Camelid Single-Domain Antibodies As an Alternative to Overcome Challenges Related to the Prevention, Detection, and Control of Neglected Tropical Diseases

Due mainly to properties such as high affinity and antigen specificity, antibodies have become important tools for biomedical research, diagnosis, and treatment of several human diseases. When the objective is to administer them for therapy, strategies are used to reduce the heterologous protein immunogenicity and to improve pharmacokinetic and pharmacodynamic characteristics. Size minimization contributes to ameliorate these characteristics, while preserving the antigen–antibody interaction site. Since the discovery that camelids produce functional antibodies devoid of light chains, studies have proposed the use of single domains for biosensors, monitoring and treatment of tumors, therapies for inflammatory and neurodegenerative diseases, drug delivery, or passive immunotherapy. Despite an expected increase in antibody and related products in the pharmaceutical market over the next years, few research initiatives are related to the development of alternatives for helping to manage neglected tropical diseases (NTDs). In this review, we summarize developments of camelid single-domain antibodies (VHH) in the field of NTDs. Particular attention is given to VHH-derived products, i.e., VHHs fused to nanoparticles, constructed for the development of rapid diagnostic kits; fused to oligomeric matrix proteins for viral neutralization; and conjugated with proteins for the treatment of human parasites. Moreover, paratransgenesis technology using VHHs is an interesting approach to control parasite development in vectors. With enormous biotechnological versatility, facility and low cost for heterologous production, and greater ability to recognize different epitopes, VHHs have appeared as an opportunity to overcome challenges related to the prevention, detection, and control of human diseases, especially NTDs.

[1]  A. Frasch,et al.  Lactose derivatives are inhibitors of Trypanosoma cruzi trans-sialidase activity toward conventional substrates in vitro and in vivo. , 2004, Glycobiology.

[2]  John A Bilello,et al.  The agony and ecstasy of "OMIC" technologies in drug development. , 2005, Current molecular medicine.

[3]  G J Ebrahim,et al.  Neglected tropical diseases , 2005, BMJ : British Medical Journal.

[4]  P. De Baetselier,et al.  Expression and extracellular release of a functional anti-trypanosome Nanobody® in Sodalis glossinidius, a bacterial symbiont of the tsetse fly , 2012, Microbial Cell Factories.

[5]  W. Gibson Faculty Opinions recommendation of Efficient targeting of conserved cryptic epitopes of infectious agents by single domain antibodies. African trypanosomes as paradigm. , 2003 .

[6]  E. Remaut,et al.  Formatted anti-tumor necrosis factor alpha VHH proteins derived from camelids show superior potency and targeting to inflamed joints in a murine model of collagen-induced arthritis. , 2006, Arthritis and rheumatism.

[7]  M. Seman,et al.  Single domain antibodies: promising experimental and therapeutic tools in infection and immunity , 2009, Medical Microbiology and Immunology.

[8]  C. Milstein,et al.  Continuous cultures of fused cells secreting antibody of predefined specificity , 1975, Nature.

[9]  J. Mucci,et al.  The trans-Sialidase from Trypanosoma cruzi Induces Thrombocytopenia during Acute Chagas' Disease by Reducing the Platelet Sialic Acid Contents , 2005, Infection and Immunity.

[10]  P. Simarro,et al.  Epidemiology of human African trypanosomiasis , 2014, Clinical epidemiology.

[11]  R. Weiss,et al.  Llama Antibody Fragments Recognizing Various Epitopes of the CD4bs Neutralize a Broad Range of HIV-1 Subtypes A, B and C , 2012, PloS one.

[12]  Christopher D Spicer,et al.  Peer-reviewed version of the manuscript published in final form at Chemical Society Reviews (2018) Peptide and protein nanoparticle conjugates: versatile platforms for biomedical applications , 2018 .

[13]  A. Frasch,et al.  Functional diversity in the trans-sialidase and mucin families in Trypanosoma cruzi. , 2000, Parasitology today.

[14]  Michael D. McLean,et al.  In Vivo Neutralization of α-Cobratoxin with High-Affinity Llama Single-Domain Antibodies (VHHs) and a VHH-Fc Antibody , 2013, PloS one.

[15]  S. Van Gucht,et al.  Post-exposure Treatment with Anti-rabies VHH and Vaccine Significantly Improves Protection of Mice from Lethal Rabies Infection , 2016, PLoS neglected tropical diseases.

