vounatsou, P. and Odermatt, P. (2013). Strongyloides stercoralis: Global Distribution and Risk Factors. PLoS Neglected Tropical Diseases 7, e2288.CrossRefGoogle ScholarPubMed
Schar, F., Inpankaew, T., Traub, R. J., Khieu, V., Dalsgaard, A., Chimnoi, W., Chhoun, C., Sok, D., Marti, H., Muth, S. and Odermatt, P. (2014). The prevalence and diversity of intestinal parasitic infections in humans and domestic animals in a rural Cambodian village. Parasitology International 63, 597603.Google Scholar
Schar, F., Giardina, F., Khieu, V., Muth, S., Vounatsou, P., Marti, H. and Odermatt, P. (2016). Occurrence of and risk factors for Strongyloides stercoralis infection in South-East Asia. Acta Tropica doi: 10.1016/j.actatropica.2015.03.008 (in press).Google Scholar
Scowden, E. B., Schaffner, W. and Stone, W. J. (1978). Overwhelming strongyloidiasis: an unappreciated opportunistic infection. Medicine (Baltimore) 57, 527544.Google Scholar
Seet, R. C., Lau, L. G. and Tambyah, P. A. (2005). Strongyloides hyperinfection and hypogammaglobulinemia. Clinical and Diagnostic Laboratory Immunology 12, 680682.Google ScholarPubMed
Siddiqui, A. A. and Berk, S. L. (2001). Diagnosis of Strongyloides stercoralis infection. Clinical Infectious Diseases 33, 10401047.Google Scholar
Stewart, D. M., Ramanathan, R., Mahanty, S., Fedorko, D. P., Janik, J. E. and Morris, J. C. (2011). Disseminated Strongyloides stercoralis infection in HTLV-1-associated adult T-cell leukemia/lymphoma. Acta Haematologica 126, 6367.CrossRefGoogle ScholarPubMed
Stone, W. J. and Schaffner, W. (1990). Strongyloides infections in transplant recipients. Seminars in Respiratory Infections 5, 5864.Google ScholarPubMed
Sugiyama, K., Hasegawa, Y., Nagasawa, T. and Hitomi, S. (2006). Exposure of medical staff to Strongyloides stercolaris from a patient with disseminated strongyloidiasis. Journal of Infection and Chemotherapy 12, 217219.Google Scholar
Sultana, Y., Jeoffreys, N., Watts, M. R., Gilbert, G. L. and Lee, R. (2013). Real-time polymerase chain reaction for detection of Strongyloides stercoralis in stool. American Journal of Tropical Medicine and Hygiene 88, 10481051.Google Scholar
Suputtamongkol, Y., Kungpanichkul, N., Silpasakorn, S. and Beeching, N. J. (2008). Efficacy and safety of a single-dose veterinary preparation of ivermectin versus 7-day high-dose albendazole for chronic strongyloidiasis. International Journal of Antimicrobial Agents 31, 4649.Google Scholar
Suputtamongkol, Y., Premasathian, N., Bhumimuang, K., Waywa, D., Nilganuwong, S., Karuphong, E., Anekthananon, T., Wanachiwanawin, D. and Silpasakorn, S. (2011). Efficacy and safety of single and double doses of ivermectin versus 7-day high dose albendazole for chronic strongyloidiasis. PLoS Neglected Tropical Diseases 5, e1044.Google Scholar
Suvarna, D., Mehta, R., Sadasivan, S., Raj, V. V. and Balakrishnan, V. (2005). Infiltrating Strongyloides stercoralis presenting as duodenal obstruction. Indian Journal of Gastroenterology 24, 173174.Google Scholar
Sykes, A. M. and McCarthy, J. S. (2011). A coproantigen diagnostic test for Strongyloides infection. PLoS Neglected Tropical Diseases 5, e955.Google Scholar
Takayanagui, O. M., Lofrano, M. M., Araugo, M. B. and Chimelli, L. (1995). Detection of Strongyloides stercoralis in the cerebrospinal fluid of a patient with acquired immunodeficiency syndrome. Neurology 45, 193194.Google Scholar
Taniuchi, M., Verweij, J. J., Noor, Z., Sobuz, S. U., Lieshout, L., Petri, W. A. Jr., Haque, R. and Houpt, E. R. (2011). High throughput multiplex PCR and probe-based detection with Luminex beads for seven intestinal parasites. American Journal of Tropical Medicine and Hygiene 84, 332337.CrossRefGoogle ScholarPubMed
ten Hove, R. J., van Esbroeck, M., Vervoort, T., van den Ende, J., van Lieshout, L. and Verweij, J. J. (2009). Molecular diagnostics of intestinal parasites in returning travellers. European Journal of Clinical Microbiology and Infectious Diseases 28, 10451053.Google Scholar
Thomas, M. C. and Costello, S. A. (1998). Disseminated strongyloidiasis arising from a single dose of dexamethasone before stereotactic radiosurgery. International Journal of Clinical Practice 52, 520521.CrossRefGoogle ScholarPubMed
Thompson, J. R. and Berger, R. (1991). Fatal adult respiratory distress syndrome following successful treatment of pulmonary strongyloidiasis. Chest 99, 772774.Google Scholar
Tiwari, S., Rautaraya, B. and Tripathy, K. P. (2012). Hyperinfection of Strongyloides stercoralis in an immunocompetent patient. Tropical Parasitology 2, 135137.Google Scholar
Toledo, R., Munoz-Antoli, C. and Esteban, J. G. (2015). Strongyloidiasis with emphasis on human infections and its different clinical forms. Advances in Parasitology 88, 165241.Google Scholar
Tullis, D. C. (1970). Bronchial asthma associated with intestinal parasites. New England Journal of Medicine 282, 370372.Google Scholar
