Skip to main content

PERSPECTIVE article

Front. Immunol., 22 November 2023
Sec. Cancer Immunity and Immunotherapy

Has the shortage of fludarabine altered the current paradigm of lymphodepletion in favor of bendamustine?

Dimitrios FilioglouDimitrios Filioglou1Muhammad Husnain,Muhammad Husnain2,3Sharad Khurana,Sharad Khurana2,3Richard J. Simpson,,&#x;Richard J. Simpson1,4,5†Emmanuel Katsanis,,,,*&#x;Emmanuel Katsanis1,2,3,4,6*†
  • 1Department of Pediatrics, University of Arizona, Tucson, AZ, United States
  • 2Department of Medicine, University of Arizona, Tucson, AZ, United States
  • 3The University of Arizona Cancer Center, Tucson, AZ, United States
  • 4Department of Immunobiology, University of Arizona, Tucson, AZ, United States
  • 5School of Nutritional Sciences and Wellness, University of Arizona, Tucson, AZ, United States
  • 6Department of Pathology, University of Arizona, Tucson, AZ, United States

The most common lymphodepletion regimen used prior to infusion of chimeric antigen receptor-T cells (CAR-T) is cyclophosphamide (CY) in combination with fludarabine (Flu) (CY-FLU). While cyclophosphamide (CY) possesses lymphotoxic effects, it concurrently preserves regulatory T cell activity, potentially affecting the efficacy of CAR-T cells. Moreover, the use of fludarabine (FLU) has been linked to neurotoxicity, which could complicate the early detection of immune effector cell-associated neurotoxicity syndrome (ICANS) observed in CAR-T cell therapy. Given the ongoing shortage of FLU, alternative lymphodepleting agents have become necessary. To date, only a limited number of studies have directly compared different lymphodepleting regimens, and most of these comparisons have been retrospective in nature. Herein, we review the current literature on lymphodepletion preceding CAR-T cell therapies for lymphoid hematologic malignancies, with a specific focus on the use of bendamustine (BEN). Recent evidence suggests that administering BEN before CAR-T cell infusion yields comparable efficacy, possibly with a more favorable toxicity profile when compared to CY-FLU. This warrants further investigation through randomized prospective studies.

Introduction

Chimeric antigen receptor-T cell (CAR-T) therapy has demonstrated remarkable efficacy in treating relapsed/refractory B-cell malignancies, including but not limited to diffuse large B-cell lymphoma (DLBCL) and B-cell acute lymphoblastic leukemia (B-ALL) (1). More recently the development of anti–B-cell maturation antigen (BCMA) CAR-T cell therapy, has shown promise in patients with multiple myeloma (MM) (2). Lymphodepletion is consistently applied prior to infusion of CAR-T cells to facilitate their expansion and persistence (3). The most common lymphodepleting regimen for commercially approved CD19-CAR-T cell therapy in young adults and children has been cyclophosphamide (CY) (500 mg/m2× 2 days) and fludarabine (FLU) (30 mg/m2 × 4 days) (CY-FLU) (4). Although several dosage alterations have been tested, the selection of this lymphodepletion approach has not undergone prospective investigation, raising the question of whether there may be more advantageous alternatives (57). Due to the extended shortage of FLU in the United States, numerous institutions, including ours, have turned to Bendamustine (BEN) as a viable alternative to CY-FLU, with promising results (8, 9).

Effects of cyclophosphamide on immune function: immunomodulatory, immunosuppressive, and lymphodepletion aspects

CY is an inactive prodrug that requires enzymatic and chemical activation by CYP450 enzymes in the liver. The resultant nitrogen mustard promotes the interstrand and intrastrand DNA crosslinking that account for its cytotoxic properties (10). CY is associated with a range of common adverse effects, including bone marrow suppression, gonadal toxicity, and carcinogenesis. Additionally, it can lead to more distinct complications such as hemorrhagic cystitis, cardiomyopathy and interstitial lung disease (10). CY is deactivated mainly by the isoform 1 of cellular aldehyde dehydrogenase (ALDH1), which is highly expressed in cells with high proliferative potential such as hematopoietic stem cells. Significantly, ALDH1 is minimally expressed in lymphocytes which provides a rationale for the use of CY as a lymphodepleting agent (11). Saida et al, investigated the use of CY in lymphodepletion and found that it eradicated lymphocytes efficiently; however, the percentage of regulatory T cells (Tregs) was significantly increased (12). Terao et al. indicated that an early rise in Tregs following Tisagenlecleucel infusion is linked to a reduced risk of cytokine release syndrome (CRS) and could potentially serve as a predictive marker. Nevertheless, despite this Treg increase, there was no discernible improvement in the overall survival (OS) of these patients (13). Indeed, it is widely recognized that Tregs suppress anti-tumor responses and have the capacity to inhibit CAR-T cell activity (14). As such, one of the intended advantages of lymphodepletion is the simultaneous reduction of all lymphocytes including Tregs (1520). CY given following hematopoietic cell transplantation (PT-CY) preserved Tregs during the early immune reconstitution period (21). The same group also reported that PT-CY led to rapid recovery of CD4+Foxp3+ T regulatory cells, which were later found to be resistant to CY via upregulation of ALDH (22). Similarly, in murine hematopoietic cell transplant models they confirmed that PT-CY preferentially recovered Tregs at day +21 while restraining the proliferation and the differentiation of alloreactive CD4+ conventional T-cells (23). While these effects ameliorate graft versus host disease (GvHD), they may simultaneously reduce the efficacy of graft versus leukemia (GvL) responses. In addition to its effects on lymphoid populations CY also influences myeloid subsets including myeloid derived suppressor cells (MDSCs), which are highly immunosuppressive and impeding their effects may improve the efficacy of adoptive immunotherapy (24). Mechanistically, MDSCs reduce the levels of l-arginine and l-tryptophan by expressing arginase-I and indoleamine 2,3-dioxygenase (IDO), thereby suppressing T cell function (25). Interestingly, the CY-derived MDSCs possessed immunosuppressive properties (15, 18). CY was reported to induce spleen colonization with an heterogenous myeloid population, including CD11b+Ly-6G+CD31+, which were found to inhibit proliferation of T cells (15, 24). In another study, conducted in tumor-bearing mice, CY was found to induce the production of CD11b+ Gr-1loLy6ChiCCR2hi monocytic MDSCs (M-MDSCs) that suppress antitumor CD4+ effector cells through the PD-1/PD-L1 axis (25). Taken together, while lymphodepletion with CY may attenuate the risk of CRS it may also dampen the anti-cancer effects of CAR-T due to its preservation of Tregs and induction of MDSC.

