Open access peer-reviewed chapter

Perspective Chapter: Prospects for Pharmacological Therapy of Hepatic Alveolar Echinococcosis

Written By

Asher John Mohan, Bhaskar Kumar Gupta and Silviya Sarah Lal

Submitted: 28 November 2022 Reviewed: 19 December 2022 Published: 26 January 2023

DOI: 10.5772/intechopen.109588

From the Edited Volume

Echinococcosis - New Perspectives

Edited by Tonay Inceboz

Chapter metrics overview

62 Chapter Downloads

View Full Metrics

Abstract

Often misdiagnosed as liver cancer at first, the Alveolar hydatid disease or hepatic alveolar echinococcosis is an uncommon but potentially harmful variant of the disease also synonymously known as Echinococcus multilocularis (E. Multilocularis). The major area being drastically affected is the liver, from where its later advances into the lung and brain, typically fatal if left untreated. Even if surgery is still the recommended course of treatment for the condition, drug therapy cannot be thwarted off and remains essential and vital for individuals with disease extremity. This chapter therefore aims to present a framework through which FDA-approved drugs and nano drug delivery technologies collaborate to manage progressive hepatic alveolar echinococcosis.

Keywords

  • alveolar hydatid disease
  • hepatic alveolar echinococcosis
  • Echinococcus multilocularis
  • FDA-approved medications
  • nano drug delivery

1. Introduction

Echinococcosis is amongst those neglected diseases that the World Health Organisation (WHO) intends to manage or mitigate by the year 2050. Significant inclinations toward overhauling the treatment, management and control of Echinococossis originate from limitations of current diagnostic techniques, toxicity and low efficacy of medication options, usually suboptimal surgical procedures and other factors [1]. As the name implies, this chapter seeks to outline the latest clinical findings regarding the management of Hepatic Alveolar Echinococcosis (HAE) also just known as Alveolar Echinococcosis (AE).

Advertisement

2. Physiology of transmission

During its life cycle, the parasitic worm primarily picks out a carnivore like dogs and their prey (rodents) to act as its definitive and intermediate hosts. Despite not being actively engaged in the propagation of the illness, humans can nonetheless serve as intermediate hosts in certain situations. The maturation of adult worms and the release of eggs in the faeces of the definitive host as mentioned above are the variables that are involved in the transmission phenomena. After being consumed by a person (through food or water), the eggs later hatch and reach the liver via the lymphatic channels. This locus is significant in terms of the eggs developing into larvae and later metastasizing into other organs of the host [1].

Advertisement

3. Prevalence

Alveoli echinococcosis has been a significant problem in northern Japan, China, Tibet and other parts of Central Asia [2, 3, 4, 5, 6, 7]. In much of Europe, both definite and intermediate hosts have now been reported to be significantly more vulnerable to the infection [8, 9]. Until recent findings from parts of Mexico, AE was not regarded as a major human hazard in North America [10]. Nonetheless, over the past 10 years, human infections have been identified in North America and other parts of Canada [11, 12].

Advertisement

4. Clinical indications

The liver is where a high proportion of the disease’s primary lesions are sited, out of which many may differ to manifest for up to 15 years. Trauma to the hepatic blood vessels or bile ducts emerges as disease symptomatology [13]. The delay in disease identification was caused by both false imaging results and acute clinical symptoms, pointing to a problem (incorrect therapeutic management) that could not be easily overlooked [14]. It has been determined that elevated levels of biological response modifiers such as interleukins and genetic variations in antigen presenting cells such as the human leukocyte antigen are positively correlated with the onset or progression of the disease in humans [15, 16]. The occurrence of AE and its rapid progression have also been linked to acquiring therapeutic immunosuppression, which results from immunosuppressant administration necessary during a liver transplant [17].

Advertisement

5. Diagnosis

Visual tests (CT scans), imaging techniques and serology are amongst the most widely employed methodologies for the confirmation of the parasite. Other than these prominent ones, southern blotting and polymerase chain reactions (PCR) are the molecular techniques involved in the diagnosis of the disease. Discussed below are the most widely employed diagnostic methods for AE.

5.1 Imaging techniques

In order to offer physicians a trustworthy source of information when making a clinical judgement, the WHO has classified the disease lesions into a variety of forms solely based on imaging data obtained in the US [18]. The effectiveness of these imaging data as a screening and follow-up aid is gaining significant attention in centres of both Asia and Europe [19, 20]. Lesions from an abscess or tumour can now be differentiated by these techniques such as Contrast-enhanced ultrasonography (CEUS) [21]. In addition to the above technique, the methodology involved in an imaging technique named FDG-positron emission tomography (FDG-PET) is used to prevent inaccurate tumour interpretations [22].

