Commentary
Chimeric antigen receptor (CAR) T cells were first clinically tested in trials against metastatic solid tumors without therapeutic benefit; however, these efforts established the necessary domains that are required for a functional CAR T-cell. With the characterization of cells within the immune system, the expression of CD19 was found to dictate B cell lineage that remains present on normal and malignant B cells [1,2]. Having such profound tissue specificity, this identified CD19 served as a promising target for antigen-specific CAR T-cell development [3]. The first clinical success was in 2010 with an advanced follicular lymphoma patient who received anti-CD19 CAR T-cell treatment with a durable response reported at 39 weeks [4]. This was rapidly followed by a separate report of treating a refractory chronic lymphocytic leukemia (CLL) patient with an anti-CD19 CAR T-cell therapy [5].
Thus began the era of CD19 CAR T-cell therapy leading to several clinical trials and the FDA approval of Yescarta® (axicabtagene ciloleucel), Kymriah® (tisagenlecleucel), Tecartus® (brexucabtagene autoleucel), and Breyanzi® (lisocabtagene maraleucel). The approved indications for these CD19 CAR T-cell therapies were for relapsed/refractory (r/r) diseases, specifically B cell acute lymphoblastic leukemia (ALL), follicular lymphoma, diffused large B-cell lymphoma, and mantle cell lymphoma [6]. What is studiously absent is CLL, one of first B cell malignancies that was a target for CD19 CAR T-cells.
CLL is the most common adult leukemia in Western populations, and although CLL can be observed in young adulthood, the disease is mostly observed at an average age of 72, with incidence increasing with increasing age [7]. With the rising numbers of “baby boomers,” there may soon be an increase in CLL numbers [8]. Chronic lymphocytic leukemia (CLL) is a complex progressive disease that can minimally affect patients before progressing to a degree where therapy is required and finally advancing to an aggressive disease that demands therapeutic intervention [9]. The precise etiology of CLL remains to be definitively explained. Genetic factors appear to strongly affect the genesis of CLL as evidenced with familial linkages and a history of challenges to the immune system (e.g., viral infections, history of atopic conditions, autoimmune phenomena) [9-12]. There are also specific chemical exposures that are correlated with CLL (herbicide exposure and tobacco usage) [9,11].
CLL is characterized by an accumulation of mature but dysfunctional B lymphocytes [7,13]. CD19 participates with the B cell receptor (BCR) to maintain a survival signal for the B cell which also results in propagation of malignant B cells. Inherited susceptibility has been localized to genes that control B cell development [9,14], whereas noninherited CLL has mutations in several pathways that are associated with aging with the mutational status of the immunoglobulin heavy variable gene as a critical differentiator for disease characterization [9,13,14]. The signaling threshold of the BCR is modulated by CD19 with the BCR/CD19 complex initiating a protein kinase driven signaling cascade [15]. Both CD19 and the BCR interact with Bruton tyrosine kinase. Initial treatment options for CLL include BTK inhibitors, phosphatidylinositol 3-kinase inhibitors, and the B-cell lymphoma 2 inhibitors, alone or combined with CD20 monoclonal antibodies [16]; however, r/r disease can occur which requires more creative treatments. CAR T-cell therapy was originally pioneered to target CLL; however, robust effects were not noted, and CAR T-cell therapy was more successfully applied to other B cell malignancies like acute lymphoblastic leukemia with blazing successes of complete remissions in over 90% of cases [17-19]. Although the currently FDA approved CD19 targeted CAR T-cell therapies have been applied to CLL, durable antitumor responses have been reported in only 26% of patients [20].
As is the case with many biochemically critical systems, B cell proliferation and activation are modulated by other receptor pathways that are also associated with the BCR, the B-cell activating factor receptor (BAFF-R)/ BAFF cascade. We submit that BAFF-R is an ideal target for CAR T-cells, specifically since BAFF-R is consistently observed in CLL [21]. BAFF-R is essential for B lymphocyte development and mature B-cell survival as well as clonal progression of malignant CLL B cells [22]. Qin et al. reported the first BAFF-R targeting CAR T-cell therapy, showing the CAR T-cells were active against several cell lines with known BAFF-R expression [23] as well as in xenograft models with the same cell lines as well as in models to mimic CD19 antigen relapse [24]. This BAFF-R CAR design, which contains a CD8 transmembrane domain and the 4-1BB and CD3 zeta costimulatory domains, is currently under evaluation in a clinical trial (NCT05370430).