Effectiveness and safety of lymphodepletion with cyclophosphamide and fludarabine

The addition of Flu to CY appears to be beneficial, improving the expansion and the persistence of CAR-T-cells (6, 26). Moreover, FLU increased disease-free survival, rate and depth of response, OS and relapse-free survival (RFS) as well as the duration of B-cell aplasia compared to CY alone (6, 2729). In fact, as evidenced by Jiang et al., absence of FLU in lymphodepletion as well as high lactate dehydrogenase (LDH) levels and low platelet counts pre-lymphodepletion were associated with higher relapse after CAR-T-cell therapy (30). Fludarabine is a purine analog prodrug that is rapidly converted in plasma to F-araA and accumulates in cells where it is phosphorylated to its active metabolite F-araATP by deoxycytidine kinase, an enzyme that is highly expressed in human lymphocytes (31). F-araA has a half-life of approximately 20 h in vivo, and its clearance depends on adequate renal function (32). In fact, creatinine clearance [CrCl(est)] below 80 ml/min was found to correlate with time to toxicity, independently of age (33). The main dose-limiting toxicities of FLU are those of other chemotherapy agents, namely myelosuppression and risk of infection. However, neurotoxicity is also of concern especially in patients with advanced disease or elderly patients with reduced renal function (34). Thus, these patients require careful monitoring and might need appropriate dose reduction. Somnolence and peripheral neuropathy occur frequently, immediately following fludarabine infusion, but are reversible and non-specific (35). Of particular importance are the late-onset (20–250 days) neurological symptoms such as progressive visual disturbances, peripheral neuropathy, ataxia, hemiparesis and dementia (34). Attention should be given not to confuse the latter with the CAR-T-cell-related immune effector cell associated neurotoxicity syndrome (ICANS), which usually occurs within 1 week following therapy, and is often associated with CRS (34). This highlights the importance of careful consideration of the fludarabine concentration used for lymphodepletion in patients receiving CD19-specific CAR-T-cell therapy. Fabrizio et al. found that the use of suboptimal fludarabine concentration (Area Under the Curve - AUC <13.8 mg × h/L) for lymphodepletion in patients with relapse-refractory (R/R) B-ALL significantly increased the risk of disease relapse however they cautioned that this should be weighed against the risk of development of toxicities with higher AUC doses (36). Analogous results were reported by Dekker et al. with the leukemia-free survival (LFS) and the duration of B cell aplasia being shorter in the FLU-underexposed (37). Similarly, Scordo et al. found that patients with aggressive B-cell NHL who received commercial axicabtagene ciloleucel (Axi-cel) an optimal AUC was associated with a lower risk of relapse/progression, however, a high AUC was associated with an increase incidence of any-grade ICANS (38). Unfortunately, the current method of FLU-dosing administration based on linear body surface area (BSA) has been found to result in variable medication exposure (39). FLU levels are not readily available in most institutions making FLU pharmacokinetics impractical for optimizing lymphodepletion whilst reducing relapse and ICANs. The ongoing global shortage of fludarabine has compelled the exploration of alternative lymphodepleting regimens (8).

