Next Article in Journal
Oxygenation of the Prefrontal Cortex during Memory Interference
Next Article in Special Issue
Immunological and Clinical Impact of Manipulated and Unmanipulated DLI after Allogeneic Stem Cell Transplantation of AML Patients
Previous Article in Journal
Therapeutic Potential of AAV1-Rheb(S16H) Transduction Against Alzheimer’s Disease
Previous Article in Special Issue
FLAMSA-RIC for Stem Cell Transplantation in Patients with Acute Myeloid Leukemia and Myelodysplastic Syndromes: A Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

The Important Role of Immunotherapies in Acute Myeloid Leukemia

1
Department of Internal Medicine, Diakonie Hospital, 70176 Stuttgart, Germany
2
Department of Internal Medicine III, University Hospital of Ulm, 89081 Ulm, Germany
J. Clin. Med. 2019, 8(12), 2054; https://doi.org/10.3390/jcm8122054
Submission received: 18 November 2019 / Accepted: 20 November 2019 / Published: 22 November 2019
(This article belongs to the Special Issue Immunotherapies for Acute Myeloid Leukemia)
This series on immunotherapies in acute myeloid leukemia (AML) aims to give readers new insights on established but also emerging immunotherapeutic approaches for AML patients. The therapeutic landscape in AML is rapidly changing, and several drugs have been developed and approved such as first and second generation FLT3 inhibitors [1,2,3], IDH1 and 2-inhibitors [4,5], demethylating agents, liposomal cytarabine and daunorubicin (CPX-351) [6], venetoclax [7,8] and the hedgehog pathway inhibitor glasdegib. However, relapse after intensive chemotherapy or allogeneic hematopoietic stem cell transplantation is one of the major obstacles impeding the complete elimination of all AML cells [9]. Thus, although the median overall survival for AML patients has increased, it still remains relatively low [10].
Therefore, immunotherapeutic approaches might be an option to prevent disease relapse and to eliminate leukemic cells or leukemic stem cells (LSC) that survive intensive treatment approaches. The efficacy of immunotherapeutic approaches has become ever more evident in solid tumors, especially immune-checkpoint inhibitors that are routinely used in several solid tumor entities, but also lymphoma [11,12].
Our focus in this special issue is different strategies of immunotherapeutic approaches in AML.
Some of the immunotherapies in the treatment of AML, such as allogeneic hematopoietic stem cell transplantation (HSCT) and donor lymphocyte infusion (DLI), have been part of routine clinical practice in the treatment of AML for a long time, whereas other immunotherapeutic approaches have only recently entered clinical practice or need to be further developed. A key aspect is the mechanisms underlying the cure of AML patients, which are based on the graft-versus-leukemia (GvL) effect, in which allogeneic T cells recognize target antigens on malignant cells by T cell approaches including DLI. An effective and well-tolerated regimen for HSCT in patients with AML and MDS is the FLAMSA-RIC regimen, and therefore novel data of this approach are presented in this issue [13].
It is very appropriate to utilize DLI after allogeneic HSCT to prevent relapse, to prolong progression-free survival, to establish full donor chimerism, and to restore the GvL effect in patients with hematological malignancies. There are different strategies to use DLI in a therapeutic setting for the treatment of morphological relapse, and also for prophylactic use in AML/MDS and DLI administered preemptively. There is also the approach of antigen-directed immunogenic and specifically stimulated and modified DLI as well as virus-specific donor T cells and third-party DLI [14].
