Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1041 word(s) 1041 2021-08-25 05:55:51 |
2 Hanging sentence was deleted -12 word(s) 1029 2021-08-26 20:54:02 | |
3 Reference formatted + formatting -12 word(s) 1029 2021-08-27 03:21:40 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Zayac, A.; Reagan, J. Acute Myeloid Leukemia for Elders. Encyclopedia. Available online: https://encyclopedia.pub/entry/13573 (accessed on 19 April 2024).
Zayac A, Reagan J. Acute Myeloid Leukemia for Elders. Encyclopedia. Available at: https://encyclopedia.pub/entry/13573. Accessed April 19, 2024.
Zayac, Adam, John Reagan. "Acute Myeloid Leukemia for Elders" Encyclopedia, https://encyclopedia.pub/entry/13573 (accessed April 19, 2024).
Zayac, A., & Reagan, J. (2021, August 26). Acute Myeloid Leukemia for Elders. In Encyclopedia. https://encyclopedia.pub/entry/13573
Zayac, Adam and John Reagan. "Acute Myeloid Leukemia for Elders." Encyclopedia. Web. 26 August, 2021.
Acute Myeloid Leukemia for Elders
Edit

Acute myeloid leukemia (AML) is an aggressive hematologic malignancy affecting about 0.5% of people in their lifetime. Over the last few decades, a growing understanding of AML has revealed it to be a heterogenous disease with a widely variable prognosis. This is largely driven by disease biology, the ability to tolerate highly toxic multi-agent chemotherapy and, in most cases, undergo allogeneic stem cell transplantation to be cured of disease.

acute myeloid leukemia older adult frontline treatment

1. Introduction

Acute myeloid leukemia (AML) is an aggressive hematologic malignancy affecting about 0.5% of people in their lifetime. Over the last few decades, a growing understanding of AML has revealed it to be a heterogenous disease with a widely variable prognosis. This is largely driven by disease biology, the ability to tolerate highly toxic multi-agent chemotherapy and, in most cases, undergo allogeneic stem cell transplantation to be cured of disease. In the best of circumstances, this is a tenuous situation with life-altering implications. Our review will focus on the characteristics of AML in “older” patients and discuss frontline management approaches for this population that can range in terms of performance status from “fit” to medically “frail”. We also discuss future directions for treatment in this disproportionately afflicted, vulnerable population.

2. AML in “Older” Adults

In older adults, monosomal karyotypes, −5 and −7, as well as other adverse cytogenetic abnormalities, including 17p, 11q, +8, and complex karyotypes predominate, while favorable cytogenetic abnormalities are uncommon [1]. In a cytogenetic analysis of CALGB-8461 that compromised patients over 60 years with predominantly de novo AML (97.5%), a complex karyotype with ≥3 abnormalities (19% of patients) and “rare aberrations” (5% of patients) were associated with lower complete remission (CR) rates, while complex karyotype with ≥5 abnormalities (15% of patients) and “rare aberrations” were also associated with inferior disease-free survival (DFS) and overall survival (OS) [2].

Despite these patients being able to tolerate intensive chemotherapy, most patients did not fare well given their age and/or cytogenetic features [3]. Interestingly, in regards to non-trial data, an analysis of the SEER database found improvements in the response rates (RR) and 12-month survival with each decade from 1977–2006 for patients who were 65–74 years old but no improvement for their 75 years of age and older counterparts despite the approval of agents to treat older patients within this timeframe [4]. This variation in survival underscores a need to discuss the prognosis and treatment of those between age 60 and 74 years old and patients 75 years old or more separately. Later in this review, we discuss the treatment of those between age 60 and 74 years old and patients 75 years old or more separately.

While not initially created for the assessment of oncology patients, they have subsequently been found to predict mortality and chemotherapy-related adverse effects [5][6]. AML and its treatment is among the most intensive stressors that a person can experience. Often, it evolves rapidly and may even require lengthy hospitalization and intensive supportive care. Several studies have gone on to assess the role and feasibility of geriatric assessments in patients receiving induction chemotherapy and even stem cell transplantation for AML in order to accurately depict the effect of induction chemotherapy on older patients [7][8][9][10][11][12][13].