Turner, S. A., Maclean, J. D., Fleckenstein, L. and Greenaway, C. (2005). Parenteral administration of ivermectin in a patient with disseminated strongyloidiasis. American Journal of Tropical Medicine and Hygiene 73, 911914.Google Scholar
Verweij, J. J., Canales, M., Polman, K., Ziem, J., Brienen, E. A., Polderman, A. M. and van Lieshout, L. (2009). Molecular diagnosis of Strongyloides stercoralis in faecal samples using real-time PCR. Transactions of the Royal Society of Tropical Medicine and Hygiene 103, 342346.Google Scholar
Vince, J. D., Ashford, R. W., Gratten, M. J. and Bana-Koiri, J. (1979). Strongyloides species infestation in young infants of papu, New Guinea: association with generalized oedema. Papua New Guinea Medical Journal 22, 120127.Google Scholar
Viney, M. E. and Lok, J. B. (2007). Strongyloides spp. WormBook: The Online Review of C. Elegans Biology 115. doi: 10.1895/wormbook.1.141.1 http://www.wormbook.org.Google ScholarPubMed
Vishwanath, S., Baker, R. A. and Mansheim, B. J. (1982). Strongyloides infection and meningitis in an immunocompromised host. American Journal of Tropical Medicine and Hygiene 31, 857858.Google Scholar
Walson, J. L., Stewart, B. T., Sangare, L., Mbogo, L. W., Otieno, P. A., Piper, B. K., Richardson, B. A. and John-Stewart, G. (2010). Prevalence and correlates of helminth co-infection in Kenyan HIV-1 infected adults. PLoS Neglected Tropical Diseases 4, e644.Google Scholar
Watts, M. R., James, G., Sultana, Y., Ginn, A. N., Outhred, A. C., Kong, F., Verweij, J. J., Iredell, J. R., Chen, S. C. and Lee, R. (2014). A loop-mediated isothermal amplification (LAMP) assay for Strongyloides stercoralis in stool that uses a visual detection method with SYTO-82 fluorescent dye. American Journal of Tropical Medicine and Hygiene 90, 306311.Google Scholar
Weiser, J. A., Scully, B. E., Bulman, W. A., Husain, S. and Grossman, M. E. (2011). Periumbilical parasitic thumbprint purpura: Strongyloides hyperinfection syndrome acquired from a cadaveric renal transplant. Transplant Infectious Disease 13, 5862.Google Scholar
Wirk, B. and Wingard, J. R. (2009). Strongyloides stercoralis hyperinfection in hematopoietic stem cell transplantation. Transplant Infectious Disease 11, 143148.Google Scholar
Wolfe, R. A., Roys, E. C. and Merion, R. M. (2010). Trends in organ donation and transplantation in the United States, 1999–2008. American Journal of Transplantation 10, 961972.CrossRefGoogle ScholarPubMed
Wurtz, R., Mirot, M., Fronda, G., Peters, C. and Kocka, F. (1994). Short report: gastric infection by Strongyloides stercoralis . American Journal of Tropical Medicine and Hygiene 51, 339340.Google Scholar
Yassin, M. A., El Omri, H., Al-Hijji, I., Taha, R., Hassan, R., Aboudi, K. A. and El-Ayoubi, H. (2010). Fatal Strongyloides stercoralis hyper-infection in a patient with multiple myeloma. Brazilian Journal of Infectious Diseases 14, 536539.Google Scholar
Yee, A., Boylen, C. T., Noguchi, T., Klatt, E. C. and Sharma, O. P. (1987). Fatal Strongyloides stercoralis infection in a patient receiving corticosteroids. Western Journal of Medicine 146, 363364.Google Scholar
Yoeli, M., Most, H., Hammond, J. and Scheinesson, G. P. (1972). Parasitic infections in a closed community. Results of a 10-year survey in Willowbrook State School. Transactions of the Royal Society of Tropical Medicine and Hygiene 66, 764776.Google Scholar
Yoshida, H., Endo, H., Tanaka, S., Ishikawa, A., Kondo, H. and Nakamura, T. (2006). Recurrent paralytic ileus associated with strongyloidiasis in a patient with systemic lupus erythematosus. Modern Rheumatology 16, 4447.CrossRefGoogle Scholar
Figure 0

Table 1. Conditions associated with hyperinfection syndrome

Figure 1

Fig. 1. Immune responses in Strongyloides stercoralis infection as a function of time after infection initiation. The infective L3 larval parasites initiate infection at skin sites and activate a variety of different cell types such as innate lymphoid cells (ILCs), macrophages (MAC), dendritic cells (DCs), natural killer cells (NK), eosinophils (Eos) and basophils/mast cells (Baso/MC). At this relatively early phase of infection (or by the time the adult worms are established in the small intestine) the parasite induces the differentiation of a small number of effector Th1/Th17 and a relatively larger number of Th2 cells which together with IgE antibody, may lead to attrition of some of the parasites. At the time of patency (when larval production occurs) there is an expansion of Th2/Th9 CD4+ cells, a further contraction of Th1/Th17 cells and the induction of alternatively activated macrophages (AAM). With the evolution of chronic longstanding infection, there is an associated expansion of IL-10- and/or TGFβ-producing regulatory T cells (Tregs) and a small contraction of Th2/Th9 cells.