Introduction and assessment of bendamustine as a lymphodepletion strategy for CAR-T cell therapies

BEN is an alkylating agent that was designed to have both antimetabolite and alkylating properties (40). Apart from creating DNA interstrands, thus impairing DNA synthesis, it has also been found to induce apoptosis and inhibit mitotic checkpoints (41, 42). Another action that separates BEN from other alkylating agents is its activation of a base excision DNA repair pathway (42). It is metabolized by the liver and not excreted by the kidneys, which makes it ideal for patients with suppressed renal function. BEN has a short half-life of only 40 min, which is much shorter compared to that of CY and FLU (~ 8.88 h and ~ 10 h respectively) (32, 43, 44). Bone marrow suppression and particularly lymphopenia are frequently seen after BEN use, with CD4+ T cells being the most affected cell population (45). A significant body of literature has emerged recently, largely from the research efforts of our group, with a primary focus on the immunomodulatory properties of BEN (46). Our investigations have delved into BEN’s effects in the pre-allogeneic hematopoietic cell transplant (HCT) conditioning setting, and as a post-transplant therapy for GvHD, specifically targeting alloreactive T-cells and Tregs (4755). PT-BEN exhibits a higher degree of lymphodepletion, which includes the elimination of Tregs, in contrast to PT-CY, and thus may also offer a distinct advantage over CY in the setting of pre-CAR-T cell lymphodepletion. We have reported that the suppressive effect of BEN in GvHD is partially due to its effects in expanding the CD11b+Gr-1+ myeloid cells (52). However, despite the BEN-induced upregulation of MDSCs, which may raise concerns about its use in lymphodepletion, we have seen an advantage in its ability to preserve GvL effects (54).

Although enhanced lymphodepletion intensity with CY-FLU may be associated with higher probability of a favorable cytokine profile (LDH, MCP-1 and IL-7), increased CAR-T expansion as well as efficacy and durability of CAR-T cell therapy, it may also increase the frequency of severe CRS and ICANs (56, 57). In fact, a post-lymphodepletion upregulation of MCP-1 and IL-7 and decrease in LDH are associated with increased probability of complete remission (58, 59). In the JULIET2 trial, BEN was used in some patients at 90 mg/m2 daily for 2 days for lymphodepletion prior to Tisagenlecleucel administration in adult patients with relapsed or refractory DLBCL. No difference in efficacy or safety was observed between patients that received BEN compared to CY-FLU (60). In another study, Ramos et al. showed that the addition of FLU 30 mg/m2/day to BEN 70 mg/m2/day given for 3 days, in patients with R/R Hodgkin lymphoma, increased the homeostatic cytokines IL-7 and IL-15 and was associated with higher CD30 CAR-T persistence and longer 1 year disease-free survival compared to BEN 90mg/m2/day for 2 days alone (61). Moreover, they found that, CRS was significantly higher in CY- based lymphodepletion compared to their BEN- based regimen with or without FLU (61). Similar results favoring the use of BEN over CY-FLU were reported by Ghilardi et al, in patients with relapsed or refractory DLBCL that received Tisagenlecleucel (62). Specifically, FLU/CY significantly reduced lymphocyte counts at the time of CAR-T infusion and was associated with higher post-lymphodepletion CRP and ferritin levels. Patients treated with CY-FLU also exhibited decreased hemoglobin levels and platelet counts when compared to those receiving BEN, resulting in a higher requirement for platelet and packed red blood cell transfusions following tisagenlecleucel infusion. Furthermore, patients in the CY-FLU group exhibited lower median neutrophil counts when compared to BEN and more Grade ≥3 neutropenia requiring more frequent administration of granulocyte colony-stimulating factor (GCSF). Conversely, patients who underwent BEN-based lymphodepletion experienced fewer occurrences of neutropenic fever, infections, and hospitalizations. BEN was also associated with reduced rates of any-grade CRS, any-grade ICANs, as well as severe ICANS when contrasted with CY-FLU. These differences were also linked to the more frequent usage of tocilizumab in CY-FLU recipients. When patients were stratified into groups with similar characteristics, it was observed that BEN-based lymphodepletion exhibited comparable efficacy to CY-FLU in terms of overall response rate and progression-free survival (PFS). Moreover, it resulted in prolonged OS. The investigators also drew the conclusion that there exists an optimal threshold level for lymphodepletion, beyond which further reduction in lymphocyte counts does not enhance CAR-T efficacy but may elevate the risk of toxicities (62). In another recent study, Garcia-Calvo et al. compared different lymphodepleting regimens as bridging therapies before either Axicabtagene Ciloleucel (axi-cel) or Tisagenlecleucel infusion in patients with R/R DLBCL (63). No difference in efficacy or safety was observed between the BEN and non-BEN-containing regimens in either CD19 CAR-T cell regimen. The rates of ICANS and CRS, all grades or grade ≥3 were similar as was non-relapse mortality. Moreover, peak CAR-T-cell expansion after infusion and area under the curve between day of infusion and day 28 were similar in BEN and non-BEN subgroups. While BEN did not provide an additional benefit in median PFS and OS, patients in the BEN subgroups were found to have a trend toward an increased complete response to CAR-T-cell therapy (63). Ahmed et al. assessed the feasibility and the efficacy of outpatient Tisagenlecleucel administration in adults with B-cell lymphoma with BEN being the most common outpatient and CY-FLU the most common inpatient lymphodepletion used. They found that outpatient lymphodepletion was associated with lower any-grade CRS and ICANS versus the inpatient while PFS and OS were similar in the two groups (64). Similar findings were observed by Ong et al. in patients with R/R aggressive B-cell lymphoma treated with Axicabtagene Ciloleucel (65). BEN was the most common lymphodepleting agent used in the outpatient setting resulting in comparable incidence of CRS, and severe ICANs, to CY-FLU, except for the occurrence of any grade ICANS, which was lower in the BEN group. In fact, patients that received BEN required lower dose of dexamethasone, supporting the finding that BEN lymphodepletion is associated with a reduced risk of neurotoxicity. Additionally, there were no significant differences in hazard ratios for disease progression, relapse, or mortality, or in the rates of severe infectious complications, despite observing higher rates of grade ≥3 neutropenia in CY-FLU-treated patients. Consequently, outpatient lymphodepletion using BEN, appears to be equally effective to CY-FLU and may represent a cost-effective, safe, and efficient alternative (65). In a recent investigation, Sidana et al. compared the two-day use of BEN 90 mg/m2/day with a three-day course of CY-FLU (300 mg/m2/day and 30 mg/m2/day respectively) before the administration of two different BCMA CAR-T cell regimens [ciltacabtagene autoleucel (cilta-cel) and idecabtagene vicleucel (ide-cel)] in patients with multiple myeloma. CAR-T cell expansion and lymphocyte recovery were found to be comparable between the lymphodepletion groups. However, both nadir absolute lymphocyte count (ALC) and ALC before the CAR-T cell infusion were found to be significantly lower in the CY-FLU group. The incidence of ICANS, CRS and ICU admissions were similar between the two regimens. Comparable rates of neutropenic fever, infections as well as use of G-CSF, IVIG, steroids, and tocilizumab were observed. Overall response rate (ORR) and PFS were not different, irrespective of the CAR-T cell product used. These investigators concluded that the use of BEN for lymphodepletion before BCMA CAR-T cell therapy demonstrates comparable safety and efficacy as CY-FLU (9). Due to the FLU shortage over the past year, we also adopted a single-agent lymphodepletion strategy using BEN at a dose of 90 mg/m² administered over two consecutive days prior to infusing commercial CAR-T products in all patients. Importantly, no unexpected adverse effects were observed in the fifteen patients treated in this manner.