In patients treated with “non-intensive” regimens, KPS < 80, an elevated fatigue index, and a diminished activity of daily living (ADL) index were associated with worse overall survival, which was seen in patients treated with best supportive care only or hypomethylating agents [10]. Utilizing the information obtained from GAs and combining it with cytogenetic and molecular information to optimally tailor individual treatment within this heterogenous group is under study and likely represents a step forward in the treatment of AML in older adults [14].

3. Consolidation Treatment

While most patients treated with induction chemotherapy have a complete remission, durability following treatment was an early issue with intensive therapy until post-remission, or consolidation, treatments were studied and found to be beneficial in sustaining these responses and potentially resulting in long-term cure. Consolidation is often achieved with either chemotherapy alone or with chemotherapy followed by hematopoietic stem cell transplantation.

However, given the complexity of the treatment schedule and multiagent approach, it is difficult to extrapolate these results to patients receiving single-agent cytarabine as a consolidation treatment. An alternative dosing schedule HIDAC has shown promise in young patients with quicker hematologic recovery, less days in the hospital, lower infection rates and no difference in survival with or without co-administration of peg-filgrastim compared with standard HIDAC dosing [15]. Further evaluation in additional randomized trials amongst older patients is needed. Notably patients treated with HMA and venetoclax do not typically undergo consolidation chemotherapy and instead remain on HMA/venetoclax as long as the response continues or toxicities are not seen.

Reduced-intensity conditioning provides an avenue for allo-SCT in older patients with lower TRM and LFS, higher relapse rates, and similar overall survival [16][17]. Allo-SCT with a HLA-matched donor, particularly from a sibling donor, appears to be a more effective consolidation method than autologous stem cell transplantation (auto-SCT) [18][19][20]. Although relapses are more common with auto-SCT, it is associated with lower treatment-related mortality and similar overall survival [18][19][20][21]. Auto-SCT remains a safe and effective consolidation approach in some older AML patients who do not have a readily available donor [22][23].

4. Measurable Residual Disease (MRD)

Measurable (previously minimal) residual disease has been a developing area of study over the last decade with significant clinical implications, and its assessment after completion of intensive therapy has even been included in recent guidelines [24][25]. Given the potential for sampling error or variation in bone marrow evaluation for morphologic evidence of persistent leukemia during or after treatment, more sensitive approaches were developed and studied [26]. The ability to detect the presence of minute numbers of cells (at least 1:10,000 and even 1:100,000) by one of three methods, multiparameter flow cytometry (MFC), next-generation sequencing (NGS), or reverse-transcriptase quantitative polymerase chain reaction (RT-qPCR) has been found to be prognostic and in other diseases (such as ALL) prompts initiation of specific alternate therapies [27][28][29][30][31][32].

Cytotoxic, targeted, and cellular therapies can lead to alterations and evolution in the molecular and genomic characteristics of any residual disease, emphasizing the importance of inclusion of NGS testing at some interval after these treatments to potentially guide future treatment options [33][34]. Additional studies are assessing combinations of these methods at various time points in the disease course and with different samples (peripheral blood vs. bone marrow) [35][36].

