Parasites and malignancies, a review 一些寄生虫也可能导致某些癌症

中文版谷歌中文翻譯(90% 準確率) | English translation
Buy/Sell Your Domains Here。在這裡購買/出售您的域名
Contact Dr. Lu for information about cancer treatments。聯繫盧博士,獲取有關癌症治療資訊。

我们都知道,一些致病细菌和病毒可能会导致癌症,如宫颈癌,肝癌和胃癌。 但是一些寄生虫也可能导致某些癌症。 这是2012年对寄生虫和癌症的评论。 我们在网站上发布这篇文章,供英文读者阅读。 我们几乎没有时间做翻译。 如果任何人可以自愿做任何翻译或编辑,请与我们联系。 谢谢。

Parasite. 2012 May; 19(2): 101–115.
Published online 2012 May 15. doi:  10.1051/parasite/2012192101
PMCID: PMC3671432

Language: English | French

Parasites and malignancies, a review, with emphasis on digestive cancer induced by Cryptosporidium parvum(Alveolata: Apicomplexa)

1 Biologie & Diversité des Pathogènes Eucaryotes Émergents (BDEEP), Centre d’Infection et d’Immunité de Lille (CIIL), INSERM U1019, CNRS UMR8402, EA4547, Université Lille Nord-de-France, Institut Pasteur de Lille France
2 Environnement et Santé, FLST, Université Catholique de Lille, Université Lille Nord-de-France France
3 Service d’Anatomie et de Cytologie Pathologiques, Groupe Hospitalier de l’Institut Catholique de Lille France
4 Instituto de Biomedicina de Sevilla, CIBER de Epidemiología y Salud Pública, Servicio de Medicina Interna, Virgen del Rocío Hospital Universitario Sevilla España
5 Parasitologie-Mycologie, Centre de Biologie, Pathologie & Génétique, Centre Hospitalier Régional et Universitaire de Lille, Université Lille Nord-de-France France
6 Cátedra de Parasitología, Escuela de Medicina, “José María Vargas”, Universidad Central de Venezuela (UCV) Caracas Venezuela
*Correspondence: Eduardo Dei-Cas, Institut Pasteur de Lille, 1, rue du Professeur Calmette, BP 245, 59019 Lille, France. E-mail: [email protected]

Abstract

The International Agency for Research on Cancer (IARC) identifies ten infectious agents (viruses, bacteria, parasites) able to induce cancer disease in humans. Among parasites, a carcinogenic role is currently recognized to the digenetic trematodes Schistosoma haematobium, leading to bladder cancer, and to Clonorchis sinensis or Opisthorchis viverrini, which cause cholangiocarcinoma. Furthermore, several reports suspected the potential association of other parasitic infections (due to Protozoan or Metazoan parasites) with the development of neoplastic changes in the host tissues. The present work shortly reviewed available data on the involvement of parasites in neoplastic processes in humans or animals, and especially focused on the carcinogenic power of Cryptosporidium parvum infection. On the whole, infection seems to play a crucial role in the etiology of cancer.

Keywords: cancer, parasite, ClonorchisCryptosporidiumOpisthorchis, protists, Schistosoma, infection

Introduction

Cancer is a leading cause of death worldwide: it accounted for 7.6 million deaths (around 13 % of all deaths) in 2008 (Globocan, 2008). More than 70 % of all cancer deaths occurred in low- and middle-income countries. Deaths from cancer worldwide are projected to continue rising, with an estimated 11 million deaths in 2030 (WHO, 2011).

Cancer is a condition in which abnormal cells divide and grow out of control and are able to invade other tissues. Changes to gene expression patterns are an important feature of cancer cells. These alterations are caused directly or indirectly by genetic or epigenetic events (Stransky et al., 2006). A recent publication of IARC (2011) classified organ, tissue or cell changes associated with cancer in three non-exclusive levels: (a) physiological level, which refers to exposurerelated modifications in the physiology and/or response of cells, tissues and organs (mitogenesis, compensatory cell division, escape from apoptosis, altered cellular adhesion, inflammation, hyperplasia, metaplasia, preneoplasia, angiogenesis, altered steroidal hormone secretion or immunity); (b) cellular level, which refers to exposure-related alterations in the signaling pathways involved in critical cellular processes related to increased risk for cancer (altered expression of genes regulating DNA repair, cyclin-dependent kinases, altered patterns of posttranslational protein modification, changes in apoptosis regulatory factors, in factors involved in DNA replication and transcription or in gap–junction-mediated intercellular communication); (c) molecular level, which refers to exposure-related changes in key cellular structures at the molecular level, including mainly genotoxicity (formation of DNA adducts, DNA strand breaks, gene mutation, chromosomal aberrations, aneuploidy, changes in DNA methylation patterns). These genetic errors which can lead to neoplastic transformation could be caused by hormones, drugs, infectious agents, chemicals, physical or mechanical trauma, and other chronic irritations (Preston-Martin et al., 1990). Among infectious agents, a broad spectrum of them have been linked to cancer, but currently the strength of causal evidence ranges from confirmed, as such Human T cell lymphotropic virus type I (HTLV-I), to speculative, as such Pneumocystis jirovecii (de la Horra, 2004).

Around the world, infection is one of the most important causes of cancer and infection-associated cancers are increasing at an alarming rate (De Martel & Franceschi, 2009). The estimated total cancer attributable to infections in the year 2002 was 1.9 million cases, or 17.8 % of the global cancer burden (Parkin, 2006). The main recognized agents are the bacterium Helicobacter pylori (5.5 % of all cancer), the human papilloma viruses (5.2 %), the hepatitis B and C viruses (4.9 %), Epstein-Barr virus (EBV) (1 %), human immunodeficiency virus (HIV) together with the human herpes virus 8 (0.9 %) and HTLV-I (0.03 %) (Parkin, 2006). However, other pathogens including parasites can also cause cancer. Among the worms (De Martel & Franceschi, 2009; IARC, 2011), the widespread digenetic trematode Schistosoma haematobium can cause urinary bladder cancer and the flukes Opisthorchis viverrini and Clonorchis sinensis were causally associated with cholangiocarcinoma in extensive areas of the Far East.

Among the parasitic protists, the association of some Apicomplexan and Flagellate species with neoplastic changes in the host tissues was suspected. However, the induction of a host cell transformation was shown experimentally only in the Apicomplexan Cryptosporidiumand Theileria. Actually, it was demonstrated recently that the Apicomplexan Cryptosporidium parvum can generate invasive cancer in gastrointestinal and biliary epithelia of SCID mice (Certad et al., 2007, 2010a, 2010b). Regarding Theileria, two species T. parva and T. annulata of cattle were shown to be able of inducing a reversible, parasite-dependent transformation of leukocytes (Dobbelaere & Rottenberg, 2003). Interestingly, many intracellular protists (Leishmania spp., Trypanosoma cruziC. parvumToxoplasma gondiiPlasmodium spp., Theileria spp.) are known to induce apoptosis inhibition (Carmen & Sinai, 2007), an effect that could be a significant step in the progression to malignancy (Lowe & Lin, 2000).

However, it has been usually difficult to identify pathogens as causative agents of cancers. The usually long latency between primary infection and cancer development is likely one of the main reasons for this remarkable difficulty (zur Hausen, 2009). For instance, the incidence of bilharzian urinary bladder cancer in various African countries peaks between the ages of 40-49 years, while infection with S. haematobium begins in childhood (as early as six months of age), and peaked usually in the second decade of life (between the ages of 5-15 years). This data suggest that bladder cancer implies a latency period of 20-30 years to develop from S. haematobium infection (IARC, 2011). Sometimes, the geographic coincidence of a specific infection with a defined type of cancer led to reveal a potential causal association. However, in the case of opportunistic pathogens (e.g. Cryptosporidium or Pneumocystis), it could still be difficult to establish if the pathogen had a causal or a consequential role, even when a specific infection-cancer association was well documented.

At present, infectious agents can be linked to pathology without meeting Koch’s postulates, Hill’s epidemiologic criteria, or even the revised criteria of Hill and Evans. Reproducible epidemiologic and laboratory data are often enough to unambiguously establish that certain infectious agents lead to some chronic outcomes, including cancer. In addition, animal models can often illustrate the plausibility of pathogenesis to humans. Also, clinical trials and surveillance can further demonstrate that preventing or treating the culprit infection avoids or eliminates the effect supporting causality (O’Connor, 2006).

In the present work, we will shortly review available data on the involvement of parasites in neoplastic processes in humans or animals, and we will focus on the carcinogenic power of C. parvum infection (Certad et al., 2007Certad et al., 2010aCertad et al., 2010b). Indeed, we don’t know if this intracellular protist is able to induce gastro-intestinal or biliary cancer in humans. However, an epidemiological study in Poland reported a high frequency (18 %) of cryptosporidiosis in patients with colorectal cancer (Sulzyc-Bielicka et al., 2007). In short, our experimental observations in the SCID mouse model, the ability of C. parvum to inhibit apoptosis in the host cell, and some reports that suggest an association of cryptosporidiosis with cancer in humans, largely justify clinical research aiming at exploring the causal involvement of C. parvum in colorectal cancer (CRC) or other digestive cancers in immunocompromised humans.

On the whole, infection seems to play a crucial role in the etiology of cancer. Actually, it was estimated that there would be 26.3 % fewer cancers in developing countries (1.5 million cases per year) and 7.7 % in developed countries (390,000 cases) if cancers associated with infectious diseases were prevented (Parkin, 2006).

