Next Article in Journal
Meiotic Recombination May Be Initiated by Copy Choice During DNA Synthesis Rather than Break/Join Mechanism
Previous Article in Journal
Comprehensive Characterization of Armoracia rusticana Roots and Leaves: Physicochemical Properties, Functional Potential, and Nutritional Composition
Previous Article in Special Issue
From Molecular Precision to Clinical Practice: A Comprehensive Review of Bispecific and Trispecific Antibodies in Hematologic Malignancies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

Special Issue: Antibody Therapy for Hematologic Malignancies

by
Jonathan P. Butchar
Division of Hematology, The Ohio State University Wexner Medical Center, 410 W, 12th Avenue, Columbus, OH 43210, USA
Int. J. Mol. Sci. 2025, 26(19), 9463; https://doi.org/10.3390/ijms26199463 (registering DOI)
Submission received: 9 September 2025 / Accepted: 25 September 2025 / Published: 27 September 2025
(This article belongs to the Special Issue Antibody Therapy for Hematologic Malignancies)
It was recently reported that hematologic malignancies represented 6.6% of total cancer cases and that the global lifetime risk of incidence was 1.67% in 2022 [1]. The economic burden was estimated at roughly 1% of global GDP in 2021 [2]. Quite expectedly, there have been great efforts toward creating therapies against these cancers. This has led to a wide variety of treatments, with new agents and modalities emerging on an essentially perpetual basis. Inhibitors of Bruton Tyrosine Kinase (BTK) are used for B-cell malignancies and continuously evolve to address resistance [3,4]. Chimeric antigen receptor (CAR) T cell therapy has shown effectiveness [5] and universal CAR T (UCAR-T) (derived from allogeneic healthy donors) has been making great strides [6]. Checkpoint inhibitors are in use and testing [7], vaccines continue to be developed [8] and PEGylated/engineered cytokines are being studied [9]. We also see continued advances with combination therapies [10,11,12,13,14].
Among these developments, there has been substantial growth of antibody-based treatments. Rituximab, approved by the US Food and Drug Administration (FDA) in 1997 [15], represented the first such agent and has shown efficacy in a range of B-cell malignancies [16]. It is a chimeric antibody consisting of murine anti-CD20 variable regions with human IgG1 and κ light chain constant regions [17]. Rituximab leads to B-cell depletion by binding CD20 on the B cells and triggering complement-dependent cytotoxicity (CDC) as well as antibody-dependent cellular cytotoxicity (ADCC) [17] and apoptosis [18,19,20]. Rituximab has now been joined by a large number of additional antibodies, from engineered anti-CD20 antibodies such as obinutuzumab [21] to cytokine-antibody fusions [22] and bispecific antibodies [23]. This Special Issue focuses on recent advances made with antibody therapy, highlighting key developments and summarizing a battery of clinical trials for leukemias and lymphomas.
Lymphoid malignancies have generally been responsive to antibody therapy, and this is reviewed by Naman et al. [16]. They provide an overview of the major types of antibodies (Figure 1). Starting with rituximab, they discuss the design, production and clinical applications of antibodies for hematologic malignancies, as well as adverse effects and resistance mechanisms. Since the first antibody, severe effects have been seen such as acute tumor lysis syndrome [24,25,26]. Indeed, even with the later-generation antibody obinutuzumab, adverse events including infusion-related reactions, thrombocytopenia and cardiac-related effects have been seen [27], along with rare but more serious severe acute thrombocytopenia [28]. As pointed out by Naman et al., however, most adverse events have been well managed over the years.
Naman et al. also address the critical topic of resistance to antibody therapy, which was documented shortly after the approval of rituximab when loss of the CD20 antigen was seen in some relapsing patients [29,30]. This antigen loss is discussed, along with other resistance mechanisms. Following this, they move forward with engineered antibodies, ADCs and bispecific antibodies. They then highlight treatment strategies, paying particular attention to combination therapy. This can entail two different antibody-based agents, an antibody plus a non-antibody drug such as lenalidomide or an antibody with a checkpoint-targeting agent. The potential for artificial intelligence (AI) to substantially impact the development of antibodies via prediction of high-immunogenicity targets is also discussed.
One factor associated with treatment resistance across multiple cancers is c-MYC overexpression [31]. To explore this in greater detail, de Jonge et al. [32] in this Special Issue used inhibitor and CRISPR approaches to identify mechanisms by which MYC could promote resistance. They found a combination of responses affected by MYC such as antigen presentation, expression of apoptosis-related proteins and effects on T-cell activation. This paves the way for multiple therapeutic approaches, which are discussed by the authors.
Roughly forty years ago, bispecific antibodies were demonstrated as a potential means of directing T cells against specific tumor-associated targets [33,34]. Here, the antibodies were created by protein crosslinking or by hybrid hybridomas. Advances in technology have greatly facilitated the generation of these antibodies, with numerous production platforms available [35]. Perhaps the most well-known bispecific antibody today is the fragment-based bispecific T-cell engager (BiTE) blinatumomab, showing great success against acute lymphocytic leukemia (ALL) [36,37]. For CLL, a relatively recent development has been a bispecific antibody targeting the aberrant IGLV3-21R110 B-cell receptor (BCR) and CD3 [38]. The same group had previously developed CAR T cells targeting the IGLV3-21R110 BCR, with the bispecific antibody created as a possible therapy for patients unable to tolerate CAR-T treatment [39].