5.2 Serology

Drawbacks of imaging techniques described above such as the inability to distinguish between an abscess and a neoplasm, difficulty in data interpretation and unreliability with respect to early diagnosis cause a shift in preference toward blood serum analysis known as serology. Furthermore, serology is more accurate as an affirmatory and reliable tool for AE cases. This is largely prevalent in developing nations where these techniques are absent [23]. Apart from these decisive advantages, patient follow up post medical or surgical treatment is also made simpler [24, 25].

5.2.1 Specific serological diagnosis of AE

This section covers the involvement of specific serological markers pertaining to the diagnosis of AE.

Excretory/secretory proteins (ESPs), also termed as parasitic antigens, are responsible for triggering the host’s immune response [26]. A member of the E. multilocularis specific ESPs, Em2, also referred to as Em2a and Em18, allows for precise discrimination between an AE and Cystic echinococcosis patient [27, 28]. Recent findings also render the role of a varied antigen named Em492 in the diagnosis of AE [29]. Similarly, showing high diagnostic performance for confirmation of AE is EM10 and its derivatives (another Echinococcus antigen) [30]. Based on molecular research done on mice, lipoprotein antigen B is the last of the few ESPs that is expressed in the disease condition [31].

5.2.2 Importance of serology both in pre and post therapy of AE

The method of Elisa to detect specific antibodies relating to AE with high sensitivities of up to 95% is employed at various laboratories. Addition of an immunoblotting technique to this ELISA maximised the diagnostic sensitivity [32]. A follow up usually done as relapses can prevail even after surgery of chronic administration of medications. This is where serology reappears back into the confirmation process where the antibody detection can be directly linked to the degree or extent of disease. A challenging situation that arises in this method occurs in the case of an immunosuppressed patient as less information is available to quantify or qualitatively analyse the antibody response. Based on a study to optimise the role of serology both for diagnosis and monitoring of pre or post therapies of AE have brought about the following results

  1. Serology can help in distinguishing between an active and inactive lesion. This holds true both in the diagnosis or pretreatment phases thus unnecessary advice for a surgery or medication can be avoided

  2. Serological methods as a follow up study post-surgery or therapy can aid in prediction of disease relapse [33].

Advertisement

6. Treatment

6.1 Invasive modus operandi

Profound resection of liver lesions constitutes the basis for the adoption of a treatment decision [34]. Even though hepatic surgery is the best course of action, significant cases of morbidity and mortality cannot be addressed even under expert supervision. This therefore exemplifies the fact as to why only one-third of the patient population develops positive outcomes from the procedure alone. Also note that the majority of geographical regions where this occurs have delayed diagnoses [35]. These drawbacks may also be attributed to the evidence that an intrusive procedure alone is unsafe for disease treatment because of its close association with complications as mentioned above [36].

In light of this, the treatment goals, and regimen for AE have now been tailored as follows.

  1. Complete occlusion of the parasitic lesion, followed by 2 years of anti-infective medication (examples discussed later).

  2. If the initial strategy fails to diminish disease burden, the period of anti-infective drugs needs an extension (upto 10 years to prevent recurrence).

  3. Lingering underlying difficulties can be tackled in aspects of reducing inflammation or obstruction in the bile duct [37]

With respect to the above-laid objectives let us dive deep into some of the pros and cons of the treatment.

An advanced parasite infection is indicated by lesions around the hepatic veins and progress into the inferior Vena cava. Such situations are resolved with the liver transplant procedure. Negative compliance with this strategic approach may be brought on by the lack of an appropriate matching donor and prolonged use of immunosuppressants [17].

As an alternative to a full transplant, the procedure known as Ex vivo liver resection can be used, in which the patient’s liver is physically removed, cleared of lesions post which it is reinserted. In cases of irresectable livers or late stages of the illness, this is favoured and produces favourable results without requiring long-term immunosuppressive treatment [38].

The other types of disease burden are hepatobiliary complications, which signify an unpleasant shift in the disease progression [39]. Ameliorative techniques used up until the turn of the century involved the percutaneous dilatation of the blocked bile ducts [40]. The contribution of such an invasive technique (termed as Perendoscopic bile duct stenting) for treating biliary obstruction is irreplaceable and is at widely being used. The desired result for individuals hospitalised for this procedure is an improvement in the quality of life [41].

6.2 Medication

Breakthroughs in AE have been made possible by faster detection, the introduction of drug therapy, and the gradual abandonment of resective procedures [42]. End-stage diagnoses, a lack of medical resources, and the failure of a specific drug or the severity of their side effects can unnerve nations going through such suffering [43].

Advertisement

7. Negative consequences in the current treatments of AE

Foretelling the outcome of AE is daunting as resection is often accompanied by chemotherapy using benzimidazoles for unquestionable periods of time [44]. Hepatotoxicity of the lot is amongst the major concerns. Along with this distress, although significantly increasing the survival rate of patients; they are capable only in reduction (static effect) but not destruction (cidal effect) of the parasite. In short, recurrence after drug withdrawal cannot be avoided [1].

