How to Select the Right Method of Lymphodepletion for your Preclinical Study

by Aliyah Weinstein, PhD | Published: November 14th, 2024

 

Key TakeawaysKey Takeaways

  • Lymphodepletion methods include genetic approaches (e.g., Rag2 Knockout, B cell-deficient strains) and non-genetic methods (e.g., chemotherapy, radiation, antibody-mediated depletion).
  • Chemotherapy and radiation target lymphoid cells; antibody-based methods allow for temporal control and can be applied to various mouse strains.
  • Rag2-deficient and Jh mice lack specific immune cells, aiding in studying immune responses and disease mechanisms.

 

Lymphodepleted mouse models play an important role in characterizing the immune cells required for particular types of immune responses, such as in the context of disease pathogenesis, vaccination, immuno-oncology, and immunotherapy. Models of lymphodepletion include genetic lymphodepletion using commercially available mouse strains such as Rag2 Knockout (KO) and B cell-deficient strains, as well as non-genetic methods including antibody-mediated depletion, chemotherapy, and radiation. Each method has unique use cases and translational relevance.

Non-Genetic Lymphodepletion

Chemotherapy and Radiation

Both chemotherapy and radiation are used clinically and preclinically for lymphodepletion1. Chemotherapy or total body irradiation at low doses are referred to as non-myeloablative regimens, as they lead to reversible myelosuppression but do target peripheral lymphoid cells, for example, to eliminate host T cells in the context of CAR-T therapy2. In the preclinical setting, immuno-oncology studies have demonstrated that lymphodepletion prior to adoptive T cell transfer and/or dendritic cell-based vaccination can lead to the rejection or stabilization of an established murine melanoma3–5. Myeloablative doses of chemotherapy or radiation are also used in the context of immunotherapy; this is used when whole bone marrow or hematopoietic stem cell transplant is the interventional therapy. Chemotherapy and radiation may be the most translationally relevant methods of lymphodepletion (or myeloablation) because they are both being used clinically in immuno-oncology and transplant settings. However, their lack of specificity and potential harsh effects can be experimental concerns.

Antibody-Based Lymphodepletion

The use of antibody-based lymphodepletion allows for temporal control of lymphodepletion, as well as for normal immune development in the mice prior to the lymphodepletion step. A major advantage of this method over genetic lymphodepletion is that it can be used with any background strain of mouse. Genetically engineered and immunodeficient mouse models are commonly used in immuno-oncology studies, so the ability to lymphodeplete on top of other targeted mutations can provide robust data in this field. For example, a recent publication from Afkhami et al. (discussed in more depth below) used antibody-based depletion of T cells in B cell-deficient Jh mice to temporally control T cell depletion following vaccination for SARS-CoV-26.

  Never Miss an Insight

Sign up to receive the latest scientific news & insights covering all areas of preclinical in vivo research.

There is translational relevance of antibody-based lymphodepletion, as well. There are many antibody-based lymphodepletion schemas in preclinical or early phase clinical trials, largely in the area of transplant biology and hematopoietic stem cell transplant7. For clinical immuno-oncology applications, trials have been ongoing to evaluate the use of antibody-based lymphodepletion over traditional chemotherapy/radiation leading up to adoptive T cell transfer8. Recent preclinical studies have evaluated the impact of radiolabeled anti-CD45 antibodies as a targeted alternative to total body irradiation, also as preparation for adoptive T cell therapy or CAR-T therapy9. Therefore, antibody-based lymphodepletion is not just an experimental intervention but also part of potential therapeutic approaches.

Genetic Lymphodepletion

Knockouts: Rag2 and Rag2/IL2rg

Rag2-deficient mice contain a mutation in the Rag2 gene that inactivates the production of the Rag2 protein – a key enzyme involved in V(D)J recombination. Because of the disruption in this process, mature B and T cells cannot form in these mice. Rag2-deficient animals are available on both the B6 and BALB/c backgrounds, making these mice versatile tools for a variety of immunologic studies.

Rag2 knockout (BALB/c) mice were used in a recent study to understand the immunosuppressive role of iNKT cells in arthritis, which is a disease driven by CD4+ T cells10. The Rag2 knockout background allowed for the adoptive transfer of autoreactive T cells in this model. Using a model of psoriatic arthritis induced by mannan injection, autoreactive CD4+ T cells were adoptively transferred into the Rag2 knockout mice with or without co-injection of iNKT cells. This tightly controlled system was used to demonstrate the specific immunosuppressive effect of iNKT cells on these autoreactive CD4+ T cells, providing more specific data than performing this study in an animal with a fully competent immune system.