In this Special Issue, Amoozgar et al. [40] provide a rich review of bi- and tri-specific antibodies that includes design, mechanisms and targets (Figure 2) and their efficacies in various leukemias and lymphomas. They then discuss adverse events and resistance mechanisms commonly seen with the use of these antibodies. Several interesting potential future directions are also mentioned, including multispecific constructs that incorporate immune modulators or checkpoint inhibitors. The latter is likely to be highly useful for CLL patients, as PD-1 expression on CLL cells is dependent on BTK and is higher in patients showing disease progression while receiving BTK inhibitor therapy [41]. Amoozgar et al. also discuss masked constructs that are cleavable by tumor-specific protease-sensitive linkers, potentially maximizing on-target effects and reducing toxicities. Finally, they delve into the world of AI, discussing AI-guided antibody design, the prediction of patient responses antibodies and treatment optimization.
For bispecifics in general, much of the focus has been on αβ T cells but γδ T cells have also been intensively investigated. These cells are MHC-independent, expandable and highly cytotoxic (reviewed in [42]). In a research article within this Special Issue, Marischen et al. [43] examined the effects of the anti-CD3/CD20 bispecific antibody mosunetuzumab on γδ T cells, comparing against obinutuzumab. Using a series of co-culture experiments, they found that mosunetuzumab was more effective at activating γδ T cells and driving stronger γδ T cell-mediated elimination of cell-line and primary-cell targets. Due to the expandable nature of γδ T cells and the established safety profile of mosunetuzumab [44,45], combined mosunetuzumab plus stimulated γδ T cells may represent a practical and powerful therapeutic modality. The authors discuss this, as well as the possibility of testing γδ T cells against additional bi- and tri-specific antibodies, which would make this approach more generalizable.
As mentioned by both Naman et al. and Amoozgar et al., safety remains a concern with bispecific antibodies. To gain a clearer understanding of this, Bayly-McCredie et al. [46] used a structured systematic review approach to evaluate the safety and efficacy of bispecific antibodies for large B-cell lymphomas (LBCLs), comparing against CAR-T therapy. Results showed a tradeoff, with bispecifics having good efficacy and tolerability and CAR-T therapy having greater efficacy with less tolerability. This not only provides impetus for further refinement of bispecifics, it indirectly hints at the contribution of economic and logistical factors in the choice of therapeutic modality, an idea brought up by Juarez-Salcedo et al. [47] in this Special Issue.
In their review, Juarez-Salcedo et al. focus on the antibody-drug conjugate, loncastuximab tesirine, as a treatment for diffuse large B-cell lymphoma (DLBCL). Here, humanized anti-CD19 is linked to SG3199, a DNA-targeting alkylating agent. Before and after its approval by the US Food and Drug Administration in 2021 [48], this ADC has received continuous attention in clinical trials. The authors discuss these trials, past and ongoing. Current trials include pairing loncastuximab with other antibodies such as rituximab, glofitamab, mosunetuzumab and polatuzumab vedotin. These combinations offer the advantage of targeting two different antigens on the tumor cells and of combining two methods of killing. Loncastuximab leads to tumor-cell death by binding DNA, while biscpecific anti-CD20 glofitamab [49] and mosunetuzumab [50] recruit T cells and the anti-CD79B ADC polatzumab vedotin disrupts microtubule formation [51]. We anxiously await the results of these trials.
Despite advances such as those reviewed above, there are still hematologic malignancies not yet fully amenable to these therapies. Damiani and Tiribelli [52] discuss a major such disease, acute myeloid leukemia (AML). They highlight several contributing factors such as difficulty identifying good target antigens, low mutational burden and low immunogenicity of candidate antigens. Despite this, antibodies are in use and testing that target antigens such as CD33, CD123 and CD371. The authors discuss these and additional antibodies, presented with a useful background section on antibody therapy in general. Specific antibodies are evaluated in ample detail, highlighting their design, targets, efficacy and caveats. Finally, the authors discuss several potential ways to improve antibody therapy for AML. These include surfaceome analysis, the use of multiple binding sites, engineered antibodies that are activated only when all targets are engaged, overcoming the immunosuppressive environment, and other interesting ideas.
Antibody therapy plays a key role in the treatment of hematologic malignancies, and antibody-based modalities will certainly improve over time as biological and computational technologies advance. Malignant cells are well known for their ability to escape both non-antibody-based [53] and antibody-based [30] treatments, so this continued progress is greatly needed. On behalf of the authors, we hope you find this Special Issue enjoyable and useful.