Summary

In summary, recent retrospective reports suggest that employing BEN lymphodepletion prior to CAR-T cell infusion may be associated with a reduction in adverse events, including CRS, ICANS, cytopenias and infections when compared to the use of CY-FLU. Importantly, this approach seems to maintain the effectiveness of CAR-T cell therapy. Additionally, BEN can be administered in an outpatient basis within a shorter timeframe, offering a cost-effective alternative. Its use has become particularly relevant during the recent period of nationwide FLU shortage. BEN’s unique immunomodulatory effects support its application as a lymphodepleting agent and merits a prospective comparison to CY-FLU through randomized clinical trials.

Data availability statement

The original contributions presented in the study are included in the article/supplementary material. Further inquiries can be directed to the corresponding author.

Author contributions

EK: Conceptualization, Investigation, Resources, Supervision, Writing – original draft, Writing – review and editing. DF: Conceptualization, Writing – original draft. MH: Writing – review and editing. SK: Writing – review and editing. RS: Writing – review and editing.

Funding

The author(s) declare that no financial support was received for the research, authorship, and/or publication of this article.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Guarini A, Radice G, Peragine N, Buracchi C, De Propris MS, Di Rocco A, et al. Long-Term host immune modulation following tisagenlecleucel administration in patients with diffuse large B-Cell lymphoma and B-Lineage acute lymphoblastic leukemia. Cancers (Basel) (2023) 15(9):2411. doi: 10.3390/cancers15092411

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Mailankody S, Matous JV, Chhabra S, Liedtke M, Sidana S, Oluwole OO, et al. Allogeneic BCMA-targeting CAR T cells in relapsed/refractory multiple myeloma: phase 1 UNIVERSAL trial interim results. Nat Med (2023) 29(2):422–9. doi: 10.1038/s41591-022-02182-7

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Amini L, Silbert SK, Maude SL, Nastoupil LJ, Ramos CA, Brentjens RJ, et al. Preparing for CAR T cell therapy: patient selection, bridging therapies and lymphodepletion. Nat Rev Clin Oncol (2022) 19(5):342–55. doi: 10.1038/s41571-022-00607-3

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Green S, Schultz L. Rational alternatives to fludarabine and cyclophosphamide-based pre-CAR lymphodepleting regimens in the pediatric and young adult B-ALL setting. Curr Oncol Rep (2023) 25(8):841–846. doi: 10.1007/s11912-023-01404-6

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Shah NN, Lee DW, Yates B, Yuan CM, Shalabi H, Martin S, et al. Long-term follow-up of CD19-CAR T-cell therapy in children and young adults with B-ALL. J Clin Oncol (2021) 39(15):1650–9. doi: 10.1200/JCO.20.02262