Parasite Protozoa and Cancer

Based on clinical and epidemiological evidences, many reports underlined a potential association between parasitic protozoan infections and cancer. Thus, the flagellate Trichomonas vaginalis was suspected to be associated with cervical (Zhang et al., 1995) and prostate cancers (Stark et al., 2009), while the Apicomplexan Toxoplasma gondii was suggested to be associated with ocular tumor, meningioma, leukemia and lymphomas (Khurana et al., 2005). It was also suggested that Plasmodium could play a co-factor role in the development of Burkitt lymphoma (Khurana et al., 2005). However, only Theileria spp. (Dobbelaere et al., 2000Dobbelaere & Rottenberg, 2003Lizundia et al., 2006Branco et al., 2010) and C. parvum (Certad et al., 2007Certad et al., 2010aCertad et al., 2010b) were clearly shown to be able of inducing a hostcell transformation associated with tumorous disease.

Trichomonas Vaginalis (Excavata: Parabasalia)

T. vaginalis is a pathogenic protozoan, sexually transmitted, which resides in the lower female genitourinary tract. T. vaginalis coexist frequently with other local infections like pneumocystosis (Duboucher et al., 2003, 2007), candidiasis, bacterial infections, HPV virus infection, the last promoting cervical tumor growth (Boyle et al., 1989). Most serological, cytological and histopathological studies have suggested an association between T. vaginalisand the risk of cervical neoplasm (Yap et al., 1995Sayed el-Ahl et al., 2002Zhang et al., 1995). But until now, cause and effect relationship has not been proven, and other studies have provided contradictory results that suggest no association between Trichomonasinfection and cancer (Chakrabarti et al., 1992).

For the last five years, trichomonosis has also been suggested as positively associated with prostate cancer in men, on the basis of the association T. vaginalis – seropositive status and prostate cancer risk (Sutcliffe et al., 2006Stark et al., 2009). This infection seemed to be associated with cancer clinical frames that progress to bony metastasis and prostate cancer death (Stark et al., 2009). The same team developed then another large study (Sutcliffe et al., 2009) in order to test the reproducibility of precedent observations and the consistency of the hypothesis of a trichomonosis-prostate cancer association. However, this second work failed to show an association between T. vaginalis serostatus and prostate cancer. Additional work is therefore warranted (Sutcliffe et al., 2009).

Theileria (Alveolata: Apicomplexa)

Intracellular parasites from Theileria genus are particularly pathogenic in cattle and lead to a lymphoproliferative disease which is often lethal. T. parva and T. annulata infections reversibly lead to the transformation of the leukocyte infected cells, which can be reversed using drug that specifically eliminate parasites (Dobbelaere & Rottenberg, 2003). In vivo, the host cell transformation is associated with uncontrolled proliferation allowing clonal expansion. T. parva infected cells can also get a metastatic phenotype leading to invasion of other host organs (Dobbelaere & Rottenberg, 2003Lüder et al., 2009).

Leukocyte metabolic pathway and molecular alterations have been investigated during Theileria infection and it has been established that multiple host-cell pathways are altered (Table I). Firstly, the anti-apoptosis signaling pathway is stimulated by the activation of the transcription factor NF-κB (Heussler et al., 2002). Secondly, the cytokine GM-CSF (= granulocyte macrophage colony stimulating factor) secretion is enhanced and itself re-stimulate infected host cell proliferation via autocrine loops. Thirdly, GM-CSF contributes to the induction of the factor c-Myc, leading to lymphocyte proliferation (Baumgartner et al., 2000Dessauge et al., 2005). Endly, Theileria dependent transformation leads to the constitutive activation of c-jun kinase (JNK) and permanent induction of activator protein 1 (AP-1) (Lizundia et al., 2006).

Table I.

Molecular factors or mechanisms linking parasite-related inflammation with cell transformation or cancer.

Theileria intracellular microorganisms reside freely in the host leukocyte in direct contact with host-cell cytoplasmic structures, and modify host-cell cytoskeleton. Parasites interact likely with host-cell microtubules during mitosis to ensure its own persistence and survival (von Schubert et al., 2010). Furthermore, Theileria alters host-cell actin dynamics, increases motility and enables infected host-cell to behave as leukocyte metastasis (Baumgartner, 2011ab).

Plasmodium Species and Toxoplasma Gondii (Alveolata: Apicomplexa)

Malaria has been suggested as a co-factor for the development of endemic Burkitt Lymphoma (eBL) that is one of the three clinical variants of this non-Hodgkin lymphoma. It is a B-cell tumorous disease characterized by a chromosomal translocation that results in deregulation of the c-Myc oncogene (Orem et al., 2007). The hypothesis of a potential role of malaria as co-factor for eBL has been supported by several epidemiological studies showing that eBL is more frequent in areas where malaria is endemic. Consistently, a recent case-control study showed a strong significant association between eBL and malaria antibodies (Carpenter et al., 2008). According to the same case-control study, malaria and Epstein Barr Virus (EBV) could act synergistically in the development of eBL. Indeed, malaria, like HIV-infection, induces polyclonal B-cell activation and hypergammaglobulinemia possibly through the stimulus by a superantigen (Bornkamm, 2009). Actually, the cysteine-rich interdomain region 1alpha (CIDR1alpha) of P. falciparum erythrocyte membrane protein 1 (PfEMP1), expressed in red blood cells, was shown to act as both a polyclonal B cell activator and an inducer of EBV lytic cycle (Chêne et al., 2007Bornkamm, 2009). However, more work is needed to clarify the etiology of eBL as even the molecular contribution of EBV to the pathogenesis of BL remains unclear.

The association of Toxoplasma infection and adenomas has been suspected in two case reports of pituitary adenoma (Zhang et al., 2002). Adenomas were found associated with T. gondii cysts among the tumor cells in patients without immunesuppression status. Potential relations between T. gondii and tumors have also been reported in ocular tumors, meningioma, leukaemia and lymphoma (Khurana et al., 2005). The role of T. gondii remains to be explored. Interestingly, T. gondii was shown to be able to induce increased host-cell (dendritic cells and macrophages) motility, an effect that could favor both in vivo spread of parasites and progression of tumorous disease (see the recent review of Baumgartner, 2011b). Paradoxically, T. gondii could inhibit tumor growth in the Lewis lung carcinoma mouse model through the induction of Th1 immune responses and antiangiogenic activity (Kim et al., 2007).

The Special Case of Cryptosporidium (Alveolata: Apicomplexa)

Cryptosporidiosis represents a major public health problem. This infection, caused by an Apicomplexan Protozoa of the genus Cryptosporidium, has been reported worldwide as a frequent cause of diarrhea, and its prevalence varies according to different regions (Xiao, 2010). In developed countries, massive Cryptosporidium foodborne and waterborne outbreaks are quite frequently reported. In developing countries, besides outbreaks, sporadic cases of cryptosporidiosis are frequent, this infection affecting mostly children under five (Xiao et al., 2004). Furthermore, cryptosporidiosis remains a clinically significant opportunistic infection in immunocompromised patients, causing potentially life-threatening diarrhea, especially in those HIV-infected subjects without access to highly active antiretroviral therapy (HAART) (Pozio & Morales, 2005). Additionally, Cryptosporidiumspecies not only infect humans, but also cause morbidity in farm animals, leading to economic losses (Sunnotel et al., 2006).

Cryptosporidium organisms reside in an intracellular parasitophorous vacuole placed at the apical site of gastrointestinal epithelial cells (Plattner & Soldati- Favre, 2008). This unusual location (Fig. 1) was currently qualified as “extracytoplasmic” (Chalmers & Davis, 2010) though others qualified it as “epicellular” (Valigurova et al., 2008). Infection of the intestinal cells can result in blunting of the intestinal villi, crypt hyperplasia and inflammation. Most Cryptosporidium species infect the epithelium of the gut but in severe infections, dissemination can occur to extra-intestinal sites (Lopez-Velez et al., 1995).

Fig. 1.

Ileocaecal adenocarcinoma induced by Cryptosporidium parvum in dexamethasone-treated SCID mice.

There were some speculations about possible associations between Cryptosporidiuminfection and neoplasia in humans. The association of cryptosporidiosis and colonic adenocarcinoma was suspected in the case of a Spanish patient carrying both, who died rapidly after the onset of symptoms (Izquierdo et al., 1988). A cryptosporidiosis case of the biliary tract clinically mimicking a pancreatic cancer in an AIDS patient was also reported (de Souza Ldo et al., 2004). Furthermore, an association between X linked immunodeficiency with hyper IgM syndrome (XHIM), and carcinoma of the liver, pancreas and biliary tree was described in children by Hayward et al. (1997). These authors proposed that the mutation responsible for this defect may favor the colonization of the biliary epithelium by different pathogens, including Cryptosporidium. A chronic infection and inflammation by this parasite will follow, perhaps being the inflammation the cause of the malignant transformation (Hayward et al., 1997).

More recently, an epidemiological study in Poland reported 18 % of cryptosporidiosis in 55 patients with colorectal cancer (Sulzyc-Bielicka et al., 2007) being this prevalence higher compared with the burden of cryptosporidiosis for the general European population (Semenza & Nichols, 2007). However, an unexpected high prevalence of Cryptosporidiuminfection was recently reported in hospitalized patients in Poland (Bajer et al., 2008). Actually, nested PCR revealed a global prevalence of 34 % in 35 patients from three Poland hospitals. In this study, prevalence was 43 % in patients apparently immunocompetent (6/14 patients), 29 % in patients with primary immunodeficiency (5/17 patients) and 25 % in patients with secondary immunodeficiency (1/4 patients) (Bajer et al., 2008).