Funding

This work was funded by National Institutes of Health/National Cancer Institute R01 1R01CA286117-01 and the Division of Hematology DSRP GR135048.

Acknowledgments

Thanks to Giovanna Merchand-Reyes and Susheela Tridandapani for critical reading of this editorial.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Sun, K.; Wu, H.; Zhu, Q.; Gu, K.; Wei, H.; Wang, S.; Li, L.; Wu, C.; Chen, R.; Pang, Y.; et al. Global landscape and trends in lifetime risks of haematologic malignancies in 185 countries: Population-based estimates from GLOBOCAN 2022. eClinicalMedicine 2025, 83, 103193. [Google Scholar] [CrossRef]
  2. Li, Z.; Chen, G.; Du, G. Global, regional, and national economic burden of hematologic malignancies (1990–2021) with projections to 2050. Front. Public Health 2025, 13, 1570792. [Google Scholar] [CrossRef]
  3. Cui, Z.X.; Yao, S.X.; Zhang, Y.X.; Lu, H.Y.; Sun, L.P.; Shi, L. Bruton’s tyrosine kinase (BTK) inhibitors: An updated patent review (2019–2024). Expert Opin. Ther. Pat. 2025, 1–25. [Google Scholar] [CrossRef]
  4. Das, D.; Xie, L.; Qiao, D.; Cao, Y.; Jiang, S.; Zheng, M.; Liu, Z.; Li, Y.; Jia, J.; Lv, Y.; et al. Discovery of Reversible, Noncovalent Bruton’s Tyrosine Kinase Inhibitors Targeting BTK C481S Mutation. ACS Med. Chem. Lett. 2025, 16, 1038–1047. [Google Scholar] [CrossRef]
  5. Cappell, K.M.; Kochenderfer, J.N. Long-term outcomes following CAR T cell therapy: What we know so far. Nat. Rev. Clin. Oncol. 2023, 20, 359–371. [Google Scholar] [CrossRef]
  6. Jiang, N.; Yang, Z.; Miao, H.; Xing, S.; Wang, S.; Li, N. Recent advances in universal chimeric antigen receptor T cell therapy. J. Hematol. Oncol. 2025, 18, 82. [Google Scholar] [CrossRef]
  7. Bilgihan, M.T.; Eryigit, A.N.; Ciftciler, R. Efficacy and Safety of Immune Checkpoint Inhibitors in Hematologic Malignancies. Clin. Lymphoma Myeloma Leuk. 2024, 24, 23–31. [Google Scholar] [CrossRef]
  8. Olsen, T.A.; Barnum, K.J.; Avigan, D.; Rosenblatt, J. Cancer vaccines in hematologic malignancy: A systematic review of the rational and evidence for clinical use. Best Pract. Res. Clin. Haematol. 2025, 38, 101650. [Google Scholar] [CrossRef]
  9. Sousa-Pimenta, M.; Dos Santos, N.R. Cytokine-based immunotherapy in hematolymphoid malignancies. Int. Rev. Cell Mol. Biol. 2025, 396, 55–79. [Google Scholar]
  10. Franklin, J.; Eichenauer, D.A.; Becker, I.; Monsef, I.; Engert, A. Optimisation of chemotherapy and radiotherapy for untreated Hodgkin lymphoma patients with respect to second malignant neoplasms, overall and progression-free survival: Individual participant data analysis. Cochrane Database Syst. Rev. 2017, 9, CD008814. [Google Scholar] [CrossRef]
  11. Watts, J.M.; Baer, M.R.; Yang, J.; Prebet, T.; Lee, S.; Schiller, G.J.; Dinner, S.N.; Pigneux, A.; Montesinos, P.; Wang, E.S.; et al. Olutasidenib alone or with azacitidine in IDH1-mutated acute myeloid leukaemia and myelodysplastic syndrome: Phase 1 results of a phase 1/2 trial. Lancet Haematol. 2023, 10, e46–e58, Erratum in Lancet Haematol. 2023, 10, e9. [Google Scholar] [CrossRef]
  12. Goldkuhle, M.; Kreuzberger, N.; von Tresckow, B.; Eichenauer, D.A.; Specht, L.; Monsef, I.; Skoetz, N. Chemotherapy alone versus chemotherapy plus radiotherapy for adults with early-stage Hodgkin’s lymphoma. Cochrane Database Syst. Rev. 2024, 12, CD007110. [Google Scholar]
  13. Jonas, B.A.; Fisch, S.C.; Curtin, P.T.; Schiller, G.J.; Jeyakumar, D.; Tzachanis, D.; Bejar, R.; Qi, L.; Wieduwilt, M.J.; Tuscano, J.M.; et al. University of California Hematologic Malignancies, C. A phase 1 trial of ibrutinib and azacitidine for higher risk myelodysplastic syndromes (University of California Hematologic Malignancies Consortium Study 1503). Leuk. Res. 2025, 155, 107717. [Google Scholar] [CrossRef]
  14. Yuan, X.; Wang, F.; Zhao, P.; Yang, B.; Yang, X.; Tian, T.; Li, B.; Liu, G.; Wang, S.; Tang, D.; et al. Efficacy and safety of CD19 combined with CD22 or CD20 chimeric antigen receptor T-cell therapy for hematological malignancies. Front. Immunol. 2025, 16, 1577360. [Google Scholar] [CrossRef]
  15. Grillo-Lopez, A.J. Rituximab: An insider’s historical perspective. Semin. Oncol. 2000, 27 (Suppl. 12), 9–16. [Google Scholar]
  16. Naman, J.; Shah, N.; Heyman, B.M. Antibody Therapy for Patients with Lymphoid Malignancies: Past and Present. Int. J. Mol. Sci. 2025, 26, 1711. [Google Scholar] [CrossRef]
  17. Reff, M.E.; Carner, K.; Chambers, K.S.; Chinn, P.C.; Leonard, J.E.; Raab, R.; Newman, R.A.; Hanna, N.; Anderson, D.R. Depletion of B cells in vivo by a chimeric mouse human monoclonal antibody to CD20. Blood 1994, 83, 435–445. [Google Scholar] [CrossRef]
  18. Harjunpaa, A.; Junnikkala, S.; Meri, S. Rituximab (anti-CD20) therapy of B-cell lymphomas: Direct complement killing is superior to cellular effector mechanisms. Scand. J. Immunol. 2000, 51, 634–641. [Google Scholar] [CrossRef]
  19. Shan, D.; Ledbetter, J.A.; Press, O.W. Apoptosis of malignant human B cells by ligation of CD20 with monoclonal antibodies. Blood 1998, 91, 1644–1652. [Google Scholar] [CrossRef]
  20. Byrd, J.C.; Kitada, S.; Flinn, I.W.; Aron, J.L.; Pearson, M.; Lucas, D.; Reed, J.C. The mechanism of tumor cell clearance by rituximab in vivo in patients with B-cell chronic lymphocytic leukemia: Evidence of caspase activation and apoptosis induction. Blood 2002, 99, 1038–1043. [Google Scholar] [CrossRef]
  21. Mossner, E.; Brunker, P.; Moser, S.; Puntener, U.; Schmidt, C.; Herter, S.; Grau, R.; Gerdes, C.; Nopora, A.; van Puijenbroek, E.; et al. Increasing the efficacy of CD20 antibody therapy through the engineering of a new type II anti-CD20 antibody with enhanced direct and immune effector cell-mediated B-cell cytotoxicity. Blood 2010, 115, 4393–4402. [Google Scholar] [CrossRef]