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Turtle CJ, Hanafi LA, Berger C, Hudecek M, Pender B, Robinson E, et al. Immunotherapy of non-Hodgkin’s lymphoma with a defined ratio of CD8+ and CD4+ CD19-specific chimeric antigen receptor-modified T cells. Sci Transl Med (2016) 8(355):355ra116. doi: 10.1126/scitranslmed.aaf8621

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Ying Z, Yang H, Guo Y, Li W, Zou D, Zhou D, et al.Relmacabtagene autoleucel (relma-cel) CD19 CAR-T therapy for adults with heavily pretreated relapsed/refractory large B-cell lymphoma in China. Cancer Med (2021) 10(3):999–1011. doi: 10.1002/cam4.3686

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Maziarz RT, Diaz A, Miklos DB, Shah NN. Perspective: an international fludarabine shortage: supply chain issues impacting transplantation and immune effector cell therapy delivery. Transplant Cell Ther (2022) 28(11):723–6. doi: 10.1016/j.jtct.2022.08.002

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Sidana S, Hosoya H, Jensen A, Liu L, Goyal A, Hovanky V, et al. Bendamustine vs. fludarabine/cyclophosphamide lymphodepletion prior to BCMA CAR-T cell therapy in multiple myeloma. Blood Cancer J (2023) 13(1):158. doi: 10.1038/s41408-023-00929-0

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Emadi A, Jones RJ, Brodsky RA. Cyclophosphamide and cancer: golden anniversary. Nat Rev Clin Oncol (2009) 6(11):638–47. doi: 10.1038/nrclinonc.2009.146

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Kastan MB, Schlaffer E, Russo JE, Colvin OM, Civin CI, Hilton J. Direct demonstration of elevated aldehyde dehydrogenase in human hematopoietic progenitor cells. Blood (1990) 75(10):1947–50. doi: 10.1182/blood.V75.10.1947.1947

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Saida Y, Watanabe S, Tanaka T, Baba J, Sato K, Shoji S, et al. Critical roles of chemoresistant effector and regulatory T cells in antitumor immunity after lymphodepleting chemotherapy. J Immunol (2015) 195(2):726–35. doi: 10.4049/jimmunol.1401468

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Terao T, Kitamura W, Fujii N, Asada N, Kamoi C, Fujiwara K, et al.Negative prognostic impact of high-dose or long-term corticosteroid use in patients with relapsed or refractory B-cell lymphoma who received tisagenlecleucel. Transplant Cell Ther (2023) 29(9):573.e571–573.e578. doi: 10.1016/j.jtct.2023.06.018

CrossRef Full Text | Google Scholar

14. Loskog A, Giandomenico V, Rossig C, Pule M, Dotti G, Brenner MK. Addition of the CD28 signaling domain to chimeric T-cell receptors enhances chimeric T-cell resistance to T regulatory cells. Leukemia (2006) 20(10):1819–28. doi: 10.1038/sj.leu.2404366

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Angulo I, de las Heras FG, García-Bustos JF, Gargallo D, Muñoz-Fernández MA, Fresno M. Nitric oxide-producing CD11b(+)Ly-6G(Gr-1)(+)CD31(ER-MP12)(+) cells in the spleen of cyclophosphamide-treated mice: implications for T-cell responses in immunosuppressed mice. Blood (2000) 95(1):212–20. doi: 10.1182/blood.V95.1.212

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Antony PA, Paulos CM, Ahmadzadeh M, Akpinarli A, Palmer DC, Sato N, et al. Interleukin-2-dependent mechanisms of tolerance and immunity in vivo. J Immunol (2006) 176(9):5255–66. doi: 10.4049/jimmunol.176.9.5255

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Antony PA, Restifo NP. CD4+CD25+ T regulatory cells, immunotherapy of cancer, and interleukin-2. J Immunother (2005) 28(2):120–8. doi: 10.1097/01.cji.0000155049.26787.45

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Mikyšková R, Indrová M, Polláková V, Bieblová J, Símová J, Reiniš M. Cyclophosphamide-induced myeloid-derived suppressor cell population is immunosuppressive but not identical to myeloid-derived suppressor cells induced by growing TC-1 tumors. J Immunother (2012) 35(5):374–84. doi: 10.1097/CJI.0b013e318255585a

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Wrzesinski C, Paulos CM, Kaiser A, Muranski P, Palmer DC, Gattinoni L, et al. Increased intensity lymphodepletion enhances tumor treatment efficacy of adoptively transferred tumor-specific T cells. J Immunother (2010) 33(1):1–7. doi: 10.1097/CJI.0b013e3181b88ffc

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Xin Yao X, Ahmadzadeh M, Lu YC, Liewehr DJ, Dudley ME, Liu F, et al. Levels of peripheral CD4(+)FoxP3(+) regulatory T cells are negatively associated with clinical response to adoptive immunotherapy of human cancer. Blood (2012) 119(24):5688–96. doi: 10.1182/blood-2011-10-386482

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Kanakry CG, Ganguly S, Bolanos Meade FJ, Zahurak ML, Kos F, Fuchs EJ, et al. Post-transplant cyclophosphamide (PT/cy) preserves effector (Teff) and regulatory (Treg) T cells during early immune reconstitution after allogeneic bone marrow transplantation. Biol Blood Marrow Transplantation (2012) 18(2, Supplement):S224–5. doi: 10.1016/j.bbmt.2011.12.065