This presents a clear conundrum regarding the need for additional AE treatment options. Therefore, selectivity, a sizable therapeutic window and a cidal rather than a static effect should be given priority when choosing a conventional drug for treatment of AE.

Advertisement

8. Pharmacological prospects for AE treatment

There is a plethora of preclinical research (both In vitro and In vivo) containing data on potential treatments for AE, such as usage of anti-infectives, anti-cancer drugs etc. However, these could not be completely depended upon due to the absence of thorough and reliable screening methods [45].

With respect to In vitro studies, the development of extensive means for specific species culture of AE larvae have brought about a change in the aforementioned conclusion, thereby opening doors for whole or reliable drug screening assays against the parasite [46, 47].

8.1 Anti-infective agents

These agents find place in AE as this condition is not restrictively a parasitic burden but also infectious in nature.

In vivo data revealed that chemotherapeutic drugs and anthelmintics, in addition to benzimidazoles, all had parasitostatic effects [48]. When used both alone (In vitro) and in conjunction with a benzimidazole (In vivo), nitazoxanide demonstrated promising outcomes against AE [49, 50]. Monotherapy and combination therapies on the contrary failed to treat AE in humans, despite the mentioned claims [51].

Tricloabendazole had the strongest In vitro antiparasitic activity of all the benzimidazole compounds evaluated. When evaluated In vitro and on additional mice models, drugs like fenbendazole, oxfendazole, and febantel were highly efficacious against the disease [52, 53].

8.2 Anti-malarial compounds

The Mefloquine compound used in treating malaria showed promising albendazole-like features when looking at an In vivo activity alone. Higher rates of reduction in the parasite burden were seen with an increase in dose of the said drug candidates. However, the latter’s incomplete pharmacokinetic data and the presence of neurotoxicity following prolonged mefloquine treatment could be a hazard to therapeutic regimens involving them [54].

8.3 Antibiotics

Clarithromycin, an antibiotic macrolide, significantly reduced the proliferation and morphological traits of E. multilocularis in vitro. Similar abnormalities were also translated on to the adult forms of the parasite. However, no suitable in vivo tests were carried out to further investigate these promising findings [55].

8.4 Other methods for assessment of potential targets

Affinity chromatography followed by MS-based sequencing can now be used to validate novel targets or other significant proteins expressed during the host–parasite encounter [56]. When it became necessary to assess the degree of harm brought on by pharmacological therapy, a technique of drug screening based on the release of the enzyme phosphoglucose isomerase was devised. This is now mostly used to anticipate how well different anti-parasitic medications can improve disease burden [57].

8.5 Cancer cell proliferation inhibitors

Reoccurring and indistinguishable characteristics between the parasitic condition and malignancies such as ongoing cell proliferation, ability to alter immune response, production of proteolytic enzymes, angiogenesis stimulation, over-expression of certain proteins, and the capability to create metastases, may find their application in usage of anti-cancer medications as a possible form of treatment [58].

One of the earliest anti-cancer compounds investigated for the potential use against AE was doxorubicin in the form of nanoparticles. Mostly because of the serious side effects that were accompanied to this moiety, this concept of therapy was dropped [59].

Later, genistein, an isoflavonoids-class of anti-tumour drug which demonstrated anti-parasitic properties, was introduced. Although highly effective, its negative impact on long-term treatment deprived it from being implemented or approved for the world scenario [60].

Another occurrence involved the research findings from the endogenous oestrogen metabolite, 2-methoxyestradiol, which caused substantial in vitro harm to the parasite. This however revealed no statistically significant differences from the conventional albendazole medication [61].

At very low concentrations, the mitogen-activated protein kinase and tyrosine-kinase inhibitors pyridinyl imidazoles and imatinib, respectively, caused parasite cell death [62, 63].

A possible therapeutic target that requires careful examination is the role glutathione-S-transferase plays in AE and other malignancies where it promotes cell proliferation. In other words, medications inhibiting cell proliferation could make way into pharmaceutical care for AE [64].

Investigation of other enzyme inhibitors, such as the proteasome inhibitor bortezomib, showed that this enzyme is a viable pharmacological target that should be pursued further [65].

With the help of early findings that require further analyses, cytostatic compounds can also be helpful moieties in AE prevention [66].

Advertisement

9. Recent lab findings

This section emphasises on the recent developments found in the research areas regarding AE.

9.1 Anti-cancer drugs

Alterations to a proto-oncogene (a gene involved in normal cell growth) may lead to the formation of oncogenes, which aids in cancer development. RAF is an example of such a proto-oncogene that regulates a variety of cellular functions, bringing about a gain in malignant properties of a normal cell. The anticancer drug, Sorafenib inhibits tumour cell proliferation and formation of new blood vessels (angiogenesis) by targeting RAF in Raf signalling pathway. The drug in question was found to exhibit strong inhibitions in conditions of multilocular echinococcosis [67].