  22. Agliardi, S.; Veronese, C.; Panzeri, F.; Palazzini, S.; Guarnieri, G.; Loiacono, S.; Martinelli, V.; Potenza, A.M.; Sbraga, E.; Rissotto, E.; et al. Immunocytokines in cancer treatment: A systematic review. Cancer Treat. Rev. 2025, 139, 102978. [Google Scholar] [CrossRef]
  23. Caserta, S.; Campo, C.; Cancemi, G.; Neri, S.; Stagno, F.; Mannina, D.; Allegra, A. Bispecific Antibodies and Antibody-Drug Conjugates in Relapsed/Refractory Aggressive Non-Hodgkin Lymphoma, Focusing on Diffuse Large B-Cell Lymphoma. Cancers 2025, 17, 2479. [Google Scholar] [CrossRef]
  24. Jensen, M.; Winkler, U.; Manzke, O.; Diehl, V.; Engert, A. Rapid tumor lysis in a patient with B-cell chronic lymphocytic leukemia and lymphocytosis treated with an anti-CD20 monoclonal antibody (IDEC-C2B8, rituximab). Ann. Hematol. 1998, 77, 89–91. [Google Scholar] [CrossRef]
  25. Winkler, U.; Jensen, M.; Manzke, O.; Schulz, H.; Diehl, V.; Engert, A. Cytokine-release syndrome in patients with B-cell chronic lymphocytic leukemia and high lymphocyte counts after treatment with an anti-CD20 monoclonal antibody (rituximab, IDEC-C2B8). Blood 1999, 94, 2217–2224. [Google Scholar] [CrossRef]
  26. Lim, L.C.; Koh, L.P.; Tan, P. Fatal cytokine release syndrome with chimeric anti-CD20 monoclonal antibody rituximab in a 71-year-old patient with chronic lymphocytic leukemia. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 1999, 17, 1962–1963. [Google Scholar]
  27. Amitai, I.; Gafter-Gvili, A.; Shargian-Alon, L.; Raanani, P.; Gurion, R. Obinutuzumab-related adverse events: A systematic review and meta-analysis. Hematol. Oncol. 2021, 39, 215–221. [Google Scholar] [CrossRef]
  28. Kou, K.; Zhou, Q.; Du, L.; Xu, F. Obinutuzumab-induced severe acute thrombocytopenia: A case report and literature review. Front. Immunol. 2025, 16, 1609862. [Google Scholar] [CrossRef]
  29. Davis, T.A.; Czerwinski, D.K.; Levy, R. Therapy of B-cell lymphoma with anti-CD20 antibodies can result in the loss of CD20 antigen expression. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 1999, 5, 611–615. [Google Scholar]
  30. Kinoshita, T.; Nagai, H.; Murate, T.; Saito, H. CD20-negative relapse in B-cell lymphoma after treatment with Rituximab. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 1998, 16, 3916. [Google Scholar]
  31. Donati, G.; Amati, B. MYC and therapy resistance in cancer: Risks and opportunities. Mol. Oncol. 2022, 16, 3828–3854. [Google Scholar] [CrossRef]
  32. de Jonge, A.V.; Csikos, T.; Eken, M.; Bulthuis, E.P.; Poddighe, P.J.; Roemer, M.G.M.; Chamuleau, M.E.D.; Mutis, T. Delineating MYC-Mediated Escape Mechanisms from Conventional and T Cell-Redirecting Therapeutic Antibodies. Int. J. Mol. Sci. 2024, 25, 12094. [Google Scholar] [CrossRef]
  33. Staerz, U.D.; Bevan, M.J. Hybrid hybridoma producing a bispecific monoclonal antibody that can focus effector T-cell activity. Proc. Natl. Acad. Sci. USA 1986, 83, 1453–1457, Erratum in Proc. Natl. Acad. Sci. USA 1986, 83, 4972.. [Google Scholar] [CrossRef]