DC-based immunotherapies also have the potential to bring about demonstrable clinical responses in AML patients, although there has not been a complete breakthrough for this type of therapy until today. Van Acker et al. have highlighted different DC strategies in AML [15].
Leukemia-associated antigens (LAAs) represent immunogenic structures to target LSC [16,17], and LAA might be relevant for the elimination of malignant cells by cytotoxic T lymphocytes. Therefore, LAAs might be a good target for specific immunotherapeutic approaches. Several LAAs have been identified in the context of malignant hematological diseases [16,18,19], and in clinical phase I/II peptide vaccination trials, some LAAs showed immunological as well as clinical responses [20,21,22,23].
In this special issue, we also elucidate antibody-based therapies in AML, such as T cell activating antibodies including immune-checkpoint inhibitors and diverse monoclonal antibodies [11,12,24]. Immune-checkpoint inhibitors have changed clinical treatment algorithms of malignant diseases such as malignant melanoma, lung cancer, as well as lymphoma. Today, immune-checkpoint inhibitors are not yet established in the routine treatment of AML but should be considered as further immunotherapeutic options in the future, especially in the context of allogeneic stem cell transplantation [24]. Further antibody-directed approaches such as unconjugated, toxin-conjugated, radio-conjugated, and multivalent formats of antibody-based therapy, are demonstrating the potential of a diverse leukemia-derived antibody strategy which is already established in acute lymphoblastic leukemia and are summarized in one section of this issue [25].
Chimeric antigen receptor T cells (CARs) are highly effective in the treatment of refractory and relapsed acute lymphoblastic leukemia, to some lower extent in aggressive lymphoma, but also in multiple myeloma [26]. However, early CAR-T cell approaches are also being tested in AML with interesting target structures, and these strategies are described in this issue [27]. Immune responses are complex and are also influenced by T cell cross-talk and communication by cytokines and the communication of leukemic cells with their microenvironment, as presented by Reikvam et al. [28] in this issue.
All of these aspects emphasize the high potential of immunotherapeutic approaches to improve the survival of AML patients in the future, where combination therapies utilizing immunotherapeutic drugs could represent further innovation strategies to further improve the treatment of AML.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Stone, R.M.; Mandrekar, S.J.; Sanford, B.L.; Laumann, K.; Geyer, S.; Bloomfield, C.D.; Thiede, C.; Prior, T.W.; Dohner, K.; Marcucci, G.; et al. Midostaurin plus Chemotherapy for Acute Myeloid Leukemia with a FLT3 Mutation. N. Engl. J. Med. 2017, 377, 454–464. [Google Scholar] [CrossRef] [PubMed]
  2. Perl, A.E.; Martinelli, G.; Cortes, J.E.; Neubauer, A.; Berman, E.; Paolini, S.; Montesinos, P.; Baer, M.R.; Larson, R.A.; Ustun, C.; et al. Gilteritinib or Chemotherapy for Relapsed or Refractory FLT3-Mutated AML. N. Engl. J. Med. 2019, 381, 1728–1740. [Google Scholar] [CrossRef] [PubMed]
  3. Cortes, J.E.; Khaled, S.; Martinelli, G.; Perl, A.E.; Ganguly, S.; Russell, N.; Kramer, A.; Dombret, H.; Hogge, D.; Jonas, B.A.; et al. Quizartinib versus salvage chemotherapy in relapsed or refractory FLT3-ITD acute myeloid leukaemia (QuANTUM-R): A multicentre, randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2019, 20, 984–997. [Google Scholar] [CrossRef]
  4. DiNardo, C.D.; Stein, E.M.; de Botton, S.; Roboz, G.J.; Altman, J.K.; Mims, A.S.; Swords, R.; Collins, R.H.; Mannis, G.N.; Pollyea, D.A.; et al. Durable Remissions with Ivosidenib in IDH1-Mutated Relapsed or Refractory AML. N. Engl. J. Med. 2018, 378, 2386–2398. [Google Scholar] [CrossRef]