9.2 Immune checkpoint inhibitor

After invasion the parasite has the capability to escape an encounter with the host immune system [68]. This immunosuppressive setting is highly influential for the viability and proliferation of the parasite [69].

It was found that Programmed Cell Death Ligand 1 abbreviated as PD-L1 plays a negative role in cancers by attenuating immune responses [70]. Studies that focused on this transmembrane protein reached the conclusion that the development of HAE was in close association with high expressions PD-L1. This could be a probable mechanism by which the parasite achieves immune escape [71].

Other supporting research was found to be in agreement with the above inference. Confirmation of this hypothesis was brought about by a control in the infection utilising immunotherapy which acted on PD-L1 pathway blockage [72, 73].

Alongside the above mechanism, an additional blocking activity on TIGIT, an inhibitory receptor led to tumour rejection. This was an indication that TIGIT and PD-1/PD-L1 blockade enhanced or synergised the anti-tumour effects of lymphocytes [74]. When In vivo studies on mice and humans were based on these mechanisms, TIGIT highlighted its potential as an immunomodulatory strategy for the treatment of AE [75, 76].

9.3 Nano drug delivery system

The rate of cure in a conventional anti-infective such as albendazole was found to be low. Poor bioavailability was found to be the reason behind the unfavourable effect. The need to alleviate this drug disadvantage led to the shift in focus for designing new formulations of the drug being discussed.

Liposomes that help in delivering active molecules to the site of action were the answer to this research problem [77, 78]. These treatments significantly reduced parasite burden in patients by increasing the mortality rate against larvae of the parasites when compared with traditional albendazole [79].

Other than liposomes, a valid suggestion for the employment of nanocrystals resulted in an increase in bioavailability of the same medication [80].

The preventive efficacies of these nanotechnology-based preparations were also found to be enhanced [81]. All these findings contribute to the fact that nanomedicinal forms of an anti-infective have developmental potential in improving anti-AE drug therapy.

Advertisement

10. Conclusions

Alveolar Echinococossis, although rare and neglected, is fatal if left untreated. The profound mode of treatment involves the resection of liver. However, we have discussed that this alone wasn’t sufficient and that supportive therapy using medications was a necessity. The severe adverse effects of traditional drugs also caused a decline in quality of life of patients. The above stated reasons brought about the need to seek further advanced treatment options for the disease such as new drug targets, elucidation of novel mechanisms involved in disease progression, various drug delivery technologies etc. The authors hence have highlighted the clinical indications and the need for specific diagnostic approaches of the disease such as imaging techniques and serology. Along with invasive modes of treatment, the additional employment of various medicaments including anthelmintics, anti-malarials, antibiotics, inhibitors of cancer proliferation, immune checkpoint inhibitors and nano drug delivery system that were found to be promising for the treatment of AE without exhibition of the adverse effects of traditional drugs were briefed. This chapter was approached with the sole purpose of exploring the various novel trends and those that are presently in action to tackle this slowly rising, yet worrisome endemic.

Acknowledgments

The authors would like to express their gratitude to People’s University, Bhanpur, Bhopal, for providing the necessary environment for completion of this chapter.

Conflict of interest

We, the authors, would like to declare that there is no conflict of interest based on the information produced.

References

  1. 1. Eckert J, Deplazes P. Biological, epidemiological, and clinical aspects of echinococcosis, a zoonosis of increasing concern. Clinical Microbiology Reviews. 2004;17(1):107-135
  2. 2. Kawamura N, Kamiyama T, Sato N, Nakanishi K, Yokoo H, Kamachi H, et al. Long-Term Results of hepatectomy for patients with alveolar echinococcosis: A single-center experience. Journal of the American College of Surgeons. 2011;212(5):804-812
  3. 3. Feng X, Qi X, Yang L, Duan X, Fang B, Gongsang Q , et al. Human cystic and alveolar echinococcosis in the Tibet Autonomous Region (TAR), China. Journal of Helminthology. 2015;89(6):671-679
  4. 4. Cai H, Guan Y, Ma X, Wang L, Wang H, Su G, et al. Epidemiology of Echinococcosis among School children in Golog Tibetan Autonomous Prefecture, Qinghai, China. The American Journal of Tropical Medicine and Hygiene. 2017;96(3):674-679
  5. 5. Abdybekova A, Sultanov A, Karatayev B, Zhumabayeva A, Shapiyeva Z, Yeshmuratov T, et al. Epidemiology of echinococcosis in Kazakhstan: An update. Journal of Helminthology. 2015;89(6):647-650
  6. 6. Torgerson PR. The emergence of echinococcosis in central Asia. Parasitology. 2013;140(13):1667-1673
  7. 7. Counotte MJ, Minbaeva G, Usubalieva J, Abdykerimov K, Torgerson PR. The Burden of Zoonoses in Kyrgyzstan: A systematic review. PLoS Neglected Tropical Diseases. 2016;10(7):e0004831
  8. 8. Vuitton DA, Demonmerot F, Knapp J, Richou C, Grenouillet F, Chauchet A, et al. Clinical epidemiology of human AE in Europe. Veterinary Parasitology. 2015;213(3-4):110-120
  9. 9. Gottstein B, Stojkovic M, Vuitton DA, Millon L, Marcinkute A, Deplazes P. Threat of alveolar echinococcosis to public health – A challenge for Europe. Trends in Parasitology. 2015;31(9):407-412
  10. 10. Deplazes P, Rinaldi L, Alvarez Rojas CA, Torgerson PR, Harandi MF, Romig T, et al. Global distribution of alveolar and cystic echinococcosis. Advances in Parasitology. 2017;95:315-493
  11. 11. Massolo A, Liccioli S, Budke C, Klein C. Echinococcus multilocularis in North America: the great unknown. Parasite. 2014;21:73
  12. 12. Catalano S, Lejeune M, Liccioli S, Verocai GG, Gesy KM, Jenkins EJ, et al. Echinococcus multilocularis in Urban Coyotes, Alberta, Canada. Emerging Infectious Diseases. 2012;18(10):1625-1628
  13. 13. Derbel F, Ben M, Hadj Hamida MB, Mazhoud J, Youssef S, Ben A, et al. Hydatid cysts of the liver - diagnosis, complications and treatment. In: Derbel F, editor. Abdominal Surgery [Internet. London, UK, London: InTech; 2012. Available from: http://www.intechopen.com/books/abdominal-surgery/hydatid-cysts-of-the-liver-diagnosis-complications-and-treatment
  14. 14. Chauchet A, Grenouillet F, Knapp J, Richou C, Delabrousse E, Dentan C, et al. Increased incidence and characteristics of alveolar echinococcosis in patients with immunosuppression-associated conditions. Clinical Infectious Diseases. 2014;59(8):1095-1104
  15. 15. Vuitton DA. Echinococcosis and allergy. Clinical Reviews in Allergy and Immunology. 2004;26(2):93-104
  16. 16. Eiermann TH, Bettens F, Tiberghien P, Schmitz K, Beurton I, Bresson-Hadni S, et al. HLA and alveolar echinococcosis. Tissue Antigens. 1998;52(2):124-129
  17. 17. Koch S, Bresson-Hadni S, Miguet JP, Crumbach JP, Gillet M, Mantion GA, et al. Experience of liver transplantation for incurable alveolar echinococcosis: A 45-case European collaborative report. Transplantation. 2003;75(6):856-863
  18. 18. Kern P, Wen H, Sato N, Vuitton DA, Gruener B, Shao Y, et al. WHO classification of alveolar echinococcosis: Principles and application. Parasitology International. 2006;55:S283-S287
  19. 19. Kratzer W. Proposal of an ultrasonographic classification for hepatic alveolar echinococcosis: Echinococcosis multilocularis Ulm classification-ultrasound. World Journal of Gastroenterology. 2015;21(43):12392
  20. 20. Graeter T, Kratzer W, Oeztuerk S, Haenle MM, Mason RA, Hillenbrand A, et al. Proposal of a computed tomography classification for hepatic alveolar echinococcosis. World Journal of Gastroenterology. 2016;22(13):3621
  21. 21. Tao S, Qin Z, Hao W, Yongquan L, Lanhui Y, Lei Y. Usefulness of gray-scale contrast-enhanced ultrasonography (SonoVue®) in diagnosing hepatic alveolar echinococcosis. Ultrasound in Medicine & Biology. 2011;37(7):1024-1028
  22. 22. Rolle AM, Soboslay PT, Reischl G, Hoffmann WH, Pichler BJ, Wiehr S. Evaluation of the metabolic activity of Echinococcus multilocularis in rodents using positron emission tomography tracers. Molecular Imaging and Biology. 2015;17(4):512-520