  34. Staerz, U.D.; Kanagawa, O.; Bevan, M.J. Hybrid antibodies can target sites for attack by T cells. Nature 1985, 314, 628–631. [Google Scholar] [CrossRef]
  35. Ma, J.; Mo, Y.; Tang, M.; Shen, J.; Qi, Y.; Zhao, W.; Huang, Y.; Xu, Y.; Qian, C. Bispecific Antibodies: From Research to Clinical Application. Front. Immunol. 2021, 12, 626616. [Google Scholar] [CrossRef]
  36. Nagorsen, D.; Kufer, P.; Baeuerle, P.A.; Bargou, R. Blinatumomab: A historical perspective. Pharmacol. Ther. 2012, 136, 334–342. [Google Scholar] [CrossRef]
  37. Topp, M.S.; Gokbuget, N.; Zugmaier, G.; Degenhard, E.; Goebeler, M.E.; Klinger, M.; Neumann, S.A.; Horst, H.A.; Raff, T.; Viardot, A.; et al. Long-term follow-up of hematologic relapse-free survival in a phase 2 study of blinatumomab in patients with MRD in B-lineage ALL. Blood 2012, 120, 5185–5187. [Google Scholar] [CrossRef]
  38. Fischer, C.; Chen, S.S.; Nimmerfroh, J.; Eugster, A.; Stucheli, S.; Schultheiss, C.; Widmer, C.; Heim, D.; Kasenda, B.; Passweg, J.; et al. IGLV3-21(R110)-directed bispecific antibodies activate T cells and promote killing in a high-risk subset of chronic lymphocytic leukemia. Haematologica 2025. [Google Scholar] [CrossRef]
  39. Markl, F.; Schultheiss, C.; Ali, M.; Chen, S.S.; Zintchenko, M.; Egli, L.; Mietz, J.; Chijioke, O.; Paschold, L.; Spajic, S.; et al. Mutation-specific CAR T cells as precision therapy for IGLV3-21(R110) expressing high-risk chronic lymphocytic leukemia. Nat. Commun. 2024, 15, 993. [Google Scholar] [CrossRef]
  40. Amoozgar, B.; Bangolo, A.; Habibi, M.; Cho, C.; Goy, A. From Molecular Precision to Clinical Practice: A Comprehensive Review of Bispecific and Trispecific Antibodies in Hematologic Malignancies. Int. J. Mol. Sci. 2025, 26, 5319. [Google Scholar] [CrossRef]
  41. Chang, A.; Pelletier, A.N.; McGuire, D.J.; Tsagiopoulou, M.; Karipidou, M.; Ayers, A.; Leal, A.M.K.; Churnetski, M.C.; O’Leary, C.B.; Switchenko, J.M.; et al. PD-1 expression identifies proliferating malignant CLL B cells and is a potential biomarker of response to BTK inhibitor therapy. Proc. Natl. Acad. Sci. USA 2025, 122, e2426935122. [Google Scholar] [CrossRef]
  42. Hayday, A.; Dechanet-Merville, J.; Rossjohn, J.; Silva-Santos, B. Cancer immunotherapy by gammadelta T cells. Science 2024, 386, eabq7248. [Google Scholar] [CrossRef]
  43. Marischen, L.; Fritsch, J.; Ilic, J.; Wahl, L.; Bertsch, T.; Knop, S.; Bold, A. Two Are Better than One: The Bi-Specific Antibody Mosunetuzumab Reveals an Improved Immune Response of Vgamma9Vdelta2 T Cells Targeting CD20 in Malignant B Cells in Comparison to the Mono-Specific Antibody Obinutuzumab. Int. J. Mol. Sci. 2025, 26, 1262. [Google Scholar] [CrossRef]
  44. Budde, L.E.; Assouline, S.; Sehn, L.H.; Schuster, S.J.; Yoon, S.S.; Yoon, D.H.; Matasar, M.J.; Bosch, F.; Kim, W.S.; Nastoupil, L.J.; et al. Single-Agent Mosunetuzumab Shows Durable Complete Responses in Patients with Relapsed or Refractory B-Cell Lymphomas: Phase I Dose-Escalation Study. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2022, 40, 481–491. [Google Scholar] [CrossRef]
