Potential Promises and Perils of Human Biological Treatments for Immunotherapy in Veterinary Oncology
Abstract
:Simple Summary
Abstract
1. Introduction
2. Tumor Immunotherapy: General Principles
3. Human Tumor Antigen Vaccination
4. Humanized Monoclonal Antibodies
5. Single-Domain Antibodies (sdAbs)
5.1. The Advantage of Being Small
5.2. Towards Clinical Applications
6. In the Future
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Klingemann, H. Immunotherapy for Dogs: Still Running behind Humans. Front. Immunol. 2021, 12, 665784. [Google Scholar] [CrossRef] [PubMed]
- Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef] [PubMed]
- Prehn, R.T.; Main, J.M. Immunity to methylcholanthrene-induced sarcomas. J. Natl. Cancer Inst. 1957, 18, 769–778. [Google Scholar]
- Segal, E.M. Immunotherapy in the frontline management of advanced and metastatic NSCLC. Am. J. Manag. Care 2021, 27, S323–S332. [Google Scholar] [CrossRef] [PubMed]
- Coulie, P.G.; Van den Eynde, B.J.; van der Bruggen, P.; Boon, T. Tumour antigens recognized by T lymphocytes: At the core of cancer immunotherapy. Nat. Rev. Cancer 2014, 14, 135–146. [Google Scholar] [CrossRef]
- Caballero, O.L.; Chen, Y.T. Cancer/testis (CT) antigens: Potential targets for immunotherapy. Cancer Sci. 2009, 100, 2014–2021. [Google Scholar] [CrossRef] [PubMed]
- Bright, R.K.; Bright, J.D.; Byrne, J.A. Overexpressed oncogenic tumor-self antigens. Hum. Vaccines Immunother. 2014, 10, 3297–3305. [Google Scholar] [CrossRef]
- Schumacher, T.N.; Scheper, W.; Kvistborg, P. Cancer Neoantigens. Annu. Rev. Immunol. 2018, 37, 173–200. [Google Scholar] [CrossRef]
- Bollineni, R.C.; Tran, T.T.; Lund-Johansen, F.; Olweus, J. Chasing neoantigens; invite naïve T cells to the party. Curr. Opin. Immunol. 2022, 75, 102172. [Google Scholar] [CrossRef]
- Linette, G.P.; Becker-Hapak, M.; Skidmore, Z.L.; Baroja, M.L.; Xu, C.; Hundal, J.; Spencer, D.H.; Fu, W.; Cummins, C.; Robnett, M.; et al. Immunological ignorance is an enabling feature of the oligo-clonal T cell response to melanoma neoantigens. Proc. Natl. Acad. Sci. USA 2019, 116, 23662–23670. [Google Scholar] [CrossRef]
- Ott, P.A.; Hu, Z.; Keskin, D.B.; Shukla, S.A.; Sun, J.; Bozym, D.J.; Zhang, W.; Luoma, A.; Giobbie-Hurder, A.; Peter, L.; et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 2017, 547, 217–221. [Google Scholar] [CrossRef] [PubMed]
- Smyth, M.J.; Thia, K.Y.T.; Street, S.E.A.; MacGregor, D.; Godfrey, D.I.; Trapani, J.A. Perforin-Mediated Cytotoxicity Is Critical for Surveillance of Spontaneous Lymphoma. J. Exp. Med. 2000, 192, 755–760. [Google Scholar] [CrossRef] [PubMed]
- Shankaran, V.; Ikeda, H.; Bruce, A.T.; White, J.M.; Swanson, P.E.; Old, L.J.; Schreiber, R.D. IFNγ and lymphocytes prevent primary tumour development and shape tumour immunogenicity. Nature 2001, 410, 1107–1111. [Google Scholar] [CrossRef] [PubMed]
- Janeway, C.A., Jr.; Bottomly, K. Signals and signs for lymphocyte responses. Cell 1994, 76, 275–285. [Google Scholar] [CrossRef]
- Jiang, Y.; Li, Y.; Zhu, B. T-cell exhaustion in the tumor microenvironment. Cell Death Dis. 2015, 6, e1792. [Google Scholar] [CrossRef]
- Zebley, C.C.; Youngblood, B. Mechanisms of T cell exhaustion guiding next-generation immunotherapy. Trends Cancer 2022, 8, 726–734. [Google Scholar] [CrossRef]
- Liu, J.; Fu, M.; Wang, M.; Wan, D.; Wei, Y.; Wei, X. Cancer vaccines as promising immuno-therapeutics: Platforms and current progress. J. Hematol. Oncol. 2022, 15, 28. [Google Scholar] [CrossRef]
- Schuler, G.; Schuler-Thurner, B.; Steinman, R.M. The use of dendritic cells in cancer immunotherapy. Curr. Opin. Immunol. 2003, 15, 138–147. [Google Scholar] [CrossRef]
- Sun, Z.; Fu, Y.X.; Peng, H. Targeting tumor cells with antibodies enhances anti-tumor immunity. Biophys. Rep. 2018, 4, 243–253. [Google Scholar] [CrossRef]
- Sterner, R.C.; Sterner, R.M. CAR-T cell therapy: Current limitations and potential strategies. Blood Cancer J. 2021, 11, 69. [Google Scholar] [CrossRef]
- Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [PubMed]
- Maeng, H.M.; Berzofsky, J.A. Strategies for developing and optimizing cancer vaccines. F1000Research 2019, 8, 654. [Google Scholar] [CrossRef] [PubMed]
- Hernandez, B.; Adissu, H.A.; Wei, B.-R.; Michael, H.T.; Merlino, G.; Simpson, R.M. Naturally Occurring Canine Melanoma as a Predictive Comparative Oncology Model for Human Mucosal and Other Triple Wild-Type Melanomas. Int. J. Mol. Sci. 2018, 19, 394. [Google Scholar] [CrossRef] [PubMed]