[16]  G. P. Smith,et al.  Filamentous fusion phage: novel expression vectors that display cloned antigens on the virion surface. , 1985, Science.

[17]  Newal K. Agnihotri Current Issues and Challenges , 2004 .

[18]  L. Wyns,et al.  Single‐domain antibody fragments with high conformational stability , 2002, Protein science : a publication of the Protein Society.

[19]  G. Chang,et al.  Development of a Humanized Antibody with High Therapeutic Potential against Dengue Virus Type 2 , 2012, PLoS neglected tropical diseases.

[20]  S. Muyldermans,et al.  Radiolabeled nanobodies as theranostic tools in targeted radionuclide therapy of cancer , 2014, Expert opinion on drug delivery.

[21]  Jiayu Liu,et al.  A GPC3-targeting Bispecific Antibody, GPC3-S-Fab, with Potent Cytotoxicity. , 2018, Journal of visualized experiments : JoVE.

[22]  Thomas Singer,et al.  The safety and side effects of monoclonal antibodies , 2010, Nature Reviews Drug Discovery.

[23]  John S. Brownstein,et al.  The global distribution and burden of dengue , 2013, Nature.

[24]  L. Wyns,et al.  Camelid immunoglobulins and nanobody technology. , 2009, Veterinary immunology and immunopathology.

[25]  S. Muyldermans,et al.  Emergence and evolution of functional heavy-chain antibodies in Camelidae. , 2003, Developmental and comparative immunology.

[26]  C. Morel,et al.  Neglected diseases: under‐funded research and inadequate health interventions , 2003, EMBO reports.

[27]  G. Leroux-Roels,et al.  Production, characterization and in vitro testing of HBcAg-specific VHH intrabodies. , 2010, The Journal of general virology.

[28]  L. Wyns,et al.  Efficient Targeting of Conserved Cryptic Epitopes of Infectious Agents by Single Domain Antibodies , 2004, Journal of Biological Chemistry.

[29]  R. Stábeli,et al.  Inhibition of the Myotoxicity Induced by Bothrops jararacussu Venom and Isolated Phospholipases A2 by Specific Camelid Single-Domain Antibody Fragments , 2016, PloS one.

[30]  S. Schenkman,et al.  Sera from chronic Chagasic patients and rodents infected with Trypanosoma cruzi inhibit trans-sialidase by recognizing its amino-terminal and catalytic domain , 1994, Infection and immunity.

[31]  R. O'Kennedy,et al.  Antibodies and antibody-derived analytical biosensors , 2016, Essays in biochemistry.

[32]  Rajendra Lingala,et al.  Generation and Characterization of an scFv Directed against Site II of Rabies Glycoprotein , 2011, Biotechnology research international.

[33]  G. Caljon,et al.  Delivery of a functional anti-trypanosome Nanobody in different tsetse fly tissues via a bacterial symbiont, Sodalis glossinidius , 2014, Microbial Cell Factories.

[34]  Serge Muyldermans,et al.  Nanobodies: natural single-domain antibodies. , 2013, Annual review of biochemistry.

[35]  S. Muyldermans,et al.  VHH, bivalent domains and chimeric Heavy chain-only antibodies with high neutralizing efficacy for scorpion toxin AahI'. , 2008, Molecular immunology.

[36]  G. Sanz,et al.  [Diagnosis, management and treatment of chronic Chagas' heart disease in areas where Trypanosoma cruzi infection is not endemic]. , 2008, Enfermedades infecciosas y microbiologia clinica.

[37]  R. Isturiz,et al.  Chagas Disease , 2021, Neglected Tropical Diseases.

[38]  W. Hol,et al.  Structures of a key interaction protein from the Trypanosoma brucei editosome in complex with single domain antibodies. , 2010, Journal of structural biology.

[39]  R K Jain,et al.  Physiological barriers to delivery of monoclonal antibodies and other macromolecules in tumors. , 1990, Cancer research.

[40]  S. Muyldermans,et al.  Nanobodies, a promising tool for species-specific diagnosis of Taenia solium cysticercosis. , 2009, International journal for parasitology.

[41]  P. Hotez,et al.  “Manifesto” for Advancing the Control and Elimination of Neglected Tropical Diseases , 2010, PLoS neglected tropical diseases.