References

  1. Pinto, A.; Zagonel, V.; Ferrara, F. Acute myeloid leukemia in the elderly: Biology and therapeutic strategies. Crit. Rev. Oncol./Hematol. 2001, 39, 275–287.
  2. Farag, S.S.; Archer, K.J.; Mrózek, K.; Ruppert, A.S.; Carroll, A.J.; Vardiman, J.W.; Pettenati, M.J.; Baer, M.R.; Qumsiyeh, M.B.; Koduru, P.R.; et al. Pretreatment cytogenetics add to other prognostic factors predicting complete remission and long-term outcome in patients 60 years of age or older with acute myeloid leukemia: Results from Cancer and Leukemia Group B 8461. Blood 2006, 108, 63–73.
  3. Frohling, S.; Schlenk, R.F.; Kayser, S.; Morhardt, M.; Benner, A.; Dohner, K.; Dohner, H.; for the German-Austrian AML Study Group. Cytogenetics and age are major determinants of outcome in intensively treated acute myeloid leukemia patients older than 60 years: Results from AMLSG trial AML HD98-B. Blood 2006, 108, 3280–3288.
  4. Thein, M.S.; Ershler, W.B.; Jemal, A.; Yates, J.W.; Baer, M.R. Outcome of older patients with acute myeloid leukemia. Cancer 2013, 119, 2720–2727.
  5. Ramjaun, A.; Nassif, M.O.; Krotneva, S.; Huang, A.R.; Meguerditchian, A.N. Improved targeting of cancer care for older patients: A systematic review of the utility of comprehensive geriatric assessment. J. Geriatr. Oncol. 2013, 4, 271–281.
  6. Takahashi, M.; Takahashi, M.; Komine, K.; Yamada, H.; Kasahara, Y.; Chikamatsu, S.; Okita, A.; Ito, S.; Ouchi, K.; Okada, Y.; et al. The G8 screening tool enhances prognostic value to ECOG performance status in elderly cancer patients: A retrospective, single institutional study. PLoS ONE 2017, 12, e0179694.
  7. Klepin, H.D.; Geiger, A.M.; Tooze, J.A.; Kritchevsky, S.B.; Williamson, J.D.; Ellis, L.R.; Levitan, D.; Pardee, T.S.; Isom, S.; Powell, B.L.; et al. The feasibility of inpatient geriatric assessment for older adults receiving induction chemotherapy for acute myelogenous leukemia. J. Am. Geriatr. Soc. 2011, 59, 1837–1846.
  8. Klepin, H.D.; Geiger, A.M.; Tooze, J.A.; Kritchevsky, S.B.; Williamson, J.D.; Pardee, T.S.; Ellis, L.R.; Powell, B.L. Geriatric assessment predicts survival for older adults receiving induction chemotherapy for acute myelogenous leukemia. Blood 2013, 121, 4287–4294.
  9. Hamaker, M.E.; Prins, M.C.; Stauder, R. The relevance of a geriatric assessment for elderly patients with a haematological malignancy—A systematic review. Leuk. Res. 2014, 38, 275–283.
  10. Deschler, B.; Ihorst, G.; Platzbecker, U.; Germing, U.; Marz, E.; de Figuerido, M.; Fritzsche, K.; Haas, P.; Salih, H.R.; Giagounidis, A.; et al. Parameters detected by geriatric and quality of life assessment in 195 older patients with myelodysplastic syndromes and acute myeloid leukemia are highly predictive for outcome. Haematologica 2013, 98, 208–216.
  11. Klepin, H.D.; Ritchie, E.; Major-Elechi, B.; Le-Rademacher, J.; Seisler, D.; Storrick, L.; Sanford, B.L.; Marcucci, G.; Zhao, W.; Geyer, S.A.; et al. Geriatric assessment among older adults receiving intensive therapy for acute myeloid leukemia: Report of CALGB 361006 (Alliance). J. Geriatr. Oncol. 2020, 11, 107–113.
  12. Saad, M.; Loh, K.P.; Tooze, J.A.; Pardee, T.S.; Ellis, L.R.; Powell, B.L.; Bhave, R.R.; Geiger, A.M.; Williamson, J.D.; Kritchevsky, S.; et al. Geriatric assessment and survival among older adults receiving postremission therapy for acute myeloid leukemia. Blood 2020, 136, 2715–2719.
