147
Views
1
CrossRef citations to date
0
Altmetric
Review

Emerging Monoclonal Antibody Therapy for the Treatment of Acute Lymphoblastic Leukemia

, &
Pages 419-431 | Published online: 18 Oct 2021

References

  • Paul S, Kantarjian H, Jabbour EJ. Adult acute lymphoblastic leukemia. Mayo Clin Proc. 2016;91(11):1645–1666. doi:10.1016/j.mayocp.2016.09.010
  • Samra B, Jabbour E, Ravandi F, et al. Evolving therapy of adult acute lymphoblastic leukemia: state-of-the-art treatment and future directions. J Hematol Oncol. 2020;13(1):70. doi:10.1186/s13045-020-00905-2
  • Czuczman MS, Olejniczak S, Gowda A, et al. Acquirement of rituximab resistance in lymphoma cell lines is associated with both global CD20 gene and protein down‐regulation regulated at the pretranscriptional and posttranscriptional levels. Clin Cancer Rec. 2008;14(5):1561‐1570.
  • Piccaluga PP, Arpinati M, Candoni A, et al. Surface antigens analysis reveals significant expression of candidate targets for immunotherapy in adult acute lymphoid leukemia. Leuk Lymphoma. 2011;52(2):325–327. doi:10.3109/10428194.2010.529206
  • Hoelzer D, Walewski J, Döhner H, et al. Improved outcome of adult Burkitt lymphoma/leukemia with rituximab and chemotherapy: report of a large prospective multicenter trial. Blood. 2014;124(26):3870–3879. doi:10.1182/blood-2014-03-563627
  • Thomas DA, O’Brien S, Faderl S, et al. Chemoimmunotherapy with a modified hyper-CVAD and rituximab regimen improves outcome in de novo Philadelphia chromosome-negative precursor B-lineage acute lymphoblastic leukemia. J Clin Oncol. 2010;28(24):3880–3889. doi:10.1200/JCO.2009.26.9456
  • Hoelzer D, Huettmann A, Kaul F, et al. Immunochemotherapy with rituximab improves molecular CR rate and outcome in CD20 B-lineage standard and high-risk patients; results of 263 CD20 patients studied prospectively in GMALL study 07/2003. Blood. 2010;116(21):170. doi:10.1182/blood.V116.21.170.170
  • Maury S, Chevert S, Thomas X, et al. Rituximab in B-lineage adult acute lymphoblastic leukemia. N Engl J Med. 2016;375(11):1044–1053. doi:10.1056/NEJMoa1605085
  • Cheson BD. Ofatumumab, a novel anti‐CD20 monoclonal antibody for the treatment of B‐cell malignancies. J Clin Oncol. 2010;28(21):3525–3530. doi:10.1200/JCO.2010.27.9836
  • Jabbour E, Richard-Carpentier G, Sasaki Y, et al. Hyper-CVAD regimen in combination with ofatumumab as frontline therapy for adults with Philadelphia chromosome-negative B-cell acute lymphoblastic leukaemia: a single-arm, Phase 2 trial. Lancet Haematol. 2020;7(7):e523–e533. doi:10.1016/S2352-3026(20)30144-7
  • Shah NN, Stevenson MS, Yuan CM, et al. Characterization of CD22 expression in acute lymphoblastic leukemia. Pediatr Blood Cancer. 2015;62(6):964‐969. doi:10.1002/pbc.25410
  • Raetz EA, Cairo MS, Borowitz MJ, et al. Chemoimmunotherapy reinduction with epratuzumab in children with acute lymphoblastic leukemia in marrow relapse: a children’s oncology group pilot study. J Clin Oncol. 2008;26(22):3756–3762. doi:10.1200/JCO.2007.15.3528
  • Raetz EA, Cairo MS, Borowitz MJ, et al. Re-induction chemoimmunotherapy with epratuzumab in relapsed acute lymphoblastic leukemia (ALL): phase II results from Children’s Oncology Group (COG) study ADVL04P2. Pediatr Blood Cancer. 2015;62(7):1171–1175. doi:10.1002/pbc.25454
  • Advani A, McDonough S, Coutre S, et al. Southwest Oncology Group Study S0910: a phase 2 trial of clofarabine/cytarabine/epratuzumab for relapsed/refractory acute lymphocytic leukemia. Br J Haematol. 2014;165(4):504–509. doi:10.1111/bjh.12778
  • Keyhani A, Huh YO, Jendiroba D, et al. Increased CD38 expression is associated with favorable prognosis in adult acute leukemia. Leuk Res. 2002;24(2):153–159. doi:10.1016/S0145-2126(99)00147-2