A new monoclonal antibody against BAFF-R has been generated that became the foundation for generation a novel BAFF-R CAR T-cell therapy that utilizes the CD28 transmembrane domain along with the CD28 and CD3 zeta costimulatory domains [25]. These MC10029 CAR T-cells have been rigorously evaluated in both in vitro and in vivo models of leukemia and lymphoma as well as in models to mimic CD19 relapse with MC10029 CAR T-cell antigen specific cytotoxicity and anti-tumor effects confirmed [25]. Aware of the literature cataloguing the limited success of CD19 CAR T-cells against CLL, we turned our attention to applying these BAFF-R targeting CAR T-cells to CLL. Showing antigen specific cytotoxicity was a critical confirmatory set of experiment; however, we were most interested in confirming activity in CLL patient derived cells. Because of reported concerns of insufficient T cells or ineffective CAR T cells in CLL patients, peripheral blood samples from CLL patients were collected and were the source for T cells that were advanced to MC10029 CAR T-cell generation and were also the source for CLL tumor cells. Luo et al. showed the efficacy of CLL patient-derived MC10029 CAR T-cells against autologous CLL tumor cells. This data and the additional data in the approved IND application are leading to a clinical trial at Mayo Clinic Florida (NCT06191887). The original BAFF-R targeting CAR T-cell design is currently in a clinical trial for patients with r/r B cell ALL (NCT04690595) or r/r B-NHL (NCT05370430). Recently, this group reported positive safety and anti-lymphoma activity in three patients with mantle cell lymphoma [26]. These encouraging results, which show that targeting BAFF-R with a CAR T-cell therapy is clinically safe and those that show efficacious and the positive results using CLL patient derived BAFF-R CAR T-cells in an autologous study, support BAFF-R is a solid target for a CAR T-cell therapeutic against CLL.
Since CD19 CAR T-cell therapy was the first-in-class for this type of immunotherapy, CD19 CAR T-cells are responsible for current expectations of durable responses and complete remissions [17-19], and with that, CD19 CAR T-cell therapy has also identified seemingly CAR T-cell specific adverse events (AE) and incidences of neurotoxicity, along the requisite AE treatment strategies. As more CAR T-cell therapies that target new antigens are being generated and approved for clinical application, we will widen and deepen our understanding of the positive and negative effects of CAR T-cells. For instance, with the recent availability of BCMA-targeting CAR T-cells, we are now seeing comparisons of immune effector cell-associated neurotoxicity syndrome (ICANS) for BCMA CAR T-cells that range from 18-20% compared to CD19 CAR T-cell that ranges of 21-60% [27]. These observations along with single-cell RNA sequencing data showing CD19 expression in mural cells may indicate an on-target mechanism for neurotoxicity [28]. Mural cells are part of the cellular network that maintains the integrity of the blood brain barrier (BBB) [29]. Disruption of the BBB may explain the edema, multifocal hemorrhage, and vascular disruption of the brain after CD19 CAR T-cell treatment [28,30]. Conversely, CD19 CAR T-cell therapy has been applied to patients with lymphoma having a secondary central nervous system involvement with favorable results that are similar to typical lymphoma patients [31,32]. This is opening the possibility of CD19 CAR T-cells treatment for primary central nervous system lymphoma (PCNSL), which currently has dismal prognosis [33].
Thus far, CD19 CAR T-cells have been the only option for B cell malignancies or other B cell pathologies and have been invaluable for many patients. With the development and clinical testing of CAR T-cells that target new antigens, we are adding options to the treatment algorithms. CLL patients are typically older patients with comorbidities common to the aging population, like diabetes, hypertension, and vascular disease [34,35]; these comorbidities can impact treatment plans. Caution should be exercised with single-mindedly applying CD19 CAR T-cell therapies when B cell malignancies and the associated patients are complex and diverse. The BAFF-R targeting CAR T-cell therapies that are entering clinical trials offer an alternative that may be ideal for specific r/r patients, like CLL patients, older patients, or patients with vascular concerns where ICANS would be problematic. Additionally, the application of CD19 CAR T-cells for lupus patients [36,37] is opening a new door for CAR T-cell application for diverse pathologies in which B cells play a role. This arena can and should be shared with BAFF-R CAR-T cells to provide additional options for patients.
References
2. Uckun FM, Jaszcz W, Ambrus JL, Fauci AS, Gajl-Peczalska K, Song CW, et al. Detailed Studies on Expression and Function of CD19 Surface Determinant by Using B43 Monoclonal Antibody and the Clinical Potential of Anti-CD19 Immunotoxins. Blood. 1988;71(1):13-29.
3. Kochenderfer JN, Feldman SA, Zhao Y, Xu H, Black MA, Morgan RA, et al. Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptor. J Immunother. 2009;32(7):689-702.
4. Kochenderfer JN, Wilson WH, Janik JE, Dudley ME, Stetler-Stevenson M, Feldman SA, et al. Eradication of B-lineage cells and regression of lymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19. Blood. 2010;116(20):4099-102.
5. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365(8):725-33.
6. Wang V, Gauthier M, Decot V, Reppel L, Bensoussan D. Systematic Review on CAR-T Cell Clinical Trials Up to 2022: Academic Center Input. Cancers (Basel). 2023;15(4):1003.
7. Kipps TJ, Stevenson FK, Wu CJ, Croce CM, Packham G, Wierda WG, et al. Chronic lymphocytic leukaemia. Nature Reviews Disease Primers. 2017;3(1):16096.
8. Hao T, Li-Talley M, Buck A, Chen W. An emerging trend of rapid increase of leukemia but not all cancers in the aging population in the United States. Sci Rep. 2019;9(1):12070.
9. Chiorazzi N, Chen SS, Rai KR. Chronic Lymphocytic Leukemia. Cold Spring Harb Perspect Med. 2021;11(2):a035220.
10. Goldin LR, Pfeiffer RM, Li X, Hemminki K. Familial risk of lymphoproliferative tumors in families of patients with chronic lymphocytic leukemia: results from the Swedish Family-Cancer Database. Blood. 2004;104(6):1850-4.
11. Hallek M, Shanafelt TD, Eichhorst B. Chronic lymphocytic leukaemia. The Lancet. 2018;391(10129):1524-37.
12. Vitale C, Boccellato E, Comba L, Jones R, Perutelli F, Griggio V, et al. Impact of Immune Parameters and Immune Dysfunctions on the Prognosis of Patients with Chronic Lymphocytic Leukemia. Cancers (Basel). 2021;13(15):3856.
13. Kharfan-Dabaja MA, Yassine F, Gadd ME, Qin H. Driving Out Chronic Lymphocytic Leukemia With CAR T Cells. Transplant Cell Ther. 2022;28(1):5-17.
14. Law PJ, Berndt SI, Speedy HE, Camp NJ, Sava GP, Skibola CF, et al. Genome-wide association analysis implicates dysregulation of immunity genes in chronic lymphocytic leukaemia. Nature Communications. 2017;8(1):14175.
15. Wang K, Wei G, Liu D. CD19: a biomarker for B cell development, lymphoma diagnosis and therapy. Experimental Hematology & Oncology. 2012;1(1):36.
16. Siddiqi T, Soumerai JD, Dorritie KA, Stephens DM, Riedell PA, Arnason J, et al. Phase 1 TRANSCEND CLL 004 study of lisocabtagene maraleucel in patients with relapsed/refractory CLL or SLL. Blood. 2022;139(12):1794-806.
17. Dourthe M-E, Rabian F, Yakouben K, Chevillon F, Cabannes-Hamy A, Méchinaud F, et al. Determinants of CD19-positive vs CD19-negative relapse after tisagenlecleucel for B-cell acute lymphoblastic leukemia. Leukemia. 2021;35(12):3383-93.
18. Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N Engl J Med. 2018;378(5):439-48.
19. Nie Y, Lu W, Chen D, Tu H, Guo Z, Zhou X, et al. Mechanisms underlying CD19-positive ALL relapse after anti-CD19 CAR T cell therapy and associated strategies. Biomark Res. 2020;8:18.
20. Fraietta JA, Lacey SF, Orlando EJ, Pruteanu-Malinici I, Gohil M, Lundh S, et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nature Medicine. 2018;24(5):563-71.
21. Rodig SJ, Shahsafaei A, Li B, Mackay CR, Dorfman DM. BAFF-R, the major B cell-activating factor receptor, is expressed on most mature B cells and B-cell lymphoproliferative disorders. Hum Pathol. 2005;36(10):1113-9.
