You're using an outdated browser. Please upgrade to a modern browser for the best experience.
Submitted Successfully!
Thank you for your contribution! You can also upload a video entry or images related to this topic. For video creation, please contact our Academic Video Service.
Version Summary Created by Modification Content Size Created at Operation
1 Monique Antionette Hartley-Brown -- 1328 2023-12-19 14:22:19 |
2 layout & references Sirius Huang Meta information modification 1328 2023-12-21 09:17:59 |

Video Upload Options

We provide professional Academic Video Service to translate complex research into visually appealing presentations. Would you like to try it?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Mo, C.C.; Hartley-Brown, M.A.; Midha, S.; Richardson, P.G. Challenge of Comparing Upfront Versus Deferred HDM-ASCT. Encyclopedia. Available online: https://encyclopedia.pub/entry/52934 (accessed on 23 December 2025).
Mo CC, Hartley-Brown MA, Midha S, Richardson PG. Challenge of Comparing Upfront Versus Deferred HDM-ASCT. Encyclopedia. Available at: https://encyclopedia.pub/entry/52934. Accessed December 23, 2025.
Mo, Clifton C., Monique A. Hartley-Brown, Shonali Midha, Paul G. Richardson. "Challenge of Comparing Upfront Versus Deferred HDM-ASCT" Encyclopedia, https://encyclopedia.pub/entry/52934 (accessed December 23, 2025).
Mo, C.C., Hartley-Brown, M.A., Midha, S., & Richardson, P.G. (2023, December 19). Challenge of Comparing Upfront Versus Deferred HDM-ASCT. In Encyclopedia. https://encyclopedia.pub/entry/52934
Mo, Clifton C., et al. "Challenge of Comparing Upfront Versus Deferred HDM-ASCT." Encyclopedia. Web. 19 December, 2023.
Challenge of Comparing Upfront Versus Deferred HDM-ASCT
Edit

The standards of care for the initial treatment of patients with newly diagnosed multiple myeloma (NDMM) who are eligible for high-dose melphalan and autologous stem cell transplantation (HDM-ASCT) include highly active triplet and quadruplet regimens based on proteasome inhibitors, immunomodulatory drugs, and monoclonal antibodies. These regimens are resulting in improved outcomes and increasingly high rates of minimal residual disease (MRD)-negative responses without HDM-ASCT as part of the upfront therapy. 

autologous stem cell transplantation genotoxicity high-dose melphalan minimal residual disease multiple myeloma transplant-eligible treatment personalization

1. Introduction

Multiple myeloma (MM) is the second most common individual hematologic malignancy [1][2], with an estimated global incidence of almost 180,000 new cases and 120,000 deaths in 2020, comprising approximately 1% of the global cancer burden [1]. The disease more commonly affects males (~56% of cases) and is generally a disease of the elderly, with the median age at diagnosis in the United States being 69 years [3]. MM exhibits substantial heterogeneity at diagnosis and throughout the disease course associated with multiple disease-related and patient-related characteristics including disease stage [4][5], cytogenetic abnormalities [6], age, and frailty [7][8], providing the context for the drive to develop personalized treatment approaches [9][10][11]. Overall survival (OS) has increased markedly over the past four decades, with the 5-year survival rate in the United States more than doubling to 59.8% [3] and the median OS in younger, fitter patients reaching approximately 10 years [12]. This is associated with the introduction and widespread adoption of high-dose melphalan plus autologous stem cell transplantation (HDM-ASCT) as a frontline therapy in eligible patients and, more importantly, the more recent development and use of numerous highly active novel agents and regimens throughout the disease course in multiple lines of therapy [13][14]. Such progress is rapid and ongoing, as evidenced by the recent approvals by the United States Food and Drug Administration (FDA) in 2021–2023 of the chimeric antigen receptor (CAR) T cell therapies idecabtagene vicleucel (ide-cel) and ciltacabtagene autoleucel (cilta-cel) [15][16], and of the bispecific antibodies teclistamab, elranatamab, and talquetamab [17][18][19].

Current Treatment of Newly Diagnosed MM (NDMM) and the Role of HDM-ASCT

The current standards of care for the treatment of NDMM are based on three classes of agents: the proteasome inhibitors (PIs; bortezomib, carfilzomib, ixazomib), the immunomodulatory drugs (lenalidomide, pomalidomide, thalidomide), and the monoclonal antibodies (mAbs; anti-CD38 mAbs daratumumab, isatuximab; anti-SLAMF7 mAb elotuzumab) [13][20]. These agents are administered in combination—typically with dexamethasone—as triplet and, increasingly, quadruplet induction therapies, and as single-agent or doublet maintenance regimens as part of frontline therapy [13][20]. Post-induction consolidation therapy depends on a patient’s eligibility for HDM-ASCT, which remains the standard approach for patients aged ≤65–70 years without contraindicating comorbidities [13][20].
The use of HDM-ASCT as a standard in transplant-eligible patients was initially established based on randomized trials versus chemotherapy in the era prior to novel agents, in which transplant resulted in improved progression-free survival (PFS) and OS [21][22]. More recently, large phase 2 and phase 3 studies incorporating triplet novel-agent induction, with or without consolidation, and maintenance therapy have further demonstrated that the addition of HDM-ASCT confers a highly significant PFS benefit [23][24][25][26][27][28]. For example, the DETERMINATION phase 3 trial showed that the addition of HDM-ASCT to lenalidomide-bortezomib-dexamethasone (RVd) induction, plus lenalidomide maintenance to progression, resulted in a nearly 2-year median PFS benefit (67.5 vs. 46.2 months) and a 35% reduction in the risk of progression (RVd-alone vs. RVd + ASCT hazard ratio [HR] 1.53) [24]. The Intergroupe Francophone du Myélome (IFM) 2009 phase 3 trial, which had a similar design but administered lenalidomide maintenance for 1 year only, also showed a substantial PFS benefit with the addition of a transplant (median PFS 47.3 vs. 35.0 months, HR 1.43), although this was markedly less than the duration of disease control seen in both arms of DETERMINATION [23].
However, the importance and feasibility of the personalization of therapy is growing [9][10][11]. In the context of increasingly active quadruplet induction regimens [29][30][31][32] and our growing understanding of the mutagenic effects of melphalan [24][33][34][35][36], as well as no OS benefit having been demonstrated with HDM-ASCT in recent studies [23][24][25][26][37], the role of HDM-ASCT as a standard approach for all-comers in transplant-eligible NDMM is being challenged. Indeed, several treatment guidelines and recommendations are including deferred HDM-ASCT as a possible option for select patients in the frontline setting (Table 1). 
Table 1. Recent treatment guidelines and recommendations for NDMM including early or deferred HDM-ASCT.

2. The Challenge of Comparing Upfront Versus Deferred HDM-ASCT

It is important to acknowledge that directly comparing outcomes with upfront or deferred HDM-ASCT is challenging due to multiple potential confounders. For example, there is an inherent immortal time bias towards patients who receive deferred HDM-ASCT as a second-line therapy, because these patients must have already received frontline therapy and must still be young and fit enough to undergo HDM-ASCT as part of their second-line treatment [42]. Early analyses suggested that there were no differences in OS between the two approaches, and this may have been due, in part, to such potential bias [42][43][44]. However, there is the potential for bias in the opposite direction too if the group receiving deferred ASCT largely includes patients with an earlier need for second-line therapy following failure of their front-line regimen, i.e., those with more aggressive relapses. Moreover, in the context of the rapidly expanding range of highly active treatment options for relapsed/refractory MM (RRMM), a PFS benefit with upfront HDM-ASCT versus a non-transplant approach could result in a delayed need for second-line therapy, during which time additional novel, active treatment options might be approved in this setting.
Furthermore, for such comparisons, trials with a lengthy follow-up are required in order to evaluate outcomes through both first- and second-line therapy, which may be substantial for transplant-eligible patients in the modern era [12]. The IFM 2009 and DETERMINATION trials provide valuable data in this regard, as both trials recommended HDM-ASCT as second-line therapy following RVd alone [24][25]. In IFM 2009, with a median follow-up of 7.5 years, 262/350 (74.9%) and 217/350 (62.0%) patients on the RVd-alone and RVd + ASCT arms, respectively, required second-line therapy, with 201/262 (76.7%) versus 49/217 (22.6%) of them having received ASCT as part of that treatment. As in earlier studies of early versus later transplant, no difference in OS (8-year rate: 60.2% vs. 62.2%) was seen between arms [23], suggesting that deferred ASCT as part of the second-line therapy represents a reasonable clinical option. This is supported by several studies demonstrating substantial efficacy with HDM-ASCT in the RRMM setting [14][45][46]. Interestingly, however, in DETERMINATION no difference in OS (5-year rate: 79.2% vs. 80.7%) was seen between RVd alone and RVd + ASCT after a median follow-up of almost 6.5 years [24], despite only 78/279 (28.0%) RVd-alone patients who had discontinued protocol therapy having received subsequent HDM-ASCT. These findings, in the context of the large PFS benefit with RVd + ASCT, suggest the possibility of competing risk impacting OS.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: Globocan Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249.
  2. Siegel, R.L.; KMiller, D.; Wagle, N.S.; Jemal, A. Cancer Statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48.
  3. NIH National Cancer Institute Surveillance, Epidemiology, and End Results Program. Cancer Stat Facts: Myeloma. 2023. Available online: https://seer.cancer.gov/statfacts/html/mulmy.html (accessed on 23 October 2023).
  4. Palumbo, A.; Avet-Loiseau, H.; Oliva, S.; Lokhorst, H.M.; Goldschmidt, H.; Rosinol, L.; Richardson, P.; Caltagirone, S.; Lahuerta, J.J.; Facon, T.; et al. Revised International Staging System for Multiple Myeloma: A Report from International Myeloma Working Group. J. Clin. Oncol. 2015, 33, 2863–2869.
  5. Greipp, P.R.; Miguel, J.S.; Durie, B.G.; Crowley, J.J.; Barlogie, B.; Blade, J.; Boccadoro, M.; Child, J.A.; Avet-Loiseau, H.; Kyle, R.A.; et al. International Staging System for Multiple Myeloma. J. Clin. Oncol. 2005, 23, 3412–3420.
  6. van de Donk, N.W.C.; Pawlyn, C.; Yong, K.L. Multiple Myeloma. Lancet 2021, 397, 410–427.
  7. Palumbo, A.; Bringhen, S.; Mateos, M.V.; Larocca, A.; Facon, T.; Kumar, S.K.; Offidani, M.; McCarthy, P.; Evangelista, A.; Lonial, S.; et al. Geriatric Assessment Predicts Survival and Toxicities in Elderly Myeloma Patients: An International Myeloma Working Group Report. Blood 2015, 125, 2068–2074.
  8. Facon, T.; Dimopoulos, M.A.; Meuleman, N.; Belch, A.; Mohty, M.; Chen, W.M.; Kim, K.; Zamagni, E.; Rodriguez-Otero, P.; Renwick, W.; et al. A Simplified Frailty Scale Predicts Outcomes in Transplant-Ineligible Patients with Newly Diagnosed Multiple Myeloma Treated in the First (Mm-020) Trial. Leukemia 2020, 34, 224–233.
  9. D’Agostino, M.; Terragna, C.; van Duin, M. Editorial: Risk Factors in Multiple Myeloma Identified before and During Treatment: Are We Ready to Personalize Treatment? Front. Oncol. 2023, 13, 1247808.
  10. Bar, N.; Firestone, R.S.; Usmani, S.Z. Aiming for the Cure in Myeloma: Putting Our Best Foot Forward. Blood Rev. 2023, 101116.
  11. Richardson, P.G.; Miguel, J.F.S.; Moreau, P.; Hajek, R.; Dimopoulos, M.A.; Laubach, J.P.; Palumbo, A.; Luptakova, K.; Romanus, D.; Skacel, T.; et al. Interpreting Clinical Trial Data in Multiple Myeloma: Translating Findings to the Real-World Setting. Blood Cancer J. 2018, 8, 109.
  12. Joseph, N.S.; Kaufman, J.L.; Dhodapkar, M.V.; Hofmeister, C.C.; Almaula, D.K.; Heffner, L.T.; Gupta, V.A.; Boise, L.H.; Lonial, S.; Nooka, A.K. Long-Term Follow-up Results of Lenalidomide, Bortezomib, and Dexamethasone Induction Therapy and Risk-Adapted Maintenance Approach in Newly Diagnosed Multiple Myeloma. J. Clin. Oncol. 2020, 38, 1928–1937.
  13. Callander, N.S.; Baljevic, M.; Adekola, K.; Anderson, L.D.; Campagnaro, E.; Castillo, J.J.; Costello, C.; Devarakonda, S.; Elsedawy, N.; Faiman, M.; et al. Nccn Guidelines(R) Insights: Multiple Myeloma, Version 3.2022. J. Natl. Compr. Cancer Netw. 2022, 20, 8–19.