  13. Scheepers, E.R.M.; Vondeling, A.M.; Thielen, N.; Griend, R.v.d.; Stauder, R.; Hamaker, M.E. Geriatric assessment in older patients with a hematologic malignancy: A systematic review. Haematologica 2020, 105, 1484–1493.
  14. Bhatt, V.R.; Wichman, C.; Al-Kadhimi, Z.S.; Koll, T.T.; Berger, A.; Armitage, J.O.; Holstein, S.A.; Gundabolu, K.; Maness, L.J. Integrating Geriatric Assessment and Genetic Profiling to Personalize Therapy Selection in Older Adults with Acute Myeloid Leukemia (AML). Blood 2019, 134, 120.
  15. Jaramillo, S.; Benner, A.; Krauter, J.; Martin, H.; Kindler, T.; Bentz, M.; Salih, H.R.; Held, G.; Köhne, C.H.; Götze, K.; et al. Condensed versus standard schedule of high-dose cytarabine consolidation therapy with pegfilgrastim growth factor support in acute myeloid leukemia. Blood Cancer J. 2017, 7, e564.
  16. Scott, B.L.; Pasquini, M.C.; Logan, B.R.; Wu, J.; Devine, S.M.; Porter, D.L.; Maziarz, R.T.; Warlick, E.D.; Fernandez, H.F.; Alyea, E.P.; et al. Myeloablative Versus Reduced-Intensity Hematopoietic Cell Transplantation for Acute Myeloid Leukemia and Myelodysplastic Syndromes. J. Clin. Oncol. 2017, 35, 1154–1161.
  17. Zeng, W.; Huang, L.; Meng, F.; Liu, Z.; Zhou, J.; Sun, H. Reduced-intensity and myeloablative conditioning allogeneic hematopoietic stem cell transplantation in patients with acute myeloid leukemia and myelodysplastic syndrome: A meta-analysis and systematic review. Int. J. Clin. Exp. Med. 2014, 7, 4357–4368.
  18. Keating, A.; DaSilva, G.; Pérez, W.S.; Gupta, V.; Cutler, C.S.; Ballen, K.K.; Cairo, M.S.; Camitta, B.M.; Champlin, R.E.; Gajewski, J.L.; et al. Autologous blood cell transplantation versus HLA-identical sibling transplantation for acute myeloid leukemia in first complete remission: A registry study from the Center for International Blood and Marrow Transplantation Research. Haematologica 2013, 98, 185–192.
  19. Saraceni, F.; Labopin, M.; Gorin, N.-C.; Blaise, D.; Tabrizi, R.; Volin, L.; Cornelissen, J.; Cahn, J.-Y.; Chevallier, P.; Craddock, C.; et al. Matched and mismatched unrelated donor compared to autologous stem cell transplantation for acute myeloid leukemia in first complete remission: A retrospective, propensity score-weighted analysis from the ALWP of the EBMT. J. Hematol. Oncol. 2016, 9, 79.
  20. Saraceni, F.; Bruno, B.; Lemoli, R.M.; Meloni, G.; Arcese, W.; Falda, M.; Ciceri, F.; Alessandrino, E.P.; Specchia, G.; Scimè, R.; et al. Autologous stem cell transplantation is still a valid option in good- and intermediate-risk AML: A GITMO survey on 809 patients autografted in first complete remission. Bone Marrow Transplant. 2016, 52, 163–166.
  21. Mizutani, M.; Hara, M.; Fujita, H.; Aoki, J.; Kanamori, H.; Ohashi, K.; Usuki, K.; Fukuda, T.; Chou, T.; Tanaka, J.; et al. Comparable outcomes between autologous and allogeneic transplant for adult acute myeloid leukemia in first CR. Bone Marrow Transplant. 2016, 51, 645–653.
  22. Oriol, A.; Ribera, J.M.; Esteve, J.; Guardia, R.; Brunet, S.; Bueno, J.; Pedro, C.; Llorente, A.; Tormo, M.; Besalduch, J.; et al. Feasibility and results of autologous stem cell transplantation in de novo acute myeloid leukemia in patients over 60 years old. Results of the CETLAM AML-99 protocol. Haematologica 2004, 89, 791–800.
  23. Heini, A.D.; Berger, M.D.; Seipel, K.; Taleghani, B.M.; Baerlocher, G.M.; Leibundgut, K.; Banz, Y.; Novak, U.; Pabst, T. Consolidation with autologous stem cell transplantation in first remission is safe and effective in AML patients above 65 years. Leuk. Res. 2017, 53, 28–34.
  24. NCCN. Clinical Practice Guidelines in Oncology (NCCN Guidelines): Acute Myeloid Leukemia; National Comprehensive Cancer Network: Fort Washington, PA, USA, 2021.
  25. Schuurhuis, G.J.; Heuser, M.; Freeman, S.; Béné, M.-C.; Buccisano, F.; Cloos, J.; Grimwade, D.; Haferlach, T.; Hills, R.K.; Hourigan, C.S.; et al. Minimal/measurable residual disease in AML: A consensus document from the European LeukemiaNet MRD Working Party. Blood 2018, 131, 1275–1291.
  26. Percival, M.-E.; Lai, C.; Estey, E.; Hourigan, C.S. Bone marrow evaluation for diagnosis and monitoring of acute myeloid leukemia. Blood Rev. 2017, 31, 185–192.
  27. Ivey, A.; Hills, R.K.; Simpson, M.A.; Jovanovic, J.V.; Gilkes, A.; Grech, A.; Patel, Y.; Bhudia, N.; Farah, H.; Mason, J.; et al. Assessment of Minimal Residual Disease in Standard-Risk AML. N. Engl. J. Med. 2016, 374, 422–433.
  28. Jongen-Lavrencic, M.; Grob, T.; Hanekamp, D.; Kavelaars, F.G.; al Hinai, A.; Zeilemaker, A.; Erpelinck-Verschueren, C.A.J.; Gradowska, P.L.; Meijer, R.; Cloos, J.; et al. Molecular Minimal Residual Disease in Acute Myeloid Leukemia. N. Engl. J. Med. 2018, 378, 1189–1199.
  29. Yoest, J.M.; Shirai, C.L.; Duncavage, E.J. Sequencing-Based Measurable Residual Disease Testing in Acute Myeloid Leukemia. Front. Cell Dev. Biol. 2020, 8, 249.
  30. Hourigan, C.S.; Dillon, L.W.; Gui, G.; Logan, B.R.; Fei, M.; Ghannam, J.; Li, Y.; Licon, A.; Alyea, E.P.; Bashey, A.; et al. Impact of Conditioning Intensity of Allogeneic Transplantation for Acute Myeloid Leukemia with Genomic Evidence of Residual Disease. J. Clin. Oncol. 2019, 38, 1273–1283.
  31. Patkar, N.; Kakirde, C.; Shaikh, A.F.; Salve, R.; Bhanshe, P.; Chatterjee, G.; Rajpal, S.; Joshi, S.; Chaudhary, S.; Kodgule, R.; et al. Clinical impact of panel-based error-corrected next generation sequencing versus flow cytometry to detect measurable residual disease (MRD) in acute myeloid leukemia (AML). Leukemia 2021, 35, 1392–1404.
  32. Gökbuget, N.; Dombret, H.; Bonifacio, M.; Reichle, A.; Graux, C.; Faul, C.; Diedrich, H.; Topp, M.S.; Brüggemann, M.; Horst, H.-A.; et al. Blinatumomab for minimal residual disease in adults with B-cell precursor acute lymphoblastic leukemia. Blood 2018, 131, 1522–1531.
  33. Morita, K.; Wang, F.; Jahn, K.; Hu, T.; Tanaka, T.; Sasaki, Y.; Kuipers, J.; Loghavi, S.; Wang, S.A.; Yan, Y.; et al. Clonal evolution of acute myeloid leukemia revealed by high-throughput single-cell genomics. Nat. Commun. 2020, 11, 5327.
  34. Hasserjian, R.P.; Steensma, D.P.; Graubert, T.A.; Ebert, B.L. Clonal hematopoiesis and measurable residual disease assessment in acute myeloid leukemia. Blood 2020, 135, 1729–1738.
  35. Tsai, C.-H.; Tang, J.-L.; Tien, F.-M.; Kuo, Y.-Y.; Wu, D.-C.; Lin, C.-C.; Tseng, M.-H.; Peng, Y.-L.; Hou, M.-F.; Chuang, Y.-K.; et al. Clinical implications of sequential MRD monitoring by NGS at 2 time points after chemotherapy in patients with AML. Blood Adv. 2021, 5, 2456–2466.
  36. Godwin, C.D.; Zhou, Y.; Othus, M.; Asmuth, M.M.; Shaw, C.M.; Gardner, K.M.; Wood, B.L.; Walter, R.B.; Estey, E.H. Acute myeloid leukemia measurable residual disease detection by flow cytometry in peripheral blood vs. bone marrow. Blood 2021, 137, 569–572.
More
Information
Subjects: Hematology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 400
Revisions: 3 times (View History)
Update Date: 27 Aug 2021
1000/1000