22. Smulski CR, Eibel H. BAFF and BAFF-Receptor in B Cell Selection and Survival. Front Immunol. 2018;9:2285.
23. Qin H, Wei G, Sakamaki I, Dong Z, Cheng WA, Smith DL, et al. Novel BAFF-Receptor Antibody to Natively Folded Recombinant Protein Eliminates Drug-Resistant Human B-cell Malignancies In Vivo. Clin Cancer Res. 2018;24(5):1114-23.
24. Qin H, Dong Z, Wang X, Cheng WA, Wen F, Xue W, et al. CAR T cells targeting BAFF-R can overcome CD19 antigen loss in B cell malignancies. Science Translational Medicine. 2019;11(511):eaaw9414.
25. Luo Y, Qie Y, Gadd ME, Manna A, Rivera-Valentin R, To T, et al. Translational development of a novel BAFF-R CAR-T therapy targeting B-cell lymphoid malignancies. Cancer Immunol Immunother. 2023;72(12):4031-47.
26. Budde LE, Del Real MM, Baird JH, Chen L, Song JY, Kambhampati S, et al. Promising Safety and Anti-Lymphoma Efficacy of Autologous Pmb-CT01 (BAFFRCAR T Cell) Therapy in a First-in-Human Phase 1 Study. Blood. 2023;142(Supplement 1):221.
27. Velasco R, Mussetti A, Villagrán-García M, Sureda A. CAR T-cell-associated neurotoxicity in central nervous system hematologic disease: Is it still a concern? Front Neurol. 2023;14:1144414.
28. Parker KR, Migliorini D, Perkey E, Yost KE, Bhaduri A, Bagga P, et al. Single-Cell Analyses Identify Brain Mural Cells Expressing CD19 as Potential Off-Tumor Targets for CAR-T Immunotherapies. Cell. 2020;183(1):126-42.e17.
29. Armulik A, Genové G, Mäe M, Nisancioglu MH, Wallgard E, Niaudet C, et al. Pericytes regulate the blood–brain barrier. Nature. 2010;468(7323):557-61.
30. Gust J, Hay KA, Hanafi L-A, Li D, Myerson D, Gonzalez-Cuyar LF, et al. Endothelial Activation and Blood–Brain Barrier Disruption in Neurotoxicity after Adoptive Immunotherapy with CD19 CAR-T Cells. Cancer Discovery. 2017;7(12):1404-19.
31. Abramson JS, McGree B, Noyes S, Plummer S, Wong C, Chen Y-B, et al. Anti-CD19 CAR T Cells in CNS Diffuse Large-B-Cell Lymphoma. New England Journal of Medicine. 2017;377(8):783-4.
32. Frigault MJ, Dietrich J, Martinez-Lage M, Leick M, Choi BD, DeFilipp Z, et al. Tisagenlecleucel CAR T-cell therapy in secondary CNS lymphoma. Blood. 2019;134(11):860-6.
33. Miyao K, Yokota H, Sakemura RL. Is CD19-directed chimeric antigen receptor T cell therapy a smart strategy to combat central nervous system lymphoma? Front Oncol. 2022;12:1082235.
34. Curovic Rotbain E, Niemann CU, Rostgaard K, Da Cunha-Bang C, Hjalgrim H, Frederiksen H. Mapping Comorbidity in CLL: Impact on Prognostic Factors, Treatment Patterns and Causes of Death. Blood. 2019;134:4285.
35. Gordon MJ, Kaempf A, Sitlinger A, Shouse G, Mei M, Brander DM, et al. The Chronic Lymphocytic Leukemia Comorbidity Index (CLL-CI): A Three-Factor Comorbidity Model. Clin Cancer Res. 2021;27(17):4814-24.
36. Mougiakakos D, Krönke G, Völkl S, Kretschmann S, Aigner M, Kharboutli S, et al. CD19-Targeted CAR T Cells in Refractory Systemic Lupus Erythematosus. N Engl J Med. 2021;385(6):567-9.
37. Mackensen A, Müller F, Mougiakakos D, Böltz S, Wilhelm A, Aigner M, et al. Anti-CD19 CAR T cell therapy for refractory systemic lupus erythematosus. Nat Med. 2022;28(10):2124-32.