CrossRef Full Text | Google Scholar

22. Kanakry CG, Ganguly S, Zahurak M, Bolaños-Meade J, Thoburn C, Perkins B, et al. Aldehyde dehydrogenase expression drives human regulatory T cell resistance to posttransplantation cyclophosphamide. Sci Transl Med (2013) 5(211):211ra157. doi: 10.1126/scitranslmed.3006960

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Hadjis AD, Nunes NS, Khan SM, Fletcher RE, Pohl AP, Venzon DJ, et al. Post-transplantation cyclophosphamide uniquely restrains alloreactive CD4(+) T-cell proliferation and differentiation after murine MHC-haploidentical hematopoietic cell transplantation. Front Immunol (2022) 13:796349. doi: 10.3389/fimmu.2022.796349

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Kodumudi KN, Weber A, Sarnaik AA, Pilon-Thomas S. Blockade of myeloid-derived suppressor cells after induction of lymphopenia improves adoptive T cell therapy in a murine model of melanoma. J Immunol (2012) 189(11):5147–54. doi: 10.4049/jimmunol.1200274

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Satoh Y, Kotani H, Iida Y, Taniura T, Notsu Y, Harada M. Supplementation of l-arginine boosts the therapeutic efficacy of anticancer chemoimmunotherapy. Cancer Sci (2020) 111(7):2248–58. doi: 10.1111/cas.14490

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Geyer MB, Rivière I, Sénéchal B, Wang X, Wang Y, Purdon TJ, et al. Safety and tolerability of conditioning chemotherapy followed by CD19-targeted CAR T cells for relapsed/refractory CLL. JCI Insight (2019) 5(9):e122627. doi: 10.1172/jci.insight.122627

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Gardner RA, Finney O, Annesley C, Brakke H, Summers C, Leger K, et al. Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults. Blood (2017) 129(25):3322–31. doi: 10.1182/blood-2017-02-769208

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Hay KA, Gauthier J, Hirayama AV, Voutsinas JM, Wu Q, Li D, et al. Factors associated with durable EFS in adult B-cell ALL patients achieving MRD-negative CR after CD19 CAR T-cell therapy. Blood (2019) 133(15):1652–63. doi: 10.1182/blood-2018-11-883710

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Turtle CJ, Hanafi LA, Berger C, Gooley TA, Cherian S, Hudecek M, et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest. (2016) 126(6):2123–38. doi: 10.1172/JCI85309

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Jiang H, Hu Y, Mei H. Consolidative allogeneic hematopoietic stem cell transplantation after chimeric antigen receptor T-cell therapy for relapsed/refractory B-cell acute lymphoblastic leukemia: who? When? Why? biomark Res (2020) 8(1):66. doi: 10.1186/s40364-020-00247-8

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Gandhi V, Plunkett W. Cellular and clinical pharmacology of fludarabine. Clin Pharmacokinet (2002) 41(2):93–103. doi: 10.2165/00003088-200241020-00002

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Lichtman SM, Etcubanas E, Budman DR, Eisenberg P, Zervos G, D’Amico P, et al. The pharmacokinetics and pharmacodynamics of fludarabine phosphate in patients with renal impairment: a prospective dose adjustment study. Cancer Invest. (2002) 20(7-8):904–13. doi: 10.1081/cnv-120005903

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Martell RE, Peterson BL, Cohen HJ, Petros WP, Rai KR, Morrison VA, et al. Analysis of age, estimated creatinine clearance and pretreatment hematologic parameters as predictors of fludarabine toxicity in patients treated for chronic lymphocytic leukemia: a CALGB (9011) coordinated intergroup study. Cancer Chemother Pharmacol (2002) 50(1):37–45. doi: 10.1007/s00280-002-0443-5

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Lowe KL, Mackall CL, Norry E, Amado R, Jakobsen BK, Binder G. Fludarabine and neurotoxicity in engineered T-cell therapy. Gene Ther (2018) 25(3):176–91. doi: 10.1038/s41434-018-0019-6

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Spriggs DR, Stopa E, Mayer RJ, Schoene W, Kufe DW. Fludarabine phosphate (NSC 312878) infusions for the treatment of acute leukemia: phase I and neuropathological study. Cancer Res (1986) 46(11):5953–8.