Furthermore, previous works had associated cryptosporidiosis with the development of benign tumors in naturally or experimentally infected vertebrates. Thus, Cryptosporidiumorganisms were detected in aural-pharyngeal polyps in naturally infected iguanas by using optical or transmission electron microscopy (Fitzgerald et al., 1998Uhl et al., 2001). Another study reported the presence of Cryptosporidium organisms in intestinal polyps of naturally infected sheep (Gregory et al., 1987). C. parvum organisms were found to be associated with cystic hyperplasia of the colonic mucosa in experimentally infected nude mice (Heine et al., 1984). None of these studies described the presence of malignant or even pre-malignant lesions.

Nevertheless, the occurrence of dysplastic changes classified as low grade intraepithelial neoplasia of bile ducts was previously reported in a model of IFN-γ knockout mice infected with C. parvum (Stephens et al., 1999). The authors of this study suggested that the response of these mice to Cryptosporidium infection may model the initial steps towards the development of cholangitis and bile duct-related cancer often observed in patients with immunodeficiency (Stephens et al., 1999). These findings were consistent with the observations of Hayward et al. (mentioned above), in relation to XHIM (Hayward et al., 1997).

More recently, the ability of C. parvum to induce gastrointestinal neoplastic changes was established experimentally in severe combined immunodeficiency (SCID) mice (Certad et al., 2007Certad et al., 2010aCertad et al., 2010b). Herein, adenomas with low or high-grade intraepithelial, intramucosal or invasive neoplasia associated with numerous C. parvumlife stages were detected in different areas of the digestive tract (Fig. 1) including stomach, duodenum and ileocaecal region. The first time, such lesions were observed unexpectedly after 46 days post-infection (Certad et al., 2007) in C. parvum (Iowa strain) chronically infected SCID mice submitted to oral dexamethasone (Dex) administration. These findings were recorded using inoculum between 105 and 108 oocysts. The use of Ki-67 supported the neoplastic nature of the described Cryptosporidium-induced epithelial transformation (Certad et al., 2010a), and showed that potential neoplastic alterations begin before the detecting of histopathological lesions by using standard stains.

A highly significant correlation was found between the extension of cryptosporidiosis and the severity of neoplastic lesions in either Dex-treated or untreated SCID mice (Certad et al., 2010a). The ability of other C. parvum strains to induce digestive neoplasia in Dextreated SCID mice was also explored. Thus, SCID mice infected with C. parvum TUM1 strain developed a fulminant cryptosporidiosis associated with intramucosal adenocarcinoma, which is considered an early histological sign of invasive cancer (Certad et al., 2010b). Thus, the ability of C. parvum to induce neoplasia was not restricted to a specific C. parvum strain. In contrast, the species C. muris revealed unable to induce neoplastic changes in the SCID mouse model (Certad et al., 2007). Reasons of this different expression of the disease according to Cryptosporidium species are unclear, though variability in pathogenicity according to Cryptosporidium species, strains, and likely to other factors, were already reported (Okhuysen & Chappell, 2002).

No data about the mechanism of C. parvum-induced neoplasia are available. Several observations in our study suggest that combination of C. parvum with Dex administration is involved in the generation of these significant histological changes in digestive epithelia of SCID mice. This steroid potentially alters innate immunity (Franchimont, 2004Stojadinovic et al., 2007) in C. parvum infected animals. Consistently, a higher C. parvum oocyst shedding was found in Dex-treated SCID mice presenting neoplastic lesions (Certad et al., 2010a). However, even lesions with low grade dysplasia induced by C. parvum should be considered as a putative precursor to digestive carcinoma. Actually, colorectal tumorigenesis is believed to involve sequential genetic changes leading to both initiation of the neoplastic process and progression from normal epithelium to carcinoma (Luperchio & Schauer, 2001). Taken together, higher rates of oocyst shedding seemed to be associated with an earlier onset and more rapid evolution of neoplastic lesions (Certad et al., 2010a). SCID mice infected with lower inoculum could reach similar rates of both oocyst shedding and severity of lesions, but they may require a longer period (Benamrouz et al., 2011, unpublished; Certad et al., 2010b).

Recent studies have reported about the capacity of some protozoa to interfere with signaling pathways of the host cell (Heussler et al., 2001Carmen & Sinai, 2007) (Table I). It is well known that T. parva, another Apicomplexan protist, is responsible for a lymphoproliferative disorder of cattle (Dobbelaere et al., 2000) (see above). This organism infects and transforms bovine lymphocytes resulting in tumors with metastatic/invasive potential by different mechanisms: (a) inhibition of apoptosis that induces host cell survival; (b) induction of proliferation of the host cell; (c) induction of leukocyte survival by apoptosis inhibiting (Lüder et al., 2009).

Interestingly, inhibition of apoptosis (Table I) was also reported in other Apicomplexan Protozoa including Cryptosporidium (Heussler et al., 2001Carmen & Sinai, 2007). Using an in vitro model of biliary cryptosporidiosis, C. parvum was shown to be able of activating NF-κB pathway in directly infected cells, preventing the induction of cell death after infection (Chen et al., 2005). NF-κB family of transcription factors regulates the activation of a number of intracellular survival signals including the c-Myc protooncogene (Chen et al., 2001). Indeed, activation of NF-κB has been observed in many cancers, including colon cancer (Naugler & Karin, 2008). The delay in the activation of NF-κB suggests that C. parvum-induced NF-κB activation is not directly associated to the attachment and invasion of the cell by the parasite (Chen et al., 2001). However, it has been found in in vitro studies that C. parvum, depending on its developmental stage, not only inhibits but it can modulate host-cell apoptosis, inhibiting apoptosis at the trophozoite stage and promoting this process at the sporozoite and merozoite stages (Mele et al., 2004). Modulation of apoptotic pathways was also investigated by microarray analysis in an in vitro model using human ileocaecal HCT8 cells. Genome wide expression profiling revealed high proportion of apoptosis genes regulated during C. parvum infection (Liu et al., 2009). Analysis of the apoptosis gene transcript profile suggests a biphasic control of host cell apoptosis. BCL2 and c-Myc genes showed an altered expression during C. parvum infection in HCT8 cells (Liu et al., 2009). Apoptosis prevention probably benefits the parasite by stabilizing the host cell long enough to permit the completion of the life cycle (Heussler et al., 2001). Indeed, resistance to apoptosis could be an essential step in the progression to malignancy (Lowe & Lin, 2000).

Interestingly, it is well known that the parasite induces modifications of the host actin cytoskeleton of intestinal epithelial cells (Table I) though little information is available about the significance of the host actin remodeling process (Plattner & Soldati-Favre, 2008). It has been reported, however, that intercellular and cell matrix adhesion molecules are involved in regulation of cell polarity, differentiation, proliferation and migration. Particularly, in colorectal cancer, cells lose actin cytoskeletal organization and normal cell adhesion when they become invasive (Buda & Pignatelli, 2004). Consistently, alterations in host cell cytoskeleton have been described as a consequence of Theileria infection (Lüder et al., 2009) (see above). Therefore, persistent infection with C. parvum could be a risk for gastrointestinal neoplasia. Unfortunately, understanding the biology of this parasite has been hampered by the lack of transfection techniques and to the absence of reliable in vitromodels.

Research on the topic could be worthwhile since the incidence of Cryptosporidium infection seems to increase not only amongst immunosuppressed patients but also in the general population worldwide. In the United States a dramatic increase in the incidence of cryptosporidiosis in recent years was reported, mainly due to a substantial rise in community outbreaks (Yoder & Beach, 2010). Moreover, low infectious threshold and the chlorine resistance of oocysts make Cryptosporidium ideally suited for transmission through drinking and recreational water, food, person to person and animal to person contact, facilitating the expansion of the infection in the community (Yoder & Beach, 2010).

The Worms That Induce Cancer in Humans

The causal involvement of parasitic worms in human cancer was firstly suspected on the basis of epidemiological data, especially on the geographic coincidence of a specific infection with a particular type of cancer. At present, IARC (2011) classified S. haematobiumC. sinensis and O. viverrini in group 1 (infectious agents definitely “carcinogenic to humans”), while Schistosoma japonicum was classed in the group 2B (agents “probably carcinogenic to humans”) and Schistosoma mansoni and Opisthorchis felineus were assigned to the group 3 (agents “not classifiable as to its carcinogenicity to humans”).

Schistosoma Haematobium (Digenea: Schistosomatidae)

S. haematobium is usually recognized as a definite cause of urinary bladder cancer (Creusy et al., 2010). The relative risk for bladder cancer in S. haematobium infected patients has been estimated between 1.8 and 23.5 (Parkin, 2006). About 3 % of urinary bladder cancers around the world are attributable to S. haematobium (de Martel & Franceschi 2009).

Digenea trematodes of Schistosoma genus belong to Schistosomatidae, the sole trematode family whose members are gonochoric worms (with separated sexes). S. haematobium adult worms dwell inside blood vessels of the perivesical venous plexus. As in other schistosomes, the body of S. haematobium male worm forms a groove or gynaecophoric channel, in which it permanently holds the thinner and longer female. The S. haematobium female produces hundreds of eggs per day throughout her life (usually three-five years). Successive reinfections are usual in endemic areas, where the prevalence of urinary schistosomiasis, especially in children, can be very high. The eggs (145 μm × 60 μm) with a typical terminal spine pass through the walls of the blood vessels, and through the ureteral or bladder wall to be excreted with urine. However, about half of the released eggs do not reach the bladder lumen and are carried back into the liver by the bloodstream and/or trapped in the bladder wall tissues. Retained eggs provoke a granulomatous inflammatory response that enhances local tissue ulceration, being the main cause of disease in the human host. Additionally, eggs provoke pseudopolyposis of the bladder and ureteral walls; chronic lesions can then evolve into fibrosis and carcinoma (de Martel & Franceschi, 2009).

Frequently associated with inflammatory response provoked by the eggs there is conversion of the transitional epithelium to a squamous metaplastic epithelium, which has a much greater proliferative potential (Abol-Enein, 2008). As a consequence, most people with chronic schistosomiasis and bladder cancer develop squamous cell carcinoma (SCC) rather than transitional cell carcinoma (TCC) (Cohen et al., 1992). Consistently, high prevalence of SCC of bladder was observed in geographic areas where S. haematobium is endemic (e.g. Egypt, Iraq, Zambia, Malawi, Kuwait) (Mostafa et al., 1999). The carcinogenic role of S. haematobium was also supported by several case-control studies and by experimental studies in animals (sees for review Vennervald & Polman, 2009).

Regarding mechanisms, several studies indicate that the carcinogenic power of S. haematobium is a multifactorial and multistage process (IARC, 2011) where several processes are involved. Many reviews were devoted to the topic for the last years (Rosin et al., 1994aRosin et al., 1994bMostafa et al., 1999Herrera & Ostrosky- Wegman, 2001Mayer & Fried, 2007Vennervald & Polman, 2009). Main data may be summarized as follows (see also Table I):

  1. Local inflammation, regenerative process and increased cell proliferation (Cohen et al., 1990, 1991) over long periods could provide more opportunities for the expression of spontaneous DNA alterations (Abol-Enein, 2008). Consistently, S. haematobiuminfection was found to be strongly associated with an increased risk of metaplasia or hyperkeratosis. Actually, in endemic areas, prevalence of inflammation (39 %), hyperkeratosis (30 %), metaplasia (33 %) and frank atypia (0.4 %) were much higher than in nonendemic ones (IARC, 2011).
  2. Schistosomiasis-induced inflammatory cells have been shown to participate in the metabolic activation of procarcinogens, such as aflatoxins, and in the formation of carcinogenic nitrosamines from nitric oxide (Rosin et al., 1994aMostafa et al., 1994). In addition, the often associated chronic bacteriuria produces also nitrosamines, including N-butyl-N-(4-hydroxybutyl) nitrosamine, a known bladder carcinogen in rodents and dogs (Hicks et al., 1982El-Bolkainy, 1983). What’s more, urinary stasis due to S. haematobium infection further contribute to locally concentrate nitrosamines and other endogenous carcinogens, favoring their absorption from urine to the bladder epithelium (Bhagwandeen, 1976). Nitrosamine was apparently an important factor to the development of neoplastic changes in baboons experimentally infected with S. haematobium, though alone, nitrosamine was unable to induce neoplasia (Hicks et al., 1980; IARC, 2011).
  3. The inflammatory response around the eggs generates genotoxic factors and products that may cause genomic instability as well as stimulate a proliferative response of the host cells to repair tissue damage caused by the inflammation. Reactive oxygen species (ROS) have a role in the etiology of cancer, and inflammatory cells, such as eosinophils, which are an important endogenous source of oxygen radicals, are found in high numbers in urine from people infected with S. haematobium (many sources, see Vennervald & Polman, 2009, for review). Consistently, levels of 8-hydroxy-2’-deoxyguanosine (8-OHdG), a critical biomarker of oxidative stress and carcinogenesis, were markedly increased in urinary bladder cell carcinomas associated with schistosomiasis when compared to non-schistosomiasis-associated cancers. DNA-repair genes 8-oxyguanine-DNA-glycosylase and apurinic/ apyrimidinic endonuclease were strongly overexpressed.
  4. Also, increased formation of DNA single strand breaks, due to oxidative damage, and higher inducible nitric oxide synthase was found in bladder SCC-associated with schistosomiasis when comparing with non-schistosomiasis-associated cancers (Salim et al., 2008).
  5. Genetic instability of host cells may lead to modifications in the regulation of tumor-suppressor genes and oncogenes interfering with the control of cell proliferation (Vennervald & Polman, 2009). Thus, mutations in the tumour-suppressor gene p53 have been observed more frequently in patients with schistosomiasis- associated bladder cancer than in patients with bladder cancer not related to schistosomiasis (Mostafa et al., 1999). Elevated levels of expression of oncoproteins, such as Bcl-2 (Chaudhary et al., 1997Swellam et al., 2004) and p53 (Chaudhary et al., 1997) have also been observed in schistosomiasis-associated bladder cancer (Vennervald & Polman, 2009).
  6. DNA alkylation damage indicated by increased levels of O6-methyldeoxyguanosine (O6-Me-dG) has been demonstrated in tissue samples from schistosomiasis- associated bladder cancer patients. Consistently, O6-alkylguanine-DNA alkyltransferase (ATase) activity was significantly higher in normal bladders than in bladders with cancers and negatively correlated with the levels of O6-Me-dG (Badawi et al., 1992, 1994).
  7. Schistosomiasis-associated urinary bladder cancer samples had more genes methylated than non-schistosomal bladder cancer samples in a study in Egyptian patients (Gutiérrez et al., 2004).
  8. Although biomarkers may not indicate overt cancer (Vennervald & Polman, 2009), the results from a study in Ghana showed a close correlation between BLCA-4 (a nuclear matrix protein involved in gene regulation and produced only in neoplastic cells), quantitative nuclear grading (QNG) and severe urinary bladder damage such as bladder wall masses and polyps. The correlation was the strongest in individuals > 30 years of age, for whom infection levels were low. Furthermore, a total of 62 out of 73 cytopathology Papanicolaoustained smears showed squamous metaplasia (Eissa et al., 2007).

Regarding mutagenesis, in vitro studies did not succeed to show a mutagenic power of neither worm nor egg extracts (Osada et al., 2005). On the whole, nitric oxide produced by the inflammatory response provoked by S. haematobium eggs, and alkylation of DNA by N-nitroso compounds did emerge as important causal factors of the DNA severe alterations related with S. haematobium-associated urinary bladder cancer (IARC, 2011).

Schistosoma Mansoni and S. Japonicum (Digenea: Schistosomatidae)

Although there are not a clear association between cancer and S. mansoni infection geographical areas, S. mansoni parasitism was linked with liver, colorectal cancer and prostate cancer (Vennervald & Polman, 2009). Moreover, Brazilian pathologists have found giant follicular lymphomas in 1577 spleens removed from schistosomiasis mansoni patients with severe portal hypertension (Andrade et al., 1971de Andrade et al., 1982). The explanation of such observations remains unclear. Patients with schistosomiasis mansoni showed decreased hepatic levels of cytochrome p450, cytochrome b-5 and NADPH cytochrome C reductase. Such alterations could cause metabolites of aflatoxin to increase significantly, suggesting that S. mansoni could potentiate the effect of carcinogens (Habib et al., 2006Vennervald & Polman, 2009).

The association between S. mansoni infection and hepato-cellular carcinoma (HCC) could also be indirect. Patients with S. mansoni have higher incidence of HBV or HCV infection (two viruses associated with cancerogenesis) than noninfected controls (Halim et al., 1999). The high exposure of schistosomiasis patients to HBV and HCV could be partly explained by transmission of these viruses during blood transfusion (via contaminated blood, syringes and needles) consecutive to hematemesis, which is a relatively frequent complication of hepatosplenic schistosomiasis, (Darwish et al., 1993).

Regarding S. japonicum, its role in cancer occurrence is less clear, although this parasite has been associated with both liver and colorectal cancer. Some epidemiological and clinical studies in China and Japan support its role as one of the risk factors in HCC formation (Khurana et al., 2005). Several epidemiological surveys have indicated a geographical association between S. japonicum infection and cancer of the liver and colon, but many of these studies did not consider other factors such as viral hepatitis, aflatoxin and other dietary carcinogens. On the whole, though an association between colon cancer and S. japonicumhas been suggested as early as in the 60’, there has been no consistent evidence of S. japonicum as a carcinogen in humans.

Opisthorchis Viverrini and Clonorchis Sinensis (Digenea: Opisthorchiidae)

C. sinensisO. viverrini and O. felineus are closely related fish-borne zoonotic Digenetic trematodes, which infect humans and various fish-eating animals. C. sinensis and O. viverriniremain an important public health problem in many endemic areas in South-East Asia where they infect more than 20 million of persons (Khurana et al., 2005). Indeed about 15 million people are infected with C. sinensis (rural areas of the Republic of Korea, Democratic People’s Republic of Korea, and China) and eight million with O. viverrini (Southeast Asian countries such as Thailand, Laos, Viet Nam and Cambodia) (Vennervald & Polman, 2009). Opisthorchiidoses due to infections with these two species were also reported in immigrants living in developed countries for many years (Attwood & Chou, 1978Schmidt et al., 1982Vernes et al., 1982). Actually, these fishborne flukes can persist for longtime in the host liver: life expectance of O. viverrini and C. sinensis are about ten years and 26 years, respectively (Attwood & Chou, 1978Sithithaworn & Haswell-Elkins, 2003).

In addition to their association with hepatobiliary disease, these liver flukes are considered as major etiological agents of cholangiocarcinoma (CCA) (Parkin, 2006). After evaluating epidemiological studies, case series, and case control studies, O. viverrini and C. sinensiswere considered definitely carcinogenic by the IARC (2011). Thus, nowadays, hepatobiliary opisthorchiasis is the strongest link which could be occurred between a human malignancy and a Metazoan infectious agent (Laha et al., 2007).