  23. 23. Stojkovic M, Gottstein B, Junghanss T. Echinococcosis. In: Manson’s Tropical Infectious Diseases. 2014;23:795-819
  24. 24. Fujimoto Y, Ito A, Ishikawa Y, Inoue M, Suzuki Y, Ohhira M, et al. Usefulness of recombinant Em18-ELISA to evaluate efficacy of treatment in patients with alveolar echinococcosis. Journal of Gastroenterology. 2005;40(4):426-431
  25. 25. Ma L, Ito A, Yhan L, Wang X-G, Yao Y-Q , Yu DG, et al. Alveolar echinococcosis: Em2plus-ELISA™ and Em18-Western blots for follow-up after treatment with albendazole. Transactions of the Royal Society of Tropical Medicine and Hygiene. 1997;91(4):476-478
  26. 26. Qiu Y, Shen S, Yang Y, Wang W. An excretory protein of Echinococcus multilocularis inhibits complement classical pathway activation. Infection and Drug Resistance. 2022;15:555-568
  27. 27. Gottstein B. Purification and characterization of a specific antigen from Echinococcus multilocularis. Parasite Immunology. 1985;7(3):201-212
  28. 28. Xiao N, Mamuti W, Yamasaki H, Sako Y, Nakao M, Nakaya K, et al. Evaluation of use of recombinant Em18 and affinity-purified Em18 for serological differentiation of alveolar Echinococcosis from cystic echinococcosis and other parasitic infections. Journal of Clinical Microbiology. 2003;41(7):3351-3353
  29. 29. Walker M, Baz A, Dematteis S, Stettler M, Gottstein B, Schaller J, et al. Isolation and characterization of a secretory component of Echinococcus multilocularis metacestodes potentially involved in modulating the host-parasite interface. Infection and Immunity. 2004;72(1):527-536
  30. 30. Helbig M, Frosch P, Kern P, Frosch M. Serological differentiation between cystic and alveolar echinococcosis by use of recombinant larval antigens. Journal of Clinical Microbiology. 1993;31(12):3211-3215
  31. 31. Mamuti W, Yamasaki H, Sako Y, Nakao M, Xiao N, Nakaya K, et al. Molecular cloning, expression, and serological evaluation of an 8-kilodalton subunit of antigen B from Echinococcus multilocularis. Journal of Clinical Microbiology. 2004;42(3):1082-1088
  32. 32. Müller N, Frei E, Nuñez S, Gottstein B. Improved serodiagnosis of alveolar echinococcosis of humans using an in vitro-produced Echinococcus multilocularis antigen. Parasitology. 2007;134(6):879-888
  33. 33. Gottstein B, Lachenmayer A, Beldi G, Wang J, Merkle B, Vu XL, et al. Diagnostic and follow-up performance of serological tests for different forms/courses of alveolar echinococcosis. Food and Waterborne Parasitology. 2019;16:e00055
  34. 34. Vuitton DA, Azizi A, Richou C, Vuitton L, Blagosklonov O, Delabrousse E, et al. Current interventional strategy for the treatment of hepatic alveolar echinococcosis. Expert Review of Anti-infective Therapy. 2016;14(12):1179-1194
  35. 35. Kern P, Menezes da Silva A, Akhan O, Müllhaupt B, Vizcaychipi KA, Budke C, et al. The echinococcoses: diagnosis, clinical management and burden of disease. Advances in Parasitology. 2017;96:259-369
  36. 36. Brunetti E, Kern P, Vuitton DA. Expert consensus for the diagnosis and treatment of cystic and alveolar echinococcosis in humans. Acta Tropica. 2010;114(1):1-16
  37. 37. Wen H, Vuitton L, Tuxun T, Li J, Vuitton DA, Zhang W, et al. Echinococcosis: Advances in the 21st Century. Clinical Microbiology Reviews. 2019;32(2):e00075-e00018
  38. 38. Mantion GA, Vuitton DA. Auto-versus allo-transplantation of the liver for end-stage alveolar echinococcosis? Chinese Medical Journal. 2011;124(18):2803-2805
  39. 39. Graeter T. Hepatobiliary complications of alveolar echinococcosis: A long-term follow-up study. World Journal of Gastroenterology. 2015;21(16):4925
  40. 40. Frei P. Late biliary complications in human alveolar echinococcosis are associated with high mortality. World Journal of Gastroenterology. 2014;20(19):5881
  41. 41. Ambregna S, Koch S, Sulz MC, Grüner B, Öztürk S, Chevaux JB, et al. A European survey of perendoscopic treatment of biliary complications in patients with alveolar echinococcosis. Expert Review of Anti-infective Therapy. 2017;15(1):79-88
  42. 42. Torgerson PR, Schweiger A, Deplazes P, Pohar M, Reichen J, Ammann RW, et al. Alveolar echinococcosis: From a deadly disease to a well-controlled infection. Relative survival and economic analysis in Switzerland over the last 35 years. Journal of Hepatology. 2008;49(1):72-77