PubMed Abstract | Google Scholar

36. Fabrizio VA, Boelens JJ, Mauguen A, Baggott C, Prabhu S, Egeler E, et al. Optimal fludarabine lymphodepletion is associated with improved outcomes after CAR T-cell therapy. Blood Adv (2022) 6(7):1961–8. doi: 10.1182/bloodadvances.2021006418

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Dekker L, Calkoen FG, Jiang Y, Blok H, Veldkamp SR, De Koning C, et al. Fludarabine exposure predicts outcome after CD19 CAR T-cell therapy in children and young adults with acute leukemia. Blood Adv (2022) 6(7):1969–76. doi: 10.1182/bloodadvances.2021006700

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Scordo M, Flynn JR, Gonen M, Devlin SM, Parascondola A, Tomas AA, et al. Identifying an optimal fludarabine exposure for improved outcomes after axi-cel therapy for aggressive B-cell non-Hodgkin lymphoma. Blood Advances. (2023) 7(18):5579–85. doi: 10.1182/bloodadvances.2023010302

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Langenhorst JB, Dorlo TPC, van Maarseveen EM, Nierkens S, Kuball J, Boelens JJ, et al. Population pharmacokinetics of fludarabine in children and adults during conditioning prior to allogeneic hematopoietic cell transplantation. Clin Pharmacokinet (2019) 58(5):627–37. doi: 10.1007/s40262-018-0715-9

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Cheson BD, Leoni L. Bendamustine: mechanism of action and clinical data. Clin Adv Hematol Oncol (2011) 9(8 Suppl 19):1–11.

Google Scholar

41. Lalic H, Aurer I, Batinic D, Visnjic D, Smoljo T, Babic A. Bendamustine: A review of pharmacology, clinical use and immunological effects (Review). Oncol Rep (2022) 47(6):114. doi: 10.3892/or.2022.8325

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Leoni LM, Bailey B, Reifert J, Bendall HH, Zeller RW, Corbeil J, et al. Bendamustine (Treanda) displays a distinct pattern of cytotoxicity and unique mechanistic features compared with other alkylating agents. Clin Cancer Res (2008) 14(1):309–17. doi: 10.1158/1078-0432.CCR-07-1061

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Dennie TW, Kolesar JM. Bendamustine for the treatment of chronic lymphocytic leukemia and rituximab-refractory, indolent B-cell non-hodgkin lymphoma. Clin Ther (2009) 31:2290–311. doi: 10.1016/j.clinthera.2009.11.031

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Juma FD, Rogers HJ, Trounce JR. The pharmacokinetics of cyclophosphamide, phosphoramide mustard and nor-nitrogen mustard studied by gas chromatography in patients receiving cyclophosphamide therapy. Br J Clin Pharmacol (1980) 10(4):327–35. doi: 10.1111/j.1365-2125.1980.tb01768.x

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Gafter-Gvili A, Polliack A. Bendamustine associated immune suppression and infections during therapy of hematological Malignancies. Leuk Lymphoma. (2016) 57(3):512–9. doi: 10.3109/10428194.2015.1110748

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Stokes J, Molina MS, Hoffman EA, Simpson RJ, Katsanis E. Immunomodulatory effects of bendamustine in hematopoietic cell transplantation. Cancers (Basel) (2021) 13(7):1702. doi: 10.3390/cancers13071702

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Katsanis E, Sapp LN, Varner N, Koza S, Stea B, Zeng Y. Haploidentical bone marrow transplantation with post-transplant cyclophosphamide/bendamustine in pediatric and young adult patients with hematologic Malignancies. Biol Blood Marrow Transplant (2018) 24(10):2034–9. doi: 10.1016/j.bbmt.2018.06.007

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Katsanis E, Stea B, Kovacs K, Truscott L, Husnain M, Khurana S, et al. Feasibility and efficacy of partially replacing post-transplantation cyclophosphamide with bendamustine in pediatric and young adult patients undergoing haploidentical bone marrow transplantation. Transplant Cell Ther (2022) 28(7):390.e391–390.e310. doi: 10.1016/j.jtct.2022.04.015

CrossRef Full Text | Google Scholar

49. Molina MS, Hoffman EA, Stokes J, Kummet N, Simpson RJ, Katsanis E. Murine precursors to type 1 conventional dendritic cells induce tumor cytotoxicity and exhibit activated PD-1/PD-L1 pathway. PloS One (2022) 17(8):e0273075. doi: 10.1371/journal.pone.0273075

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Molina MS, Hoffman EA, Stokes J, Kummet N, Smith KA, Baker F, et al. Regulatory dendritic cells induced by bendamustine are associated with enhanced flt3 expression and alloreactive T-cell death. Front Immunol (2021) 12:699128. doi: 10.3389/fimmu.2021.699128

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Molina MS, Stokes J, Hoffman EA, Eremija J, Zeng Y, Simpson RJ, et al. Bendamustine conditioning skews murine host DCs toward pre-cDC1s and reduces gvHD independently of batf3. Front Immunol (2020) 11:1410. doi: 10.3389/fimmu.2020.01410

PubMed Abstract | CrossRef Full Text | Google Scholar

52. Stokes J, Hoffman EA, Molina MS, Eremija J, Larmonier N, Zeng Y, et al. Bendamustine with total body irradiation limits murine graft-versus-host disease in part through effects on myeloid-derived suppressor cells. Biol Blood Marrow Transplant (2019) 25(3):405–16. doi: 10.1016/j.bbmt.2018.10.009

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Stokes J, Hoffman EA, Molina MS, Kummet N, Simpson RJ, Zeng Y, et al. Bendamustine with total body irradiation conditioning yields tolerant T-cells while preserving T-cell-dependent graft-versus-leukemia. Oncoimmunology (2020) 9(1):1758011. doi: 10.1080/2162402X.2020.1758011