The clinical manifestations, imaging findings and pathophysiology for O. viverrini and C. sinensis infection are almost the same (Lun et al., 2005Sripa et al., 2009Sripa & Pairojkul, 2008). Vennervald & Polman (2009) have reviewed recently these aspects. The adult liver flukes are found in the biliary system, especially in the medium-sized or small intrahepatic bile ducts causing mechanical obstruction, inflammation, adenomatous hyperplasia and periductal fibrosis. Thus, in contrast with Fasciola hepatica, the Opisthorchiidae worms do not invade the hepatic parenchyma and hence, most of the disease manifestations result from the mechanical irritation and associated pathological changes caused by the worms to the biliary epithelium. Extensive and chronic infections can cause severe hepatobiliary disease manifestations such as cholangitis, cholecystitis, cholelithiasis, obstruction jaundice, hepatomegaly and fibrosis of the periportal tract (Vennervald & Polman, 2009). Worldwide, CCA (derived from the epithelial cells or cholangiocytes lining the bile ducts) is much less common than hepatocellular carcinoma (HCC, derived from hepatocytes), except for those areas in East Asia, where infection with O. viverrini or C. sinensis is widespread and highly prevalent in humans (Srivatanakul et al., 2004Porta et al., 2011).

The mechanisms of carcinogenesis due to liver fluke infection remain unclear. N-nitroso-compounds are generated locally from NO produced by inflammatory cells around the bile ducts. NO may lead to DNA damage, particularly in proliferating bile duct cells (Sripa et al., 2007). Furthermore, recent evidence suggests that O. viverrini secretes mitogenic proteins in host tissues where they might promote cell proliferation, mutagenesis and finally carcinogenesis (Laha et al., 2007Sripa et al., 2007). Moreover, the flukes induced in vitroover-expression of mRNAs encoding growth-promoting proteins including transforming growth factor (Thuwajit et al., 2006). Regarding C. sinensis, this worm releases products that induce cell proliferation, associated with up-regulation of cyclin B and transcription factor E2F1 (Kim et al., 2008). Despite the direct effect of liver fluke molecules on cell proliferation, liver fluke-induced CCA results likely from a slow indirect process closely linked to chronic inflammation that involves the activation of oxidative stress pathways (Sripa et al., 2007).

Conclusion

Some causal associations between parasites and cancers have been established already in humans and in experimental animals. More research in the field is warranted to substantiate additional links. In addition to clinical and epidemiological data, molecular factors or mechanisms linking parasite infection and cancer development need still to be investigated, especially, host chronic inflammatory response, which is a widely recognized basic mechanism for the development of most of infection-related tumors.

Developing strategies designed to prevent or appropriately treat cancer-associated parasites arise as a major issue of Public Health, especially among the bottom billion living in the poorest areas of the developing world. In these areas, where parasitic infections are highly prevalent, the underlying causes of parasiteassociated cancers can be frequently neglected. Avoiding exposure, reducing parasite transmission, and treating infection early could implement this prevention potential, reducing the global impact of some cancers, mainly but not only in developing countries. These strategies must, however, be built on sound scientific evidence.

On the other hand, in many cases, parasitic agents are widely present in humans, but only a small fraction of infected individuals develop cancer. Thus, it is also important to identify subpopulations of exposed individuals who are likely to develop cancer by developing sensitive and specific molecular markers in order to be able to implement preventive strategies. Together, parasitic determinants of cancer offer a spectrum of research and prevention possibilities that could substantially affect global health by reducing some cancers worldwide.

Acknowledgments

Research on the topic is being developed in the framework of an Agreement between Institut Pasteur de Lille and Université Catholique de Lille (UCL). French Ministry of Research (EA4547 Université de Lille 2) and the French National Research Agency (Projects No. ANR-09-ALIA-009 “ProtoFood” and ANR-10-ALIA-004 “Fish-Parasites”) provided financial support. In addition, specific support was provided by Catholic University of Lille (UCL) including a grant for S.B. G.C. was supported by a scholarship from the Consejo de Desarrollo Científico y Humanístico of the Universidad Central de Venezuela. Special thanks to Prof. P. Gosset (GHICL) for advice and help, Dr E. Baumelou and Dr C. Iliescu (Direction of Research, GHICL-FLM) for encouragement and scientific support, and to Dr L. Delhaes (Centre of Biology, Pathology and Cytogenetics, CHRU Lille, France) for critical reading of the manuscript.