  43. 43. Hemphill A, Stadelmann B, Rufener R, Spiliotis M, Boubaker G, Müller J, et al. Treatment of echinococcosis: Albendazole and mebendazole – What else? Parasite. 2014;21:70
  44. 44. Reuter S. Benzimidazoles in the treatment of alveolar echinococcosis: A comparative study and review of the literature. Journal of Antimicrobial Chemotherapy. 2000;46(3):451-456
  45. 45. Hemphill A, Stadelmann B, Scholl S, Müller J, Spiliotis M, Müller N, et al. Echinococcus metacestodes as laboratory models for the screening of drugs against cestodes and trematodes. Parasitology. 2010;137(3):569-587
  46. 46. Brehm K, Spiliotis M. Recent advances in the in vitro cultivation and genetic manipulation of Echinococcus multilocularis metacestodes and germinal cells. Experimental Parasitology. 2008;119(4):506-515
  47. 47. Zheng H, Zhang W, Zhang L, Zhang Z, Li J, Lu G, et al. The genome of the hydatid tapeworm Echinococcus granulosus. Nature Genetics. 2013;45(10):1168-1175
  48. 48. Sileslucas M, Hemphill A. Cestode parasites: Application of in vivo and in vitro models for studies on the host-parasite relationship. Advances in Parasitology. 2002;51:133-230
  49. 49. Küster T, Kriegel N, Stadelmann B, Wang X, Dong Y, Vennerstrom JL, et al. Amino ozonides exhibit in vitro activity against Echinococcus multilocularis metacestodes. International Journal of Antimicrobial Agents. 2014;43(1):40-46
  50. 50. Stettler M, Rossignol JF, Fink R, Walker M, Gottstein B, Merli M, et al. Secondary and primary murine alveolar echinococcosis: Combined albendazole/nitazoxanide chemotherapy exhibits profound anti-parasitic activity. International Journal for Parasitology. 2004;34(5):615-624
  51. 51. Tappe D, Müller A, Frosch M, Stich A. Limitations of amphotericin B and nitazoxanide in the treatment of alveolar echinococcosis. Annals of Tropical Medicine and Parasitology. 2009;103(2):177-181
  52. 52. Richter D, Richter J, Grüner B, Kranz K, Franz J, Kern P. In vitro efficacy of triclabendazole and clorsulon against the larval stage of Echinococcus multilocularis. Parasitology Research. 2013;112(4):1655-1660
  53. 53. Küster T, Stadelmann B, Aeschbacher D, Hemphill A. Activities of fenbendazole in comparison with albendazole against Echinococcus multilocularis metacestodes in vitro and in a murine infection model. International Journal of Antimicrobial Agents. 2014;43(4):335-342
  54. 54. Küster T, Stadelmann B, Hermann C, Scholl S, Keiser J, Hemphill A. In Vitro and In Vivo efficacies of mefloquine-based treatment against alveolar Echinococcosis. Antimicrobial Agents and Chemotherapy. 2011;55(2):713-721
  55. 55. Mathis A, Wild P, Boettger EC, Kapel CMO, Deplazes P. Mitochondrial Ribosome as the target for the macrolide antibiotic clarithromycin in the Helminth Echinococcus multilocularis. Antimicrobial Agents and Chemotherapy. 2005;49(8):3251-3255
  56. 56. The Taenia solium Genome Consortium, Tsai IJ, Zarowiecki M, Holroyd N, Garciarrubio A, Sanchez-Flores A, et al. The genomes of four tapeworm species reveal adaptations to parasitism. Nature. 2013;496(7443):57-63
  57. 57. Stadelmann B, Scholl S, Muller J, Hemphill A. Application of an in vitro drug screening assay based on the release of phosphoglucose isomerase to determine the structure-activity relationship of thiazolides against Echinococcus multilocularis metacestodes. Journal of Antimicrobial Chemotherapy. 2010;65(3):512-519
  58. 58. Klinkert MQ , Heussler V. The use of anticancer drugs in antiparasitic chemotherapy. Mini Reviews in Medicinal Chemistry. 2006;6(2):131-143
  59. 59. Liance M, Nemati F, Bories C, Couvreur P. Experience with doxorubicin-bound polyisohexylcyanoacrylate nanoparticles on murine alveolar echinococcosis of the liver. International Journal for Parasitology. 1993;23(3):427-429
  60. 60. Naguleswaran A, Spicher M, Vonlaufen N, Ortega-Mora LM, Torgerson P, Gottstein B, et al. In vitro metacestodicidal activities of genistein and other isoflavones against Echinococcus multilocularis and Echinococcus granulosus. Antimicrobial Agents and Chemotherapy. 2006;50(11):3770-3778
  61. 61. Spicher M, Naguleswaran A, Ortega-Mora LM, Müller J, Gottstein B, Hemphill A. In vitro and in vivo effects of 2-methoxyestradiol, either alone or combined with albendazole, against Echinococcus metacestodes. Experimental Parasitology. 2008;119(4):475-482
  62. 62. Gelmedin V, Caballero-Gamiz R, Brehm K. Characterization and inhibition of a p38-like mitogen-activated protein kinase (MAPK) from Echinococcus multilocularis: Antiparasitic activities of p38 MAPK inhibitors. Biochemical Pharmacology. 2008;76(9):1068-1081