- Bergman, P.J.; McKnight, J.; Novosad, A.; Charney, S.; Farrelly, J.; Craft, D.; Wulderk, M.; Jeffers, Y.; Sadelain, M.; Hohenhaus, A.E.; et al. Long-Term Survival of Dogs with Advanced Malignant Melanoma after DNA Vaccination with Xenogeneic Human Tyrosinase: A Phase I Trial1. Clin. Cancer Res. 2003, 9, 1284–1290. [Google Scholar] [PubMed]
- Liao, J.C.; Gregor, P.; Wolchok, J.D.; Orlandi, F.; Craft, D.; Leung, C.; Houghton, A.N.; Bergman, P.J. Vaccination with human tyrosinase DNA induces antibody responses in dogs with advanced melanoma. Cancer Immun. 2006, 6, 8. [Google Scholar]
- Bergman, P.J.; Camps-Palau, M.A.; McKnight, J.A.; Leibman, N.F.; Craft, D.M.; Leung, C.; Liao, J.; Riviere, I.; Sadelain, M.; Hohenhaus, A.E.; et al. Development of a xenogeneic DNA vaccine program for canine malignant melanoma at the Animal Medical Center. Vaccine 2006, 24, 4582–4585. [Google Scholar] [CrossRef]
- Grosenbaugh, D.A.; Leard, A.T.; Bergman, P.J.; Klein, M.K.; Meleo, K.; Susaneck, S.; Hess, P.R.; Jankowski, M.K.; Jones, P.D.; Leibman, N.F.; et al. Safety and efficacy of a xenogeneic DNA vaccine encoding for human tyrosinase as adjunctive treatment for oral malignant melanoma in dogs following surgical excision of the primary tumor. Am. J. Vet. Res. 2011, 72, 1631–1638. [Google Scholar] [CrossRef]
- McLean, J.L.; Lobetti, R.G. Use of the melanoma vaccine in 38 dogs: The South African experience. J. S. Afr. Vet. Assoc. 2015, 86, 1246. [Google Scholar] [CrossRef]
- Ottnod, J.M.; Smedley, R.C.; Walshaw, R.; Hauptman, J.G.; Kiupel, M.; Obradovich, J.E. A retrospective analysis of the efficacy of Oncept vaccine for the adjunct treatment of canine oral malignant melanoma. Vet. Comp. Oncol. 2013, 11, 219–229. [Google Scholar] [CrossRef]
- Treggiari, E.; Grant, J.P.; North, S.M. A retrospective review of outcome and survival following surgery and adjuvant xenogeneic DNA vaccination in 32 dogs with oral malignant melanoma. J. Vet. Med. Sci. 2016, 78, 845–850. [Google Scholar] [CrossRef]
- Verganti, S.; Berlato, D.; Blackwood, L.; Amores-Fuster, I.; Polton, G.A.; Elders, R.; Doyle, R.; Taylor, A.; Murphy, S. Use of Oncept melanoma vaccine in 69 canine oral malignant melanomas in the UK. J. Small Anim. Pract. 2017, 58, 10–16. [Google Scholar] [CrossRef] [PubMed]
- Turek, M.; LaDue, T.; Looper, J.; Nagata, K.; Shiomitsu, K.; Keyerleber, M.; Buchholz, J.; Gieger, T.; Hetzel, S. Multimodality treatment including ONCEPT for canine oral melanoma: A retrospective analysis of 131 dogs. Vet. Radiol. Ultrasound 2020, 61, 471–480. [Google Scholar] [CrossRef] [PubMed]
- Berry, A.; Hayes, A.; Schiavo, L.; Dobson, J. Multimodal Treatment of a Canine Lingual Melanoma Using a Combination of Immunotherapy and a Tyrosine Kinase Inhibitors. Vet. Sci. 2022, 9, 54. [Google Scholar] [CrossRef] [PubMed]
- Ilieva, K.M.; Cheung, A.; Mele, S.; Chiaruttini, G.; Crescioli, S.; Griffin, M.; Nakamura, M.; Spicer, J.F.; Tsoka, S.; Lacy, K.E.; et al. Chondroitin Sulfate Proteoglycan 4 and Its Potential as an Antibody Immunotherapy Target across Different Tumor Types. Front. Immunol. 2018, 8, 01911. [Google Scholar] [CrossRef] [PubMed]
- Riccardo, F.; Iussich, S.; Maniscalco, L.; Lorda Mayayo, S.; La Rosa, G.; Arigoni, M.; De Maria, R.; Gattino, F.; Lanzardo, S.; Lardone, E.; et al. CSPG4-Specific Immunity and Survival Prolongation in Dogs with Oral Malignant Melanoma Immunized with Human CSPG4 DNA. Clin. Cancer Res. 2014, 20, 3753–3762. [Google Scholar] [CrossRef]
- Piras, L.A.; Riccardo, F.; Iussich, S.; Maniscalco, L.; Gattino, F.; Martano, M.; Morello, E.; Lorda Mayayo, S.; Rolih, V.; Garavaglia, F.; et al. Prolongation of survival of dogs with oral malignant melanoma treated by en bloc surgical resection and adjuvant CSPG4-antigen electrovaccination. Vet. Comp. Oncol. 2017, 15, 996–1013. [Google Scholar] [CrossRef]
- Giacobino, D.; Camerino, M.; Riccardo, F.; Cavallo, F.; Tarone, L.; Martano, M.; Dentini, A.; Iussich, S.; Lardone, E.; Franci, P.; et al. Difference in outcome between curative intent vs marginal excision as a first treatment in dogs with oral malignant melanoma and the impact of adjuvant CSPG4-DNA electrovaccination: A retrospective study on 155 cases. Vet. Comp. Oncol. 2021, 19, 651–660. [Google Scholar] [CrossRef]
- Sardesai, N.Y.; Weiner, D.B. Electroporation delivery of DNA vaccines: Prospects for success. Curr. Opin. Immunol. 2011, 23, 421–429. [Google Scholar] [CrossRef]
- Chulpanova, D.S.; Solovyeva, V.V.; Kitaeva, K.V.; Dunham, S.P.; Khaiboullina, S.F.; Rizvanov, A.A. Recombinant Viruses for Cancer Therapy. Biomedicines 2018, 6, 94. [Google Scholar] [CrossRef]