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Stokes J, Hoffman EA, Zeng Y, Larmonier N, Katsanis E. Post-transplant bendamustine reduces GvHD while preserving GvL in experimental haploidentical bone marrow transplantation. Br J Haematol (2016) 174(1):102–16. doi: 10.1111/bjh.14034

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Stokes J, Simpson RJ, Katsanis E. Commentary: post-transplantation cyclophosphamide uniquely restrains alloreactive CD4+ T-cell proliferation and differentiation after murine MHC-haploidentical hematopoietic cell transplantation. Front Immunol (2022) 13:887648. doi: 10.3389/fimmu.2022.887648

PubMed Abstract | CrossRef Full Text | Google Scholar

56. Hay KA. Cytokine release syndrome and neurotoxicity after CD19 chimeric antigen receptor-modified (CAR-) T cell therapy. Br J Haematol (2018) 183(3):364–74. doi: 10.1111/bjh.15644

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Holland EM, Molina JC, Dede K, Moyer D, Zhou T, Yuan CM, et al. Efficacy of second CAR-T (CART2) infusion limited by poor CART expansion and antigen modulation. J Immunother Cancer (2022) 10(5):e004483. doi: 10.1136/jitc-2021-004483

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Bethge WA, Martus P, Schmitt M, Holtick U, Subklewe M, von Tresckow B, et al. GLA/DRST real-world outcome analysis of CAR T-cell therapies for large B-cell lymphoma in Germany. Blood (2022) 140(4):349–58. doi: 10.1182/blood.2021015209

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Hirayama AV, Gauthier J, Hay KA, Voutsinas JM, Wu Q, Gooley T, et al. The response to lymphodepletion impacts PFS in patients with aggressive non-Hodgkin lymphoma treated with CD19 CAR T cells. Blood (2019) 133(17):1876–87. doi: 10.1182/blood-2018-11-887067

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Schuster SJ, Bishop MR, Tam CS, Waller EK, Borchmann P, McGuirk JP, et al. Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med (2019) 380(1):45–56. doi: 10.1056/NEJMoa1804980

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Ramos CA, Grover NS, Beaven AW, Lulla PD, Wu M-F, Ivanova A, et al. Anti-CD30 CAR-T cell therapy in relapsed and refractory hodgkin lymphoma. J Clin Oncol (2020) 38(32):3794–804. doi: 10.1200/JCO.20.01342

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Ghilardi G, Chong EA, Svoboda J, Wohlfarth P, Nasta SD, Williamson S, et al. Bendamustine is safe and effective for lymphodepletion before tisagenlecleucel in patients with refractory or relapsed large B-cell lymphomas. Ann Oncol (2022) 33(9):916–28. doi: 10.1016/j.annonc.2022.05.521

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Garcia-Calvo GII, Navarro V, Lopez AAM, Bastos Oreiro M, Rejeski K, Aleksandra Jalowiec K, et al. Bendamustine-containing regimens can be safely used as bridging in large b-cell lymphoma patients receiving chimeric antigen receptor T-cell therapy. Hemasphere (2023) 7(Suppl: e48751b3.):P1151. doi: 10.1097/01.HS9.0000971500.48751.b3

CrossRef Full Text | Google Scholar

64. Ahmed N, Wesson W, Mushtaq MU, Porter DL, Nasta SD, Brower J, et al. Patient characteristics and outcomes of outpatient tisagenlecleucel recipients for B cell non-hodgkin lymphoma. Transplant Cell Ther (2023) 29(7):449.e441–449.e447. doi: 10.1016/j.jtct.2023.04.019

CrossRef Full Text | Google Scholar

65. Ong SY, Pak S, Mei M, Wang Y, Popplewell L, Baird JH, et al. Bendamustine lymphodepletion is a well-tolerated alternative to fludarabine and cyclophosphamide lymphodepletion for axicabtagene ciloleucel therapy for aggressive B-cell lymphoma. Am J Hematol (2023) 98(11):1751–1761. doi: 10.1002/ajh.27069

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: fludarabine, cyclophosphamide, bendamustine, lymphodepletion, CAR (chimeric antigen receptor) T cells

Citation: Filioglou D, Husnain M, Khurana S, Simpson RJ and Katsanis E (2023) Has the shortage of fludarabine altered the current paradigm of lymphodepletion in favor of bendamustine? Front. Immunol. 14:1329850. doi: 10.3389/fimmu.2023.1329850

Received: 30 October 2023; Accepted: 08 November 2023;
Published: 22 November 2023.

Edited by:

Matthew Mei, City of Hope National Medical Center, United States

Reviewed by:

Tamer Othman, City of Hope National Medical Center, United States

Copyright © 2023 Filioglou, Husnain, Khurana, Simpson and Katsanis. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Emmanuel Katsanis, ekatsani@arizona.edu

ORCID: Richard J. Simpson, orcid.org/0000-0002-7064-6881
Emmanuel Katsanis, orcid.org/0000-0003-1466-6965

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.