References

  • Abol-Enein H.Infection: is it a cause of bladder cancer? Scand. J. Urol. Nephrol., 2008, 218, S79–S84 [PubMed]
  • Andrade Z.A. & Abreu W.N.Follicular lymphoma of the spleen in patients with hepatosplenic schistosomiasis mansoniAm. J. Trop. Med. Hyg., 1971, 20, 237–243[PubMed]
  • Attwood H.D. & Chou S.T.The longevity of Clonorchis sinensisPathology, 1978, 10, 153–156 [PubMed]
  • Badawi A.F., Mostafa M.H., Aboul-Azm T, Haboubi N.Y., O’Connor P.J. & Cooper D.P.Promutagenic methylation damage in bladder DNA from patients with bladder cancer associated with schistosomiasis and from normal individualsCarcinogenesis, 1992, 13,877–881 [PubMed]
  • Badawi A.F., Cooper D.P., Mostafa M.H., Aboul-Azm T., Barnard R., Margison G.P. & O’Connor P.J.O6-alkylguanine-DNA alkyltransferase activity in schistosomiasis-associated human bladder cancerEur. J. Cancer, 1994, 30A, 1314–1319 [PubMed]
  • Bajer A., Bednarska M., Cacciò S.M., Wolska-Kuśnierz B., Heropolitanska-Pliszka E., Bernatowska E., Wielopolska M., Paziewska A., Welc-Faleciak R. & Siński E.Genotyping of Cryptosporidium isolates from human clinical cases in PolandParasitol. Res., 2008, 103, 37–42 [PubMed]
  • Barakat F.M., Mcdonald V., Foster G.R., Tovey M.G. & Korbel D.S.Cryptosporidium parvum infection rapidly induces a protective innate immune response involving type I interferonJ. Infect. Dis., 2009, 200, 1548–1555 [PubMed]
  • Baumgartner M., Chaussepied M., Moreau M.F., Werling D., Davis W.C., Garcia A. & Langsley G.Constitutive PI3-K activity is essential for proliferation, but not survival, of Theileria parva-transformed B cellsCell. Microbiol., 2000, 2, 329–339 [PubMed]
  • Baumgartner M.Enforcing host cell polarity: an apicomplexan parasite strategy towards disseminationCurr. Opin. Microbiol., 2011a, 14, 436–444 [PubMed]
  • Baumgartner M.Theileria annulata promotes Src kinase-dependent host cell polarization by manipulating actin dynamics in podosomes and lamellipodiaCell. Microbiol., 2011b, 13, 538–553 [PubMed]
  • Bhagwandeen S.B.Schistosomiasis and carcinoma of the bladder in ZambiaS. Afr. Med. J., 1976, 50, 1616–1620 [PubMed]
  • Bornkamm G.W.Epstein-Barr virus and the pathogenesis of Burkitt’s lymphoma: more questions than answersInt. J. Cancer, 2009, 124, 1745–1755 [PubMed]
  • Boyle C.A., Lowell D.M., Kelsey J.L., Livolsi V.A. & Boyle K.E.Cervical intraepithelial neoplasia among women with papillomavirus infection compared to women with Trichomonas infectionCancer, 1989, 64, 168–172 [PubMed]
  • Branco S., Orvalho J., Leitão A., Pereira I., Malta M., Mariano I., Carvalho T., Baptista R., Shiels B.R. & Peleteiro M.C.Fatal cases of Theileria annulata infection in calves in Portugal associated with neoplastic-like lymphoid cell proliferationJ. Vet. Sci., 2010, 11, 27–34 [PMC free article] [PubMed]
  • Buda A., Pignatelli M.Cytoskeletal network in colon cancer: from genes to clinical applicationInt. J. Biochem. Cell. Biol., 2004, 36, 759–765 [PubMed]
  • Carmen J.C. & Sinai A.P.Suicide prevention: disruption of apoptotic pathways by protozoan parasitesMol. Microbiol., 2007, 64, 904–916 [PubMed]
  • Carpenter L.M., Newton R., Casabonne D., Ziegler J., Mbulaiteye S., Mbidde E., Wabinga H., Jaffe H. & Beral V.Antibodies against malaria and Epstein-Barr virus in childhood Burkitt lymphoma: a case-control study in UgandaInt. J. Cancer, 2008, 122, 1319–1323 [PubMed]
  • Certad G., Ngouanesavanh T., Guyot K., Gantois N., Chassat T., Mouray A., Fleurisse L., Pinon A., Cailliez J.C., Deicas E. & Creusy C.Cryptosporidium parvum, a potential cause of colic adenocarcinomaInfect. Agent. Cancer,2007, 2, 22. [PMC free article][PubMed]
  • Certad G., Creusy C., Guyot K., Mouray A., Chassat T., Delaire B., Pinon A., Sitja-Bobadilla A., Alvarez-Pellitero P., Praet M., Cuvelier C. & Dei-Cas E.Fulminant cryptosporidiosis associated with digestive adenocarcinoma in SCID mice infected with Cryptosporidium parvum TUM1 strainInt. J. Parasitol., 2010a, 40, 1469–1475[PubMed]
  • Certad G., Creusy C., Ngouanesavanh T., Guyot K., Gantois N., Mouray A., Chassat T., Flament N., Fleurisse L., Pinon A., Delhaes L. & Dei-Cas E.Development of Cryptosporidium parvum induced gastro-intestinal neoplasia in SCID mice: severity of lesions is correlated with infection intensityAm. J. Trop. Med. Hyg., 2010b, 82, 257–265 [PMC free article] [PubMed]
  • Chakrabarti R.N., Dutta K., Sarkhel T. & Maity S.Cytologic evidence of the association of different infective lesions with dysplastic changes in the uterine cervixEur. J. Gynaecol. Oncol., 1992, 13, 398–402 [PubMed]
  • Chalmers R.M. & Davies A.P.Minireview: clinical cryptosporidiosisExp. Parasitol., 2010, 124, 138–146 [PubMed]
  • Chaudhary K.S., Lu Q.L., Abel P.D., Khandan-Nia N., Shoma A.M., Baz M., Stamp G.W. & Lalani E.N.Expression of bcl-2 and p53 oncoproteins in schistosomiasis-associated transitional and squamous cell carcinoma of urinary bladderBr. J. Urol., 1997, 245, 78–84 [PubMed]
  • Chen X.M., Levine S.A., Splinter P.L., Tietz P.S., Ganong A.L., Jobin C., Gores G.J., Paya C.V., Larusso N.F.Cryptosporidium parvum activates nuclear factor kappaB in biliary epithelia preventing epithelial cell apoptosisGastroenterology, 2001, 120, 1774–1783 [PubMed]
  • Chen X.M., O’Hara S.P., Nelson J.B., Splinter P.L., Small A.J., Tietz P.S., Limper A.H. & Larusso N.F.Multiple TLRs are expressed in human cholangiocytes and mediate host epithelial defense responses to Cryptosporidium parvum via activation of NF-kappaBJ. Immunol., 2005, 175, 7447–7456 [PubMed]
  • Chêne A., Donati D., Guerreiro-Cacais A.O., Levitsky V., Chen Q., Falk K.I., Orem J., Kironde F., Wahlgren M. & Bejarano M.T.A molecular link between malaria and Epstein-Barr virus reactivationPLoS Pathog., 2007, 3, e80. [PMC free article][PubMed]
  • Cohen S.M. & Johansson S.L.Epidemiology and etiology of bladder cancerUrol. Clin. N. Am., 1992, 19, 421–428 [PubMed]
  • Creusy C., Certad G., GuyoT K. & Dei-Cas E.Parasites and oncogenesis with a special reference to gastro-intestinal neoplasia induced by Cryptosporidium parvumInDetection of bacteria, viruses, parasites and fungi. Viola-Magni M., (ed.), NATO Series Science for Peace, Springer, Berlin, 2010, 381–388
  • Darwish M.A., Raouf T.A., Rushdy P., Constantine N.T., Rao M.R. & Edelman R.Risk factors associated with a high seroprevalence of hepatitis C virus infection in Egyptian blood donorsAm. J. Trop. Med. Hyg., 1993, 49, 440–447 [PubMed]
  • Andrade D.R., Ishioka S., Câmara-Lopes L.H. & Meira J.A.Association of hepatosplenic schistosomiasis mansoni and histiocytic lymphomaArq. Gastroenterol., 1982, 19, 77–80 [PubMed]
  • La Horra C., Varela J.M., Fernandez-Alonso J., Medrana F.J., Respandiza N., Montes-Cano M.A. & Calderone E.J.Association between human-Pneumocystis infection and small-cell lung carcinomaEur. J. Clin. Invest., 2004, 34229–235 [PubMed]
  • Martel C. & Franceschi S.Infections and cancer: established associations and new hypothesesCrit. Rev. Oncol. Hematol., 2009, 70, 183–194 [PubMed]
  • Souza Ldo R., Rodrigues M.A., Morceli J., Kemp R. & Mendes R.P.Cryptosporidiosis of the biliary tract mimicking pancreatic cancer in an AIDS patientRev. Soc. Bras. Med. Trop., 2004, 37, 182–185 [PubMed]
  • Deng M., Lancto C.A. & Abrahamsen M.S.Cryptosporidium parvum regulation of human epithelial cell gene expressionInt. J. Parasitol., 2004, 34, 73–82 [PubMed]
  • Dessauge F., Lizundia R. & Langsley G.Constitutively activated CK2 potentially plays a pivotal role in Theileria-induced lymphocyte transformationParasitology, 2005, 130, 37–44 [PubMed]
  • Dobbelaere D. & Heussler V.Transformation of leukocytes by Theileria parva and T. annulataAnnu. Rev. Microbiol., 1999, 53, 1–42 [PubMed]
  • Dobbelaere D.A., Fernandez P.C. & Heussler V.T.Theileria parva: taking control of host cell proliferation and survival mechanismsCell. Microbiol., 2000, 2, 91–99[PubMed]
  • Dobbelaere D.A. & Rottenberg S.Theileria-induced leukocyte transformationCurr. Opin. Microbiol., 2003, 6, 377–382 [PubMed]
  • Duboucher C., Noël C., Durand-Joly I., Gerbod D., Delgado-Viscogliosi P., Jouveshomme S., Leclerc C., Cartolano G.L., Dei-Cas E., Capron M. & Viscogliosi E.Pulmonary coinfection by Trichomonas vaginalis and Pneumocystis sp. as a novel manifestation of AIDSHum. Pathol., 2003, 34,508–511 [PubMed]
  • Duboucher C., Caby S., Chabé M., Gantois N., Delgado-Viscogliosi P., Pierce R., Capron M., Dei-Cas E. & Viscogliosi E.Human pulmonary trichomonosesPresse Med., 2007, 36, 835–839 [PubMed]
  • Eissa S., Swellam M., Ali-Labib R., Mansour A., El-Malt O. & Tash F.M.Detection of telomerase in urine by three methods: evaluation of diagnostic accuracy for bladder cancerJ. Urol., 2007, 178, 1068–1072 [PubMed]
  • El-Bolkainy M.N.Schistosomiasis and bladder cancerInThe pathology of bladder cancer. Bryan G.T. & Cohen S.H. (eds), CRC Press, Boca Raton, FL, 1983, 57–90
  • El-Sheikh S.S., Madaan S., Alhasso A., Abel P., Stamp G. & Lalani E.N.Cyclooxygenase-2: a possible target in Schistosoma-associated bladder cancerBJU Int., 2001, 88, 921–927 [PubMed]