Several studies have also used Rag2-deficient mice to study the influence of innate lymphoid cells (ILCs) versus lymphoid cells in gut barrier damage11,12. Rag2 deficiency stalls the development of T and B cells, but not ILCs. One study looked at specific bacterial populations in the gut microbiome that protect against barrier damage, and which immune cells are required for this protection. The E. coli isolate GDAR2-2 drives ILC3 expansion in the gut and protects against C. rodentium infection and colitis in Rag2-deficient (B6) animals, suggesting that this protection is driven by ILC3s.12 In another study, to test the hypothesis that IL-33 drives ILC-mediated protection against amebic colitis, Rag2 knockout (B6) mice were treated with recombinant hIL-33 followed by amebic challenge, and mice were still protected from colitis. When the Rag2/IL2rg double KO model, which is deficient T cells, B cells, NK cells, and ILCs, was used, protection was lost.11

Indeed, Taconic’s Rag2/IL2rg double knockout model is useful for elucidating not just the influence of ILCs, but also of NK cells, when compared to the Rag2 knockout. One study used the biologic differences between these two strains to show that host NK cells were required to completely protect MB49 bladder carcinoma-bearing mice from death in conjunction with adoptive cell transfer13.

Of note, many older studies use the scid mouse, which is also deficient in T and B cells. However, compared to the Rag2-deficient models, the scid mutation is “leaky,” meaning that some functional T and B cells will in fact develop in mice bearing this mutation. Another key limitation of the scid models is their radio- and chemo-sensitivity due to the role of Prkdc in DNA damage repair.

B Cell-Deficient Strains (Jh) 

Jh mice are also available on both the B6 and BALB/c backgrounds. These mice contain a deletion of all four JH gene segments in the Ig heavy chain locus. Because of this deletion, B cell development in these mice is drastically altered and the mice do not present with any IgM or IgG antibodies in the serum. These mice do exhibit normal T cell differentiation, and therefore this model can be used to distinguish between the influence of these two lymphoid subpopulations.

Your experimental goals will determine the best model of lymphodepletion for you to use. In a recent study evaluating the efficacy and mechanism of action of a novel COVID-19 vaccine, Afkhami et al. showed that vaccinated Jh (BALB/c) mice did not clear mouse-adapted SARS-CoV-2 from the lung compared to lymphoreplete animals6. This group also used a T cell antibody-depleting cocktail in this study, which was used to demonstrate the relative impact of B vs. T cells at different time points following vaccination. The combination of genetic and antibody-based approaches allowed the researchers to elucidate both the initiation of the vaccine response and the interaction between B and T cells. Using a Rag2-deficient animal genetically deficient in both B and T cells would not have allowed for the nuanced discoveries described in this publication. 

Conclusion

There are many ways to lymphodeplete an animal in preparation for your preclinical study. First, determining the questions that you hope to answer with your study will inform which method(s) should be selected as well as which background strain should be used. Not discussed here are super immunodeficient mice such as those in Taconic’s NOG portfolio. These mice not only lack lymphocytes but also have deficiencies in innate immune cells such as dendritic cells and macrophages and therefore go beyond the scope of lymphodepletion discussed herein.


References:

  1. Anasetti C, Mulé JJ. To ablate or not to ablate? HSCs in the T cell driver’s seat. J Clin Invest. 2007;117(2):306-310. doi:10.1172/jci30973
  2. Nissani A, Lev-Ari S, Meirson T, Jacoby E, Asher N, Ben-Betzalel G, Itzhaki O, Shapira-Frommer R, Schachter J, Markel G, Besser MJ. Comparison of non-myeloablative lymphodepleting preconditioning regimens in patients undergoing adoptive T cell therapy. J Immunother Cancer. 2021;9(5):e001743. doi:10.1136/jitc-2020-001743
  3. Koike N, Kuhn L, Pilon-Thomas SA, Mulé JJ. Adoptive Immunotherapy Combined With A Dendritic Cell-Based Vaccine Results in the Rejection of Established Tumor in A Murine Melanoma Model. J Immunother. 2005;28(6):614. doi:10.1097/01.cji.0000190953.25503.49
  4. Nelson MH, Bowers JS, Bailey SR, Diven MA, Fugle CW, Kaiser ADM, Wrzesinski C, Liu B, Restifo NP, Paulos CM. Toll-like receptor agonist therapy can profoundly augment the antitumor activity of adoptively transferred CD8+ T cells without host preconditioning. J Immunother Cancer. 2016;4(1):6. doi:10.1186/s40425-016-0110-8
  5. Gattinoni L, Finkelstein SE, Klebanoff CA, Antony PA, Palmer DC, Spiess PJ, Hwang LN, Yu Z, Wrzesinski C, Heimann DM, Surh CD, Rosenberg SA, Restifo NP. Removal of homeostatic cytokine sinks by lymphodepletion enhances the efficacy of adoptively transferred tumor-specific CD8+ T cells. J Exp Medicine. 2005;202(7):907-912. doi:10.1084/jem.20050732
  6. Afkhami S, D’Agostino MR, Zhang A, Stacey HD, Marzok A, Kang A, Singh R, Bavananthasivam J, Ye G, Luo X, Wang F, Ang JC, Zganiacz A, Sankar U, Kazhdan N, Koenig JFE, Phelps A, Gameiro SF, Tang S, Jordana M, Wan Y, Mossman KL, Jeyanathan M, Gillgrass A, Medina MFC, Smaill F, Lichty BD, Miller MS, Xing Z. Respiratory mucosal delivery of next-generation COVID-19 vaccine provides robust protection against both ancestral and variant strains of SARS-CoV-2. Cell. 2022;185(5):896-915.e19. doi:10.1016/j.cell.2022.02.005
  7. Griffin JM, Healy FM, Dahal LN, Floisand Y, Woolley JF. Worked to the bone: antibody-based conditioning as the future of transplant biology. J Hematol Oncol. 2022;15(1):65. doi:10.1186/s13045-022-01284-6
  8. Louis CU, Straathof K, Bollard CM, Gerken C, Huls MH, Gresik MV, Wu MF, Weiss HL, Gee AP, Brenner MK, Rooney CM, Heslop HE, Gottschalk S. Enhancing the in vivo expansion of adoptively transferred EBV-specific CTL with lymphodepleting CD45 monoclonal antibodies in NPC patients. Blood. 2009;113(11):2442-2450. doi:10.1182/blood-2008-05-157222
  9. Dawicki W, Allen KJH, Garg R, Geoghegan EM, Berger MS, Ludwig DL, Dadachova E. Targeted lymphodepletion with a CD45-directed antibody radioconjugate as a novel conditioning regimen prior to adoptive cell therapy. Oncotarget. 2020;11(39):3571-3581. doi:10.18632/oncotarget.27731
  10. Zhao M, Svensson MND, Venken K, Chawla A, Liang S, Engel I, Mydel P, Day J, Elewaut D, Bottini N, Kronenberg M. Altered thymic differentiation and modulation of arthritis by invariant NKT cells expressing mutant ZAP70. Nat Commun. 2018;9(1):2627. doi:10.1038/s41467-018-05095-7
  11. Uddin MJ, Leslie JL, Burgess SL, Oakland N, Thompson B, Abhyankar M, Revilla J, Frisbee A, Donlan AN, Kumar P, Jr WAP. The IL-33-ILC2 pathway protects from amebic colitis. Mucosal Immunol. 2022;15(1):165-175. doi:10.1038/s41385-021-00442-2
  12. Wu WJH, Kim M, Chang LC, Assie A, Saldana-Morales FB, Zegarra-Ruiz DF, Norwood K, Samuel BS, Diehl GE. Interleukin-1β secretion induced by mucosa-associated gut commensal bacteria promotes intestinal barrier repair. Gut Microbes. 2022;14(1):2014772. doi:10.1080/19490976.2021.2014772
  13. Perez-Diez A, Joncker NT, Choi K, Chan WFN, Anderson CC, Lantz O, Matzinger P. CD4 cells can be more efficient at tumor rejection than CD8 cells. Blood. 2007;109(12):5346-5354. doi:10.1182/blood-2006-10-051318

Related Resources

Flyer

Jh Mouse

Antibody-deficient C57BL/6 and BALB/c mice for immunology and immuno-oncology research. The Jh mouse permits clinically-relevant dosing of Immunogenic test articles in syngeneic tumor model systems

service

White Paper

Genetically Engineered Models for Advanced Syngeneic Tumor Modeling

This white paper examines the GEMs available for syngeneic studies and highlights the benefits of each.

service

Get In Touch

Book a complimentary consultation

Taconic Biosciences' model generation team has produced about 5,000 models in the last 15 years, developing a globally-recognized reputation for advancing the work of in vivo researchers. Our scientific program managers are here to help you navigate the complexities of model generation.

If you need immediate assistance, please contact Customer Service:

Taconic Corporate Offices

Email: info@taconic.com

Phone: +1 (518) 697-3900

273 Hover Ave., Germantown, NY 12526

North American Customer Service

Email: info@taconic.com

Phone:
+1 (518) 697-3915 

Toll-free:
+1 (888) 822-6642


Hours:
(Monday - Friday): 7 a.m. - 6 p.m. ET

European Customer Service

Email: info@taconic.com

Phone (Europe and Denmark):
+45 70 23 04 05 

Phone (Germany):
+49 214 50 68 023 

Hours: (Monday - Friday):
7 a.m. - 5 p.m. CET