  5. Stein, E.M.; DiNardo, C.D.; Pollyea, D.A.; Fathi, A.T.; Roboz, G.J.; Altman, J.K.; Stone, R.M.; DeAngelo, D.J.; Levine, R.L.; Flinn, I.W.; et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood 2017, 130, 722–731. [Google Scholar] [CrossRef]
  6. Lancet, J.E.; Uy, G.L.; Cortes, J.E.; Newell, L.F.; Lin, T.L.; Ritchie, E.K.; Stuart, R.K.; Strickland, S.A.; Hogge, D.; Solomon, S.R.; et al. CPX-351 (cytarabine and daunorubicin) Liposome for Injection Versus Conventional Cytarabine Plus Daunorubicin in Older Patients With Newly Diagnosed Secondary Acute Myeloid Leukemia. J. Clin. Oncol. 2018, 36, 2684–2692. [Google Scholar] [CrossRef]
  7. DiNardo, C.D.; Pratz, K.; Pullarkat, V.; Jonas, B.A.; Arellano, M.; Becker, P.S.; Frankfurt, O.; Konopleva, M.; Wei, A.H.; Kantarjian, H.M.; et al. Venetoclax combined with decitabine or azacitidine in treatment-naive, elderly patients with acute myeloid leukemia. Blood 2019, 133, 7–17. [Google Scholar] [CrossRef]
  8. Wei, A.H.; Strickland, S.A., Jr.; Hou, J.Z.; Fiedler, W.; Lin, T.L.; Walter, R.B.; Enjeti, A.; Tiong, I.S.; Savona, M.; Lee, S.; et al. Venetoclax Combined With Low-Dose Cytarabine for Previously Untreated Patients With Acute Myeloid Leukemia: Results From a Phase Ib/II Study. J. Clin. Oncol. 2019, 37, 1277–1284. [Google Scholar] [CrossRef]
  9. Lee, C.J.; Savani, B.N.; Mohty, M.; Gorin, N.C.; Labopin, M.; Ruggeri, A.; Schmid, C.; Baron, F.; Esteve, J.; Giebel, S.; et al. Post-remission strategies for the prevention of relapse following allogeneic hematopoietic cell transplantation for high-risk acute myeloid leukemia: Expert review from the Acute Leukemia Working Party of the European Society for Blood and Marrow Transplantation. Bone Marrow Transplant. 2018. [Google Scholar] [CrossRef]
  10. Dohner, H.; Estey, E.; Grimwade, D.; Amadori, S.; Appelbaum, F.R.; Buchner, T.; Dombret, H.; Ebert, B.L.; Fenaux, P.; Larson, R.A.; et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017, 129, 424–447. [Google Scholar] [CrossRef] [PubMed]
  11. Annibali, O.; Crescenzi, A.; Tomarchio, V.; Pagano, A.; Bianchi, A.; Grifoni, A.; Avvisati, G. PD-1 /PD-L1 checkpoint in hematological malignancies. Leuk. Res. 2018, 67, 45–55. [Google Scholar] [CrossRef] [PubMed]
  12. Gravbrot, N.; Gilbert-Gard, K.; Mehta, P.; Ghotmi, Y.; Banerjee, M.; Mazis, C.; Sundararajan, S. Therapeutic Monoclonal Antibodies Targeting Immune Checkpoints for the Treatment of Solid Tumors. Antibodies 2019, 8, 51. [Google Scholar] [CrossRef] [PubMed]
  13. Owattanapanich, W.; Ungprasert, P.; Wais, V.; Kungwankiattichai, S.; Bunjes, D.; Kuchenbauer, F. FLAMSA-RIC for Stem Cell Transplantation in Patients with Acute Myeloid Leukemia and Myelodysplastic Syndromes: A Systematic Review and Meta-Analysis. J. Clin. Med. 2019, 8, 1437. [Google Scholar] [CrossRef] [PubMed]
  14. Greiner, J.; Götz, M.; Bunjes, D.; Hofmann, S.; Wais, V. Immunological and clinical impact of manipulated and unmanipulated DLI after allogeneic stem cell transplantation (allo-SCT) of AML patients. J. Clin. Med. 2019. submitted for publication. [Google Scholar]
  15. Van Acker, H.H.; Versteven, M.; Lichtenegger, F.S.; Roex, G.; Campillo-Davo, D.; Lion, E.; Subklewe, M.; Van Tendeloo, V.F.; Berneman, Z.N.; Anguille, S. Dendritic Cell-Based Immunotherapy of Acute Myeloid Leukemia. J. Clin. Med. 2019, 8, 579. [Google Scholar] [CrossRef]
  16. Anguille, S.; Van Tendeloo, V.F.; Berneman, Z.N. Leukemia-associated antigens and their relevance to the immunotherapy of acute myeloid leukemia. Leukemia 2012, 26, 2186–2196. [Google Scholar] [CrossRef]
  17. Schneider, V.; Zhang, L.; Rojewski, M.; Fekete, N.; Schrezenmeier, H.; Erle, A.; Bullinger, L.; Hofmann, S.; Gotz, M.; Dohner, K.; et al. Leukemic progenitor cells are susceptible to targeting by stimulated cytotoxic T cells against immunogenic leukemia-associated antigens. Int. J. Cancer 2015, 137, 2083–2092. [Google Scholar] [CrossRef]