  • Ferlay J., Shin H.R., Bray F., Forman D., Mathers C. & Parkin D.M.GLOBOCAN 2008 v1.2Cancer incidence and mortality worldwide: IARC CancerBase No. 10 (Internet). International Agency for Research on Cancer, Lyon, France, 2010. Available from: http://globocan.iarc.fr
  • Fitzgerald S.D., Moisan P.G. & Bennett R.Aural polyp associated with cryptosporidiosis in an iguana (Iguana iguana)J. Vet. Diagn. Invest., 1998, 10, 179–180 [PubMed]
  • Franchimont D.Overview of the actions of glucocorticoids on the immune response: a good model to characterize new pathways of immunosuppression for new treatment strategiesAnn. N. Y. Acad. Sci., 2004, 1024, 124–137 [PubMed]
  • Gregory M.W., Catchpole J., Pittilo R.M. & Norton C.C.Ovine coccidiosis: observations on “oocyst patches” and polyps innaturally-acquired infectionsInt. J. Parasitol., 1987, 17, 1113–1124 [PubMed]
  • Gutiérrez M.I., Siraj A.K., Khaled H., Koon N., El-Rifai W. & Bhatia K.CpG island methylation in Schistosoma– and non-Schistosoma-associated bladder cancerMod. Pathol., 2004, 17, 1268–1274 [PubMed]
  • Habib S.L., Said B., Awad A.T., Mostafa M.H. & Shank R.C.Novel adenine adducts, N7-guanine-AFB1 adducts, and p53 mutations in patients with schistosomiasis and aflatoxin exposureCancer Detect. Prev., 2006, 30, 491–498 [PubMed]
  • Halim A.B., Garry R.F., Dash S. & Gerber M.A.Effect of schistosomiasis and hepatitis on liver diseaseAm. J. Trop. Med. Hyg., 1999, 60, 915–920 [PubMed]
  • Hayward A.R., Levy J., Facchetti F., Notarangelo L., Ochs H.D., Etzioni A., Bonnefoy J.Y., Cosyns M. & Weinberg A.Cholangiopathy and tumors of the pancreas, liver, and biliary tree in boys with X-linked immunodeficiency with hyper-IgMJ. Immunol., 1997, 158, 977–983 [PubMed]
  • Heine J., Moon H.W. & Woodmansee D.B.Persistent Cryptosporidium infection in congenitally athymic (nude) miceInfect. Immun., 1984, 43, 856–859 [PMC free article] [PubMed]
  • Herrera L.A. & Ostrosky-Wegman P.Do helminths play a role in carcinogenesis? Trends Parasitol., 2001, 17, 172–175 [PubMed]
  • Heussler V.T., Kuenzi P. & Rottenberg S.Inhibition of apoptosis by intracellular protozoan parasitesInt. J. Parasitol., 2001, 31, 1166–1176 [PubMed]
  • Heussler V.T., Rottenberg S., Schwab R., Küenzi P., Fernandez P.C., Mckellar S., Shiels B., Chen Z.J., Orth K., Wallach D. & Dobbelaere D.A.Hijacking of host cell IKK signalosomes by the transforming parasite TheileriaScience, 2002, 298, 1033–1036 [PubMed]
  • Hicks R.M., James C. & Webbe G.Effect of Schistosoma haematobium and N-butyl-N-(4-hydroxybutyl)nitrosamine on the development of urothelial neoplasia in the baboonBr. J. Cancer, 1980, 42, 730–755 [PMC free article] [PubMed]
  • Hicks R.M., Ismail M.M., Walters C.L., Beecham P.T., Rabie M.F. & Alamy M.A.Association of bacteriuria and urinary nitrosamine formation with Schistosoma haematobium infection in the Qalyub area of EgyptTrans. R. Soc. Trop. Med. Hyg., 1982, 76, 519–527 [PubMed]
  • Iarc – International Agency For Research on Cancer Monograph on the evaluation of carcinogenic risks to humans. Part B: Biological agents. World Health Organization, International Agency for Research on Cancer Volume. 2011, Volume 100, 1–487
  • Izquierdo J., Antunez I., Calderon M.T., Perez Giraldo C. & Munoz Sanz A.Diarrhea caused by Cryptosporidium and colonic neoplasiaRev. Clin. Esp., 1988, 182,393–394[PubMed]
  • Jittimanee J., Sermswan R.W., Puapairoj A., Maleewong W. & Wongratanacheewin S.Cytokine expression in hamsters experimentally infected with Opisthorchis viverriniParasite Immunol., 2007, 29, 159–167 [PubMed]
  • Khurana S., Dubey M.L. & Malla N.Association of parasitic infections and cancersIndian J. Med. Microbiol., 2005, 23, 74–79 [PubMed]
  • Kim J.O., Jung S.S., Kim S.Y., Kim T.Y., Shin D.W., Lee J.H. & Lee Y.H.Inhibition of Lewis lung carcinoma growth by Toxoplasma gondii through induction of Th1 immune responses and inhibition of angiogenesisJ. Korean Med. Sci., 2007, 22, S38–S46 [PMC free article] [PubMed]
  • Kim E.M., Kim J.S., Choi M.H., Hong S.T. & Bae Y.M.Effects of excretory/secretory products from Clonorchis sinensis and the carcinogen dimethylnitrosamine on the proliferation and cell cycle modulation of human epithelial HEK293T cellsKorean J. Parasitol., 2008, 46, 127–132 [PMC free article] [PubMed]
  • Laha T., Pinlaor P., Mulvenna J., Sripa B., Sripa M., Smout M.J., Gasser R.B., Brindley P.J. & Loukas A.Gene discovery for the carcinogenic human liver flukeOpisthorchis viverrini. BMC Genomics, 2007, 8, 189 [PMC free article] [PubMed]
  • Laurent F., Kagnoff M.F., Savidge T.C., Naciri M. & Eckmann L.Human intestinal epithelial cells respond to Cryptosporidium parvum infection with increased prostaglandin H synthase 2 expression and prostaglandin E2 and F2alpha productionInfect. Immun., 1998, 66, 1787–1790 [PMC free article] [PubMed]
  • Laurent F., Eckmann L., Savidge T.C., Morgan G., Theodos C., Naciri M. & Kagnoff M.F.Cryptosporidium parvum infection of human intestinal epithelial cells induces the polarized secretion of C-X-C chemokinesInfect. Immun., 1997, 65, 5067–5073 [PMC free article] [PubMed]
  • Liu J., Deng M., Lancto C.A., Abrahamsen M.S., Rutherford M.S. & Enomoto S.Biphasic modulation of apoptotic pathways in Cryptosporidium parvum-infected human intestinal epithelial cellsInfect. Immun., 2009, 77, 837–849 [PMC free article][PubMed]
  • Lizundia R., Chaussepied M., Huerre M., Werling D., Santo J.P. & Langsley G.c-Jun NH2-terminal kinase/c-Jun signaling promotes survival and metastasis of B lymphocytes transformed by TheileriaCancer Res., 2006, 66, 6105–6110 [PubMed]
  • Lopez-Velez R., Tarazona R., Garcia Camacho A., Gomez-Mampaso E., Guerrero A., Moreira V. & Villanueva R.Intestinal and extraintestinal cryptosporidiosis in AIDS patientsEur. J. Clin. Microbiol. Infect. Dis., 1995, 14, 677–681 [PubMed]
  • Lowe S.W. & Lin A.W.Apoptosis in cancerCarcinogenesis, 2000, 21, 485–495[PubMed]
  • Lüder C.G., Stanway R.R., Chaussepied M., Langsley G. & Heussler V.T.Intracellular survival of apicomplexan parasites and host cell modificationInt. J. Parasitol., 2009, 39, 163–173 [PubMed]
  • Lun Z.R., Gasser R.B., Lai D.H., Li A.X., Zhu X.Q., Yu X.B. & Fang Y.Y.Clonorchiasis: a key foodborne zoonosis in ChinaLancet Infect. Dis., 2005, 5, 31–41 [PubMed]
  • Luperchio S.A. & Schauer D.B.Molecular pathogenesis of Citrobacter rodentium and transmissible murine colonic hyperplasiaMicrobes Infect., 2001, 3, 333–340[PubMed]
  • Maillot C., Gargala G., Delaunay A., Ducrotte P., Brasseur P., Ballet J.J. & Favennec L.Cryptosporidium parvum infection stimulates the secretion of TGF-beta, IL-8 and RANTES by Caco-2 cell lineParasitol. Res., 2000, 86, 947–949 [PubMed]
  • Mayer D.A. & Fried B.The role of helminth infections in carcinogenesisAdv. Parasitol., 2007, 65, 239–296 [PubMed]
  • Mele R., Gomez Morales M.A., Tosini F. & Pozio E.Cryptosporidium parvum at different developmental stages modulates host cell apoptosis in vitroInfect. Immun., 2004, 72, 6061–6067 [PMC free article] [PubMed]
  • Mohsen M.A., Hassan A.A., El-Sewedy S.M., Aboul-Azm T., Magagnotti C., Fanelli R. & Airoldi L.Biomonitoring of N-nitroso compounds, nitrite and nitrate in the urine of Egyptian bladder cancer patients with or without Schistosoma haematobiuminfectionInt. J. Cancer, 1999, 82, 789–794 [PubMed]
  • Mostafa M.H., Helmi S., Badawi A.F., Tricker A.R., Spiegelhalder B. & Preussmann R.Nitrate, nitrite and volatile N-nitroso compounds in the urine of Schistosoma haematobium and Schistosoma mansoni infected patientsCarcinogenesis, 1994, 15, 619–625 [PubMed]
  • Mostafa M.H., Sheweita S.A. & O’Connor P.J.Relationship between schistosomiasis and bladder cancerClin. Microbiol. Rev., 1999, 12, 97–111 [PMC free article][PubMed]
  • Naugler W.E. & Karin M.NF-kappaB and cancer-identifying targets and mechanismsCurr. Opin. Genet. Dev., 2008, 18, 19–26 [PMC free article] [PubMed]
  • Ninlawan K., O’Hara S.P., Splinter P.L., Yongvanit P., Kaewkes S., Surapaitoon A., Larusso N.F. & Sripa B.Opisthorchis viverrini excretory/secretory products induce toll-like receptor 4 upregulation and production of interleukin 6 and 8 in cholangiocyteParasitol. Int., 2010, 59, 616–621 [PMC free article] [PubMed]
  • O’Connor S.M., Taylor C.E. & Hughes J.M.Emerging infectious determinants of chronic diseasesEmerg. Infect. Dis., 2006, 12, 1051–1057 [PMC free article][PubMed]
  • Okhuysen P.C. & Chappell C.L.Cryptosporidium virulence determinants-are we there yet? Int. J. Parasitol., 2002, 32, 517–525 [PubMed]
  • Orem J., Mbidde E.K., Lambert B., Sanjose S. & Weiderpass E.Burkitt’s lymphoma in Africa, a review of the epidemiology and etiologyAfr. Health Sci., 2007, 7, 166–175 [PMC free article] [PubMed]
  • Osada Y., Kumagai T., Masuda K., Suzuki T. & Kanazawa T.Mutagenicity evaluation of Schistosoma spp. extracts by the umu-test and V79/HGPRT gene mutation assayParasitol. Int., 2005, 11, 175–183 [PubMed]
  • Parkin D.M.The global health burden of infection-associated cancers in the year 2002Int. J. Cancer, 2006, 118, 3030–3044 [PubMed]
  • Pinlaor S., Hiraku Y., Ma N., Yongvanit P., Semba R., Oikawa S., Murata M., Sripa B., Sithithaworn P. & Kawanishi S.Mechanism of NO-mediated oxidative and nitrative DNA damage in hamsters infected with Opisthorchis viverrini: a model of inflammation-mediated carcinogenesisNitric Oxide, 2004, 11, 175–183 [PubMed]