- Oladejo, M.; Paterson, Y.; Wood, L.M. Clinical Experience and Recent Advances in the Development of Listeria-Based Tumor Immunotherapies. Front. Immunol. 2021, 12, 642316. [Google Scholar] [CrossRef]
- Wolf, B.J.; Princiotta, M.F. Processing of Recombinant Listeria monocytogenes Proteins for MHC Class I Presentation Follows a Dedicated, High-Efficiency Pathway. J. Immunol. 2013, 190, 2501–2509. [Google Scholar] [CrossRef] [PubMed]
- Villanueva, M.S.; Sijts, A.J.; Pamer, E.G. Listeriolysin is processed efficiently into an MHC class I-associated epitope in Listeria monocytogenes-infected cells. J. Immunol. 1995, 155, 5227–5233. [Google Scholar] [CrossRef] [PubMed]
- Mason, N.J.; Gnanandarajah, J.S.; Engiles, J.B.; Gray, F.; Laughlin, D.; Gaurnier-Hausser, A.; Wallecha, A.; Huebner, M.; Paterson, Y. Immunotherapy with a HER2-Targeting Listeria Induces HER2-Specific Immunity and Demonstrates Potential Therapeutic Effects in a Phase I Trial in Canine Osteosarcoma. Clin. Cancer Res. 2016, 22, 4380–4390. [Google Scholar] [CrossRef]
- Ehrhart, N.P.; Ryan, S.D.; Fan, T.M. 24—Tumors of the Skeletal System. In Withrow and MacEwen’s Small Animal Clinical Oncology, 5th ed.; Withrow, S.J., Vail, D.M., Page, R.L., Eds.; W.B. Saunders: Saint Louis, MO, USA, 2013; pp. 463–503. [Google Scholar] [CrossRef]
- Brady, J.V.; Troyer, R.M.; Ramsey, S.A.; Leeper, H.; Yang, L.; Maier, C.S.; Goodall, C.P.; Ruby, C.E.; Albarqi, H.A.M.; Taratula, O.; et al. A Preliminary Proteomic Investigation of Circulating Exosomes and Discovery of Biomarkers Associated with the Progression of Osteosarcoma in a Clinical Model of Spontaneous Disease. Transl. Oncol. 2018, 11, 1137–1146. [Google Scholar] [CrossRef] [PubMed]
- Troyer, R.M.; Ruby, C.E.; Goodall, C.P.; Yang, L.; Maier, C.S.; Albarqi, H.A.; Brady, J.V.; Bathke, K.; Taratula, O.; Mourich, D.; et al. Exosomes from Osteosarcoma and normal osteoblast differ in proteomic cargo and immunomodulatory effects on T cells. Exp. Cell Res. 2017, 358, 369–376. [Google Scholar] [CrossRef] [PubMed]
- Wycislo, K.L.; Fan, T.M. The Immunotherapy of Canine Osteosarcoma: A Historical and Systematic Review. J. Vet. Intern. Med. 2015, 29, 759–769. [Google Scholar] [CrossRef]
- Wilk, S.S.; Zabielska-Koczywąs, K.A. Molecular Mechanisms of Canine Osteosarcoma Metastasis. Int. J. Mol. Sci. 2021, 22, 3639. [Google Scholar] [CrossRef]
- Selmic, L.E.; Burton, J.H.; Thamm, D.H.; Withrow, S.J.; Lana, S.E. Comparison of Carboplatin and Doxorubicin-Based Chemotherapy Protocols in 470 Dogs after Amputation for Treatment of Appendicular Osteosarcoma. J. Vet. Intern. Med. 2014, 28, 554–563. [Google Scholar] [CrossRef]
- Angstadt, A.Y.; Motsinger-Reif, A.; Thomas, R.; Kisseberth, W.C.; Guillermo Couto, C.; Duval, D.L.; Nielsen, D.M.; Modiano, J.F.; Breen, M. Characterization of canine osteosarcoma by array comparative genomic hybridization and RT-qPCR: Signatures of genomic imbalance in canine osteosarcoma parallel the human counterpart. Genes Chromosomes Cancer 2011, 50, 859–874. [Google Scholar] [CrossRef]
- Fossey, S.L.; Liao, A.T.; McCleese, J.K.; Bear, M.D.; Lin, J.; Li, P.-K.; Kisseberth, W.C.; London, C.A. Characterization of STAT3 activation and expression in canine and human osteosarcoma. BMC Cancer 2009, 9, 81. [Google Scholar] [CrossRef]
- Gardner, H.L.; Sivaprakasam, K.; Briones, N.; Zismann, V.; Perdigones, N.; Drenner, K.; Facista, S.; Richholt, R.; Liang, W.; Aldrich, J.; et al. Canine osteosarcoma genome sequencing identifies recurrent mutations in DMD and the histone methyltransferase gene SETD2. Commun. Biol. 2019, 2, 266. [Google Scholar] [CrossRef] [PubMed]
- Marley, K.; Bracha, S.; Seguin, B. Osteoprotegerin activates osteosarcoma cells that co-express RANK and RANKL. Exp. Cell Res. 2015, 338, 32–38. [Google Scholar] [CrossRef] [PubMed]
- Paoloni, M.; Davis, S.; Lana, S.; Withrow, S.; Sangiorgi, L.; Picci, P.; Hewitt, S.; Triche, T.; Meltzer, P.; Khanna, C. Canine tumor cross-species genomics uncovers targets linked to osteosarcoma progression. BMC Genom. 2009, 10, 625. [Google Scholar] [CrossRef] [PubMed]
- Rowell, J.L.; McCarthy, D.O.; Alvarez, C.E. Dog models of naturally occurring cancer. Trends Mol. Med. 2011, 17, 380–388. [Google Scholar] [CrossRef]
- Flint, A.F.; U’Ren, L.; Legare, M.E.; Withrow, S.J.; Dernell, W.; Hanneman, W.H. Overexpression of the erbB-2 Proto-oncogene in Canine Osteosarcoma Cell Lines and Tumors. Vet. Pathol. 2004, 41, 291–296. [Google Scholar] [CrossRef]