  18. Greiner, J.; Schmitt, M.; Li, L.; Giannopoulos, K.; Bosch, K.; Schmitt, A.; Dohner, K.; Schlenk, R.F.; Pollack, J.R.; Dohner, H.; et al. Expression of tumor-associated antigens in acute myeloid leukemia: Implications for specific immunotherapeutic approaches. Blood 2006, 108, 4109–4117. [Google Scholar] [CrossRef]
  19. Greiner, J.; Ono, Y.; Hofmann, S.; Schmitt, A.; Mehring, E.; Gotz, M.; Guillaume, P.; Dohner, K.; Mytilineos, J.; Dohner, H.; et al. Mutated regions of nucleophosmin 1 elicit both CD4(+) and CD8(+) T-cell responses in patients with acute myeloid leukemia. Blood 2012, 120, 1282–1289. [Google Scholar] [CrossRef]
  20. Schmitt, M.; Schmitt, A.; Rojewski, M.T.; Chen, J.; Giannopoulos, K.; Fei, F.; Yu, Y.; Gotz, M.; Heyduk, M.; Ritter, G.; et al. RHAMM-R3 peptide vaccination in patients with acute myeloid leukemia, myelodysplastic syndrome, and multiple myeloma elicits immunologic and clinical responses. Blood 2008, 111, 1357–1365. [Google Scholar] [CrossRef]
  21. Rezvani, K.; Yong, A.S.; Mielke, S.; Savani, B.N.; Musse, L.; Superata, J.; Jafarpour, B.; Boss, C.; Barrett, A.J. Leukemia-associated antigen-specific T-cell responses following combined PR1 and WT1 peptide vaccination in patients with myeloid malignancies. Blood 2008, 111, 236–242. [Google Scholar] [CrossRef] [PubMed]
  22. Greiner, J.; Schmitt, A.; Giannopoulos, K.; Rojewski, M.T.; Gotz, M.; Funk, I.; Ringhoffer, M.; Bunjes, D.; Hofmann, S.; Ritter, G.; et al. High-dose RHAMM-R3 peptide vaccination for patients with acute myeloid leukemia, myelodysplastic syndrome and multiple myeloma. Haematologica 2010, 95, 1191–1197. [Google Scholar] [CrossRef] [PubMed]
  23. Rezvani, K.; Yong, A.S.; Mielke, S.; Jafarpour, B.; Savani, B.N.; Le, R.Q.; Eniafe, R.; Musse, L.; Boss, C.; Kurlander, R.; et al. Repeated PR1 and WT1 peptide vaccination in Montanide-adjuvant fails to induce sustained high-avidity, epitope-specific CD8+ T cells in myeloid malignancies. Haematologica 2011, 96, 432–440. [Google Scholar] [CrossRef] [PubMed]
  24. Giannopoulos, K. Targeting Immune Signaling Checkpoints in Acute Myeloid Leukemia. J. Clin. Med. 2019, 8, 236. [Google Scholar] [CrossRef]
  25. Williams, B.A.; Law, A.; Hunyadkurti, J.; Desilets, S.; Leyton, J.V.; Keating, A. Antibody Therapies for Acute Myeloid Leukemia: Unconjugated, Toxin-Conjugated, Radio-Conjugated and Multivalent Formats. J. Clin. Med. 2019, 8, 1261. [Google Scholar] [CrossRef]
  26. Majzner, R.G.; Mackall, C.L. Clinical lessons learned from the first leg of the CAR T cell journey. Nat. Med. 2019, 25, 1341–1355. [Google Scholar] [CrossRef]
  27. Hofmann, S.; Schubert, M.L.; Wang, L.; He, B.; Neuber, B.; Dreger, P.; Muller-Tidow, C.; Schmitt, M. Chimeric Antigen Receptor (CAR) T Cell Therapy in Acute Myeloid Leukemia (AML). J. Clin. Med. 2019, 8, 200. [Google Scholar] [CrossRef]
  28. Reikvam, H.; Aasebo, E.; Brenner, A.K.; Bartaula-Brevik, S.; Gronningsaeter, I.S.; Forthun, R.B.; Hovland, R.; Bruserud, O. High Constitutive Cytokine Release by Primary Human Acute Myeloid Leukemia Cells Is Associated with a Specific Intercellular Communication Phenotype. J. Clin. Med. 2019, 8, 970. [Google Scholar] [CrossRef] [Green Version]

Share and Cite

MDPI and ACS Style

Greiner, J. The Important Role of Immunotherapies in Acute Myeloid Leukemia. J. Clin. Med. 2019, 8, 2054. https://doi.org/10.3390/jcm8122054

AMA Style

Greiner J. The Important Role of Immunotherapies in Acute Myeloid Leukemia. Journal of Clinical Medicine. 2019; 8(12):2054. https://doi.org/10.3390/jcm8122054

Chicago/Turabian Style

Greiner, Jochen. 2019. "The Important Role of Immunotherapies in Acute Myeloid Leukemia" Journal of Clinical Medicine 8, no. 12: 2054. https://doi.org/10.3390/jcm8122054

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