  • Mirgh S, Ahmed R, Agrawal N. Will Daratumumab be the next game changer in early thymic precursor-acute lymphoblastic leukaemia? Br J Haematol. 2019;187(2):e33–e35. doi:10.1111/bjh.16154
  • Krejcik J, Casneuf T, Nijhof IS, et al. Daratumumab depletes CD38+ immune regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma. Blood. 2016;128(3):384–394. doi:10.1182/blood-2015-12-687749
  • Bride KL, Vincent TL, Im SY, et al. Preclinical efficacy of daratumumab in T cell acute lymphoblastic leukemia. Blood. 2018;131(9):995–999. doi:10.1182/blood-2017-07-794214
  • Bonda A, Punatar S. Daratumumab at the frontiers of post-transplant refractory T acute lymphoblastic leukemia-a worthwhile strategy? Bone Marrow Transplant. 2018;53(11):1487–1489. doi:10.1038/s41409-018-0222-5
  • Ofran Y, Ringelstein-Harlev S, Slouzkey I, et al. Daratumumab for eradication of minimal residual disease in high-risk advanced relapse of T-cell/CD19/CD22-negative acute lymphoblastic leukemia. Leukemia. 2020;34(1):293–295. doi:10.1038/s41375-019-0548-z
  • Hu Y, Turner MJ, Shields J, et al. Investigation of the mechanism of action of alemtuzumab in a human CD52 transgenic mouse model. Immunology. 2009;128(2):260–270. doi:10.1111/j.1365-2567.2009.03115.x
  • Angiolillo AL, Yu AL, Reaman G, et al. A phase II study of Campath-1H in children with relapsed or refractory acute lymphoblastic leukemia: a Children’s Oncology Group report. Pediatr Blood Cancer. 2009;53(6):978–983. doi:10.1002/pbc.22209
  • Stock W, Sanford B, Lozanski G, et al. Alemtuzumab can be incorporated into front-line therapy of adult acute lymphoblastic leukemia (ALL): final phase I results of a Cancer and Leukemia Group B Study (CALGB 10102). Blood. 2009;114(22):838. doi:10.1182/blood.V114.22.838.838
  • Silva A, Laranjeira ABA, Martins LR, et al. IL-7 contributes to the progression of human T-cell acute lymphoblastic leukemias. Cancer Res. 2011;71(14):4780–4789. doi:10.1158/0008-5472.CAN-10-3606
  • Canté-Barrett K, Spijkers-Hagelstein JAP, Buijs-Gladdines JGCAM, et al. MEK and PI3K-AKT inhibitors synergistically block activated IL7 receptor signaling in T-cell acute lymphoblastic leukemia. Leukemia. 2016;30(9):1832–1843. doi:10.1038/leu.2016.83
  • Akkapeddi P, Fragoso R, Hixon JA, et al. A fully human anti-IL-7Rα antibody promotes antitumor activity against T-cell acute lymphoblastic leukemia. Leukemia. 2019;33(9):2155–2168. doi:10.1038/s41375-019-0434-8
  • Bargou R, Leo E, Zugmaier G, et al. Tumor regression in cancer patients by very low doses of a T cell engaging antibody. Science. 2008;321(5891):974–977. doi:10.1126/science.1158545
  • Topp MS, Gokburger N, Stein AS, et al. Phase II trial of the anti-CD19 bispecific T cell-engager blinatumomab shows hematologic and molecular remissions in patients with relapsed or refractory B-precursor acute lymphoblastic leukemia. J Clin Oncol. 2014;32(37):4134–4140. doi:10.1200/JCO.2014.56.3247
  • Topp MS, Kufer P, Gokbuget N, et al. Targeted therapy with the T-cell-engaging antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival. J Clin Oncol. 2011;29(18):2493–2498. doi:10.1200/JCO.2010.32.7270
  • Himabukuro-Vornhagen A, Gödel P, Subklewe M, et al. Cytokine release syndrome. J Iimmunother Cancer. 2018;56:6.
  • Topp MS, Gokbuget N, Stein A, et al. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicenter, single-arm, phase 2 study. Lancet Oncol. 2015;16(1):57–66. doi:10.1016/S1470-2045(14)71170-2
  • Caimi PF, Ahmed N, Rojas P, et al. Prophylactic tocilizumab before CD3/4-1bb anti-CD19 car-T cell infusion decreases incidence of severe crs without increased risk of neurotoxicity. Cytotherapy. 2020;22(5):S16–S17.
  • Kantarjan H, Stein A, Gokbuget N, et al. Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N Engl J Med. 2017;376(9):836–847. doi:10.1056/NEJMoa1609783
  • Foa R, Bassan R, Vitale A, et al. Dasatinib-blinatumomab for ph-positive acute lymphoblastic leukemia in adults. N Engl J Med. 2020;383(17):1613–1623. doi:10.1056/NEJMoa2016272
  • Kantarjian HM, De Angelo DJ, Stelljes M, et al. Inotuzumab ozogamicin for acute lymphoblastic leukemia. N Engl J Med. 2016;375(8):740–753. doi:10.1056/NEJMoa1509277
  • Kantarjian HM, De Angelo DJ, Stelljes M, et al. Inotuzumab ozogamincin versus standard of care in relapsed or refractory acute lymphoblastic leukemia: final report and long-term survival follow-up from the randomized, Phase 3 INO-VATE study. Cancer. 2019;125(14):2474–2487. doi:10.1002/cncr.32116
  • Jabbour E, Ravandi F, Kebriael P, et al. Salvage chemoimmunotherapy with inotuzumab ozogamicin combined with mini-hyper-CVD for patients with relapsed or refractory Philadelphia chromosome-negative acute lymphoblastic leukemia: a phase 2 clinical trial. JAMA Oncol. 2018;4(2):230–234. doi:10.1001/jamaoncol.2017.2380
  • Sasaki K, Kantarijan HM, Ravandi F, et al. Sequential combination of low-intensity chemotherapy (mini-hyper-CVD) plus inotuzumab ozogamicin with or without blinatumomab in patients with relapsed/refractory Philadelphia chromosome-negative acute lymphoblastic leukemia (ALL): a phase 2 trial. Blood. 2019;132(Supplement 1):553. doi:10.1182/blood-2018-99-115162
  • Kantarjian HM, Ravandi F, Short NJ, et al. Inotuzumab ozogamicin in combination with low-intensity chemotherapy for older patients with Philadelphia chromosome-negative acute lymphoblastic leukaemia: a single-arm, phase 2 study. Lancet Oncol. 2018;19(2):240–248. doi:10.1016/S1470-2045(18)30011-1
  • Roberts AW, Davids MS, Pagel JM, et al. Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia. N Engl J Med. 2016;374(4):311–322. doi:10.1056/NEJMoa1513257
  • DiNardo CD, Pratz K, Pullarkat V, et al. Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood. 2018. doi:10.1182/blood-2018-08-868752
  • Jain N, Lamb AV, O’Brien S, et al. Early T-cell precursor acute lymphoblastic leukemia/lymphoma (ETP-ALL/LBL) in adolescents and adults: a high-risk subtype. Blood. 2016;127(15):1863–1869. doi:10.1182/blood-2015-08-661702
  • Chonghaile TN, Roderick JE, Glenfield C, et al. Maturation stage of T-cell acute lymphoblastic leukemia determines BCL-2 versus BCL-XL dependence and sensitivity to ABT-199. Cancer Discov. 2014;4(9):1074–1087. doi:10.1158/2159-8290.CD-14-0353
  • Richard-Carpentier G, Jabbour E, Short NJ, et al. Clinical experience with venetoclax in combination with chemotherapy for relapsed or refractory t-cell acute lymphoblastic leukemia.. Clin Lymphoma Myeloma Leuk. 2020;20(4):212–218. doi:10.1016/j.clml.2019.09.608
  • Lacayo NJ, Pullarkat VA, Stock W, et al. Safety and efficacy of venetoclax in combination with navitoclax in adult and pediatric relapsed/refractory acute lymphoblastic leukemia and lymphoblastic lymphoma. Blood. 2019;134.
  • Frey NV, Porter DL. Cytokine release syndrome with novel therapeutics for acute lymphoblastic leukemia. Am J Hematol. 2019;94(S1):S24–S27. doi:10.1002/ajh.25442
  • Brentjens RJ, Santos E, Nikhamin Y, et al. Genetically targeted T cells eradicate systemic acute lymphoblastic leukemia xenografts. Clin Cancer Res. 2007;13(18):5426–5435. doi:10.1158/1078-0432.CCR-07-0674
  • Lee DW, Kochenderfer JN, Stetler-Stevenson M, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a Phase 1 dose-escalation trial. Lancet. 2015;385(9967):517–528.
  • Davila ML, Riviere I, Wang X, et al. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6(224):224ra25
  • Teachey DT, Lacey SF, Shaw PA, et al. Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov. 2016;6(6):664–679. doi:10.1158/2159-8290.CD-16-0040
  • Turtle CJ, Hanafi LA, Berger C, et al. CD19 CAR-T cells of defined CD4+: CD8+composition in adult B cell ALL patients. J Clin Invest. 2016;126(6):2123–2138. doi:10.1172/JCI85309
  • Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371(16):1507–1517. doi:10.1056/NEJMoa1407222
  • Maude SL, Laetsch TW, Buechner J, et al. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018;378(5):439–448. doi:10.1056/NEJMoa1709866
  • Hunter BD, Jacobson CA. Car t-cell associated neurotoxicity: mechanisms, clinicopathologic correlates, and future directions. J Natl Cancer Inst. 2019;111(7):646–654. doi:10.1093/jnci/djz017
  • Fry TJ, Shah NN, Orentas RJ, et al. CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nat Med. 2018;24(1):20–28. doi:10.1038/nm.4441
  • Amrolia PJ, Wynn R, Hough RE, et al. Phase I study of AUTO3, a bicistronic chimeric antigen receptor (CAR) T-cell therapy targeting CD19 and CD22, in pediatric patients with relapsed/refractory Bcell acute lymphoblastic leukemia (r/r B-ALL): Amelia Study. Blood. 2019;134(Supplement_1):2620. doi:10.1182/blood-2019-123424
  • Schultz LM, Muffly LS, Spiegel JY, et al. Phase I trial using CD19/CD22 bispecific CAR T cells in pediatric and adult acute lymphoblastic leukemia (ALL). Blood. 2019;134(Supplement_1):744. doi:10.1182/blood-2019-129411