- Shahabi, V.; Seavey, M.M.; Maciag, P.C.; Rivera, S.; Wallecha, A. Development of a live and highly attenuated Listeria monocytogenes-based vaccine for the treatment of Her2/neu-overexpressing cancers in human. Cancer Gene Ther. 2011, 18, 53–62. [Google Scholar] [CrossRef]
- Musser, M.L.; Berger, E.P.; Tripp, C.D.; Clifford, C.A.; Bergman, P.J.; Johannes, C.M. Safety evaluation of the canine osteosarcoma vaccine, live Listeria vector. Vet. Comp. Oncol. 2021, 19, 92–98. [Google Scholar] [CrossRef]
- Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef]
- Tawbi, H.A.; Schadendorf, D.; Lipson, E.J.; Ascierto, P.A.; Matamala, L.; Castillo Gutiérrez, E.; Rutkowski, P.; Gogas, H.J.; Lao, C.D.; De Menezes, J.J.; et al. Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma. N. Engl. J. Med. 2022, 386, 24–34. [Google Scholar] [CrossRef]
- Ma, W.; Xue, R.; Zhu, Z.; Farrukh, H.; Song, W.; Li, T.; Zheng, L.; Pan, C.-x. Increasing cure rates of solid tumors by immune checkpoint inhibitors. Exp. Hematol. Oncol. 2023, 12, 10. [Google Scholar] [CrossRef]
- Sharma, P.; Allison, J.P. Dissecting the mechanisms of immune checkpoint therapy. Nat. Rev. Immunol. 2020, 20, 75–76. [Google Scholar] [CrossRef] [PubMed]
- Maekawa, N.; Konnai, S.; Ikebuchi, R.; Okagawa, T.; Adachi, M.; Takagi, S.; Kagawa, Y.; Nakajima, C.; Suzuki, Y.; Murata, S.; et al. Expression of PD-L1 on canine tumor cells and enhancement of IFN-γ production from tumor-infiltrating cells by PD-L1 blockade. PLoS ONE 2014, 9, e98415. [Google Scholar] [CrossRef] [PubMed]
- Stevenson, V.B.; Perry, S.N.; Todd, M.; Huckle, W.R.; LeRoith, T. PD-1, PD-L1, and PD-L2 Gene Expression and Tumor Infiltrating Lymphocytes in Canine Melanoma. Vet. Pathol. 2021, 58, 692–698. [Google Scholar] [CrossRef] [PubMed]
- Coy, J.; Caldwell, A.; Chow, L.; Guth, A.; Dow, S. PD-1 expression by canine T cells and functional effects of PD-1 blockade. Vet. Comp. Oncol. 2017, 15, 1487–1502. [Google Scholar] [CrossRef] [PubMed]
- Hartley, G.; Elmslie, R.; Dow, S.; Guth, A. Checkpoint molecule expression by B and T cell lymphomas in dogs. Vet. Comp. Oncol. 2018, 16, 352–360. [Google Scholar] [CrossRef] [PubMed]
- Choi, J.W.; Withers, S.S.; Chang, H.; Spanier, J.A.; De La Trinidad, V.L.; Panesar, H.; Fife, B.T.; Sciammas, R.; Sparger, E.E.; Moore, P.F.; et al. Development of canine PD-1/PD-L1 specific monoclonal antibodies and amplification of canine T cell function. PLoS ONE 2020, 15, e0235518. [Google Scholar] [CrossRef] [PubMed]
- Nemoto, Y.; Shosu, K.; Okuda, M.; Noguchi, S.; Mizuno, T. Development and characterization of monoclonal antibodies against canine PD-1 and PD-L1. Vet. Immunol. Immunopathol. 2018, 198, 19–25. [Google Scholar] [CrossRef]
- Hartley, G.; Faulhaber, E.; Caldwell, A.; Coy, J.; Kurihara, J.; Guth, A.; Regan, D.; Dow, S. Immune regulation of canine tumour and macrophage PD-L1 expression. Vet. Comp. Oncol. 2017, 15, 534–549. [Google Scholar] [CrossRef]
- Mason, N.J.; Chester, N.; Xiong, A.; Rotolo, A.; Wu, Y.; Yoshimoto, S.; Glassman, P.; Gulendran, G.; Siegel, D.L. Development of a fully canine anti-canine CTLA4 monoclonal antibody for comparative translational research in dogs with spontaneous tumors. mAbs 2021, 13, 2004638. [Google Scholar] [CrossRef]
- Pantelyushin, S.; Ranninger, E.; Guerrera, D.; Hutter, G.; Maake, C.; Markkanen, E.; Bettschart-Wolfensberger, R.; Rohrer Bley, C.; Läubli, H.; vom Berg, J. Cross-Reactivity and Functionality of Approved Human Immune Checkpoint Blockers in Dogs. Cancers 2021, 13, 785. [Google Scholar] [CrossRef]
- Nascimento, C.; Urbano, A.C.; Gameiro, A.; Ferreira, J.; Correia, J.; Ferreira, F. Serum PD-1/PD-L1 Levels, Tumor Expression and PD-L1 Somatic Mutations in HER2-Positive and Triple Negative Normal-Like Feline Mammary Carcinoma Subtypes. Cancers 2020, 12, 1386. [Google Scholar] [CrossRef] [PubMed]
- Wagner, K.S.; Stickings, P.; White, J.M.; Neal, S.; Crowcroft, N.S.; Sesardic, D.; Efstratiou, A. A review of the international issues surrounding the availability and demand for diphtheria antitoxin for therapeutic use. Vaccine 2009, 28, 14–20. [Google Scholar] [CrossRef] [PubMed]
- Silverstein, A.M. Clemens Freiherr von Pirquet: Explaining immune complex disease in 1906. Nat. Immunol. 2000, 1, 453–455. [Google Scholar] [CrossRef] [PubMed]
- Smith, S.L. Ten years of Orthoclone OKT3 (muromonab-CD3): A review. J. Transpl. Coord. Off. Publ. N. Am. Transpl. Coord. Organ. 1996, 6, 109–119, quiz 120–121. [Google Scholar] [CrossRef]