  14. Kumar, S.; Baizer, L.; Callander, N.S.; Giralt, S.A.; Hillengass, J.; Freidlin, B.; Hoering, A.; Richardson, P.G.; Schwartz, E.I.; Reiman, A.; et al. Gaps and Opportunities in the Treatment of Relapsed-Refractory Multiple Myeloma: Consensus Recommendations of the Nci Multiple Myeloma Steering Committee. Blood Cancer J. 2022, 12, 98.
  15. Munshi, N.C.; Anderson, L.D., Jr.; Shah, N.; Madduri, D.; Berdeja, J.; Lonial, S.; Raje, N.; Lin, Y.; Siegel, D.; Oriol, A.; et al. Idecabtagene Vicleucel in Relapsed and Refractory Multiple Myeloma. N. Engl. J. Med. 2021, 384, 705–716.
  16. Berdeja, J.G.; Madduri, D.; Usmani, S.Z.; Jakubowiak, A.; Agha, M.; Cohen, A.D.; Stewart, A.K.; Hari, P.; Htut, M.; Lesokhin, A.; et al. Ciltacabtagene Autoleucel, a B-Cell Maturation Antigen-Directed Chimeric Antigen Receptor T-Cell Therapy in Patients with Relapsed or Refractory Multiple Myeloma (Cartitude-1): A Phase 1b/2 Open-Label Study. Lancet 2021, 398, 314–324.
  17. Moreau, P.; Garfall, A.L.; van de Donk, N.; Nahi, H.; San-Miguel, J.F.; Oriol, A.; Nooka, A.K.; Martin, T.; Rosinol, L.; Chari, A.; et al. Teclistamab in Relapsed or Refractory Multiple Myeloma. N. Engl. J. Med. 2022, 387, 495–505.
  18. Lesokhin, A.M.; Tomasson, M.H.; Arnulf, B.; Bahlis, N.J.; Prince, H.M.; Niesvizky, R.; Rodriotaguez-Otero, P.; Martinez-Lopez, J.; Koehne, G.; Touzeau, C.; et al. Elranatamab in Relapsed or Refractory Multiple Myeloma: Phase 2 Magnetismm-3 Trial Results. Nat. Med. 2023, 29, 2259–2267.
  19. Chari, A.; Minnema, M.C.; Berdeja, J.G.; Oriol, A.; van de Donk, N.; Rodriguez-Otero, P.; Askari, E.; Mateos, M.V.; Costa, L.J.; Caers, J.; et al. Talquetamab, a T-Cell-Redirecting Gprc5d Bispecific Antibody for Multiple Myeloma. N. Engl. J. Med. 2022, 387, 2232–2244.
  20. Dimopoulos, M.A.; Moreau, P.; Terpos, E.; Mateos, M.V.; Zweegman, S.; Cook, G.; Delforge, M.; Hajek, R.; Schjesvold, F.; Cavo, M.; et al. Multiple Myeloma: Eha-Esmo Clinical Practice Guidelines for Diagnosis, Treatment and Follow-up(Dagger). Ann. Oncol. 2021, 32, 309–322.
  21. Child, J.A.; Morgan, G.J.; Davies, F.E.; Owen, R.G.; Bell, S.E.; Hawkins, K.; Brown, J.; Drayson, M.T.; Selby, P.J.; Party Medical Research Council Adult Leukaemia Working. High-Dose Chemotherapy with Hematopoietic Stem-Cell Rescue for Multiple Myeloma. N. Engl. J. Med. 2003, 348, 1875–1883.
  22. Koreth, J.; Cutler, C.S.; Djulbegovic, B.; Behl, R.; Schlossman, R.L.; Munshi, N.C.; Richardson, P.G.; Anderson, K.C.; Soiffer, R.J.; Alyea, E.P., 3rd. High-Dose Therapy with Single Autologous Transplantation Versus Chemotherapy for Newly Diagnosed Multiple Myeloma: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Biol. Blood Marrow Transpl. 2007, 13, 183–196.
  23. Perrot, A.; Lauwers-Cances, V.; Cazaubiel, T.; Facon, T.; Caillot, D.; Clement-Filliatre, L.; Macro, M.; Decaux, O.; Belhadj, K.; Mohty, M.; et al. Early Versus Late Autologous Stem Cell Transplant in Newly Diagnosed Multiple Myeloma: Long-Term Follow-up Analysis of the Ifm 2009 Trial. Blood 2020, 136 (Suppl. S1), 39.
  24. Richardson, P.G.; Jacobus, S.J.; Weller, E.A.; Hassoun, H.; Lonial, S.; Raje, N.S.; Medvedova, E.; McCarthy, P.L.; Libby, E.N.; Voorhees, P.M.; et al. Triplet Therapy, Transplantation, and Maintenance until Progression in Myeloma. N. Engl. J. Med. 2022, 387, 132–147.
  25. Attal, M.; Lauwers-Cances, V.; Hulin, C.; Leleu, X.; Caillot, D.; Escoffre, M.; Arnulf, B.; Macro, M.; Belhadj, K.; Garderet, L.; et al. Lenalidomide, Bortezomib, and Dexamethasone with Transplantation for Myeloma. N. Engl. J. Med. 2017, 376, 1311–1320.
  26. Gay, F.; Musto, P.; Rota-Scalabrini, D.; Bertamini, L.; Belotti, A.; Galli, M.; Offidani, M.; Zamagni, E.; Ledda, A.; Grasso, M.; et al. Carfilzomib with Cyclophosphamide and Dexamethasone or Lenalidomide and Dexamethasone Plus Autologous Transplantation or Carfilzomib Plus Lenalidomide and Dexamethasone, Followed by Maintenance with Carfilzomib Plus Lenalidomide or Lenalidomide Alone for Patients with Newly Diagnosed Multiple Myeloma (Forte): A Randomised, Open-Label, Phase 2 Trial. Lancet Oncol. 2021, 22, 1705–1720.
  27. Cavo, M.; Gay, F.; Beksac, M.; Pantani, L.; Petrucci, M.T.; Dimopoulos, M.A.; Dozza, L.; van der Holt, B.; Zweegman, S.; Oliva, S.; et al. Autologous Haematopoietic Stem-Cell Transplantation Versus Bortezomib-Melphalan-Prednisone, with or without Bortezomib-Lenalidomide-Dexamethasone Consolidation Therapy, and Lenalidomide Maintenance for Newly Diagnosed Multiple Myeloma (Emn02/Ho95): A Multicentre, Randomised, Open-Label, Phase 3 Study. Lancet Haematol. 2020, 7, e456–e468.
  28. Yong, K.; Wilson, W.; de Tute, R.M.; Camilleri, M.; Ramasamy, K.; Streetly, M.; Sive, J.; Bygrave, C.A.; Benjamin, R.; Chapman, M.; et al. Upfront Autologous Haematopoietic Stem-Cell Transplantation Versus Carfilzomib-Cyclophosphamide-Dexamethasone Consolidation with Carfilzomib Maintenance in Patients with Newly Diagnosed Multiple Myeloma in England and Wales (Cardamon): A Randomised, Phase 2, Non-Inferiority Trial. Lancet Haematol. 2023, 10, e93–e106.
  29. Costa, L.J.; Chhabra, S.; Medvedova, E.; Dholaria, B.R.; Schmidt, T.M.; Godby, K.N.; Silbermann, R.; Dhakal, B.; Bal, S.; Giri, S.; et al. Daratumumab, Carfilzomib, Lenalidomide, and Dexamethasone with Minimal Residual Disease Response-Adapted Therapy in Newly Diagnosed Multiple Myeloma. J. Clin. Oncol. 2022, 40, 2901–2912.
  30. Landgren, O.; Hultcrantz, M.; Diamond, B.; Lesokhin, A.M.; Mailankody, S.; Hassoun, H.; Tan, C.; Shah, U.A.; Lu, S.X.; Salcedo, M.; et al. Safety and Effectiveness of Weekly Carfilzomib, Lenalidomide, Dexamethasone, and Daratumumab Combination Therapy for Patients with Newly Diagnosed Multiple Myeloma: The Manhattan Nonrandomized Clinical Trial. JAMA Oncol. 2021, 7, 862–868.
  31. Moreau, P.; Attal, M.; Hulin, C.; Arnulf, B.; Belhadj, K.; Benboubker, L.; Bene, M.C.; Broijl, A.; Caillon, H.; Caillot, D.; et al. Bortezomib, Thalidomide, and Dexamethasone with or without Daratumumab before and after Autologous Stem-Cell Transplantation for Newly Diagnosed Multiple Myeloma (Cassiopeia): A Randomised, Open-Label, Phase 3 Study. Lancet 2019, 394, 29–38.
  32. Voorhees, P.M.; Kaufman, J.L.; Laubach, J.; Sborov, D.W.; Reeves, B.; Rodriguez, C.; Chari, A.; Silbermann, R.; Costa, L.J.; Anderson, L.D., Jr.; et al. Daratumumab, Lenalidomide, Bortezomib, and Dexamethasone for Transplant-Eligible Newly Diagnosed Multiple Myeloma: The Griffin Trial. Blood 2020, 136, 936–945.
  33. Maura, F.; Weinhold, N.; Diamond, B.; Kazandjian, D.; Rasche, L.; Morgan, G.; Landgren, O. The Mutagenic Impact of Melphalan in Multiple Myeloma. Leukemia 2021, 35, 2145–2150.
  34. Samur, M.K.; Roncador, M.; Samur, A.A.; Fulciniti, M.; Bazarbachi, A.H.; Szalat, R.; Shammas, M.A.; Sperling, A.S.; Richardson, P.G.; Magrangeas, F.; et al. High-Dose Melphalan Treatment Significantly Increases Mutational Burden at Relapse in Multiple Myeloma. Blood 2023, 141, 1724–1736.
  35. Rustad, E.H.; Yellapantula, V.; Leongamornlert, D.; Bolli, N.; Ledergor, G.; Nadeu, F.; Angelopoulos, N.; Dawson, K.J.; Mitchell, T.J.; Osborne, R.J.; et al. Timing the Initiation of Multiple Myeloma. Nat. Commun. 2020, 11, 1917.
  36. Radivoyevitch, T.; Dean, R.M.; Shaw, B.E.; Brazauskas, R.; Tecca, H.R.; Molenaar, R.J.; Battiwalla, M.; Savani, B.N.; Flowers, M.E.D.; Cooke, K.R.; et al. Risk of Acute Myeloid Leukemia and Myelodysplastic Syndrome after Autotransplants for Lymphomas and Plasma Cell Myeloma. Leuk. Res. 2018, 74, 130–136.
  37. Straka, C.; Schaefer-Eckart, K.; Hertenstein, B.; Bassermann, F.; Salwender, H.; Langer, C.; Krönke, J.; Kull, M.; Schilling, G.; Schieferdecker, A.; et al. Long-Term Outcome of a Prospective Randomized Trial Comparing Continuous Lenalidomide/Dexamethasone with Lenalidomide/Dexamethasone Induction, Mel140 with Autologous Blood Stem Cell Transplantation and Single Agent Lenalidomide Maintenance in Patients of Age 60-75 Years with Newly Diagnosed Multiple Myeloma. Blood 2022, 140 (Suppl. S1), 287–288.
  38. Sive, J.; Cuthill, K.; Hunter, H.; Kazmi, M.; Pratt, G.; Smith, D.; British Society of Haematology. Guidelines on the Diagnosis, Investigation and Initial Treatment of Myeloma: A British Society for Haematology/UK Myeloma Forum Guideline. Br. J. Haematol. 2021, 193, 245–268.
  39. Dhakal, B.; Shah, N.; Kansagra, A.; Kumar, A.; Lonial, S.; Garfall, A.; Cowan, A.; Poudyal, B.S.; Costello, C.; Gay, F.; et al. Astct Clinical Practice Recommendations for Transplantation and Cellular Therapies in Multiple Myeloma. Transpl. Cell Ther. 2022, 28, 284–293.
  40. Rajkumar, S.V. Multiple Myeloma: 2022 Update on Diagnosis, Risk Stratification, and Management. Am. J. Hematol. 2022, 97, 1086–1107.
  41. mSMART. Treatment Guidelines: Multiple Myeloma. 2023. Available online: https://www.msmart.org/mm-treatment-guidelines (accessed on 23 October 2023).
  42. Gandolfi, S.; Vekstein, C.; Laubach, J.P.; O’Brien, A.; Masone, K.; Munshi, N.C.; Anderson, K.C.; Richardson, P.G. The Evolving Role of Transplantation in Multiple Myeloma: The Need for a Heterogeneous Approach to a Heterogeneous Disease. Clin. Adv. Hematol. Oncol. 2018, 16, 564–574.
  43. Dunavin, N.C.; Wei, L.; Elder, P.; Phillips, G.S.; Benson, D.M., Jr.; Hofmeister, C.C.; Penza, S.; Greenfield, C.; Rose, K.S.; Rieser, G.; et al. Early Versus Delayed Autologous Stem Cell Transplant in Patients Receiving Novel Therapies for Multiple Myeloma. Leuk. Lymphoma 2013, 54, 1658–1664.
  44. Remenyi, P.; Varga, G.; Mikala, G.; Reti, M.; Gopcsa, L.; Batai, A.; Csukly, