Journal of Infection and Molecular Biology

Download PDF Download ePUB
JIMB_MH20160608160644-R2_ Hossain and Bhuiyan

 

Review Article

 

 

Hookworm Infection: A Neglected Tropical Disease of Mankind

 

Muhammed Hossain*, Md. Jamal Uddin Bhuiyan

Department of Parasitology, Faculty of Veterinary and Animal Science, Sylhet Agricultural University, Sylhet-3100, Bangladesh.

 

Abstract | Hookworm infection is one of the neglected tropical disease which poses a global disease-burden by infecting 576 millions people around the world. Majority of hookworm infections are harbored by childern and adults. It is endemic in developing countries such as Asia, Africa, Southeast Asia, Bangladesh, Central and South America. Hookworm infection threats the mankind and cause anemia, hypo-albuminemia and malnutrition but other known effects include intellectual, cognitive impairment and stunted growth in children. Hookworm diagnosis is industrious as it has no gold standard diagnostic techniques, however, the ELISA shows moderate sensitivity and specificity to detect hookworm specific antibody titer in serum. Likewise, PCR method considered reliable for DNA detection in fecal samples.Lately, there is a drastic advancement in our realization about this widespread parasite. Advances in molecular biology had led to the identification of a variety of new molecules from hookworms, which have importance either in the molecular pathogenesis of hookworm infection or in the host-parasite relationship. Benzimidazole anthleminthics like albendazole, levamisole, mebendazole and pyrantel pamoate are the current corner stone for helminths treatment. This review aim to discuss currently published research on epidemiology, pathogenesis, clinical manifestations, diagnosis, treatment, immune mechanism, prevention and control.

 

Keywords | Hookworm, Neglected, Prevention and review

 

Editor | Tahir Yaqub, University of Veterinary and Animal Sciences, Lahore, Pakistan.

Received | September 09, 2016; Accepted | October 14, 2016; Published | November 07, 2016

*Correspondence | Muhammed Hossain, Department of Parasitology, Faculty of Veterinary and Animal Science, Sylhet Agricultural University, Sylhet-3100, Bangladesh; Email: bmhossain34sau@gmail.com

Citation | Hossain M and Bhuiyan MJU (2016). Hookworm infection: A neglected tropical disease of mankind. J. Inf. Mol. Biol. 4(2): 24-43.

DOI | http://dx.doi.org/10.14737/journal.jimb/2016/4.2.24.43

ISSN (Online) | 2307-5465; ISSN (Print) | 2307-5716

Copyright © 2016 Hossain and Bhuiyan. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

 

Hookworm infection caused by Necator americanus and Ancylostoma duodenale are blood feeding nematode (Yulan et al., 2009) has been reported worldwide, especially among people in tropical and subtropical countries with low socio-economic status (Halpenny et al., 2013; Furst et al., 2012; Bethony et al., 2006). It is one of the most important parasites of soil-transmitted helminths (STHs) group. It has been estimated that 576 million people around the world are infected with hookworm in Sub-Saharan Africa, the Pacific Islands, India, Southeast Asia, Latin America and the Caribbean (De Silva et al., 2003a). Nowadays hookworm infection is among the most important tropical diseases in humans; the use of disability-adjusted life years as a quantitative measure of the burden of disease reveals that this infection out ranks African trypanosomiasis, dengue, Chagas disease, schistosomiasis and leprosy (Hotez et al., 2003). Several studies ensured its continuous existence in Bangladesh (Khair et al., 2016; Hossain et al., 2015; Sultana et al., 2012; Gilgen et al., 2001; Hall et al., 1994; Ali et al., 1985) along with strongyloidiasis (Hossain and Bhuiyan, 2016; Hossain et al., 2016). N. americanus has been found to be more predominant worldwide than A. duodenale (Jiraanankul et al., 2011). High-risk groups suffering from hookworm disease are children and pregnant women (Liabsuetrakul et al., 2009). Human acquires hookworm infection via third stage infective larvae (L3) penetrating the intact skin (Yulan et al., 2009; Liabsuetrakul et al., 2009; Tomono et al., 2003). The larvae migrate to heart and lung and then move to trachea where they have been swallowed. After going through two molts, larvae develop into blood feeding adult worms; where the female worms start to produce eggs that are excreted out through feces (Cline et al., 1984; Maxwell et al., 1987). The eggs hatch in moist soil and produce larvae that develop to the L3 stage after two molts, completing the hookworm life cycle (Craig and Scott, 2014; Bungiro and Cappello, 2004).

 

 

The hookworm infection mainly causes anaemia, hypo-albuminemia and malnutrition but other known effects include intellectual, cognitive impairment and stunted growth in children (Jardim-Botelho et al., 2008; Bethony et al., 2006; Brooker et al., 2006; Ekiz et al., 2005; Hotez et al., 2004; Sakti et al., 1999; Hotez, 1989; Albonico et al., 1998; Stoltzfus et al., 1997) and in pregnant women anemia supported by hookworm (Ndyomugyenyi et al., 2008). In previous decade prevention and control of hookworm infection has been globally neglected because most individuals with light to moderate hookworm infection generally are asymptomatic. The development of effective hookworm control is possible because of the availability of proven, cost effective and logistically feasible intervention strategies. Besides, for STH infections, regular periodic chemotherapy using benzimidazole anthleminthics of school-aged children delivered through the school system is the main intervention strategy (Hotez et al., 2005; Bundy et al., 2005; Awasthi et al., 2003) and also need to pay heed on sanitation and hygiene (Gruber et al., 2013; Bartram and Cairncross, 2010). Understanding where at risk populations live is fundamental for appropriate resource allocation and cost effective control. In particular, it allows for reliable estimation of the overall drug needs of programs and efficient geographical targeting of control efforts (Brooker and Michael, 2000). The school-based deworming programs could effectively reduce the hookworm infection of children (Wikagul et al., 2005; De Silva et al., 2003) but could miss positive adult cases. Considerable efforts need to develop a vaccine against hookworm and thus far more than 20 proteins have been explored as potential vaccine antigen targets (Bungiro et al., 2004; Hotez et al., 2008). However, there is still a long way to go before an effective hookworm vaccine might eventually become available. Metabolic profiling pursues a systematic biological approach and can deepen our understanding of metabolic responses of an organism to stimulate, such as disease, physiological changes and genetic modification (Nicholson et al., 1999). This review focused on current published research on improved diagnostic techniques for hookworm detection and immune mechanism thought to be responsible for infection along with epidemiology, pathogenesis, clinical manifestations, diagnosis, treatment, prevention and control.

 

Geographical Distribution

 

Hookworm is one of the important parasites of the soil-transmitted helminths group. Thus approximately one-half of the people of Southeast Asia living in poverty have one or more soil-transmitted helminths infection (Hotez et al., 2015). Necator americanus is the predominant hookworm worldwide, except in some defined locations where A. duodenale is focally endemic (Hotez et al., 2004). The distribution of worm burdens among different human hosts is highly over dispersed so that often only 10 percent of the infected population carries 70 percent of the worms (Bundy, 1995).

 

The major regions endemic for N. americanus include South and Southwest China (Hotez, 2002), South India (Yadla et al., 2003), Southeast Asia, sub-Saharan Africa, and Central and South America (Hotez et al., 2003) (Figure 1). Coastal areas of these regions are especially associated with high Necator transmission (Lwambo et al., 1992).

 

The A. duodenale predominant regions include more northerly latitudes of South and West China (e.g., Anhui, Sichuan Provinces) and India (e.g., Kanpur). The A. duodenale may survive in these harsher climates because of its ability to undergo arrested development in host tissues during periods of dryness or cold. A. duodenale infections also occur in Egypt, Northern Australia and in a few localities in Latin America including Northern Argentina, Paraguay (Labiano-Abello et al., 1999), Peru and in a region bordering El Salvador and Honduras (Knight et al., 1981).

 

HOST RANGE

 

N. americanus is generally considered the only member of this genus to infect humans. This species has also been recovered occasionally from non-human primates (Orihel, 1971; Michaud et al., 2003). It has been further suggested that the pig may serve as a transport host for N. americanus (Steenhard et al., 2000). A. duodenale is the only significant human hookworm of the genus Ancylostoma. Ancylostoma ceylanicum parasite of cats is infecting human as a zoonosis in some regions of Asia but it is not associated with host blood loss in humans (Carroll and Grove, 1986) and therefore is not considered as major pathogen (Hotez, 1995). High proportion of cases with A. ceylanicum infections, an unique zoonotic hookworm infection found in ASEAN countries, especially Malaysia, Thailand, Cambodia and Lao PDR (Ngui et al., 2012a; Ngui et al., 2012b). In northeastern Australia, the dog hookworm A. caninum has been reported to cause both eosinophilic enteritis and aphthous ileitis syndromes (Prociv and Croese, 1996; Landmann and Prociv, 2003). The natural history of zoonotic A. caninum infection has been extensively reviewed recently and will not be considered further (Prociv and Croese, 1996; Prociv, 1997). There is evidence that some persons are predisposed to a heavy (or light) hookworm burden owing to either genetic or exposure factors (Quinnell et al., 2001; Williams et al., 1997).

 

Epidemiological Patterns by Age and Sex

 

Children can be infected with hookworm as young as 6 month old (Brooker et al., 1999). The infection prevalence typically rises gradually with increasing age to adulthood (Shiferaw and Mengistu, 2015; Abossie et al., 2014; Wegayehu et al., 2013). Interestingly, recent evidence from studies of populations in China and Southeast Asia suggest that peak prevalence is observed among the middle aged or even individuals over the age of 60 years (Sengchanh et al., 2011; Lili et al., 2000; Gandhi et al., 2001; Bethony et al., 2002). In contrast, there appears to be considerable variation in age intensity profiles seen for hookworm (Bundy and Keymer, 1991). In West Africa, for example, convex age intensity profiles are observed (Udonsi, 1984; Behnke et al., 2000) while in China intensity increases continues to rise throughout life and is highest among the elderly (Gandhi et al., 2001; Bethony et al., 2002a).

 

An exception to this trend is the study in China, which shows that worm burdens tend to continuously increase with age (Ye et al., 1994). The observation that the elderly are at risk for heavy hookworm burdens has potentially important implications for the rapidly changing demographics in the developing world. It has been suggested that males are commonly infected with hookworm than females (Bundy, 1988a) which is suggested to reflect differential susceptibility to infection arising from immunosuppression associated with male hormones (Poulin, 1996; Moore and Wilson, 2002).

 

In particular, hookworm infection is more prevalent among adults occupational exposure is likely to be important where males are more commonly infected than females (Khair et al., 2016; Behnke et al., 2000) an observation in Mali explained by the fact that males are involved in constructing houses which frequently incorporate human faeces into materials to strengthen household structure. In contrast, in South China (Sengchanh et al., 2011; Gandhi et al., 2001; Bethony et al., 2002a) and Vietnam (Needham et al., 1998) females exhibit higher hookworm intensities among specific age groups. Because in Vietnam, elderly women were observed to be responsible for most of the night soil use and the higher infection levels could be explained, in this instance on occupational exposure (Humphries et al., 1997). However, Sex based differences in infection patterns may not always reflect occupational exposure. For example, in Hainan, China women exhibit a higher prevalence and intensity of hookworm compared to males despite the absence of night soil use (Abossie et al., 2014; Needham et al., 1998; Gandhi et al., 2001; Bethony et al., 2002).

 

Risk Factors

 

Agent Factors

 

Hookworms are nematodes belonging to the family Ancylostomatidae, a part of the super-family Strongyloidea. The two major genera that affect humans are Necator and Ancylostoma characterized by the presence of either teeth or cutting plates that line the adult parasite buccal capsule (Hotez, 1995). The two species that account for almost all human infections, A. duodenale and N. americanus are highly host specific and occur in most warm temperate regions (Beaver and Cupp, 1984). Developmentally arrested and can live in the soil for weeks if there is appropriate warmth, shade and moisture (Brooker et al., 2004) and can cause human infection. The surface protein of the organism is supposed to be antigenic and causes infection. The World Health Organization defines moderate intensity infections as those with 2000-3999 eggs per gram of feces and heavy intensity infections as those with 4,000 or more eggs per gram (Montresor et al., 2002). Whereas the intensities for the former peak in childhood and adolescence, hookworm intensity usually either steadily rises in intensity with age in adulthood (Hotez et al., 2004; Bethony et al., 2002). The biological basis for this observation is unknown (Olatunde and Onyemelukw, 1994).

 

Household, Socio-economic and Occupational Risk Factors

 

As the transmission of hookworm engages contamination of the environment by hookworm eggs, it is expected that risk factors for infection may include poor personal hygiene (Traub et al., 2004; Asaolu and Ofoezie, 2003; Gunawardena et al., 2005), low educational attainment (Mihrshahi et al., 2009; Liabsuetrakul et al., 2009) and household sanitation (Ensink et al., 2005) and unfinished house floor (Soares et al., 2011; Pullan et al., 2010), which in turn are influenced by differences in socio-economic status (Halpenny et al., 2013; Furst et al., 2012; Balen et al., 2011; Brooker et al., 2004; Traub et al., 2004). Some studies have demonstrated that hookworm infection is associated with the absence of a latrine (Hossain et al., 2015; Wang et al., 2012; Olsen et al., 2001; Chongsuvivatwong et al., 1996) and low socio-economic status (Holland et al., 1988). A study in Kenya showed that variation in household income and education level of the head of household were not associated with any helminths infection (Olsen et al., 2001) conversely a study in Nigeria exposed level of education is significantly associated with hookworm infection (Adeniyi et al., 2015; Quihui et al., 2006; Nematian et al., 2004). Hookworm has also been noted to be more common in families who are involved with agricultural pursuits, attributed to widespread use of faeces as night soil fertilizer (Humphries et al., 1997) and among vegetable growers and farmers (Kirwan et al., 2009; Conde et al., 2007; Hotez et al., 1997). When faeces are used deliberately, heavy fecal pollution of plantations occurs where few toilets are available resulting in high levels of hookworm infection (Schad et al., 1983). Foot ware is also risk factors for hookworm infection (Sandy et al., 2014; Alemu et al., 2003; Ratnayaka and Wang, 2012) because walking in barefoot have high chance of hookworm infection (Shiferaw and Mengistu, 2015; Abate et al., 2013; Alemu et al., 2011). For instance, in India, Bangladesh and Sri Lanka, high rates of infection are observed among workers and their families in the tea gardens (Hossain et al., 2015; Gilgen et al., 2001).

 

Seasonal and Environmental Factors

 

Since larval stages of hookworm for motility, rate of development and survival are dependent on the surrounding environmental humidity, temperature and ultra-violet light and geographical differences in transmission will be influenced by these factors and related factors such as rainfall, soil type and altitude (Bongi et al., 2005; Chandler, 1929). Chandler concluded that 20-30°C was optimal for transmission with larvae reaching maturity in five days, with the lower limit lying between 8-10°C and the upper limit 40-45°C. The above of temperatures 35-40°C arrests development and even death occurs (Nwosu, 1978; Udonsi and Atata, 1987; Smith and Schad, 1990) and at temperatures of 35°C larvae of N. americanus all dead, with the highest cumulative hatching rates obtained at 30°C (Udonsi and Atata, 1987).

 

The lower thermal limit of hookworm in the tropics is often determined by altitude KwaZulu-Natal Province, South Africa that hookworm transmission is confined to the coastal plain below 150 m above sea level (Appleton and Gouws, 1996; Appleton et al., 1999; Mabaso et al., 2003). Above these altitudes low temperatures (<20°C) limit the transmission of the parasite (Yu, 1995). It has been noted, that A. duodenale will occur in some areas where N. americanus L3 cannot survive during the winter months. It has been postulated that the unique ability of A. duodenale L3 to undergo arrested development in the human host, may allow this species to survive during the cold winter months (Schad et al., 1973). Since hookworms are unable to survive desiccation, the amount of rainfall in an area is also an important factor influencing hookworm transmission both spatially and temporally. The spatial relationship between rainfall and hookworm prevalence is well established (Brooker and Michael, 2000). In some endemic areas, hookworm infection exhibits marked seasonality (Khanum et al., 2014; Cook et al., 2009). For A. duodenale which undergoes arrested development following the transmission during the rainy season, new infections appear after 8-10 months (Schad et al., 1973).

 

Studies in West Africa show that populations of L3 larvae are highest during the rainy season (Udonsi et al., 1980) and faecal egg counts are highest 2-7 months after the rainy season (Knight and Merrett, 1981; Nwosu, 1981). However, although seasonal fluctuations in transmission occur, such fluctuations may be of little significance to the overall persistence of hookworm populations (Anderson, 1982). A final environmental factor that may influence the transmission of hookworm is soil type. It has long been believed that hookworm thrive in areas with sandy soils because of the small particle size and well aerated texture of sandy soil. So although infective larvae quickly die on the surface of sandy soil in direct sunlight, they are able to rapidly migrate into the soil and during the rains are able to migrate vertically as moisture permits. In contrast, clay soils inhibit larval migration. Early evidence of an association between soil type and hookworm was provided by studies in the southern American States (Brooker and Michael, 2000). High prevalence of hookworm was significantly associated with well drained sandy soil types (Mabaso et al., 2003; Saathoff et al., 2002) whereas low prevalence was associated with clay soils.

 

HISTORY AND MORPHOLOGY

 

In 1838, Dubini provided the detail description of the worm now known as A. duodenale after examining specimens taken from a woman who had died in Milan. Little importance seemed to be attached to these observations until 1880 when an epidemic of anaemia occurred amongst the miners digging the St. Gotthard tunnel. By 1903, the British Home Secretary fearing widespread health and no doubt economic effects on the national mining industry, commissioned a report on Ankylostomiasis in Westphalian Collieries. Hookworm infections were well established in the USA and in due course the Rockefeller Sanitary Commission for the eradication of hookworm disease was established (Hegner et al., 1938; Ettling, 1990; Crompton and Whitehead, 1993). The overall information of hookworm given (Table 1).

 

Table 1: Life history characteristics of human hookworm infections (Crompton & Whitehead, 1993)

 

Traits

A. duodenale

N. americanus

Adult worm size (mm)

Male

8-11

7-9

Female

10-13

9-11

Adult life span (years)

1

3-5

Sex ratio (M:F)

1:1

1.5:1

Pre-patent period (days)

53

49-56

Fecundity (eggs/female/day)

10000-25000

5000-10000

Optimum temp (°C) for

free living larval stages

20-27

28-32

Route of infection

O, P, T

P

Arrested development

Yes

No

 

Based on information from Hoagland & Schad (1978) and Beaver & Jung (1985); O: Oral route; P: Percutaneous route; T: Trans-placental route; Schad & Banwell (1984) review evidence to suggest that A. duodenale may also exploit the trans-mammary route of infection as does A. caninum (Miller, 1981)

 

Eggs of hookworms (A. duodenale and N. americanus) are similar in morphology. They are colorless and oval in shape, measuring about 65 x 40μm. The eggs contain an ovum which appears unsegmented (Sianto et al., 2009). In stool more than 12 hours old, larvae may be seen inside the egg. The L3 stage of larvae 600μm of length (Brooker et al., 2004).

 

 

TRANSMISSION

 

The highest rates of hookworm transmission occur in the world’s coastal regions, where infective third stage larvae can migrate freely in sandy soils and where temperature and moisture are optimal for viability of larvae (Mabaso et al., 2003). Acquiring hookworm infection is directly related to exposure to soil where filariform larvae, the infective stage live in and penetrate human skin (Liabsuetrakul et al., 2009; Tomono et al., 2003; Traub et al., 2004) when get opportunity (Figure 2). Human acquire hookworm when the infective larval stages (known as third stage larvae or L3) living in the soil either penetrate through the skin epidermis (Haas et al., 2005) (both N. americanus and A. duodenale) or when they are ingested (for A. duodenale) (Zeehaida et al., 2011; Olsen et al., 2009; Brooker et al., 2004). It has also been reported that N. americanus L3 will invade the buccal epithelium if they enter through the mouth (Nagahana et al., 1963). Exceptionally, larvae may be transmitted through fomites. For instance, if washed clothes are dried on the ground, larvae may creep on the textile from surrounding soil, resulting in an infestation when the piece of clothing is put on (Tomovic et al., 2008).

 

BIOLOGY OF HOOKWORM

 

The life cycle of A. duodenale was first elucidated by (Looss, 1901), and N. americanus was discovered in the Western Hemisphere (Chandler, 1929; Stiles, 1902). The life cycle of hookworms is direct (Hoagland and Schad, 1978; Schad and Banwell, 1984). Hookworms live in the small intestine and feed on host mucosa and blood (Roche and Layrisse, 1966). Female worms produce eggs which are passed out through stool (Augustine, 1922) to be embryonated in the soil. Under favourable condition (adequate but not excessive moisture; warmth, 25-28°C, shade) larvae hatch within 1 to 2 days. The rhabditiform larvae grow in the faeces in the soil (Cline et al., 1984) and after 5 to 10 days (and two molts) they become filariform (third-stage) larvae that are infective (Cort and Payne, 1922) and feeds on organic debris and microorganisms. These infective larvae can survive 3 to 4 weeks in favourable environmental conditions.

 

On contact with the human host, the larvae penetrate the skin (Logan, 2009) and are carried through the blood vessels to the heart and then to the lungs. They penetrate from the pulmonary capillaries into the pulmonary alveoli, migrate up the airways and pass down the esophagus through the stomach to the duodenum where the hookworms mature (Craig and Scott, 2014; Maxwell, 1987). Male locates female then they mate and eggs appear in the faeces. The L3 can live for several weeks in the soil until they exhaust their lipid metabolic reserves. Following host entry, the L3 receive a signal present in mammalian serum and tissue that causes them to resume development and secrete bioactive polypeptides (Hotez et al., 1993; Hawdon and Hotez, 1996). Resumption of development is cGMP dependent and involves a muscurinic neuronal pathway, which is similar to the one used for Caenorhabditis elegans dauer recovery (Hawdon and Datu, 2003). Among the major proteins secreted by host activated hookworm L3 is a zinc containing metallo-protease of the astacin class (Zhan et al., 2003) and two cysteine-rich secretory proteins known as ancylostoma secreted proteins, which belong to the pathogenesis related protein super family (Hotez and Hawdon, 1996; Hotez et al., 2003).

 

There are significant biological differences between the two major human hookworms (Hoagland and Schad, 1978; Hotez, 1995). N. americanus is smaller than A. duodenale and produces fewer eggs and causes less blood loss (Albonico et al., 1998). Therefore, some investigators believe that N. americanus more adept at immune invasion, produces less blood loss and better adapted to human parasitism (Hoagland and Schad, 1978; Pritchard and Brown, 2001). A. duodenale is associated with greater intestinal blood loss than any other hookworm. This accounts for the observation, best documented in Tanzania, that the species of hookworm being transmitted in a community strongly influences the burden of iron deficiency anaemia in the community (Albonico et al., 1998). However, N. americanus is more widespread worldwide and therefore, more significant as a cause of disease burden. Unlike N. americanus and A. duodenale also has the unique ability to undergo arrested development in humans (Ekiz et al., 2005; Schad et al., 1973) and may under certain conditions; enter human mammary glands during pregnancy prior to lactogenic transmission (Hotez, 1989; Yu et al., 1995). The occurrence of neonatal ancylostomiasis has been documented the best in Asia and Africa (Yu et al., 1995).

 

CLINICAL FEATURES

 

Acute Infection

 

The repeated exposure of N. americanus and A. duodenale L3 through the skin can result cutaneous syndrome known as “ground itch” (Brooker et al., 2004). Sometime sleep disturbance because of intense itching (Jackson et al., 2006). This comprises a pruritic erythematous papulo-vesicular rash. Ground itch appears most commonly on the hands and feet (Figure 3). In contrast A. braziliense L3 results in cutaneous larva migrants (CLM) which is characterized by serpiginous burrows appearing most frequently on the feet, buttocks and abdomen (Blackwell and Vega-Lopez, 2001). It is not known whether other animal hookworm such as A. caninum significant causes of CLM (Landmann and Prociv, 2003). The increased frequency of CLM among travellers returning from the Caribbean resorts and among residents along the Atlantic and Gulf coasts of the United States (Yosipovitch et al., 2002). A second form of CLM associated with folliculitis has also been reported (Sakai et al., 2008; Caumes et al., 2002).

 

 

Following the entry of human hookworm L3 undergo larval pulmonary migration, which can be accompanied by cough, sore throat and fever (Miller, 1979). Pulmonary hookworm infection resembles Loffler’s pneumonitis because of its association with lung eosinophil. Increased circulating levels of IgE occur in response to migrations of third stage larvae in the lungs and intestines (Maxwell et al., 1987). Hookworm pneumonitis is usually not severe although it may last for more than a month until the L3 leave the lungs and enter into the gastrointestinal tract following swallow. Entry of hookworm L3 entry into the gastrointestinal tract and their development into adult hookworms frequently results in epigastric pain (Anyaeze, 2003) which precedes the appearance of eggs in the faeces (Maxwell et al., 1987). When A. duodenale infection occurs via the oral route, the early L3 migrations sometimes produce a syndrome which is characterized by nausea, vomiting, pharyngeal irritation, cough, dyspnea and hoarseness (Harada, 1962).

 

Hypo-proteinemia caused by Hookworm

 

The consequences of hookworm infection are loss of protein (Betson et al., 2012) by plasma being directly ingested by the adult worm (Smith et al., 2010; Albonico and Savioli, 1997), hookworm rarely contributes to malnutrition with the important exception of iron deficiency anaemia. However, in some areas of high transmission with heavy worm burdens hookworm induced protein loss is substantial and may result in hypo-proteinemia leading to edema and even anasarca (Hotez, 2002). In addition, hookworm associated protein loss results in weight loss among vulnerable populations and consequently hookworm infected pregnant women in Sierra Leone show weight gain following treatment (Torlesse, 1999).

 

Hookworm Induced Anaemia

 

The major clinical manifestation of human hookworm infection is intestinal blood loss (Roche and Layrisee, 1966; Miller, 1979; Crompton and Stephenson, 1990). Heavy hookworm infections or moderate infections induce iron deficiency and microcytic hypochromic anaemia (Smith and Brooker, 2010; King et al., 2005; Lone et al., 2004; Beaver et al., 1984). The attachment of hookworms cutting organs to the intestinal mucosa and sub-mucosa and the subsequent rupture of intestinal capillaries and arterioles cause blood loss due to secretion of antiplatelet agents by the parasite helps to maintain continuous oozing of blood at the hookworm attachment site and the free flow of blood through the parasite alimentary canal (Stanssens et al., 1996; del Valle et al., 2003).

 

Table 2: Hookworm and host blood loss. The data are abstracted from (Holland, 1987, 1989; Pawlowski et al., 1991; Crompton, 2001) who give details of sources of information and techniques used to make measurements and estimates)

 

Traits

A. duodenale

N. americanus

Intestinal blood loss in ml per worm per day, mean (range)

0.15

(0.05-0.30)

0.03

(0.01-0.04)

Number of worms causing a blood loss of 1ml/day

5 (4-7)

25

(14-50)

Blood loss (ml/day) per 1000 epg stool

Mean±SD

2.2

(1.54-2.86)

4.4±2.16

1.3

(0.82-2.24)

2.2±1.01

Iron loss (mg/day) per 1000 epg stool

0.76-1.35

0.45-0.65

Worm burden responsible for 1000 epg stool

11

32

 

The free hemoglobin is digested through the concerted action of aspartic, cysteine and metallo-hemoglobinases (Jones and Hotez, 2002; Williamson et al., 2003a; 2003b). Hookworm burdens of 40 to 160 worms are usually sufficient to cause anemia, although this depends on host iron stores (Lwambo et al., 1992) and the species of hookworm (Albonico et al., 1998). A strong association between intensity of infection and anaemia, with an increasing severity of anaemia at higher intensities of hookworm infection (Jackson, 1987; Sill et al. 1987; Shulman et al., 1996; Egwunyenga et al., 2001).Hookworm has traditionally been considered relatively unimportant in contributing towards the anaemia among pre-school children, where malaria is the important etiological factor to cause anemia (Brooker et al., 1999).

 

Perinatal Hookworm Infection

 

The hookworm anemia of pregnant women causes adverse maternal-fetal consequences including prematurity, low birth weight and impaired lactation (Miller, 1979). Hookworm infection during pregnancy has been reported to result in lactogenic transmission to neonate (Yu et al., 1995). This occurs because A. duodenale L3 can arrest their development in human tissues (Schad et al., 1973) with parturition the L3 enter the mammary glands and milk. Neonatal infection resulting from vertical transmission of hookworm results in severe disease associated with profound anaemia (Yu et al., 1995).

 

School Performance and Productivity in Adulthood

 

Severe and chronic infection with hookworm during children’s development also has consequences for their cognitive performance and ultimately their educational achievement (Jinabhai et al., 2001). Recent studies conducted throughout the developing world have provided evidence that school children infected with helminths perform poorly in tests of cognitive function (Shang, 2011; Jardim-Botelho et al., 2008; Watkins and Pollitt, 1997; Drake et al., 2000). The effect on cognitive function may occur as a result of combination of symptoms associated with infection, namely iron deficiency anaemia (IDA) and growth retardation (WHO, 2006; Grantham-McGregor and Ani, 2001; Mendez and Adair, 1999; Lozoff, 1990). A recent study among Bangladeshi tea pluckers found a negative association between intensity of hookworm and both hemoglobin and measures of labor productivity and that hemoglobin and productivity were positively associated (Gilgen et al., 2001).

 

Effects of Helminths in Allergic Patients

 

The global increase in allergy especially in urban areas (Pearce et al., 2007) has led researchers to propose a modified hygiene hypothesis in which the decline in helminths infection is associated with an increase in allergic diseases (Rook, 2009). A number of studies show that communities with helminths infection have reduced rates of allergy (van den Biggelaar et al., 2000; Cooper et al., 2003; Rodrigues et al., 2008) and the evidence that people with hookworm have less asthma (Leonardi-Bee et al., 2006; Scrivener et al., 2001; Flohr et al., 2010) has inspired researchers to use experimental infections on asthma patients (Feary et al., 2010). It is proposed that the active suppression of Th2 responses by helminths has a bystander effect on concurrent allergic responses (McSorley and Maizels, 2012). The other side of these phenomena is that anti-helminths treatment programmes risk increased rates of allergic disease and this has already been demonstrated in a number of intervention studies (Lynch et al., 1993; van den Biggelaar et al., 2004; Flohr et al., 2009).

 

DIAGNOSIS

 

There is no gold standard for diagnosis of hookworm infection and diagnosis is often delayed or overlooked due to patients presenting with non-specific gastrointestinal complaints. Nowadays widely taken samples for hookworm diagnosis are blood and fresh stool. Several diagnostic methods have been compared to detect the presence of hookworm including stool examination by Harada Mori filter paper culture, Kato-Katz thick smear, sodium acetate-acetic acid-formalin (SAF) solution, ether concentration method and the FLOTAC technique and Polymerase Chain Reaction (PCR).

 

Direct Smear

 

This method involves the identification of hookworm egg or larvae under microscope from fresh stool samples by normal saline, Eosin or Lugol’s Iodine as emulsifying agents (Cheesbrough, 1982) and Kato-Katz thick smear was prepared from each stool sample on microscope slides using 41.7 mg punched plastic templates (Katz et al., 1972), sodium acetate-acetic acid-formalin (SAF) solution (Bogoch et al., 2006; Marti and Escher, 1990), ether concentration method (Allen and Ridley, 1970) and the FLOTAC technique (Cringoli, 2006).

 

Cultural Techniques

 

Harada Mori culture techniques provide the morphological identification of hookworm larvae which first introduced by (Harada and Mori, 1955) then it is widely used (Vonghachack et al., 2015; Banu et al., 2013; Steinmann et al., 2007). This culture method is more effecitve than direct saline smear and ether concentration techniques (Marchi and Cantos, 2003).

 

Serology

 

Diagnosis of hookworm could be done by serological methods especially in patients with eosinophilia or mildly symptomatic patients. The serological methods determine the presence of hookworm antibody in the serum of the human hosts. The antibody could be determined by Enzyme Linked Immuno Sorbent Assay (ELISA) and Western Blot Analysis (WBA) (Brooker et al., 2004) as low titers of hookworm specific antibodies are noted in heavy infection along with a low or normal eosinophil count.

 

PCR

 

DNA extraction: Bead beating technique a conventional method for DNA extraction (Salonen et al., 2010) from raw stool samples but now sophisticated methods like QIAGEN DNeasy Blood & Tissue Kit are developed and used widely (Qiagen Inc., Valencia, CA).

 

Nested and Real Time PCR: The nested PCR method established for detection of hookworm. Purified DNA template was used for amplification in a DNA thermal cycler using a genus specific primer set as described by (Yong et. al., 2007). The rDNAregion comprising the first and second internal transcribed spacers plus the 5.8 S gene and approximately 50 nucleotides of the 28S rRNAwas amplified using oligonucleotide primers NC5 >: 5’-GTAGGTGAACCTGCGGAAGGATCATT-3’ (forward) and < NC2:5’-TTAGTTTCTTTTCCTCCGCT-3’ (reverse) designed to regions of the 18 S and 28 S genes, respectively (Gasser et al., 1996), and found to be conserved across a range of strong ylid nematodes. A real-time PCR method

 

Table 3: Treatment of hookworm [Modified from the Medical Letter on Drugs and Therapeutics, Drugs for Parasitic Infections (Anonymous, 2004). In children of 1-2 years the dose of albendazole is 200 mg instead of 400 mg, based on a recommendation in the Report of the WHO informal consultation on the use of praziquantel during pregnancy and lactation and albendazole/mebendazole in children less than 24 months (Kabatereine et al., 2007; Koukouknari et al., 2006; Montresor et al., 2003; WHO, 2002)]

 

Infection

Drugs

Dose

Adult

Child

Hookworm

Albendazole

400 mg once

400 mg once

Mebendazole

100 mg twice a day for 3 days

100 mg twice a day for 3 days

Pyrantel pamoate

11 mg/kg (maximum dose 1gm) for 3 days

11 mg/kg (maximum dose 1gm) for 3 days

Levamisole

2.5 mg/kg once;

repeat after 7 days in heavy infection

2.5 mg/kg once;

repeat after 7 days in heavy infection

 

developed by (Verweij et al., 2009) to detect hookworm DNA in fecal samples utilizing a primer and probe set.

 

TREATMENT

 

Benzimidazole anthleminthics are the current corner stone for helminths treatment because of their broad spectrum of activity, low cost, high efficacy and ease of administration (Savioli et al., 2002). Four anthleminthics are available for the treatment of hookworm such as albendazole, levamisole, mebendazole and pyrantel pamoate (Table 3). A single oral dose of ivermectin (200μg per kg body weight) kills the migrating larvae effectively (Caumes, 2003).

 

Varying cure and egg reducation rates have also been reported for levamisole and pyrantel pamoate (Utzinger and Keiser, 2004). The repeated round of pyrantel pamoate increases resistence to hookworm infection (Black et al., 2010). The anthelminthic treatment reduced intensities of hookworm and other helminths infection and improved haemoglobin among school children (Stephenson et al., 1989). Allergy of the patients misdirected anti-parasitic response of hypersensitive individuals (Artis et al., 2012). Not only children can benefit from treatment, in a recent study among pregnant women in Sierra Leone anthelmintic treatment increased haemoglobin concentration by 6.6 g/L, relative to controls (Torlesse and Hodges, 2000). In Sri Lanka treatment improved both the health of mothers and their birth outcomes (de Silva et al., 1999). Moreover, in areas where hookworm is endemic, reinfection often occurs within just a few months after deworming with the use of a benzimidazole anthelmintic (Albonico et al., 1995). In some cases, treatments are required three times a year to improve the iron status of the host (Stoltzfus et al., 1998; de Silva, 2003).

 

IMMUNE RESPONSES TO HOOK-WORM INFECTION

 

The complex life cycle of the hookworm offers numerous opportunities for the parasite and host to interact at the molecular level. Helminths infections are known to exert strong immune-modulatory effects on their mammalian hosts (Danilowicz-Luebert et al., 2011). Immunological responses to hookworm infection in both human and experimental animal hosts have extensively reviewed (Behnke, 1991; Loukas and Prociv, 2001). For instance, N. americanus will mature in hamsters but there is a wide change on the number of L3 that develop in adult hamsters (Rose and Behnke, 1990; Xue et al., 2003) and which frequently acquire resistance and do not develop patent infections (Rajasekariah et al., 1985; Xue et al., 2003). Likewise, A. duodenale will develop in dogs only with the administration of exogenous steroids (Leiby et al., 1987). This may be because the two species that account for almost all human infections, A. duodenale and N. americanus are highly host specific (Beaver et al., 1984). Hookworm based immune therapy resulted suppression of pro-inflammatory anti-gliadin immune-response and induction of systemic and mucosal type 2 cytokine response (Gaze et al., 2012) although overt suppression of clinical disease was not observed (Daveson et al., 2011).

 

The aspect of human immune response against hookworm infection is antibody levels to crude larval and adult soluble extracts or adult excretory/secretory products (Loukas and Prociv, 2001); often hookworm secretory/excretory products suppress intestinal colitis (Ferreira et al., 2013). The methods used to identify antibody responses (Sarles, 1938; Otto et al., 1942; Sheldon and Groover, 1942) and detailed analysis of Ig subclasses by enzyme-linked immune-sorbent assays (ELISA) and Western Blot analysis. These analyses have shown that extensive and vigorous antibody responses of all five of the human immunoglobulin (Ig) isotypes are mounted against crude antigen preparations in naturally infected individuals (Carr and Pritchard, 1987; Behnke, 1991; Loukas and Prociv, 2001). As antibody isotypes also differ in their biological properties, including their ability to mediate or block the killing of helminths (Shackelford et al., 1988; Khalife et al., 1989; Dunne et al., 1993).

 

Along with most helminths, the antibody response to hookworm consists predominantly of the Th2 antibody isotypes, IgG1, IgG4 and IgE with most attention on IgE because during hookworm infection serum levels of IgE increase 100-fold (Jarrett and Bazin, 1974). IgE participates in an orchestrated IgE network (Sutton and Gould, 1993; Garraud et al., 2003) with activation of this system often leading to cellular (mast cell, basophil and eosinophil) degranulation with toxicity against helminths (Garraud et al., 2003). The interpretation for the high levels of non-specific IgE found in the serum if an infected individual is a reduction in the risk of anaphylaxis (Hagan, 1993). The small proportion of the serum IgE that is raised against N. americanus is highly specific (Pritchard and Walsh, 1995) epitopes than other immunoglobulin isotypes.

 

During human intestinal infection with canine hookworm A. caninum, IgE responses proved to be more specific than the IgG responses to adult antigens with selected patients generating detectable levels of IgE but not IgG to a diagnostic A. caninum antigen (Jiz et al., 2009; Loukas et al., 1994).The IgG1 and IgG4 level are also elevated in hookworm infection with IgG4 against L3 suggested to be a marker of active infection with N. americanus (Palmer et al., 1996) and A. duodenale (Xue et al., 2000). The role of IgG4 is poorly understood although like IgE it is up regulated in atopic conditions and helminths allergic infections (Capron, 2011). It is thought to down regulate the immune response by competitively inhibiting IgE mediated mechanisms; i.e. blocking mast cell activation (Rihet et al., 1991) and IgG4 is stronger among other immunoglobulin (Geiger et al., 2011).

 

Adult hookworm also induces the production of secretory IgE, IgG and IgM but not IgA and the levels of these Ig return to normal after anthelminthic treatment. The absence of secretory IgA may reflect the secretion of hookworm proteases that specifically cleave IgA (Loukas and Prociv, 2001). Despite the extensive antibody response to infection, there is limited conclusive evidence that these antibodies offer any protection (Pritchard et al., 1995) by either significantly reducing larval or adult hookworm numbers, similar to that found for schistosome infections (Hagan et al., 1991; Dunne et al., 1992) and parasite burden because of the numerous uncontrolled variables such as exposure, behavioral modifications and co-infection with other helminths (Woolhouse, 1992; 1993).

 

The hallmark feature of the immune response to helminths infection is peripheral blood eosinophilia (Loukas and Prociv, 2001). Eosinophils predominate in the inflammatory responses to hookworm L3 in tissues and with sufficient larval dose, can be reflected in peripheral blood eosinophilia (Geiger et al., 2008; Behnke, 1991). Circulating eosinophils from human volunteers infected with N. americanus were functional and secreted superoxide (White et al., 1986). Peripheral eosinophil responses in experimental human infections with either N. americanus (Maxwell et al., 1987) or A. doudenale (Nawalinski and Schad, 1974) were boosted greatly by the arrival and development of worms in the gut and evidence suggests that these cells can kill infected larval stages but not the adults of most helminths species investigated (Meeusen and Balic, 2000). Mast cell degranulation in response to IgE allergen interaction plays a critical role in local mobilization and activation of eosinophils. While mast cell proteases degrade cuticular collagens of adult N. americanus (McKean and Pritchard, 1989) and are considered crucial to the host response to hookworms.

 

Eosinophilia, mastocytosis and IgE stimulation are the important three main immune alterations observed during a hookworm infection in humans. But the overall immune responses of human hosts to hookworm infection are remarkably similar to infections with other helminths; dominated by the production of T-helper2 (Th2) cytokines interleukin (IL-4, IL-5, IL-9, IL-10 and IL-13) which are consistent with the development of strong IgE, eosinophil and mast cell responses mentioned above. Therefore, the inherent ability of helminths to induce Th2 responses lead to elucidation of the underlying mechanisms in lung, skin or gut (Obata-Ninomiya et al., 2013; Harvie et al., 2013; Harvie et al., 2010; Knott et al., 2007) that lead to Th2 responses and in terms of understanding Th2 response function (MacDonald et al., 2002) along with increased mucus and fluid production to eject the parasites (Madden et al., 2004). A concomitant observation was the high level of IL-10 compared to other cytokines (IL-4, IL-5 and IL-13) that accompany chronic hookworm infection and decline that accompanies treatment (Turner et al., 2013; MacDonald et al., 2002) by using anthelmintic.

 

Experimental systems have demonstrated that the host protection to nematode infection may be a CD4+ T cell-dependent process with the IL-4 secreted by these cells has an essential or very important role in the process (Finkelman et al., 1997; Lawrence et al., 1998). Hookworm antigen induces cell apoptosis by intrinsic mitochondrial pathway and induces generation of suppressor CD4+ and CD8+ T cells (Gazzinelli-Guimaraes et al., 2013; Cuellar et al., 2009). However, the steps from IL-4 secretion to worm elimination are not still clear. Much of the basic cellular immunology of hookworm in humans remains to be done, especially the elucidation of the roles of cytokines, chemokine and cell surface markers during infection.

 

STRATEGIES FOR CONTROL AND PREVENTION

 

Because of its high transmission potential, hookworm has proven to be extremely difficult to eliminate or eradicate in areas of poverty and poor sanitation (Brooker et al., 2004). The interruption of transmission cycle is another key components of STH specially hookworm control (Hawdon, 2014; Strunz et al., 2014; Anderson et al., 2014; Truscott et al., 2014). The current strategy to control hookworm is chemotherapy although it alone cannot remove hookworm infection (Freeman et al., 2013) but with support of health education (Tomono et al., 2003), improved water and sanitation (Greene et al., 2012; Freeman and Clasen, 2011; Asaolu and Ofoezie, 2003) and socio-economic status (Mihrshahi et al., 2009). Beyond saying improved sanitation and hygiene are essential for the long term control of parasitic diseases. However, the availability of latrine facilities is associated with lower hookworm intensities (Chongsuvivatwong et al., 1996), the impact of introducing sanitation on infection levels may only be evident after decades (Esrey et al., 1991) and may not be completely effective. Despite associations between hookworm infection and use of footwear to protect from exposure to infective larvae, there is debate as to whether promotion of footwear is an effective control strategy (Albonico et al., 1999). The long time required for improved sanitation and behaviour change, for a quick-acting and medium term measure to control helminths infection is chemotherapy (Kabatereine et al., 2007; Koukouknari et al., 2006; Albonico et al., 1999) mentioned in (Table 3). The use of anti-hookworm vaccine with anthelmintic drugs brought benefits (Hotez et al., 2003). Efforts led by the World Health Organization have focused on annual, twice-yearly or thrice-yearly doses in school because the heaviest intensities of STH infections are most commonly encountered in school-age children (World Bank, 2003). Treatment of potable water also can reduce the transmission of hookworm (Clasen et al., 2007).

 

CONCLUSION

 

Hookworm infection is still a topic of great concern because of its high morbidity. The public health importance of hookworm infections are very little of practical significance is known about the details of how the parasites interact with their hosts including the nature and effectiveness of the immune responses that are generated. Interim it is warranted to investigate the risk factors involved in hookworm infection and screen patients from endemic areas prior to receiving chemotherapy. It is also essential to highlight that plethora of prevention effort in endemic countries such as health education campaigns on the disease, proper sanitation through appropriate disposal of fecal material, regular deworming and the use of protective footwear are achievable goals for reducing the occurrence of hookworm.

 

Acknowledgments

 

The authors acknowledge the contributions of his fellow research team for their sincere suggestion during the writing of whole manuscript.

 

Conflict of interests

 

The Authors declares no conflict of interest.

 

Authors’ contribution

 

The first author M Hossain did data management task, wrote the whole manuscript and made all the relevant correction during revision. MJU Bhuiyan helped in the supervision of the whole manuscript, and suggested for relevant correction of common error in written manuscript.

 

REFERENCES

 

  • Abate A, Kibret B, Teklu T, Yalew A (2013). Cross-Sectional Study on the prevalence of intestinal parasites and associated risk factors in Teda health centre, Northwest Ethiopia. ISRN Parasitol. 1(2): 1-5.
  • Abossie A, Seid M (2014). Assessment of the prevalence of intestinal parasitosis and associated risk factors among primary school children in Chencha town, Southern Ethiopia. BMC Public Health. 14: 166. http://dx.doi.org/10.1186/1471-2458-14-166
  • Adeniyi KA, Oseihie II, Chiedozie KO, Adedapo BA (2015). Prevalence of helminth infestation during pregnancy and its association with maternal anemia and low birth weight. Int. J. Gynecol. Obstetrics. 129: 199-202. http://dx.doi.org/10.1016/j.ijgo.2014.12.002
  • Albonico M, Savioli L (1997). Hookworm infection and disease: advances for control. Ann. Ist. Super Sanita. 33: 567-579.
  • Albonico M, Smith PG, Ercole E (1995). Rate of reinfection with intestinal nematodes after treatment of children with mebendazole or albendazole in a highly endemic area. Trans. R. Soc. Trop. Med. Hyg. 89: 538-41. http://dx.doi.org/10.1016/0035-9203(95)90101-9
  • Albonico M, Stoltzfus RJ, Ercole E, Cancrini G (1998). Epidemiological evidence for a differential effect of hookworm species, A. duodenale or N. americanus, on iron status of children. Int. J. Epidemiol. 27: 530-537. http://dx.doi.org/10.1093/ije/27.3.530
  • Alemu A, Atnafu A, Addis Z, Teklu T, Mathewos B (2011). Soil transmitted helminths and S. mansoni infections among school children in Zarima Town, Northwest Ethiopia. BMC Infect. Dis. 11: 189-96. http://dx.doi.org/10.1186/1471-2334-11-189
  • Ali S, Barbhuiya M, Rahman A, Chowdhury S (1985). Incidence of hookworm among the workers in tea garden. Bangladesh Med. Res. Council Bull. 11: 69-74.
  • Allen AVH, Ridley DS (1970). Further observations on the formol-ether concentration technique for faecal parasites. J. Clin. Pathol. 23: 545-546. http://dx.doi.org/10.1136/jcp.23.6.545
  • Anderson RM, Truscott J, Hollingsworth TD (2014). The coverage and frequency of mass drug administration required to eliminate persistent transmission of soil-transmitted helminths. Trans. R. Soc. L. B. Biol. Sci. 369: 43-5. http://dx.doi.org/10.1098/rstb.2013.0435
  • Anderson RM (1982). The population dynamics and control of hookworm and roundworm infection. In: Anderson, RM., editor. Population Dynamics of Infectious Diseases: Theory and Applications. London: Chapman and Hall. Pp. 67-109. http://dx.doi.org/10.1007/978-3-642-68635-1
  • Anonymous (2004). Drugs for parasitic infections. Med Lett Drugs Ther August. http://www.themedicalletter.com/restricted/articles/ w1189c.pdf (accessed Nov 12, 2005).
  • Anyaeze CM (2003). Reducing burden of hookworm disease in the management of upper abdominal pain in the tropics. Trop. Doctor. 33: 174-175.
  • Appleton CC, Gouws E (1996). The distribution of common intestinal nematodes along an altitudinal transect in Kwa-Zulu Natal, South Africa. Ann. Trop. Med. Parasitol. 90: 181-188.
  • Appleton CC, Maurihungirire M, Gouws E (1999). The distribution of helminth infections along the coastal plain of Kwazulu-Natal province, South Africa. Ann. Trop. Med. Parasitol. 93: 859-868. http://dx.doi.org/10.1080/00034989957862
  • Artis D, Maizels RM, Finkelman FD (2012). Forum: Immunology: Allergy challenged. Nature. 484: 458-459. http://dx.doi.org/10.1038/484458a
  • Asaolu SO, Ofoezie IE (2003). The role of health education and sanitation in the control of helminth infections. Acta Trop. 86: 283-294. http://dx.doi.org/10.1016/S0001-706X(03)00060-3
  • Augustine DL (1922). Investigations on the control of hookworm disease. X. Experiments on the length of life of infective hookworm larvae in soil. Am. J. Hyg. 2: 177-187.
  • Awasthi S, Bundy DAP, Savioli L (2003). Helminthic infections. Br. Med. J. 23: 431-433. http://dx.doi.org/10.1136/bmj.327.7412.431
  • Balen J, Raso G, Li YS, Williams GM (2011). Risk factors for helminths infections in a rural and a peri-urban setting of the Dongting Lake area, People’s Republic of China. Int. J. Parasitol. 41: 1165-73. http://dx.doi.org/10.1016/j.ijpara.2011.07.006
  • Banu SS, Ahmed BN, Banu SG and Ameen KH (2013). Prevalence of Soil Transmitted Helminthes (STHs) Infection among Children Aged 2-17 Years in Urban and Rural Areas of Dhaka District in Bangladesh. Bangladesh J. Med. Microbiol. 5: 16-22. http://dx.doi.org/10.3329/bjmm.v5i2.16933
  • Bartram J, Cairncross S (2010). Hygiene, sanitation, and water: forgotten foundations of health. PLoS Med. 7: e1000367. http://dx.doi.org/10.1371/journal.pmed.1000367
  • Beaver PC, jung RC (1985). Animal Agents and Vectors of Human Disease, 5th Ed. Philadelphia, Lea & Febiger.
  • Beaver PC, Jung RC, and Cupp EW (1984). Clinical parasitology, 9th 700 Loukas and Prociv Clin. Microbiol. Rev. ed. Pp. 269-301. Lea & Febiger, Philadelphia, Pa.
  • Behnke JM, De Clercq D, Vercruysse J (2000). The epidemiology of human hookworm infections in the southern region of Mali. Trop. Med. Int. Health. 5: 343-354. http://dx.doi.org/10.1046/j.1365-3156.2000.00553.x
  • Behnke J (1991). Human parasitic diseases: Hookworm infections. In: Gilles, HM., Ball, P., editors. Immunology. Amsterdam: Elsevier. Pp. 93-155.
  • Bethony J, Chen J, Xiao S, Zhan B (2002). Emerging patterns of hookworm infections: Infl uence of aging on the intensity of Necator infection in Hainan Province, People’s Republic of China. Clin. Infect. Dis. 35: 1336-1344. http://dx.doi.org/10.1086/344268
  • Bethony J, Brooker S, Hotez PJ (2006). Soil-transmitted helminth infections: ascariasis, trichuriasis, and hookworm. Lancet. 367: 1521-1532. http://dx.doi.org/10.1016/S0140-6736(06)68653-4
  • Betson M, Kabatereine NB, Stothard JR (2012). Use of Fecal Occult Blood Tests as Epidemiologic Indicators of Morbidity Associated with Intestinal Schistosomiasis during Preventive Chemotherapy in Young Children. Am. J. Trop. Med. Hyg. 87: 694-700. http://dx.doi.org/10.4269/ajtmh.2012.12-0059
  • Black CL, Mwinzi PN, Colley DG (2010). Influence of exposure history on the immunology and development of resistance to human S. mansoni. PLoS Negl. Trop. Dis. 4: e637. http://dx.doi.org/10.1371/journal.pntd.0000637
  • Blackwell V, Vega-Lopez F (2001). Cutaneous larva migrans: Clinical features and management of 44 cases in the returning traveler. Br. J. Dermatol. 145: 434-437. http://dx.doi.org/10.1046/j.1365-2133.2001.04406.x
  • Bogoch II, Raso G, Utzinger J (2006). Differences in microscopic diagnosis of helminths and intestinal protozoa among diagnostic centres. Eur. J. Clin. Microbiol. Infect. Dis. 25: 344-347. http://dx.doi.org/10.1007/s10096-006-0135-x
  • Bongi S, Morel A (2005). Understanding small scale providers of sanitation services: A case study of Kibera, Nairobi, Kenya: Water and Sanitation Program.
  • Brooker S, Michael E (2000). The potential of geographical information systems and remote sensing in the epidemiology and control of human helminth infections. Adv. Parasitol. 47: 245-288. http://dx.doi.org/10.1016/s0065-308x(00)47011-9
  • Brooker S, Peshu N, Snow RW (1999). Epidemiology of hookworm infection and its contribution to anaemia among pre-school children on the Kenyan coast. Trans. R. Soc. Trop. Med. Hyg. 93: 240-246. http://dx.doi.org/10.1016/S0035-9203(99)90007-X
  • Brooker S, Peshu N, Marsh K, Snow RW (1999). Epidemiology of hookworm infection and its contribution to anaemia among pre-school children on the Kenyan coast. Trans. R. Soc. Trop. Med. Hyg. 93:240-246. http://dx.doi.org/10.1016/S0035-9203(99)90007-X
  • Brooker S, Bethony J, Hotez PJ (2004). Human hookworm infection in the 21st century. Adv. Parasitol. 58: 197-288. http://dx.doi.org/10.1016/S0065-308X(04)58004-1
  • Brooker S, Clements AC, Bundy DA (2006). Global epidemiology, ecology and control of soil-transmitted helminth infections. Adv. Parasitol. 62: 221-261. http://dx.doi.org/10.1016/S0065-308X(05)62007-6
  • Bundy DAP (1988a). Sexual effects on parasite infection. Parasitol. Today. 4: 186-189. http://dx.doi.org/10.1016/0169-4758(88)90076-2
  • Bundy DAP (1995). Epidemiology and transmission of intestinal helminths. In: Farthing MJG, Keusch GT, Wakelin D, eds. Enteric infection 2: intestinal helminths. New York: Chapman & Hall Medical. Pp. 5-24.
  • Bundy D, Shaeffer S, Sembene M (2005). Chapter 61, School Based Health and Nutrition Programs. In: editors. Disease Control Priorities for Developing Countries. Oxford: Oxford University Press.
  • Bundy DAP, Keymer AE (1991). The epidemiology of hookworm infection; Human Parasitic Diseases Volume 4. Hookworm Infections. Amsterdam: Elsevier. Pp. 157-178.
  • Bungiro R, Cappello M (2004). Hookworm infection: new developments and prospects for control. Curr. Opin. Infect. Dis. 17: 421-426. http://dx.doi.org/10.1097/00001432-200410000-00006
  • Capron M (2011). Effect of parasite infection on allergic disease. Allergy. 66: 16-18. http://dx.doi.org/10.1111/j.1398-9995.2011.02624.x
  • Carroll SM, Grove D (1986). Experimental infection of humans with A. ceylanicum: Clinical, parasitological, hematological and immunological findings. Trop. Geo. Med. 38: 38-45.
  • Caumes E (2003). Treatment of cutaneous larva migrans and Toxocara infection. Fundam. Clin. Pharmacol. 17: 213-216. http://dx.doi.org/10.1046/j.1472-8206.2003.00172.x
  • Caumes E, Ly F, Bricaire F (2002). Cutaneous larva migrans with folliculitis: report of seven cases and review of the literature. Br. J. Dermatol. 146: 314-316. http://dx.doi.org/10.1046/j.0007-0963.2001.04531.x
  • Chandler AC (1929). Hookworm disease: Its distribution, biology, epidemiology, pathology, diagnosis, treatment and control: Macmillan.
  • Cheesbrough M (1982). Medical laboratory manual for tropical countries. Central African J. Med. 21: 48.
  • Chongsuvivatwong V, Greater A, Duerawee M (1996). Predictors for the risk of hookworm infection: experience from endemic villages in southern Thailand. Trans. R. Soc. Trop. Med. Hyg. 90: 630–633. http://dx.doi.org/10.1016/S0035-9203(96)90412-5
  • Clasen T, Schmidt WP, Cairncross S (2007). Interventions to improve water quality for preventing diarrhoea: systematic review and meta-analysis. BMJ. 33: 782. http://dx.doi.org/10.1136/bmj.39118.489931.BE
  • Cline BL, Little MD, Bartholomew RK, Halsey NA (1984). Larvicidal activity of albendazole against N. americanus in human volunteers. Am. J. Trop. Med. Hyg. 33: 387-394.
  • Conde JF, Feldman SR, Arcury TA (2007). Cutaneous larva migrans in a migrant latino farmworker. J. Agro Med. 12: 45-48. http://dx.doi.org/10.1300/J096v12n02_05
  • Cook DM, Swanson RC, Booth GM (2009). A Retrospective analysis of prevalence of gastrointestinal Parasites among school children in the Palajunoj ValleyGuatemala. J. Health Popul. Nutr. 27: 31-40. http://dx.doi.org/10.3329/jhpn.v27i1.3321
  • Cooper PJ, Chico ME, Griffin GE, Nutman TB (2003). Reduced risk of atopy among school-age children infected with geohelminth parasites in a rural area of the tropics. J. Allergy Clin. Immunol. 111: 995-1000. http://dx.doi.org/10.1067/mai.2003.1348
  • Cort WW, Payne GC (1922). Investigations on the control of hookworm disease. VI. A study on the effect of hookworm control measures on soil pollution and infestation in a sugar estate. Am. J. Hyg. 2: 107-148.
  • Craig JM, Scott AL (2014). Helminths in the lungs. Parasite Immunol. 36: 463-474. http://dx.doi.org/10.1111/pim.12102
  • Cringoli G (2006). FLOTAC, a novel apparatus for a multivalent faecal egg count technique. Parassitol. 48: 381-384.
  • Crompton DWT (2001). The public health significance of hookworm disease. Parasitol. 121: 39-50. http://dx.doi.org/10.1017/S0031182000006454
  • Crompton DWT, whitehead RR (1993). Hookworm infections and human iron metabolism. Parasitol. 107: 137-145. http://dx.doi.org/10.1017/S0031182000075569
  • Crompton DWT, Stephenson LS (1990). Hookworm infection, nutritional status and productivity. In Hookworm Disease. Pp. 231-264. London and Philadelphia, Taylor and Francis Ltd.
  • Cuellar C, Wu W, Mendez S (2009). The hookworm tissue inhibitor of metalloproteases (Ac-TMP-1) modifies dendritic cell function and induces generation of CD4 and CD8 suppressor T cells. PLoS Negl. Trop. Dis. 3: e439. http://dx.doi.org/10.1371/journal.pntd.0000439
  • Danilowicz-Luebert E, Steinfelder S, Hartmann S (2011). Modulation of specific and allergy-related immune responses by helminths. J. Biomed. Biotechnol. 8: 157-8.
  • Daveson AJ, Loukas A, Croese J (2011). Effect of hookworm infection on wheat challenge in celiac disease: a randomised double-blinded placebo controlled trial. PLoS One. 6: e17366. http://dx.doi.org/10.1371/journal.pone.0017366
  • de Silva NR (2003). Impact of mass chemotherapy on the morbidity due to soil-transmitted nematodes. Acta. Trop. 86: 197-214. http://dx.doi.org/10.1016/S0001-706X(03)00035-4
  • de Silva NR, Sirisena JL, Gunasekera DP, Ismail MM, de Silva HJ (1999). Effect of mebendazole therapy during pregnancy on birth outcome. Lancet. 353: 1145-1149. http://dx.doi.org/10.1016/S0140-6736(98)06308-9
  • De Silva NR, Brooker S, Hotez PJ, Savioli L (2003a). Soil-transmitted helminth infections: updating the global picture. Trends Parasitol. 19: 547-551. http://dx.doi.org/10.1016/j.pt.2003.10.002
  • del Valle A, Jones BF, Cappello M (2003b). Isolation and molecular cloning of a secreted hookworm platelet inhibitor from adult A. caninum. Mol. Biochem. Parasitol. 129: 167-177. http://dx.doi.org/10.1016/S0166-6851(03)00121-X
  • Despommier DD, Hotez PJ, Knirsch C (2000). Parasitic diseases. 4th Ed. New York: Apple Tree Productions.
  • Drake LJ, Jukes MCH, Sternberg RJ, Bundy DAP (2000). Geohelminth infections (Ascariasis, Trichuriasis and Hookworm): cognitive and developmental impacts. Seminars Pediatric Infect. Dis. 11: 245-251. http://dx.doi.org/10.1053/spid.2000.9638
  • Dunne DW, Richardson BA, Thorne KJ, Butterworth AE (1993). The use of mouse/ human chimaeric antibodies to investigate the roles of different antibody isotypes, including IgA2, in the killing of Schistosoma mansoni schistosomula by eosinophils. Parasite Immunol. 15: 181-185. http://dx.doi.org/10.1111/j.1365-3024.1993.tb00598.x
  • Egwunyenga AO, Nmorsi OPG, Duhlinska-Popova DD (2001). Plasmodium/intestinal helminth coinfections among pregnant Nigerian women. Memórias do Instituto Oswaldo Cruz. 96: 1055-1059. http://dx.doi.org/10.1590/S0074-02762001000800005
  • Ekiz C, Agaoglu L, Karakas Z, Gurel N, Yalcin I (2005). The effect of iron deficiency anemia on the function of the immune system. Hematol. J. 5: 579-83. http://dx.doi.org/10.1038/sj.thj.6200574
  • Ensink JH, van der Hoek W, Amerasinghe FP (2005). High risk of hookworm infection among wastewater farmers in Pakistan. Trans. R. Soc. Trop. Med. Hyg. 99: 809-818. http://dx.doi.org/10.1016/j.trstmh.2005.01.005
  • Ettling J (1990). The role of the Rockefeller Foundation in hookworm research and control. In Hookworm Disease. Pp. 3-14. London and Philadelphia, Taylor and Francis, Ltd.
  • Farthing MJG, Keusch GT, Wakelin D (1995). Enteric Infection: Mechanisms, Manifestations, and Management. Vol. II. London: Chapman and Hall. Pp. 129-150.
  • Feary JR, Venn AJ, Pritchard DI, Britton JR (2010). Experimental hookworm infection: a randomized placebo-controlled trial in asthma. Clin. Exp. Allergy. 40: 299-306. http://dx.doi.org/10.1111/j.1365-2222.2009.03433.x
  • Ferreira I, Smyth D, Giacomin P, Ruyssers N (2013). Hookworm excretory/secretory products induce interleukin-4 (IL-4)+ IL-10+ CD4+ T cell responses and suppress pathology in a mouse model of colitis. Infect. Immun. 81: 2104-11. http://dx.doi.org/10.1128/IAI.00563-12
  • Flohr C, Quinnell RJ, and Britton J (2009). Do helminth parasites protect against atopy and allergic disease? Clin. Exp. Allergy. 39: 20-32. http://dx.doi.org/10.1111/j.1365-2222.2008.03134.x
  • Flohr C, Tuyen LN, Quinnell RJ, Pritchard DI, Britton J (2010). Reduced helminth burden increases allergen skin sensitization but not clinical allergy: A randomized, double-blind, placebo671 controlled trial in Vietnam. Clin. Exp. Allergy. 40: 131-42.
  • Freeman MC, Clasen T (2011). Assessing the impact of a school-based safe water intervention on household adoption of point-of-use water treatment practices in southern India. Am. J. Trop. Med. Hyg. 84: 370-378. http://dx.doi.org/10.4269/ajtmh.2011.10-0361
  • Freeman MC, Ogden S, Addiss DG, Amnie AG (2013). Integration of water, sanitation, and hygiene for the prevention and control of neglected tropical diseases: A rationale for inter-sectoral collaboration. PLoS Negl. Trop. Dis. 7: e2439. http://dx.doi.org/10.1371/journal.pntd.0002439
  • Furst T, Keiser J, Utzinger J (2012). Global burden of human food-borne trematodiasis: a systematic review and meta-analysis. Lancet Infect. Dis. 12: 210-21. http://dx.doi.org/10.1016/S1473-3099(11)70294-8
  • Gandhi NS, Chen JZ, Shuhua X, Hawdon JM, Hotez PJ (2001). Epidemiology of Necator americanus hookworm infections in Xiulongkan Village, Hainan Province, China: High prevalence and intensity among middle-aged and elderly residents. J. Parasitol. 87: 739-743. http://dx.doi.org/10.2307/3285128
  • Garraud O, Perraut R, Riveau G, Nutman TB (2003). Class and subclass selection in parasite-specific antibody responses. Trends Parasitol. 19: 300-304. http://dx.doi.org/10.1016/S1471-4922(03)00139-9
  • Gasser RB, Stewart LE,Speare R (1996). Genetic markers in ribosomal DNA for hookworm identification. Acta. Trop. 62: 15-21. http://dx.doi.org/10.1016/S0001-706X(96)00015-0
  • Gaze S, McSorley HJ, Daveson J, Croese J, Loukas A (2012). Characterising the mucosal and systemic immune responses to experimental human hookworm infection. PLoS Pathog. 8: e1002520. http://dx.doi.org/10.1371/journal.ppat.1002520
  • Gazzinelli-Guimaraes PH, Dhom-Lemos LC, Ricci ND (2013). Cell apoptosis induced by hookworm antigens: a strategy of immunomodulation. Front Biosci. 5: 662-75.
  • Geiger SM, Alexander ND, Bethony JM (2011). N. americanus and helminths co-infections: further down-modulation of hookworm-specific type 1 immune responses. PLoS Negl. Trop. Dis. 5: e1280. http://dx.doi.org/10.1371/journal.pntd.0001280
  • Geiger SM, Fujiwara RT, Bethony JM (2008). Early stage-specific immune responses in primary experimental human hookworm infection. Microbes Infect. 10: 1524-1535. http://dx.doi.org/10.1016/j.micinf.2008.09.003
  • Gilgen DD, Mascie-Taylor CG and Rosetta LL (2001). Intestinal helminths infections, anaemia and labour productivity of female tea pluckers in Bangladesh. Trop. Med. Int. Health. 6: 449-57. http://dx.doi.org/10.1046/j.1365-3156.2001.00729.x
  • Grantham-McGregor S, Ani C (2001). A review of studies on the effect of iron deficiency on cognitive development in children. J. Nutr. 131: 649-666.
  • Greene LE, Freeman MC, Moe C (2012). Impact of a school-based hygiene promotion and sanitation intervention on pupil hand contamination in Western Kenya: a cluster randomized trial. Am. J. Trop. Med. Hyg. 87: 385-393. http://dx.doi.org/10.4269/ajtmh.2012.11-0633
  • Gruber JS, Reygadas F, Nelson K (2013). A stepped wedge, cluster randomized trial of a household UV-disinfection and safe storage drinking water intervention in rural Baja California Sur, Mexico. Am. J. Trop. Med. Hyg. 89: 238-245. http://dx.doi.org/10.4269/ajtmh.13-0017
  • Gunawardena GS, Karunaweera ND, Ismail MM (2005). Effects of climatic, socio-economic and behavioural factors on the transmission of hookworm (N. americanus) on two low country plantations in Sri Lanka. Ann. Trop. Med. Parasitol. 99: 601-609. http://dx.doi.org/10.1179/136485905X51436
  • Haas W, Haberl B, Stiegeler P, Syafruddin (2005). Behavioural strategies used by the hook worms N. americanus and A. duodenale to find, recognize and invade the human host. Parasitol. Res. 95: 30-39. http://dx.doi.org/10.1007/s00436-004-1257-7
  • Hagan P (1993). IgE and protective immunity to helminth infections. Parasite Immunology. 15:1-4. http://dx.doi.org/10.1111/j.1365-3024.1993.tb00565.x
  • Hagan P, Blumenthal UJ, Simpson AJ, Wilkins HA (1991). Human IgE, IgG4 and resistance to reinfection with Schistosoma haematobium. Nature. 349: 243-245. http://dx.doi.org/10.1038/349243a0
  • Hall A, Conway DJ, Anwar KS, Rahman ML (1994). Strongyloides stercoralis in an urban slum community in Bangladesh: factors independently associated with infection. Trans. R. Soc. Trop. Med. Hyg. 88: 527-530. http://dx.doi.org/10.1016/0035-9203(94)90146-5
  • Halpenny CM, Paller C, Valdes VE, Scott ME (2013). Regional, household and individual factors that influence soil transmitted helminth reinfection dynamics in preschool children from rural indigenous Panama. PLoS Negl. Trop. Dis. 7: e2070. http://dx.doi.org/10.1371/journal.pntd.0002070
  • Harada Y, Mori O (1955). A new method for culturing hookworm. Yonago Acta. Med. 1: 177-179.
  • Harada Y (1962). Wakana disease and hookworm allergy. Yonago Acta. Med. 6: 109-118.
  • Harvie M, Camberis M, Le Gros G (2013). Development of CD4 T cell dependent immunity against N. brasiliensis infection. Front Immunol. 4: 74. http://dx.doi.org/10.3389/fimmu.2013.00074
  • Harvie M , Camberis M, Tang SC, Delahunt B (2010). The lung is an important site for priming CD4 T-cell-mediated protective immunity against gastrointestinal helminth parasites. Infect. Immunol. 78: 3753-3762. http://dx.doi.org/10.1128/IAI.00502-09
  • Hawdon JM (2014). Controlling soil-transmitted helminths: Time to think inside the box? J. Parasitol. 100: 166-88. http://dx.doi.org/10.1645/13-412.1
  • Hawdon JM, Datu B, Crowell M (2003). Molecular cloning of a novel multidomain Kunitz-type proteinase inhibitor from the hookworm A. caninum. J. Parasitol. 89: 402-407. http://dx.doi.org/10.1645/0022-3395(2003)089[0402:MCOANM]2.0.CO;2
  • Hawdon JM, Jones BF, Hoffman DR, Hotez PJ (1996). Cloning and characterization of Ancylostoma-secreted protein. A novel protein associated with the transition to parasitism by infective hookworm larvae. J. Biol. Chem. 22: 6672-6678.
  • Hegner R, Root FM, augustine DI, huff CG (1938). Parasitology. New York, Appleton Century Crofts Inc.
  • Hoagland KE, Schad GA (1978). N. americanus and A. duodenale: life history parameters and epidemiological implications of two sympathetic species. Exp. Parasitol. 44: 36-49. http://dx.doi.org/10.1016/0014-4894(78)90078-4
  • Holland CV (1987). Hookworm infection. In Impact of Helminth Infections on Human Nutrition. (Stephenson LS & Holland, CV) Pp. 128-160. London and Philadelphia. Taylor and Francis Ltd.
  • Holland CV (1989). An assessment of the impact of four intestinal nematode infections on human nutrition. Clinic. Nutr. 8: 239-250.
  • Holland CV, Taren DL, Tiffany J, Rivera G (1988). Intestinal helminthiases in relation to the socioeconomic environment of Panamanian children. Soc. Sci. Med. 26: 209-213. http://dx.doi.org/10.1016/0277-9536(88)90241-9
  • Hossain M and Bhuiyan MJU (2016). Overview of Strongyloidiasis: A Neglected Tropical Disease. J. Adv. Parasitol. 3(3): 93-103. http://dx.doi.org/10.14737/journal.jap/2016/3.3.93.103
  • Hossain M, Alam MDS, Khair M, Sayeed MDA, Bhuiyan MJU (2016). Prevalence and Risk Factors of Strongyloides stercoralis infection in selected tea garden of Sylhet, Bangladesh. J. Trop. Dis. 4: 206. http://dx.doi.org/10.4172/2329-891X.1000206
  • Hossain M (2015). Prevalence and risk factors of soil transmitted helminths (STHs) infection among tea garden community in Sylhet and slum dwellers of Dhaka city, Bangladesh. MS, Dissertation, Sylhet Agricultural University, Sylhet.
  • Hotez PJ (2002). China’s hookworms. China Q. 172: 1029-1041. http://dx.doi.org/10.1017/S0009443902000608
  • Hotez PJ, Bottazzi ME, Goodenow MM (2015). Neglected Tropical Diseases among the Association of Southeast Asian Nations (ASEAN): Overview and Update. PLoS Negl. Trop. Dis. 9(4): e0003575. http://dx.doi.org/10.1371/journal.pntd.0003575
  • Hotez PJ, Feng Z, Davis GM (1997). Emerging and reemerging helminthiases and the public health of China. Emerging Infect. Dis. 3: 303-310. http://dx.doi.org/10.3201/eid0303.970306
  • Hotez PJ, Hawdon J, Schad GA (1993). Hookworm larval amphiparatenesis: the Caenorhabditis elegans Dafc paradigm. Parasitol. Today. 9: 23-6. http://dx.doi.org/10.1016/0169-4758(93)90159-D
  • Hotez PJ, Zhan B and Bethony JM (2003). Progress in the development of a recombinant vaccine for human hookworm disease: The Human Hookworm Vaccine Initiative. Int. J. Parasitol. 33: 1245-58. http://dx.doi.org/10.1016/S0020-7519(03)00158-9
  • Hotez PJ, Brooker S, Bethony JM, Xiao S (2004). Hookworm infection. New Eng. J. Med. 351: 799-807. http://dx.doi.org/10.1056/NEJMra032492
  • Hotez PJ, Bethony JM, Loukas A (2008). Multivalent anthelminthic vaccine to prevent hookworm and schistosomiasis. Expert Rev. Vaccines. 7: 745-752. http://dx.doi.org/10.1586/14760584.7.6.745
  • Hotez PJ (1989). Hookworm disease in children. Pediatric Infect. Dis. J. 8: 516-520. http://dx.doi.org/10.1097/00006454-198908000-00009
  • Hotez PJ, Bundy DAP, Savioli L (2005). Disease Control Priorities in Developing Countries. 2nd Edition. WHO, World Bank, NIH, Oxford University Press.
  • Hotez PJ (1996). Human hookworm infection: experience from endemic villages in southern Thailand. Trans. R. Soc. Trop. Med. Hyg. 90: 630-633. http://dx.doi.org/10.1016/S0035-9203(96)90412-5
  • Humphries DL, Stephenson LS, Khanh LT (1997). The use of human faeces for fertilizer is associated with increased intensity of hookworm infection in Vietnamese women. Trans. R. Soc. Trop. Med. Hyg. 91: 518-520. http://dx.doi.org/10.1016/S0035-9203(97)90007-9
  • Jackson A, Heukelbach J, Harms G, Feldmeier H (2006). A study in a community in Brazil in which cutaneous larva migrans is endemic. Clin. Infect. Dis. 43: 13-18. http://dx.doi.org/10.1086/505221
  • Jackson RT, Jackson LC (1987). Biological and behvioural contributors to anemia during pregnancy in Liberia, West Africa. Human Biol. 59: 585-597.
  • Jardim-Botelho A, Raff S, Gazzinelli MF (2008). Hookworm, A. lumbricoides infection and polyparasitism associated with poor cognitive performance in Brazilian schoolchildren. Trop. Med. Int. Health. 13: 994-1004. http://dx.doi.org/10.1111/j.1365-3156.2008.02103.x
  • Jarrett E, Bazin H (1974). Elevation of total serum IgE in rats following helminth parasite infection. Nature. 251: 613-614. http://dx.doi.org/10.1038/251613a0
  • Jinabhai CC, Taylor M, Sullivan KR (2001). A randomized controlled trial of the effect of antihelminthic treatment and micronutrient fortification on health status and school performance of rural primary school children. Ann. Trop. Paediatrics. 21: 319-33. http://dx.doi.org/10.1080/07430170120093508
  • Jiraanankul V, Aphijirawat W, Traub RJ, Leelayoova S (2011). Incidence and risk factors of hookworm infection in a rural community of central Thailand. Am. J. Trop. Med. Hyg. 84: 594-598. http://dx.doi.org/10.4269/ajtmh.2011.10-0189
  • Jiz M, Friedman JF, Olveda R, Acosta L, Kurtis JD (2009). Immunoglobulin E (IgE) responses to paramyosin predict resistance to reinfection with Schistosoma japonicum and are attenuated by IgG4. Infect. Immunol. 77: 2051-2058. http://dx.doi.org/10.1128/IAI.00012-09
  • Jones BF, Hotez PJ (2002). Molecular cloning and characterization of Ac-mep-1, a developmentally regulated gut luminal metalloendopeptidase from adult A. caninum hookworms. Mol. Biochem. Parasitol. 119: 107-116. http://dx.doi.org/10.1016/S0166-6851(01)00409-1
  • Kabatereine NB, Brooker S, Fleming FM (2007). Impact of a national helminth control programme on infection and morbidity in Ugandan school children. Bull. World Health Organ. 85: 91-9. http://dx.doi.org/10.2471/BLT.06.030353
  • Katz N, Chaves A, Pellegrino J (1972). A simple device for quantitative stool thick-smear technique in Schistosomiasis mansoni. Rev. Inst. Med. Trop. Sao Paulo. 14: 397-400.
  • Khair M, Khanum H., Hossain M., Alam MS (2016). Prevalence, Risk Factors and Comparative Diagnosis of Soil Transmitted Helminths (STH) in children of slum areas of Dhaka and tea garden areas of Sylhet. 12th Biennial Conference, BAU, Bangladesh, Abstract. Pp. 55
  • Khanum H, Rahman F, Zaman RF (2014). Occurrence of intestinal parasites among the teachers, students and staffs of Dhaka University. J. Asiatic Soc. Bangladesh Sci. 39: 239-246. http://dx.doi.org/10.3329/jasbs.v39i2.17863
  • King CH, Dickman K, Tisch DJ (2005). Reassessment of the cost of chronic helmintic infection: a meta-analysis of disability-related outcomes in endemic schistosomiasis. Lancet. 365: 1561-9. http://dx.doi.org/10.1016/S0140-6736(05)66457-4
  • Kirwan P, Asaolu S, Smith H, Holland C (2009). Soil-transmitted helminth infections in Nigerian children aged 0–25 months. J. Helminthol. 83: 261-266. http://dx.doi.org/10.1017/S0022149X08201252
  • Knight R, Merrett TG (1981). Hookworm infection in rural Gambia. Seasonal changes, morbidity and total IgE levels. 75: 299-314.
  • Koukouknari A, Kazibwe F, Tukahebwa EM (2006). Morbidity indicators of Schistosoma mansoni: relationship between infection and anemia in Ugandan school children before and after praziquantel and albendazole chemotherapy. Am. J. Trop. Med. Hyg. 75: 278-86.
  • Labiano-Abello N, Wilson ML, Hotez PJ (1999). Epidemiology of hookworm infection in Itagua, Paraguay: a cross sectional study. Memórias do Instituto Oswaldo Cruz. 94: 583-586. http://dx.doi.org/10.1590/S0074-02761999000500003
  • Landmann JK, Prociv P (2003). Experimental human infection with the dog hookworm, A. caninum. Med. J. Aus. 178: 69-71.
  • Leiby DA, el Naggar HM, Schad GA (1987). Thirty generations of A. duodenale in laboratory reared beagles. J. Parasitol. 73: 844-848. http://dx.doi.org/10.2307/3282429
  • Leonardi-Bee J, Pritchard D, Britton J (2006). Asthma and current intestinal parasite infection: systematic review and meta-analysis. Am. J. Respir. Crit. Care Med. 174: 514-23. http://dx.doi.org/10.1164/rccm.200603-331OC
  • Liabsuetrakul T, Bavonnarongdet P, Buadung A (2009). Epidemiology and the effect of treatment of soil-transmitted helminthiasis in pregnant women in southern Thailand. 40: 211-222.
  • Lili Z, Bingxiang Z, Hong T, Hotez P, Zhen F (2000). Epidemiology of human geohelminth infections (ascariasis, trichuriasis and necatoriasis) in Lushui and Puer Counties, Yunnan Province, China. Southeast Asian J. Trop. Med. Public Health. 31: 448-453.
  • Logan M (2009). Methods in Improving the Quality of N. americanus larvae for use in Therapeutic Applications. Honours thesis. James Cook University, Townsville.
  • Lone FW, Qureshi RN, Emanuel F (2004). Maternal anaemia and its impact on perinatal outcome. Trop. Med. Int. Health. 9: 486-90. http://dx.doi.org/10.1111/j.1365-3156.2004.01222.x
  • Looss A (1901). On the penetration of Ancylostoma larvae into the human skin. Centralblatt Backteriol. Parasitenkunde. 29: 733-739.
  • Loukas A, Opdebeeck J, Croese J, Prociv P, (1994). Immunologic incrimination of A. caninum as a human enteric pathogen. Am. J. Trop. Med. Hyg. 50: 69-77.
  • Loukas A, Prociv P (2001). Immune responses in hookworm infections. Clinic. Microbiol. Rev. 14: 689-703. http://dx.doi.org/10.1128/CMR.14.4.689-703.2001
  • Lozoff B (1990). Has iron deficiency been shown to cause altered behaviour in infants?. In: Dobbing, J., editor. Brain, Behaviour and Iron in the Infant. London: Springer-Verlag, Pp. 107-131. http://dx.doi.org/10.1007/978-1-4471-1766-7_11
  • Lwambo N, Bundy D, Medley G (1992). A new approach to morbidity risk assessment in hookworm endemic communities. Epidemiol. Infect. 108: 469-481. http://dx.doi.org/10.1017/S0950268800049980
  • Mabaso MLH, Appleton CC, Hughes JC, Gouws E (2003). The effect of soil type and climate on hookworm (N. americanus) distribution in KwaZulu-Natal, South Africa. Trop. Med. Int. Health. 8: 722-727. http://dx.doi.org/10.1046/j.1365-3156.2003.01086.x
  • Madden KB, Yeung KA, Gause WC, Finkelman FD (2004). Enteric nematodes induce stereotypic STAT6-dependent alterations in intestinal epithelial cell function. J. Immunol. 172: 5616-5621. http://dx.doi.org/10.4049/jimmunol.172.9.5616
  • Marchi BJ, Cantos GA (2003). Evaluation of techniques for the diagnosis of S. stercoralis in human immunodeficiency virus (HIV) positive and HIV negative individuals in the city of Itajai, Brazil. Braz. J. Infect. Dis. 7: 402-408.
  • Marti H, Escher E (1990). SAF-an alternative fixation solution for parasitological stool specimens. Schweiz. Med. Wochenschr. 120: 1473-1476.
  • Maxwell C, Hussain R, Schad GA, Ottesen EA (1987). The clinical and immunological responses of normal human volunteers to low dose hookworm (N. americanus) infection. Am. J. Trop. Med. Hyg. 37: 126-134.
  • McKean PG, Pritchard DI (1989). The action of a mast cell protease on the cuticular collagens of N. americanus. Parasite Immunol. 11: 293-297. http://dx.doi.org/10.1111/j.1365-3024.1989.tb00667.x
  • McSorley HJ, Maizels RM (2012). Helminth infections and host immune regulation. Clinic. Microbiol. Rev. 25: 585-608. http://dx.doi.org/10.1128/CMR.05040-11
  • Meeusen EN, Balic A (2000). Do eosinophils have a role in the killing of helminth parasites? Parasitol. Today. 16: 95-101. http://dx.doi.org/10.1016/S0169-4758(99)01607-5
  • Mendez MA, Adair LS (1999). Severity and timing of stunting in the first two years of life affect performance on cognitive tests in late childhood. J. Nutr. 129: 1555-1562.
  • Michaud C, Ique C, Montoya E, Gozalo A (2003). A survey for helminth parasites in feral New World non-human primate populations and its comparasiton with parasitiological data from man in the region. J. Med. Primatol. 32: 341-345. http://dx.doi.org/10.1046/j.1600-0684.2003.00037.x
  • Michelle LK, Klaus IM, Paul RG, Hui W, Paul SF (2007). Impaired resistance in early secondary Nippostrongylus brasiliensis infections in mice with defective eosinophilopoeisis. Int. J. Parasitol. 37: 1367-1378. http://dx.doi.org/10.1016/j.ijpara.2007.04.006
  • Mihrshahi S, Casey GJ, Biggs BA (2009). The effectiveness of 4 monthly albendazole treatment in the reduction of soil-transmitted helminth infections in women of reproductive age in Viet Nam. Int. J. Parasitol. 39: 1037-1043. http://dx.doi.org/10.1016/j.ijpara.2009.01.013
  • Miller TA (1979). Hookworm infection in man. Adv. Parasitol. 17: 315-384. http://dx.doi.org/10.1016/S0065-308X(08)60552-7
  • Miller GC (1981). Helminths and the transmammary route of infection. Parasitol. 82: 335-342. http://dx.doi.org/10.1017/S0031182000057073
  • Montresor A, Awashti S, Crompton DWT (2003). Use of benzimidazoles in children younger than 24 months for the treatment of soil-transmitted helminthiasis. Acta. Trop. 86: 223-232. http://dx.doi.org/10.1016/S0001-706X(03)00042-1
  • Montresor A, Crompton DWT, Gyorkos TW, Savioli L (2002). Helminth control in school-age children: A guide for managers of control programmes. Geneva: World Health Organization. Available: http:⁄⁄www.who.int/wormcontrol/documents/helminth_control/en/ (Accessed 26 January 2005).
  • Moore SL, Wilson K (2002). Parasites as a Viability Cost of Sexual Selection in Natural Populations of Mammals. Sci. 297: 2015-2018. http://dx.doi.org/10.1126/science.1074196
  • Nagahana M, Tanabe K, Okamoto K, Ito S, Fukutome S (1963). Experimental infection of three cases of human beings with N. americanus larvae through the mucous membrane of the mouth. Jap. J. Parasitol. 12: 162-167.
  • Nawalinski TA, Schad GA (1974). Arrested development in A. duodenale: Course of a self-induced infection in man. Am. J. Trop. Med. Hyg. 23: 895-898.
  • Ndyomugyenyi R, Kabatereine N, Olsen A, Magnussen P (2008). Malaria and hookworm infections in relation to haemoglobin and serum ferritin levels in pregnancy in Masindi district, western Uganda. Trans. R. Soc. Trop. Med. Hyg. 102: 130-6. http://dx.doi.org/10.1016/j.trstmh.2007.09.015
  • Needham C, Kim HT, Hall A, Bundy DAP (1998). Epidemiology of soil-transmitted nematode infections in Ha Nam Province, Vietnam. Trop. Med. Int. Health. 3: 904-912. http://dx.doi.org/10.1046/j.1365-3156.1998.00324.x
  • Nematian J, Nematian E, Gholamrezanezhad A, Asgari AA (2004). Prevalence of intestinal parasitic infections and their relation with socio-economic factors and hygienic habits in Tehran primary school students. Acta Trop. 92: 179-86. http://dx.doi.org/10.1016/j.actatropica.2004.06.010
  • Ngui R, Ching LS, Kai TT, Roslan MA, Lim YA (2012a). Molecular identification of human hookworm infections in economically disadvantaged communities in Peninsular Malaysia. Am. J. Trop. Med. Hyg. 86: 837-842. http://dx.doi.org/10.4269/ajtmh.2012.11-0446
  • Ngui R, Lim YA, Traub R, Mahmud R, Mistam MS (2012b). Epidemiological and genetic data supporting the transmission of A. ceylanicum among human and domestic animals. PLoS Negl. Trop. Dis. 6: e1522.
  • Nicholson JK, Lindon JC, Holmes E (1999). ‘Metabonomics’: understanding the metabolic responses of living systems to pathophysiological stimuli via multivariate statistical analysis of biological NMR spectroscopic data. Xenobiotica. 29: 1181-1189. http://dx.doi.org/10.1080/004982599238047
  • Nwosu ABC (1978). Investigations into the free-living phase of the cat hookworm life-cycle. Zeitschrift fur Parasitenkunde. 56: 243-249. http://dx.doi.org/10.1007/BF00931717
  • Nwosu ABC (1981). The community ecology of soil-transmitted helminth infection of humans in a hyperendemic area of southern Nigeria. Ann. Trop. Med. Parasitol. 75: 197-203. http://dx.doi.org/10.1080/00034983.1981.11687428
  • Obata-Ninomiya K, K Ishiwata, Tsutsui H (2013). The skin is an important bulwark of acquired immunity against intestinal helminths. J. Exp. Med. 210: 2583-2595. http://dx.doi.org/10.1084/jem.20130761
  • Olatunde BO, Onyemelukwe GC (1994). Immunosuppression in Nigerians with hookworm infection. Afr. J. Med. Sci. 23: 221-225.
  • Olsen A, Samuelsen H, Onyango-Ouma W (2001). A study of risk factors for intestinal helminth infections using epidemiological and anthropological approaches. J. Biosoc. Sci. 33: 569-584. http://dx.doi.org/10.1017/S0021932001005697
  • Olsen A, van Lieshout L, Polman K, Steinmann P (2009). Strongyloidiasis-the most neglected of the neglected tropical diseases? Trans. R. Soc. Trop. Med. Hyg. 103: 967-72. http://dx.doi.org/10.1371/journal.pntd.0000533
  • Orihel TC (1971). N. americanus infection in primates. J. Parasitol. 57: 117–21. http://dx.doi.org/10.2307/3277764
  • Otto G, Schugman N, Groover M (1942). A precipitin reaction resulting from N. americanus larvae in serum from hookworm infected individuals. Proceed. Helminthol. Soc. Washington DC. 9: 25-26.
  • Palmer DR, Bradley M, Bundy DAP (1996). IgG4 responses to antigens of adult N. americanus: potential for use in large-scale epidemiological studies. Bull. World Health Org. 74: 381-386
  • Pawlowski ZS, Schad GA, Stott GJ (1991). Hookworm Infection and Anaemia. Geneva, World Health Organisation.
  • Pearce N, Aït-Khaled N, Mitchell E, Robertson C (2007). Worldwide trends in the prevalence of asthma symptoms: phase III of the International Study of Asthma and Allergies in Childhood (ISAAC). Thorax. 62: 758-766. http://dx.doi.org/10.1136/thx.2006.070169
  • Poulin R (1996). Sexual inequalities in helminth infections: a cost of being male? Am. Naturalist. 147: 287-295. http://dx.doi.org/10.1086/285851
  • Pritchard DI, Brown A (2001). Is N. americanus approaching a mutualistic symbiotic relationship with humans? Trends Parasitol. 17: 169-172. http://dx.doi.org/10.1016/S1471-4922(01)01941-9
  • Pritchard DI, Walsh EA (1995). The specificity of the human IgE response to N. americanus. Parasite Immunol. 17: 605-607. http://dx.doi.org/10.1111/j.1365-3024.1995.tb01005.x
  • Prociv P (1997). Pathogenesis of human hookworm infection: insights from a ‘new’ zoonosis. Chemic. Immunol. 66: 62-98. http://dx.doi.org/10.1159/000058666
  • Prociv P, Croese J (1996). Human enteric infection with A. caninum: hookworms reappraised in the light of a “new” zoonosis. Acta Trop. 62: 23-44. http://dx.doi.org/10.1016/S0001-706X(96)00016-2
  • Pullan RL, Kabatereine NB, Quinnell RJ, Brooker S (2010). Spatial and genetic epidemiology of hookworm in a rural community in Uganda. PLoS Negl. Trop. Dis. 4: e713. http://dx.doi.org/10.1371/journal.pntd.0000713
  • Quihui L, Valencia ME, Hagan P, Morales G (2006). Role of the employment status and education ofmothers in the prevalence of intestinal parasitic infections in Mexican rural schoolchildren. BMC Public Health. 6: 225. http://dx.doi.org/10.1186/1471-2458-6-225
  • Quinnell RJ, Griffin J, Raiko A, Pritchard DI (2001). Predisposition to hookworm infection in Papua New Guinea. Trans. R. Soc. Trop. Med. Hyg. 95: 139-42. http://dx.doi.org/10.1016/S0035-9203(01)90138-5
  • Rajasekariah GR, Deb BN, Dhage KR, Bose S (1985). Site of resistance to N. americanus in hamsters. Acta Trop. 42: 333-340.
  • Ratnayaka RMKT, Wang ZJ (2012). Prevalence and effect of personal hygiene on transmission of helminth infection among primary schoolchildren living in slums. Int. J. Multidiscip. Res. 2: 1-12.
  • Rihet P, Demeure CE, Prata A, Dessein AJ (1991). Evidence for an association between human resistance to Schistosoma mansoni and high anti-larval IgE levels. Eu. J. Immunol. 21: 2679-86. http://dx.doi.org/10.1002/eji.1830211106
  • Roche M, Layrisse M (1966). The nature and causes of hookworm anaemia. Am. J. Trop. Med. Hyg. 15: 1031-110.
  • Roche M, Layrisse M (1966). The nature and causes of “hookworm anemia”. Am. J. Trop. Med. Hyg. 15: 1029-1102.
  • Rodríguez E, Anadón AM, Gárate T, Ubeira FM (2008). Novel sequences and epitopes of diagnostic value derived from the Anisakis simplex Ani s 7 major allergen. Allergy. 63: 219-225. http://dx.doi.org/10.1111/j.1398-9995.2007.01564.x
  • Rook GAW (2009). Review series on helminths, immune modulation and the hygiene hypothesis: the broader implications of the hygiene hypothesis. Immunol. 126: 3-11. http://dx.doi.org/10.1111/j.1365-2567.2008.03007.x
  • Rose RA, Behnke JM (1990). N. americanus in the DSN hamster: density-dependent expulsion of adult worms during primary infection. Parasitol. 100: 469-478. http://dx.doi.org/10.1017/S0031182000078781
  • Saathoff E (2002). PhD Thesis. University of Copenhagen. Geohelminth and Schistosoma haematobium infection in school children from rural northern KwaZulu-Natal, South Africa.
  • Sakai H, Otsubo S, Yamasaki H, Kagei N, Iizuka H (2008). Multiple papules and nodules on the face and neck caused by the larvae of an unknown nematode: a non-creeping type eruption. J. Am. Acad. Dermatol. 58: 668-670. http://dx.doi.org/10.1016/j.jaad.2007.06.023
  • Sakti H, Nokes C, Hendratno S, Hall A, Bundy DA, Satoto (1999). Evidence for an association between hookworm infection and cognitive function in Indonesian school children. Trop. Med. Int. Health. 4: 322-334. http://dx.doi.org/10.1046/j.1365-3156.1999.00410.x
  • Salonen A, Nikkila J, Immonen O, de Vos WM (2010). Comparative analysis of fecal DNA extraction methods with phylogenetic microarray: effective recovery of bacterial and archaeal DNA using mechanical cell lysis. J. Microbiol. Methods. 81: 127-134. http://dx.doi.org/10.1016/j.mimet.2010.02.007
  • Sandy S, Sumarni S, Soeyoko (2014). Foot wear as a risk factor of hookworm infection in elementary school students. Univ. Med. 33: 133-40
  • Sarles MP (1938). The in vitro action of immune rat serum on the nematode, Nippostrongylus muris. J. Infect. Dis. 62: 337-348. http://dx.doi.org/10.1093/infdis/62.3.337
  • Savioli L, Stansfield S, Neira M, Shein AM (2002). Schistosomiasis and soil-transmitted helminth infections: forging control efforts. Trans. R. Soc. Trop. Med. Hyg. 96: 577-579. http://dx.doi.org/10.1016/S0035-9203(02)90316-0
  • Schad GA, Chowdhury AB, Thomas J, Tonascia A (1973). Arrested development in human hookworm infections: an adaptation to a seasonally unfavourable external environment. Sci. 180: 52-54. http://dx.doi.org/10.1126/science.180.4085.502
  • Schad GA, Banwell JG (1984). Hookworms. In Tropical and Geographical Medicine. Pp. 359-372. New York, McGraw-Hill Book Co.
  • Schad GA, Nawalinski TA, Kochar V, Cross JH (1983). Human ecology and the distribution and abundance of hookworm populations. Human ecology and infectious diseases. London: Academic Press. Pp. 187-223. http://dx.doi.org/10.1016/B978-0-12-196880-9.50013-0
  • Schad GA, Warren SK (1990). Hookworm disease current status and new directions. London: Taylor and Francis. Pp. 231-264.
  • Sengchanh K, Manithong V, Peter O, Boungnong B (2011). Soil-transmitted helminth infections and risk factors in preschool children in southern rural Lao People’s Democratic Republic. Trans. R. Soc. Trop. Med. Hyg. 105: 160-166. http://dx.doi.org/10.1016/j.trstmh.2010.11.011
  • Shackelford PG, Nelson SJ, Palma AT, Nahm MH (1988). Human antibodies to group A streptococcal carbohydrate. Ontogeny, subclass restriction, and clonal diversity. J. Immunol. 140: 3200-3205.
  • Shang Y (2011) Burden of diseases on soil-transmitted helminth infections among school-age children in China. Doctoral Thesis, National Institute of Parasitic Diseases, Chinese Centre for Disease Control and Prevention. Shanghai, China.
  • Sheldon AJ, Groover MEJ (1942). An experimental approach to the problem of acquired immunity in human hookworm (N. americanus) infections. Am. J. Hyg. 36: 183-186.
  • Shiferaw MB, Mengistu AD (2015). Helminthiasis: Hookworm Infection Remains a Public Health Problem in Dera District, South Gondar, Ethiopia. PLoS One. 10(12): e0144588. http://dx.doi.org/10.1371/journal.pone.0144588
  • Shulman CE, Graham WJ, Snow RW, Marsh K (1996). Malaria is an important cause of anaemia in primigravidae: evidence from a district hospital in coastal Kenya. Trans. R. Soc. Trop. Med. Hyg. 90: 535-539. http://dx.doi.org/10.1016/S0035-9203(96)90312-0
  • Sill PR, Hill AVS, Igo JD (1987). Multifactorial aetiology of anaemia of pregnancy in Port Moresby, Papua New Guinea. Papua New Guinea Med. J. 30: 193-198
  • Smith G, Schad GA (1990). A. duodenale and N. americanus: Effect of temperature on egg development and mortality. Parasitol. 99: 127-132. http://dx.doi.org/10.1017/S0031182000061102
  • Smith JL, Brooker S (2010). Impact of hookworm infection and deworming on anaemia in non-pregnant populations: a systematic review. Trop. Med. Int. Health. 15: 776-95. http://dx.doi.org/10.1111/j.1365-3156.2010.02542.x
  • Soares Magalhaes RJ, Barnett AG, Clements AC (2011). Geographical analysis of the role of water supply and sanitation in the risk of helminth infections of children in West Africa. Proceed. Nat. Acad. Sci. USA. 108: 284-90. http://dx.doi.org/10.1073/pnas.1106784108
  • Stanssens P, Bergum PW, Lasters I, Vlasuk GP (1996). Anticoagulant repertoire of the hookworm A. caninum. Proceed. Nat. Acad. Sci. USA. 93: 2149-2154. http://dx.doi.org/10.1073/pnas.93.5.2149
  • Steenhard NR, Storey PA, Nansen P, Polderman AM (2000). The role of pigs as transport hosts of the human helminths Oesophagostomum bifurcum and N. americanus. Acta Trop. 76: 125-130. http://dx.doi.org/10.1016/S0001-706X(00)00077-2
  • Steinmann P, Zhou XN, Wang XZ, Utzinger J (2007). Occurrence of Strongyloides stercoralis in Yunnan Province, China, and comparison of diagnostic methods. PLoS Negl. Trop. Dis. 1: 75. http://dx.doi.org/10.1371/journal.pntd.0000075
  • Stiles CW (1902). A new species of hookworm (Uncinaria americana) parasitic in man. Harvard University press, pages: 111-113.
  • Stoltzfus RJ, Albonico M, Schulze KJ, Savioli L (1998). Effects of the Zanzibar school-based deworming program on iron status of children. Am. J. Clin. Nutr. 68: 179-86.
  • Stoltzfus RJ, Chwaya HM, Albonico M, Savioli L (1997). Epidemiology of iron deficiency anemia in Zanzibari schoolchildren: the importance of hookworms. Am. J. Clin. Nutr. 65: 153-159.
  • Strunz EC, Addiss DG, Stocks ME, Utzinger J, Freeman MC (2014). Water, sanitation, hygiene, and soil-transmitted helminth infection: a systematic review and meta-analysis. PLoS Med. 11: e1001620. http://dx.doi.org/10.1371/journal.pmed.1001620
  • Sultana Y, Gilbert GL, Ahmed BN, Lee R (2012). Strongyloidiasis in a high risk community of Dhaka, Bangladesh. Trans. R. Soc. Trop. Med. Hyg. 106: 756-762. http://dx.doi.org/10.1016/j.trstmh.2012.08.011
  • Sutton BJ, Gould HJ (1993). The human IgE network. Nature. 366: 421-428. http://dx.doi.org/10.1038/366421a0
  • Tomono N, Anantaphruti MT, Leerapan P, Kojima S, Looareesuwan S (2003). Risk factors of helminthiases among school children in southern Thailand. Southeast Asian J. Trop. Med. Public Health. 34: 264-268.
  • Tomović M, Skiljević D, Vukićević J, Pavlović MD, Medenica L (2008). Two cases of probable endogenous extensive cutaneous larva migrans in Serbia. Acta Dermatovenerol Alp Panonica Adriat. 17: 37-40.
  • Torlesse H, Hodges M (2000). Anthelminthic treatment and haemoglobin concentrations during pregnancy. Lancet. 356: 1083. http://dx.doi.org/10.1016/S0140-6736(00)02738-0
  • Torlesse H (1999). PhD Dissertation. University of Glasgow. Parasitic infections and anaemia during pregnancy in Sierra Leone.
  • Traub RJ, Robertson ID, Irwin P, Andrew Thompson RC (2004). The prevalence, intensities and risk factors associated with geohelminth infection in tea-growing communities of Assam, India. Trop. Med. Int. Health. 9: 688-701. http://dx.doi.org/10.1111/j.1365-3156.2004.01252.x
  • Truscott JE, Hollingsworth TD, Brooker SJ, Anderson RM (2014). Can chemotherapy alone eliminate the transmission of soil transmitted helminths? Parasit. Vectors. 7: 266. http://dx.doi.org/10.1186/1756-3305-7-266
  • Turner JE, PJ Morrison, Wilhel C (2013). IL-9-mediated survival of type 2 innate lymphoid cells promotes damage control in helminth-induced lung inflammation. J. Exp. Med. 210: 2951-2965. http://dx.doi.org/10.1084/jem.20130071
  • Udonsi JK (1984). N. americanus infection: a cross-sectional study of a rural community in relation to some clinical symptoms. Ann. Trop. Med. Parasitol. 78: 443-444.
  • Udonsi JK, Atata G (1987). N. americanus: temperature, pH, light, and larval development, longevity, and desiccation tolerance. Exp. Parasitol. 63: 136-142. http://dx.doi.org/10.1016/0014-4894(87)90154-8
  • Udonsi JK, Nwosu ABC, Anya AO (1980). Necator americanus: Population Structure, Distribution, and Fluctuations in Population Densities of Infective Larvae in Contaminated Farmlands. Fur Parasitenkunde. 63: 251-259. http://dx.doi.org/10.1007/BF00931987
  • Utzinger J, Keiser J (2004). Schistosomiasis and soil-transmitted helminthiasis: common drugs for treatment and control. Expert Opin. Pharmacotherapy. 5: 263-286. http://dx.doi.org/10.1517/14656566.5.2.263
  • Van den Biggelaar AHJ, Kremsner PG, Yazdanbakhsh M (2004). Long-term treatment of intestinal helminths increases mite skin-test reactivity in Gabonese schoolchildren. J. Infect. Dis. 189: 892-900. http://dx.doi.org/10.1086/381767
  • Van den Biggelaar AH, Deelder AM, Kremsner PG, Yazdanbakhsh M (2000). Decreased atopy in children infected with Schistosoma haematobium: a role for parasite-induced interleukin-10. Lancet. 356: 1723-1727. http://dx.doi.org/10.1016/S0140-6736(00)03206-2
  • Verweij J, Canales M, Polderman A (2009). Molecular diagnosis of Strongyloides stercoralis in faecal samples using real-time PCR. Trans. R. Soc. Trop. Med. Hyg. 103: 342-6. http://dx.doi.org/10.1016/j.trstmh.2008.12.001
  • Vonghachack Y, Sayasone S, Odermatt P (2015). Epidemiology of Strongyloides stercoralis on Mekong islands in southern Laos. Acta Trop. 141: 289-294. http://dx.doi.org/10.1016/j.actatropica.2014.09.016
  • Wang X, Zhang L, Luo R, Wang G, Chen Y (2012). Soil-Transmitted Helminth Infections and Correlated Risk Factors in Preschool and School-Aged Children in Rural Southwest China. PLoS One. 7: e45939. http://dx.doi.org/10.1371/journal.pone.0045939
  • Watkins WE, Pollitt E (1997). “Stupidity or Worms”: do intestinal worms impair mental performance? Psychol. Bull. 121: 171-191. http://dx.doi.org/10.1037/0033-2909.121.2.171
  • Wegayehu T, Tsalla T, Seifu B, Teklu T (2013). Prevalence of intestinal parasitic infections among highland and lowland dwellers in Gamo Area, South Ethiopia. BMC Public Health. 13: 151. http://dx.doi.org/10.1186/1471-2458-13-151
  • White CJ, Maxwell CJ, Gallin JI (1986). Changes in the structural and functional properties of human eosinophils during experimental hookworm infection. J. Infect. Dis. 154: 778-783. http://dx.doi.org/10.1093/infdis/154.5.778
  • WHO (2002). Report of the WHO informal consultation on the use of praziquantel during pregnancy/lactation and albendazole/ mebendazole in children under 24 months. 8–9 April. http:// whqlibdoc.who.int/hq/2003/WHO_CDS_CPE_PVC_2002.4.pdf.
  • WHO (2006). Child Growth Standards. Geneva, Switzerland: World Health Organization. 2006.
  • Williams-Blangero S, Blangero J, Bradley M (1997). Quantitative genetic analysis of susceptibility to hookworm infection in a population from rural Zimbabwe. Hum. Biol. 69: 201-8.
  • Williamson AL, Brindley PJ, Knox DP, Hotez PJ, Loukas A (2003a). Digestive proteases of blood feeding nematodes and other parasites. Trends Parasitol. 19: 417-423. http://dx.doi.org/10.1016/S1471-4922(03)00189-2
  • Williamson AL, Brindley PJ, Zhan B, Loukas A (2003b). Hookworm aspartic protease, Na-APR-2 cleaves human haemoglobin and serum proteins in a host-specific fashion. J. Infect. Dis. 187: 484-494. http://dx.doi.org/10.1086/367708
  • Woolhouse ME (1992). A theoretical framework for the immunoepidemiology of helminth infection. Parasite Immunol. 14: 563-578. http://dx.doi.org/10.1111/j.1365-3024.1992.tb00029.x
  • Woolhouse ME (1993). A theoretical framework for immune responses and predisposition to helminths infection. Parasite Immunol. 15: 583-594.
  • World Bank (2003). School deworming at a glance. Washington (DC): World Bank. Pp. 4.
  • Xue HC, Wang Y, Zhan B, Drake L, Feng Z, Hotez PJ (2000). Epidemiology of human ancylostomiasis among rural villagers in Nanlin County, Anhui Province, China. Southeast Asian J. Trop. Med. Public Health. 31: 736-741.
  • Xue J, Liu S, Ren HN, Li TH, Hotez P, Xiao SH (2003). Necator americanus: maintenance through one hundred generations in golden hamsters (Mesocricetus auratus). I. Hotst sex-associated differences in hookworm burden and fecundity. Exp. Parasitol. 104: 62-66. http://dx.doi.org/10.1016/S0014-4894(03)00094-8
  • Yadla S, Sen HG, Hotez PJ (2003). An epidemiological study of ancylostomiasis in a rural area of Kanpur District Uttar Pradesh, India. Indian J. Public Health. 47: 53-60.
  • Ye XP, Wu ZX, Sun FH (1994). The population biology and control of N. americanus in a village community in south-eastern China. Ann. Trop. Med. Parasitol. 88: 635-643.
  • Yong TS, Lee JH, Sim S, Lee J, Min DY, Chai JY, Rim HJ (2007). Differential diagnosis of Trichostrongylus and hookworm eggs via PCR using ITS-1 sequence. Korean J. Parasitol. 45: 69-74. http://dx.doi.org/10.3347/kjp.2007.45.1.69
  • Yosipovitch G, Sugeng MW, Seow CS, Goh CL (2002). Widespread and unusual presentations of cutaneous larva migrans acquired in tropical sandy beach resorts. J. Eu. Acad. Dermatol. Venereal. 16: 284-285. http://dx.doi.org/10.1046/j.1468-3083.2002.00487_1.x
  • Yu SH, Jian ZX, Xu LQ (1995). Infantile hookworm disease in China. A review. Acta Trop. 59: 265-270. http://dx.doi.org/10.1016/0001-706X(95)00089-W
  • Yulan W, Xiao S, Xue J, Utzinger J, Elaine H (2009). Systems Metabolic Effects of a N. americanus Infection in Syrian Hamster. J. Proteome Res. 8: 5442-5450. http://dx.doi.org/10.1021/pr900711j
  • Zeehaida M, Zairi NZ, Maimunah A, Madihah B (2011). Strongyloides stercoralis in common vegetables and herbs in Kota Bharu, Kelantan, Malaysia. Trop. Biomed. 28: 188-93.
  • Zhan B, Liu Y, Badamchian M, Hawdon JM, Hotez PJ (2003). Molecular characterization of the Ancylostoma-secreted protein (ASP) family from the adult stage of Ancylostoma caninum. Int. J. Parasitol. 33: 897-907. http://dx.doi.org/10.1016/S0020-7519(03)00111-5
  •