- Sgro, C. Side-effects of a monoclonal antibody, muromonab CD3/orthoclone OKT3: Bibliographic review. Toxicology 1995, 105, 23–29. [Google Scholar] [CrossRef] [PubMed]
- Kimball, J.A.; Norman, D.J.; Shield, C.F.; Schroeder, T.J.; Lisi, P.; Garovoy, M.; O’Connell, J.B.; Stuart, F.; McDiarmid, S.V.; Wall, W. The OKT3 antibody response study: A multicentre study of human anti-mouse antibody (HAMA) production following OKT3 use in solid organ tranplantation. Transpl. Immunol. 1995, 3, 212–221. [Google Scholar] [CrossRef]
- Ducourau, E.; Mulleman, D.; Paintaud, G.; Miow Lin, D.C.; Lauféron, F.; Ternant, D.; Watier, H.; Goupille, P. Antibodies toward infliximab are associated with low infliximab concentration at treatment initiation and poor infliximab maintenance in rheumatic diseases. Arthritis Res. Ther. 2011, 13, R105. [Google Scholar] [CrossRef]
- van den Bemt, B.J.F.; den Broeder, A.A.; Wolbink, G.J.; Hekster, Y.A.; van Riel, P.L.C.M.; Benraad, B.; van den Hoogen, F.H.J. Anti-infliximab antibodies are already detectable in most patients with rheumatoid arthritis halfway through an infusioncycle: An open-label pharmacokinetic cohort study. BMC Musculoskelet. Disord. 2011, 12, 12. [Google Scholar] [CrossRef]
- Baert, F.; Noman, M.; Vermeire, S.; Van Assche, G.; D’Haens, G.; Carbonez, A.; Rutgeerts, P. Influence of Immunogenicity on the Long-Term Efficacy of Infliximab in Crohn’s Disease. N. Engl. J. Med. 2003, 348, 601–608. [Google Scholar] [CrossRef]
- Haraoui, B.; Cameron, L.; Ouellet, M.; White, B. Anti-infliximab antibodies in patients with rheumatoid arthritis who require higher doses of infliximab to achieve or maintain a clinical response. J. Rheumatol. 2006, 33, 31–36. [Google Scholar]
- Bou-Assaly, W.; Mukherji, S. Cetuximab (erbitux). AJNR. Am. J. Neuroradiol. 2010, 31, 626–627. [Google Scholar] [CrossRef] [PubMed]
- O’Neil, B.H.; Allen, R.; Spigel, D.R.; Stinchcombe, T.E.; Moore, D.T.; Berlin, J.D.; Goldberg, R.M. High Incidence of Cetuximab-Related Infusion Reactions in Tennessee and North Carolina and the Association with Atopic History. J. Clin. Oncol. 2007, 25, 3644–3648. [Google Scholar] [CrossRef] [PubMed]
- Chung, C.H.; Mirakhur, B.; Chan, E.; Le, Q.-T.; Berlin, J.; Morse, M.; Murphy, B.A.; Satinover, S.M.; Hosen, J.; Mauro, D.; et al. Cetuximab-induced anaphylaxis and IgE specific for galactose-alpha-1,3-galactose. N. Engl. J. Med. 2008, 358, 1109–1117. [Google Scholar] [CrossRef]
- Carson, A.S.; Gardner, A.; Iweala, O.I. Where’s the Beef? Understanding Allergic Responses to Red Meat in Alpha-Gal Syndrome. J. Immunol. 2022, 208, 267–277. [Google Scholar] [CrossRef] [PubMed]
- Vaisman-Mentesh, A.; Gutierrez-Gonzalez, M.; DeKosky, B.J.; Wine, Y. The Molecular Mechanisms That Underlie the Immune Biology of Anti-drug Antibody Formation Following Treatment with Monoclonal Antibodies. Front. Immunol. 2020, 11, 01951. [Google Scholar] [CrossRef]
- Jeglum, K.A. Chemoimmunotherapy of Canine Lymphoma with Adjuvant Canine Monoclonal Antibody 231. Vet. Clin. N. Am. Small Anim. Pract. 1996, 26, 73–85. [Google Scholar] [CrossRef]
- Beirão, B.C.B.; Raposo, T.; Jain, S.; Hupp, T.; Argyle, D.J. Challenges and opportunities for monoclonal antibody therapy in veterinary oncology. Vet. J. 2016, 218, 40–50. [Google Scholar] [CrossRef]
- Hwang, W.Y.; Foote, J. Immunogenicity of engineered antibodies. Methods 2005, 36, 3–10. [Google Scholar] [CrossRef]
- Michels, G.M.; Walsh, K.F.; Kryda, K.A.; Mahabir, S.P.; Walters, R.R.; Hoevers, J.D.; Martinon, O.M. A blinded, randomized, placebo-controlled trial of the safety of lokivetmab (ZTS-00103289), a caninized anti-canine IL-31 monoclonal antibody in client-owned dogs with atopic dermatitis. Vet. Dermatol. 2016, 27, 505-e136. [Google Scholar] [CrossRef]
- Van Brussel, L.; Moyaert, H.; Escalada, M.; Mahabir, S.P.; Stegemann, M.R. A masked, randomised clinical trial evaluating the efficacy and safety of lokivetmab compared to saline control in client-owned dogs with allergic dermatitis. Vet. Dermatol. 2021, 32, 477-e131. [Google Scholar] [CrossRef]
- Souza, C.P.; Rosychuk, R.A.W.; Contreras, E.T.; Schissler, J.R.; Simpson, A.C. A retrospective analysis of the use of lokivetmab in the management of allergic pruritus in a referral population of 135 dogs in the western USA. Vet. Dermatol. 2018, 29, 489-e164. [Google Scholar] [CrossRef] [PubMed]
- Gearing, D.P.; Huebner, M.; Virtue, E.R.; Knight, K.; Hansen, P.; Lascelles, B.D.X.; Gearing, R.P.; Drew, A.C. In Vitro and In Vivo Characterization of a Fully Felinized Therapeutic Anti-Nerve Growth Factor Monoclonal Antibody for the Treatment of Pain in Cats. J. Vet. Intern. Med. 2016, 30, 1129–1137. [Google Scholar] [CrossRef] [PubMed]
- Lascelles, B.D.X.; Knazovicky, D.; Case, B.; Freire, M.; Innes, J.F.; Drew, A.C.; Gearing, D.P. A canine-specific anti-nerve growth factor antibody alleviates pain and improves mobility and function in dogs with degenerative joint disease-associated pain. BMC Vet. Res. 2015, 11, 101. [Google Scholar] [CrossRef] [PubMed]
- Gearing, D.P.; Virtue, E.R.; Gearing, R.P.; Drew, A.C. A fully caninised anti-NGF monoclonal antibody for pain relief in dogs. BMC Vet. Res. 2013, 9, 226. [Google Scholar] [CrossRef] [PubMed]
- Igase, M.; Nemoto, Y.; Itamoto, K.; Tani, K.; Nakaichi, M.; Sakurai, M.; Sakai, Y.; Noguchi, S.; Kato, M.; Tsukui, T.; et al. A pilot clinical study of the therapeutic antibody against canine PD-1 for advanced spontaneous cancers in dogs. Sci. Rep. 2020, 10, 18311. [Google Scholar] [CrossRef]
- Maekawa, N.; Konnai, S.; Takagi, S.; Kagawa, Y.; Okagawa, T.; Nishimori, A.; Ikebuchi, R.; Izumi, Y.; Deguchi, T.; Nakajima, C.; et al. A canine chimeric monoclonal antibody targeting PD-L1 and its clinical efficacy in canine oral malignant melanoma or undifferentiated sarcoma. Sci. Rep. 2017, 7, 8951. [Google Scholar] [CrossRef]
- Maekawa, N.; Konnai, S.; Asano, Y.; Sajiki, Y.; Deguchi, T.; Okagawa, T.; Watari, K.; Takeuchi, H.; Takagi, S.; Hosoya, K.; et al. Exploration of serum biomarkers in dogs with malignant melanoma receiving anti-PD-L1 therapy and potential of COX-2 inhibition for combination therapy. Sci. Rep. 2022, 12, 9265. [Google Scholar] [CrossRef]
- Maekawa, N.; Konnai, S.; Nishimura, M.; Kagawa, Y.; Takagi, S.; Hosoya, K.; Ohta, H.; Kim, S.; Okagawa, T.; Izumi, Y.; et al. PD-L1 immunohistochemistry for canine cancers and clinical benefit of anti-PD-L1 antibody in dogs with pulmonary metastatic oral malignant melanoma. npj Precis. Oncol. 2021, 5, 10. [Google Scholar] [CrossRef]
- Hamers-Casterman, C.; Atarhouch, T.; Muyldermans, S.; Robinson, G.; Hammers, C.; Songa, E.B.; Bendahman, N.; Hammers, R. Naturally occurring antibodies devoid of light chains. Nature 1993, 363, 446–448. [Google Scholar] [CrossRef]
- Greenberg, A.S.; Avila, D.; Hughes, M.; Hughes, A.; McKinney, E.C.; Flajnik, M.F. A new antigen receptor gene family that undergoes rearrangement and extensive somatic diversification in sharks. Nature 1995, 374, 168–173. [Google Scholar] [CrossRef]
- Bathula, N.V.; Bommadevara, H.; Hayes, J.M. Nanobodies: The Future of Antibody-Based Immune Therapeutics. Cancer Biother. Radiopharm. 2021, 36, 109–122. [Google Scholar] [CrossRef] [PubMed]
- Muyldermans, S. Applications of Nanobodies. Annu. Rev. Anim. Biosci. 2021, 9, 401–421. [Google Scholar] [CrossRef] [PubMed]
- Saga, T.; Neumann, R.D.; Heya, T.; Sato, J.; Kinuya, S.; Le, N.; Paik, C.H.; Weinstein, J.N. Targeting cancer micrometastases with monoclonal antibodies: A binding-site barrier. Proc. Natl. Acad. Sci. USA 1995, 92, 8999–9003. [Google Scholar] [CrossRef] [PubMed]
- Juweid, M.; Neumann, R.; Paik, C.; Perez-Bacete, M.J.; Sato, J.; van Osdol, W.; Weinstein, J.N. Micropharmacology of Monoclonal Antibodies in Solid Tumors: Direct Experimental Evidence for a Binding Site Barrier. Cancer Res. 1992, 52, 5144–5153. [Google Scholar] [PubMed]
- Cruz, E.; Kayser, V. Monoclonal antibody therapy of solid tumors: Clinical limitations and novel strategies to enhance treatment efficacy. Biologics 2019, 13, 33–51. [Google Scholar] [CrossRef]
- Thurber, G.M.; Zajic, S.C.; Wittrup, K.D. Theoretic Criteria for Antibody Penetration into Solid Tumors and Micrometastases. J. Nucl. Med. 2007, 48, 995–999. [Google Scholar] [CrossRef]
- Arbabi Ghahroudi, M.; Desmyter, A.; Wyns, L.; Hamers, R.; Muyldermans, S. Selection and identification of single domain antibody fragments from camel heavy-chain antibodies. FEBS Lett. 1997, 414, 521–526. [Google Scholar] [CrossRef]
- Debie, P.; Lafont, C.; Defrise, M.; Hansen, I.; van Willigen, D.M.; van Leeuwen, F.W.B.; Gijsbers, R.; D’Huyvetter, M.; Devoogdt, N.; Lahoutte, T.; et al. Size and affinity kinetics of nanobodies influence targeting and penetration of solid tumours. J. Control. Release 2020, 317, 34–42. [Google Scholar] [CrossRef]
- Bannas, P.; Lenz, A.; Kunick, V.; Well, L.; Fumey, W.; Rissiek, B.; Haag, F.; Schmid, J.; Schütze, K.; Eichhoff, A.; et al. Molecular imaging of tumors with nanobodies and antibodies: Timing and dosage are crucial factors for improved in vivo detection. Contrast Media Mol. Imaging 2015, 10, 367–378. [Google Scholar] [CrossRef]
- Beltrán Hernández, I.; Rompen, R.; Rossin, R.; Xenaki, K.T.; Katrukha, E.A.; Nicolay, K.; van Bergen en Henegouwen, P.; Grüll, H.; Oliveira, S. Imaging of Tumor Spheroids, Dual-Isotope SPECT, and Autoradiographic Analysis to Assess the Tumor Uptake and Distribution of Different Nanobodies. Mol. Imaging Biol. 2019, 21, 1079–1088. [Google Scholar] [CrossRef]
- Bordeau, B.M.; Yang, Y.; Balthasar, J.P. Transient Competitive Inhibition Bypasses the Binding Site Barrier to Improve Tumor Penetration of Trastuzumab and Enhance T-DM1 Efficacy. Cancer Res. 2021, 81, 4145–4154. [Google Scholar] [CrossRef] [PubMed]
- Rossotti, M.A.; Bélanger, K.; Henry, K.A.; Tanha, J. Immunogenicity and humanization of single-domain antibodies. FEBS J. 2021, 289, 4304–4327. [Google Scholar] [CrossRef] [PubMed]
- Ackaert, C.; Smiejkowska, N.; Xavier, C.; Sterckx, Y.G.J.; Denies, S.; Stijlemans, B.; Elkrim, Y.; Devoogdt, N.; Caveliers, V.; Lahoutte, T.; et al. Immunogenicity Risk Profile of Nanobodies. Front. Immunol. 2021, 12, 632687. [Google Scholar] [CrossRef] [PubMed]
- Holland, M.C.; Wurthner, J.U.; Morley, P.J.; Birchler, M.A.; Lambert, J.; Albayaty, M.; Serone, A.P.; Wilson, R.; Chen, Y.; Forrest, R.M.; et al. Autoantibodies to Variable Heavy (VH) Chain Ig Sequences in Humans Impact the Safety and Clinical Pharmacology of a VH Domain Antibody Antagonist of TNF-α Receptor 1. J. Clin. Immunol. 2013, 33, 1192–1203. [Google Scholar] [CrossRef]
- Papadopoulos, K.P.; Isaacs, R.; Bilic, S.; Kentsch, K.; Huet, H.A.; Hofmann, M.; Rasco, D.; Kundamal, N.; Tang, Z.; Cooksey, J.; et al. Unexpected hepatotoxicity in a phase I study of TAS266, a novel tetravalent agonistic Nanobody® targeting the DR5 receptor. Cancer Chemother. Pharmacol. 2015, 75, 887–895. [Google Scholar] [CrossRef]
- Arbabi-Ghahroudi, M. Camelid Single-Domain Antibodies: Promises and Challenges as Lifesaving Treatments. Int. J. Mol. Sci. 2022, 23, 5009. [Google Scholar] [CrossRef]
- Jovčevska, I.; Muyldermans, S. The Therapeutic Potential of Nanobodies. BioDrugs 2020, 34, 11–26. [Google Scholar] [CrossRef]
- Wang, J.; Kang, G.; Yuan, H.; Cao, X.; Huang, H.; de Marco, A. Research Progress and Applications of Multivalent, Multispecific and Modified Nanobodies for Disease Treatment. Front. Immunol. 2022, 12, 838082. [Google Scholar] [CrossRef]
- Scully, M.; Cataland, S.R.; Peyvandi, F.; Coppo, P.; Knöbl, P.; Kremer Hovinga, J.A.; Metjian, A.; de la Rubia, J.; Pavenski, K.; Callewaert, F.; et al. Caplacizumab Treatment for Acquired Thrombotic Thrombocytopenic Purpura. N. Engl. J. Med. 2019, 380, 335–346. [Google Scholar] [CrossRef]
- Bartunek, J.; Barbato, E.; Heyndrickx, G.; Vanderheyden, M.; Wijns, W.; Holz, J.-B. Novel Antiplatelet Agents: ALX-0081, a Nanobody Directed towards von Willebrand Factor. J. Cardiovasc. Transl. Res. 2013, 6, 355–363. [Google Scholar] [CrossRef]
- Ishiwatari-Ogata, C.; Kyuuma, M.; Ogata, H.; Yamakawa, M.; Iwata, K.; Ochi, M.; Hori, M.; Miyata, N.; Fujii, Y. Ozoralizumab, a Humanized Anti-TNFα NANOBODY® Compound, Exhibits Efficacy Not Only at the Onset of Arthritis in a Human TNF Transgenic Mouse but Also during Secondary Failure of Administration of an Anti-TNFα IgG. Front. Immunol. 2022, 13, 853008. [Google Scholar] [CrossRef] [PubMed]
- Papp, K.A.; Weinberg, M.A.; Morris, A.; Reich, K. IL17A/F nanobody sonelokimab in patients with plaque psoriasis: A multicentre, randomised, placebo-controlled, phase 2b study. Lancet 2021, 397, 1564–1575. [Google Scholar] [CrossRef] [PubMed]
- Van Roy, M.; Ververken, C.; Beirnaert, E.; Hoefman, S.; Kolkman, J.; Vierboom, M.; Breedveld, E.; ‘t Hart, B.; Poelmans, S.; Bontinck, L.; et al. The preclinical pharmacology of the high affinity anti-IL-6R Nanobody® ALX-0061 supports its clinical development in rheumatoid arthritis. Arthritis Res. Ther. 2015, 17, 135. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.; Liu, Y.; Zhao, Z.; Li, Y.; Mustafa, B.; Chen, Z.; Barve, A.; Jain, A.; Yao, X.; Li, G.; et al. Discovery of Anti-PD-L1 Human Domain Antibodies for Cancer Immunotherapy. Front. Immunol. 2022, 13, 838966. [Google Scholar] [CrossRef]
- Zhang, F.; Wei, H.; Wang, X.; Bai, Y.; Wang, P.; Wu, J.; Jiang, X.; Wang, Y.; Cai, H.; Xu, T.; et al. Structural basis of a novel PD-L1 nanobody for immune checkpoint blockade. Cell Discov. 2017, 3, 17004. [Google Scholar] [CrossRef]
- Broos, K.; Lecocq, Q.; Xavier, C.; Bridoux, J.; Nguyen, T.T.; Corthals, J.; Schoonooghe, S.; Lion, E.; Raes, G.; Keyaerts, M.; et al. Evaluating a Single Domain Antibody Targeting Human PD-L1 as a Nuclear Imaging and Therapeutic Agent. Cancers 2019, 11, 872. [Google Scholar] [CrossRef]