  • Pinlaor S., Hiraku Y., Yongvanit P., Tada-Oikawa S., Ma N., Pinlaor P., Sithithaworn P., Sripa B., Murata M., Oikawa S. & Kawanishi S.iNOS-dependent DNA damage via NFkappaB expression in hamsters infected with Opisthorchis viverrini and its suppression by the antihelminthic drug praziquantelInt. J. Cancer, 2006, 119, 1067–1072 [PubMed]
  • Plattner F. & Soldati-Favre D.Hijacking of host cellular functions by the ApicomplexaAnnu. Rev. Microbiol., 2008, 62, 471–487 [PubMed]
  • Porta C., Riboldi E. & Sica A.Mechanisms linking pathogens-associated inflammation and cancerCancer Lett., 2011, 305, 250–262 [PubMed]
  • Pozio E. & Morales M.A.The impact of HIV-protease inhibitors on opportunistic parasitesTrends Parasitol., 2005, 21, 58–63 [PubMed]
  • Preston-Martin S., Pike M.C., Ross R.K., Jones P.A. & Henderson B.E.Increased cell division as a cause of Human cancerCancer Res., 1990, 50, 7415–7421 [PubMed]
  • Raziuddin S., Masihuzzaman M., Shetty S. & Ibrahim A.Tumor necrosis factor alpha production in schistosomiasis with carcinoma of urinary bladderJ. Clin. Immunol., 1993, 13, 23–29 [PubMed]
  • Rosin M.P., Anwar W.A. & Ward A.J.Inflammation, chromosomal instability, and cancer: the schistosomiasis modelCancer Res., 1994a, 54, 1929s–1933 s [PubMed]
  • Rosin M.P., Saad El Din Zaki S., Ward A.J. & Anwar W.A.Involvement of inflammatory reactions and elevated cell proliferation in the development of bladder cancer in schistosomiasis patientsMutat. Res., 1994b, 305, 283–292 [PubMed]
  • Salim E., Morimura K., Menesi A., El-Lity M., Fukushima S. & Wanibuchi H.Elevated oxidative stress and DNA damage and repair levels in urinary bladder carcinomas associated with schistosomiasisInt. J. Cancer, 2008, 123, 601–608[PubMed]
  • Sayed El-Ahl S.A., El-Wakil H.S., Kamel N.M. & Mahmoud M.S.A preliminary study on the relationship between Trichomonas vaginalis and cervical cancer in Egyptian womenJ. Egypt. Soc. Parasitol., 2002, 32, 167–178 [PubMed]
  • Schmidt L., Lepers M., Brion M., Abdellatifi M.H., Dei-Cas E., Vernes A. & Fourrier A.Opisthorchiidoses chez les réfugiés de l’Asie du Sud-Est dans le Nord de la FranceMed. Mal. Infect., 1982, 12, 442–446
  • Schmuckli-Maurer J., Kinnaird J., Pillai S., Hermann P., Mckellar S., Weir W., Dobbelaere D. & Shiels B.Modulation of NF-kappaB activation in Theileria annulata-infected cloned cell lines is associated with detection of parasite-dependent IKK signalosomes and disruption of the actin cytoskeletonCell. Microbiol., 2010, 12, 158–173 [PubMed]
  • Schneider I., Haller D., Kullmann B., Beyer D., Ahmed J.S. & Seitzer U.Identification, molecular characterization and subcellular localization of a Theileria annulata parasite protein secreted into the host cell cytoplasmParasitol. Res., 2007, 101, 1471–1482[PubMed]
  • Semenza J.C. & Nichols G.Cryptosporidiosis surveillance and water-borne outbreaks in EuropeEurosurveillance, 2007, 12, 120–123 [PubMed]
  • Sithithaworn P. & Haswell-Elkins M.Epidemiology of Opisthorchis viverriniActa Trop., 1988, 88, 187–194 [PubMed]
  • Sripa B., Kaewkes S., Sithithaworn P., Mairiang E., Laha T., Smout M., Pairojkul C., Bhudhisawasdi V., Tesana S., Thinkamrop B., Bethony J.M., Loukas A. & Brindley P.J.Liver fluke induces cholangiocarcinomaPLoS Med., 2007, 4, e201. [PMC free article] [PubMed]
  • Sripa B. & Pairojkul C.Cholangiocarcinoma: lessons from ThailandCurr. Opin. Gastroenterol., 2008, 24(3), 349–356 [PMC free article] [PubMed]
  • Sripa B., Mairiang E., Thinkhamrop B., Laha T., Kaewkes S., Sithithaworn P., Tessana S., Loukas A., Brindley P.J. & Bethony J.M.Advanced periductal fibrosis from infection with the carcinogenic human liver fluke Opisthorchis viverrini correlates with elevated levels of interleukin-6Hepatology, 2009, 50, 1273–1281 [PMC free article] [PubMed]
  • Srivatanakul P., Sriplung H. & Deerasamee S.Epidemiology of liver cancer: an overviewAsian Pac. J. Cancer Prev., 2004, 5, 118–125 [PubMed]
  • Stark J.R., Judson G., Alderete J.F., Mundodi V., Kucknoor A.S., Giovannucci E.L., Platz E.A., Sutcliffe S., Fall K., Kurth T., Ma J., Stampfer M.J. & Mucci L.A.Prospective study of Trichomonas vaginalis infection and prostate cancer incidence and mortality: Physicians’ Health StudyJ. Natl. Cancer Inst., 2009, 101, 1368–1369 [PMC free article] [PubMed]
  • Stephens J., Cosyns M., Jones M. & Hayward A.Liver and bile duct pathology following Cryptosporidium parvum infection of immunodeficient miceHepatology, 1999, 30, 27–35 [PubMed]
  • Stojadinovic O., Lee B., Vouthounis C., Vukelic S., Pastar I., Blumenberg M., Brem H. & Tomic-Canic M.Novel genomic effects of glucocorticoids in epidermal keratinocytes: inhibition of apoptosis, interferon-gamma pathway, and wound healing along with promotion of terminal differentiationJ. Biol. Chem., 2007, 282, 4021–4034 [PubMed]
  • Stransky N., Vallot C., Reyal F., Bernard-Pierrot I., Medina S.G., Segraves R., Rycke Y., Elvin P., Cassidy A., Spraggon C., Graham A., Southgate J., Asselain B., Allory Y., Abbou C.C., Albertson D.G., Thiery J.P., Chopin D.K., Pinkel D. & Radvanyi F.Regional copy number-independent deregulation of transcription in cancerNat. Genet., 2006, 38, 1386–1396 [PubMed]
  • Sulzyc-Bielicka V., Kuzna-Grygiel W., Kolodziejczyk L., Bielicki D., Kladny J., Stepien-Korzonek M. & Telatynskasmieszek B.Cryptosporidiosis in patients with colorectal cancerJ. Parasitol., 2007, 93, 722–724 [PubMed]
  • Sunnotel O., Lowery C.J., Moore J.E., Dooley J.S., Xiao L., Millar B.C., Rooney P.J. & Snelling W.J.CryptosporidiumLett. Appl. Microbiol., 2006, 43, 7–16 [PubMed]
  • Sutcliffe S., Giovannucci E., Alderete J.F., Chang T.H., Gaydos C.A., Zenilman J.M., Marzo A.M., Willett W.C. & Platz E.A.Plasma antibodies against Trichomonas vaginalis and subsequent risk of prostate cancerCancer Epidemiol. Biomarkers Prev., 2006, 15, 939–945 [PubMed]
  • Sutcliffe S., Alderete J.F., Till C., Goodman P.J., Hsing A.W., Zenilman J.M., Marzo A.M. & Platz E.A.Trichomonosis and subsequent risk of prostate cancer in the Prostate Cancer Prevention TrialInt. J. Cancer, 2009, 124, 2082–2087 [PMC free article][PubMed]
  • Swellam M., Abd-Elmaksoud N., Halim M.H., Khatab H. & Khiry H.Incidence of Bcl-2 expression in bladder cancer: relation to schistosomiasisClin. Biochem., 2004, 37, 798–802 [PubMed]
  • Thuwajit C., Thuwajit P., Uchida K., Daorueang D., Kaewkes S., Wongkham S. & Miwa M.Gene expression profiling defined pathways correlated with fibroblast cell proliferation induced by Opisthorchis viverrini excretory/secretory productWorld J. Gastroenterol., 2006, 12, 3585–3592 [PMC free article] [PubMed]
  • Uhl E.W., Jacobson E., Bartick T.E., Micinilio J. & Schimdt R.Aural-pharyngeal polyps associated with Cryptosporidium infection in three iguanas (Iguana iguana)Vet. Pathol., 2001, 38, 239–242 [PubMed]
  • Valigurova A., Jirku M., Koudela B., Gelnar M., Modry D. & Slapeta J.Cryptosporidia: epicellular parasites embraced by the host cell membraneInt. J. Parasitol., 2008, 38, 913–922 [PubMed]
  • Vennervald B.J. & Polman K.Helminths and malignancyParasite Immunol., 2009, 31, 686–696 [PubMed]
  • Vernes A., Abdellatifi M., Dei-Cas E., Poirriez J., Abdelmalek R.Y., Leroux N., Colin J.J., Brion M. & Fourrier A.Parasitoses et hémoglobinopathies chez 1 170 réfugiés du Sud Est asiatiqueNouv. Press. Med., 1982, 11, 2687–2691 [PubMed]
  • Schubert C., Xue G., Schmuckli-Maurer J., Woods K.L., Nigg E.A. & Dobbelaere D.A.The transforming parasite Theileria co-opts host cell mitotic and central spindles to persist in continuously dividing cellsPLoS Biol., 2010, 8, e1000499. [PMC free article] [PubMed]
  • Who – World Health Organization CancerFact Sheet , 2011,(297), http://www.who.int/mediacentre/factsheets/ fs297/en/index.html
  • Xiao L., Fayer R., Ryan U. & Upton S.J.Cryptosporidium taxonomy: recent advances and implications for public healthClin. Microbiol. Rev., 2004, 17, 72–97 [PMC free article] [PubMed]
  • Xiao L.Molecular epidemiology of cryptosporidiosis: an updateExp. Parasitol., 2010, 124, 80–89 [PubMed]
  • Yap E.H., Ho T.H., Chan Y.C., Thong T.W., Ng G.C., Ho L.C. & Singh M.Serum antibodies to Trichomonas vaginalis in invasive cervical cancer patientsGenitourin. Med., 1995, 71, 402–404 [PMC free article] [PubMed]
  • Yoder J.S. & Beach M.J.Cryptosporidium surveillance and risk factors in the United StatesExp. Parasitol., 2010, 124, 31–39 [PubMed]
  • Zhang Z.F., Graham S., Yu S.Z., Marshall J., Zielezny M., Chen Y.X., Sun M., Tang S.L., Liao C.S., Xu J.L. & Yang X.Z.Trichomonas vaginalis and cervical cancer. A prospective study in ChinaAnn. Epidemiol., 1995, 5, 325–332 [PubMed]
  • Zhang X., Li Q., Hu P., Cheng H. & Huang G.Two case reports of pituitary adenoma associated with Toxoplasma gondii infection. J. Clin. Pathol., 2002, 55, 965–966 [PMC free article] [PubMed]
  • Zur Hausen H.The search for infectious causes of human cancers: where and whyVirology, 2010, 392, 1–10 [PubMed]
$$$ If you are interested in a writer or editor position, check out here.We are hiring. $$$

101

No Responses

Write a response

five + nine =