[42]  S. Muyldermans,et al.  Nanobodies®: proficient tools in diagnostics , 2010, Expert review of molecular diagnostics.

[43]  S. Muyldermans,et al.  The specific variable domain of camel heavy-chain antibodies is encoded in the germline. , 1998, Journal of molecular biology.

[44]  L. Wyns,et al.  Selection and identification of single domain antibody fragments from camel heavy‐chain antibodies , 1997, FEBS letters.

[45]  L. Álvarez-Vallina,et al.  Antibody engineering: facing new challenges in cancer therapy , 2005, Acta Pharmacologica Sinica.

[46]  V. Labas,et al.  A one‐step exclusion‐binding procedure for the purification of functional heavy‐chain and mammalian‐type γ‐globulins from camelid sera , 2009, Biotechnology and applied biochemistry.

[47]  Justin K H Liu The history of monoclonal antibody development – Progress, remaining challenges and future innovations , 2014, Annals of medicine and surgery.

[48]  F. Ducancel,et al.  Molecular engineering of antibodies for therapeutic and diagnostic purposes , 2012, mAbs.

[49]  J. Scheller,et al.  ELPylated anti-human TNF therapeutic single-domain antibodies for prevention of lethal septic shock. , 2011, Plant biotechnology journal.

[50]  J. Kolkman,et al.  Nanobodies - from llamas to therapeutic proteins. , 2010, Drug discovery today. Technologies.

[51]  P. Kennedy,et al.  Human African trypanosomiasis of the CNS: current issues and challenges. , 2004, The Journal of clinical investigation.

[52]  M. Arbabi-Ghahroudi Camelid Single-Domain Antibodies: Historical Perspective and Future Outlook , 2017, Front. Immunol..

[53]  S. Muyldermans,et al.  Naturally occurring antibodies devoid of light chains , 1993, Nature.

[54]  S. Hussain,et al.  Description of a Nanobody-based Competitive Immunoassay to Detect Tsetse Fly Exposure , 2015, PLoS neglected tropical diseases.

[55]  L. Wyns,et al.  Recognition of antigens by single-domain antibody fragments: the superfluous luxury of paired domains. , 2001, Trends in biochemical sciences.

[56]  Y. Michotte,et al.  Using microdialysis to analyse the passage of monovalent nanobodies through the blood–brain barrier , 2012, British journal of pharmacology.

[57]  Janice M. Reichert,et al.  Antibodies to watch in 2017 , 2016, mAbs.

[58]  E. Goldman,et al.  Selection and Characterization of Anti-Dengue NS1 Single Domain Antibodies , 2018, Scientific Reports.

[59]  Romain Girod,et al.  Differential Susceptibilities of Aedes aegypti and Aedes albopictus from the Americas to Zika Virus , 2016, PLoS neglected tropical diseases.

[60]  D. Ecker,et al.  The therapeutic monoclonal antibody market , 2015, mAbs.

[61]  A. de Marco Biotechnological applications of recombinant single-domain antibody fragments , 2011, Microbial cell factories.

[62]  Chunlai Jiang,et al.  Single Domain Antibody Multimers Confer Protection against Rabies Infection , 2013, PloS one.

[63]  R. Stábeli,et al.  Novel Camelid Antibody Fragments Targeting Recombinant Nucleoprotein of Araucaria hantavirus: A Prototype for an Early Diagnosis of Hantavirus Pulmonary Syndrome , 2014, PloS one.

[64]  S. Muyldermans,et al.  Experimental therapy of African trypanosomiasis with a nanobody-conjugated human trypanolytic factor , 2006, Nature Medicine.

[65]  E. Clercq Hope and hype , 2006, Nature Medicine.

[66]  B. Devreese,et al.  Generation of a Nanobody Targeting the Paraflagellar Rod Protein of Trypanosomes , 2014, PloS one.

[67]  Vicki Sifniotis,et al.  The state‐of‐play and future of antibody therapeutics☆ , 2017, Advanced drug delivery reviews.

[68]  Jay P. Graham,et al.  Challenges and opportunities associated with neglected tropical disease and water, sanitation and hygiene intersectoral integration programs , 2015, BMC Public Health.

[69]  S. Muyldermans,et al.  A bispecific nanobody to provide full protection against lethal scorpion envenoming , 2010, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[70]  Michael S. Diamond,et al.  Molecular Insight into Dengue Virus Pathogenesis and Its Implications for Disease Control , 2015, Cell.

[71]  T. Baral Immunobiology of African Trypanosomes: Need of Alternative Interventions , 2010, Journal of biomedicine & biotechnology.

[72]  Lode Wyns,et al.  Crystal structure of a camel single-domain VH antibody fragment in complex with lysozyme , 1996, Nature Structural Biology.

[73]  A. Varki,et al.  Trans-sialidase from Trypanosoma cruzi catalyzes sialoside hydrolysis with retention of configuration. , 2000, Glycobiology.

[74]  A. Frasch,et al.  Relevance of the Diversity among Members of the Trypanosoma Cruzi Trans-Sialidase Family Analyzed with Camelids Single-Domain Antibodies , 2008, PloS one.

[75]  Gary Walsh,et al.  Biopharmaceutical benchmarks , 2000, Nature Biotechnology.

[76]  S. Muyldermans,et al.  Dual Beneficial Effect of Interloop Disulfide Bond for Single Domain Antibody Fragments* , 2011, The Journal of Biological Chemistry.

[77]  J. Coura Chagas disease: what is known and what is needed--a background article. , 2007, Memorias do Instituto Oswaldo Cruz.

[78]  A. Frasch,et al.  The trans‐sialidase from Trypanosoma cruzi triggers apoptosis by target cell sialylation , 2006, Cellular microbiology.

[79]  Gary Walsh,et al.  Biopharmaceutical benchmarks 2014 , 2014, Nature Biotechnology.

[80]  Ginés Sanz,et al.  Diagnóstico, manejo y tratamiento de la cardiopatía chagásica crónica en áreas donde la infección por Trypanosoma cruzi no es endémica , 2007 .

[81]  M. Z. Siddiqui Monoclonal Antibodies as Diagnostics; an Appraisal , 2010, Indian journal of pharmaceutical sciences.

[82]  F. Seiler,et al.  [Structure and function of immunoglobulins]. , 1982, Beitrage zu Infusionstherapie und klinische Ernahrung.

[83]  P. Vanlandschoot,et al.  Protective Effect of Different Anti-Rabies Virus VHH Constructs against Rabies Disease in Mice , 2014, PloS one.

[84]  Jaap Goudsmit,et al.  Novel Human Monoclonal Antibody Combination Effectively Neutralizing Natural Rabies Virus Variants and Individual In Vitro Escape Mutants , 2005, Journal of Virology.

[85]  R. Weiss,et al.  Llama-Derived Single Domain Antibodies to Build Multivalent, Superpotent and Broadened Neutralizing Anti-Viral Molecules , 2011, PloS one.

[86]  L. Cavacini,et al.  Structure and function of immunoglobulins. , 2010, The Journal of allergy and clinical immunology.

[87]  Serge Muyldermans,et al.  Efficient tumor targeting by single‐domain antibody fragments of camels , 2002, International journal of cancer.

[88]  A. Frasch,et al.  Trypanosoma cruzi surface mucins: host-dependent coat diversity , 2006, Nature Reviews Microbiology.

[89]  L. Wyns,et al.  Comparison of llama VH sequences from conventional and heavy chain antibodies. , 1997, Molecular immunology.

[90]  Jaideep P. Sundaram,et al.  Health Innovation Networks to Help Developing Countries Address Neglected Diseases , 2005, Science.

[91]  Yuesheng Li,et al.  Generation of a Novel Bacteriophage Library Displaying scFv Antibody Fragments from the Natural Buffalo Host to Identify Antigens from Adult Schistosoma japonicum for Diagnostic Development , 2015, PLoS neglected tropical diseases.

[92]  E. Stokstad Taming rabies. , 2017, Science.

[93]  W. Ye,et al.  Development of VHH Antibodies against Dengue Virus Type 2 NS1 and Comparison with Monoclonal Antibodies for Use in Immunological Diagnosis , 2014, PloS one.

[94]  A. Frasch,et al.  Mice infected with Trypanosoma cruzi produce antibodies against the enzymatic domain of trans-sialidase that inhibit its activity , 1994, Infection and immunity.

[95]  Michael P Barrett,et al.  The trypanosomiases , 2003, The Lancet.

[96]  A. Frasch,et al.  Antibodies inhibiting Trypanosoma cruzi trans-sialidase activity in sera from human infections. , 1994, The Journal of infectious diseases.