  63. 63. Hemer S, Brehm K. In vitro efficacy of the anticancer drug imatinib on Echinococcus multilocularis larvae. International Journal of Antimicrobial Agents. 2012;40(5):458-462
  64. 64. Liebau E. Molecular cloning, expression and characterization of a recombinant glutathione S-transferase from Echinococcus multilocularis. Molecular and Biochemical Parasitology. 1996;77(1):49-56
  65. 65. Stadelmann B, Aeschbacher D, Huber C, Spiliotis M, Müller J, Hemphill A. Profound activity of the anti-cancer drug bortezomib against Echinococcus multilocularis metacestodes identifies the proteasome as a novel drug target for cestodes. PLoS Neglected Tropical Diseases. 2014;8(12):e3352
  66. 66. Hübner C, Wiehr S, Kocherscheidt L, Wehrl H, Pichler BJ, Schmid A, et al. Effects of in vitro exposure of Echinococcus multilocularis metacestodes to cytostatic drugs on in vivo growth and proliferation of the parasite. Parasitology Research. 2010;107(2):459-463
  67. 67. Eberhard DA, Johnson BE, Amler LC, Goddard AD, Heldens SL, Herbst RS, et al. Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non–small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. Journal of Clinical Oncology. 2005;23(25):5900-5909
  68. 68. Zhang Q , Ye JR, Ma HM, Wu JJ, Jiang T, Zheng H. Role of immune tolerance in BALB/c mice with anaphylactic shock after Echinococcus granulosus infection. Immunologic Research. 2016;64(1):233-241
  69. 69. Bakhtiar NM, Spotin A, Mahami-Oskouei M, Ahmadpour E, Rostami A. Recent advances on innate immune pathways related to host–parasite cross-talk in cystic and alveolar echinococcosis. Parasites Vectors. 2020;13(1):232
  70. 70. Han Y, Liu D, Li L. PD-1/PD-L1 pathway: Current researches in cancer. American Journal of Cancer Research. 2020;10(3):727-742
  71. 71. La X, Zhang F, Li Y, Li J, Guo Y, Zhao H, et al. Upregulation of PD-1 on CD4+CD25+T cells is associated with immunosuppression in liver of mice infected with Echinococcus multilocularis. International Immunopharmacology. 2015;26(2):357-366
  72. 72. Bellanger A, Courquet S, Pallandre J, Godet Y, Millon L. Echinococcus multilocularis vesicular fluid induces the expression of immune checkpoint proteins in vitro. Parasite Immunology [Internet]. 2020;42(6):e12711
  73. 73. Jebbawi F, Bellanger A, Lunström-Stadelmann B, Rufener R, Dosch M, Goepfert C, et al. Innate and adaptive immune responses following PD-L1 blockade in treating chronic murine alveolar echinococcosis. Parasite Immunology. 2021;43(8):e12834
  74. 74. Ge Z, Peppelenbosch MP, Sprengers D, Kwekkeboom J. TIGIT, the next step towards successful combination immune checkpoint therapy in cancer. Frontiers in Immunology. 2021;12:699895
  75. 75. Zhang C, Lin R, Li Z, Yang S, Bi X, Wang H, et al. Immune exhaustion of T cells in alveolar echinococcosis patients and its reversal by blocking checkpoint receptor TIGIT in a murine model. Hepatology. 2020;71(4):1297-1315
  76. 76. Zhang C, Wang H, Li J, Hou X, Li L, Wang W, et al. Involvement of TIGIT in natural killer cell exhaustion and immune escape in patients and mouse model with liver Echinococcus multilocularis infection. Hepatology. 2021;74(6):3376-3393
  77. 77. Yue X, Dai Z. Liposomal nanotechnology for cancer theranostics. Current Medicinal Chemistry. 2018;25(12):1397-1408
  78. 78. Akbarzadeh A, Rezaei-Sadabady R, Davaran S, Joo SW, Zarghami N, Hanifehpour Y, et al. Liposome: Classification, preparation, and applications. Nanoscale Research Letters. 2013;8(1):102
  79. 79. Bakhtiar NM, Akbarzadeh A, Casulli A, Mahami-Oskouei M, Ahmadpour E, Nami S, et al. Therapeutic efficacy of nanocompounds in the treatment of cystic and alveolar echinococcoses: Challenges and future prospects. Parasitology Research. 2019;118(9):2455-2466
  80. 80. Pensel P, Paredes A, Albani CM, Allemandi D, Sanchez Bruni S, Palma SD, et al. Albendazole nanocrystals in experimental alveolar echinococcosis: Enhanced chemoprophylactic and clinical efficacy in infected mice. Veterinary Parasitology. 2018;251:78-84
  81. 81. Hu C, Liu Z, Liu C, Zhang Y, Fan H, Qian F. Improvement of antialveolar echinococcosis efficacy of albendazole by a novel nanocrystalline formulation with enhanced oral bioavailability. ACS Infectious Diseases. 2020;6(5):802-810

Written By

Asher John Mohan, Bhaskar Kumar Gupta and Silviya Sarah Lal

Submitted: 28 November 2022 Reviewed: 19 December 2022 Published: 26 January 2023