- Sun, Z.; Li, W.; Mellors, J.W.; Orentas, R.; Dimitrov, D.S. Construction of a Large Size Human Immunoglobulin Heavy Chain Variable (VH) Domain Library, Isolation and Characterization of Novel Human Antibody VH Domains Targeting PD-L1 and CD22. Front. Immunol. 2022, 13, 869825. [Google Scholar] [CrossRef]
- Papadopoulos, K.P.; Harb, W.; Peer, C.J.; Hua, Q.; Xu, S.; Lu, H.; Lu, N.; He, Y.; Xu, T.; Dong, R.; et al. First-in-Human Phase I Study of Envafolimab, a Novel Subcutaneous Single-Domain Anti-PD-L1 Antibody, in Patients with Advanced Solid Tumors. Oncologist 2021, 26, e1514–e1525. [Google Scholar] [CrossRef]
- Li, J.; Deng, Y.; Zhang, W.; Zhou, A.-P.; Guo, W.; Yang, J.; Yuan, Y.; Zhu, L.; Qin, S.; Xiang, S.; et al. Subcutaneous envafolimab monotherapy in patients with advanced defective mismatch repair/microsatellite instability high solid tumors. J. Hematol. Oncol. 2021, 14, 95. [Google Scholar] [CrossRef]
- Breen, M.; Modiano, J.F. Evolutionarily conserved cytogenetic changes in hematological malignancies of dogs and humans–man and his best friend share more than companionship. Chromosome Res. 2008, 16, 145–154. [Google Scholar] [CrossRef]
- Ito, D.; Frantz, A.M.; Modiano, J.F. Canine lymphoma as a comparative model for human non-Hodgkin lymphoma: Recent progress and applications. Vet. Immunol. Immunopathol. 2014, 159, 192–201. [Google Scholar] [CrossRef]
- Ponce, F.; Marchal, T.; Magnol, J.P.; Turinelli, V.; Ledieu, D.; Bonnefont, C.; Pastor, M.; Delignette, M.L.; Fournel-Fleury, C. A morphological study of 608 cases of canine malignant lymphoma in France with a focus on comparative similarities between canine and human lymphoma morphology. Vet. Pathol. 2010, 47, 414–433. [Google Scholar] [CrossRef] [PubMed]
- Ramsey, S.A.; Xu, T.; Goodall, C.; Rhodes, A.C.; Kashyap, A.; He, J.; Bracha, S. Cross-species analysis of the canine and human bladder cancer transcriptome and exome. Genes Chromosomes Cancer 2017, 56, 328–343. [Google Scholar] [CrossRef] [PubMed]
- Dhawan, D.; Hahn, N.M.; Ramos-Vara, J.A.; Knapp, D.W. Naturally-occurring canine invasive urothelial carcinoma harbors luminal and basal transcriptional subtypes found in human muscle invasive bladder cancer. PLoS Genet. 2018, 14, e1007571. [Google Scholar] [CrossRef] [PubMed]
- Knapp, D.W.; Dhawan, D.; Ramos-Vara, J.A.; Ratliff, T.L.; Cresswell, G.M.; Utturkar, S.; Sommer, B.C.; Fulkerson, C.M.; Hahn, N.M. Naturally-Occurring Invasive Urothelial Carcinoma in Dogs, a Unique Model to Drive Advances in Managing Muscle Invasive Bladder Cancer in Humans. Front. Oncol. 2020, 9, 01493. [Google Scholar] [CrossRef]
- Knapp, D.W.; Ramos-Vara, J.A.; Moore, G.E.; Dhawan, D.; Bonney, P.L.; Young, K.E. Urinary Bladder Cancer in Dogs, a Naturally Occurring Model for Cancer Biology and Drug Development. ILAR J. 2014, 55, 100–118. [Google Scholar] [CrossRef]
- Caliari, D.; Zappulli, V.; Rasotto, R.; Cardazzo, B.; Frassineti, F.; Goldschmidt, M.H.; Castagnaro, M. Triple-negative vimentin-positive heterogeneous feline mammary carcinomas as a potential comparative model for breast cancer. BMC Vet. Res. 2014, 10, 185. [Google Scholar] [CrossRef]
- De Maria, R.; Olivero, M.; Iussich, S.; Nakaichi, M.; Murata, T.; Biolatti, B.; Di Renzo, M.F. Spontaneous Feline Mammary Carcinoma Is a Model of HER2 Overexpressing Poor Prognosis Human Breast Cancer. Cancer Res. 2005, 65, 907–912. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Hambly, J.N.; Ruby, C.E.; Mourich, D.V.; Bracha, S.; Dolan, B.P. Potential Promises and Perils of Human Biological Treatments for Immunotherapy in Veterinary Oncology. Vet. Sci. 2023, 10, 336. https://doi.org/10.3390/vetsci10050336
Hambly JN, Ruby CE, Mourich DV, Bracha S, Dolan BP. Potential Promises and Perils of Human Biological Treatments for Immunotherapy in Veterinary Oncology. Veterinary Sciences. 2023; 10(5):336. https://doi.org/10.3390/vetsci10050336
Chicago/Turabian StyleHambly, Jeilene N., Carl E. Ruby, Dan V. Mourich, Shay Bracha, and Brian P. Dolan. 2023. "Potential Promises and Perils of Human Biological Treatments for Immunotherapy in Veterinary Oncology" Veterinary Sciences 10, no. 5: 336. https://doi.org/10.3390/vetsci10050336
APA StyleHambly, J. N., Ruby, C. E., Mourich, D. V., Bracha, S., & Dolan, B. P. (2023). Potential Promises and Perils of Human Biological Treatments for Immunotherapy in Veterinary Oncology. Veterinary Sciences, 10(5), 336. https://doi.org/10.3390/vetsci10050336