  45. Budde, L.E.; Sehn, L.H.; Matasar, M.; Schuster, S.J.; Assouline, S.; Giri, P.; Kuruvilla, J.; Canales, M.; Dietrich, S.; Fay, K.; et al. Safety and efficacy of mosunetuzumab, a bispecific antibody, in patients with relapsed or refractory follicular lymphoma: A single-arm, multicentre, phase 2 study. Lancet Oncol. 2022, 23, 1055–1065. [Google Scholar] [CrossRef]
  46. Bayly-McCredie, E.; Treisman, M.; Fiorenza, S. Safety and Efficacy of Bispecific Antibodies in Adults with Large B-Cell Lymphomas: A Systematic Review of Clinical Trial Data. Int. J. Mol. Sci. 2024, 25, 9736. [Google Scholar] [CrossRef]
  47. Juarez-Salcedo, L.M.; Nimkar, S.; Corazon, A.M.; Dalia, S. Loncastuximab Tesirine in the Treatment of Relapsed or Refractory Diffuse Large B-Cell Lymphoma. Int. J. Mol. Sci. 2024, 25, 7580. [Google Scholar] [CrossRef]
  48. Mullard, A. FDA approves ADC Therapeutics’ loncastuximab tesirine, ushering in a new cytotoxic payload. Nat. Rev. Drug Discov. 2021, 20, 414. [Google Scholar] [CrossRef]
  49. Cremasco, F.; Menietti, E.; Speziale, D.; Sam, J.; Sammicheli, S.; Richard, M.; Varol, A.; Klein, C.; Umana, P.; Bacac, M.; et al. Cross-linking of T cell to B cell lymphoma by the T cell bispecific antibody CD20-TCB induces IFNgamma/CXCL10-dependent peripheral T cell recruitment in humanized murine model. PLoS ONE 2021, 16, e0241091. [Google Scholar] [CrossRef]
  50. Sun, L.L.; Ellerman, D.; Mathieu, M.; Hristopoulos, M.; Chen, X.; Li, Y.; Yan, X.; Clark, R.; Reyes, A.; Stefanich, E.; et al. Anti-CD20/CD3 T cell-dependent bispecific antibody for the treatment of B cell malignancies. Sci. Transl. Med. 2015, 7, 287ra70. [Google Scholar] [CrossRef]
  51. Pfeifer, M.; Zheng, B.; Erdmann, T.; Koeppen, H.; McCord, R.; Grau, M.; Staiger, A.; Chai, A.; Sandmann, T.; Madle, H.; et al. Anti-CD22 and anti-CD79B antibody drug conjugates are active in different molecular diffuse large B-cell lymphoma subtypes. Leukemia 2015, 29, 1578–1586. [Google Scholar] [CrossRef] [PubMed]
  52. Damiani, D.; Tiribelli, M. Monoclonal Antibodies Against Myeloid Leukemia Cells: Current Knowledge and Future Directions. Int. J. Mol. Sci. 2025, 26, 4571. [Google Scholar] [CrossRef] [PubMed]
  53. Woyach, J.A.; Furman, R.R.; Liu, T.M.; Ozer, H.G.; Zapatka, M.; Ruppert, A.S.; Xue, L.; Li, D.H.; Steggerda, S.M.; Versele, M.; et al. Resistance mechanisms for the Bruton’s tyrosine kinase inhibitor ibrutinib. N. Engl. J. Med. 2014, 370, 2286–2294. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Visual representation of the antibody therapy classes. (a) A pure murine antibody, as derived from hybridoma cell lines. (b) A chimeric antibody with humanized (orange) fAb regions, improving the immunogenicity and half-life. A notable example is rituximab, which targets CD20. (c) A glycoengineered antibody with a reduced fucose Fc region (purple), improving the antibody-dependent cellular cytotoxicity (ADCC). A notable example is obinutuzumab, which also targets CD20. (d) An antibody–drug conjugate with cytotoxic payload (red). A notable example is polatuzumab vedotin, which targets CD79b and delivers a payload of monomethyl auristatin E. (e) A bispecific antibody (shown as blue versus green) with distinct FAb complementarity. A notable example is glofitamab, which targets CD20 and CD3. Binding CD3 on a CD8+ T-cell activates its cytotoxic activity toward the approximated CD20+ cell. Adopted from Naman et al. [16].
Figure 1. Visual representation of the antibody therapy classes. (a) A pure murine antibody, as derived from hybridoma cell lines. (b) A chimeric antibody with humanized (orange) fAb regions, improving the immunogenicity and half-life. A notable example is rituximab, which targets CD20. (c) A glycoengineered antibody with a reduced fucose Fc region (purple), improving the antibody-dependent cellular cytotoxicity (ADCC). A notable example is obinutuzumab, which also targets CD20. (d) An antibody–drug conjugate with cytotoxic payload (red). A notable example is polatuzumab vedotin, which targets CD79b and delivers a payload of monomethyl auristatin E. (e) A bispecific antibody (shown as blue versus green) with distinct FAb complementarity. A notable example is glofitamab, which targets CD20 and CD3. Binding CD3 on a CD8+ T-cell activates its cytotoxic activity toward the approximated CD20+ cell. Adopted from Naman et al. [16].
Ijms 26 09463 g001
Figure 2. Targeting effector cells with bispecific antibodies for cancer therapy—illustrating mechanisms of T-cell and NK-cell redirection. Adopted from Amoozgar et al. [40].
Figure 2. Targeting effector cells with bispecific antibodies for cancer therapy—illustrating mechanisms of T-cell and NK-cell redirection. Adopted from Amoozgar et al. [40].
Ijms 26 09463 g002
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.

Share and Cite

MDPI and ACS Style

Butchar, J.P. Special Issue: Antibody Therapy for Hematologic Malignancies. Int. J. Mol. Sci. 2025, 26, 9463. https://doi.org/10.3390/ijms26199463

AMA Style

Butchar JP. Special Issue: Antibody Therapy for Hematologic Malignancies. International Journal of Molecular Sciences. 2025; 26(19):9463. https://doi.org/10.3390/ijms26199463

Chicago/Turabian Style

Butchar, Jonathan P. 2025. "Special Issue: Antibody Therapy for Hematologic Malignancies" International Journal of Molecular Sciences 26, no. 19: 9463. https://doi.org/10.3390/ijms26199463

APA Style

Butchar, J. P. (2025). Special Issue: Antibody Therapy for Hematologic Malignancies. International Journal of Molecular Sciences, 26(19), 9463. https://doi.org/10.3390/ijms26199463

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop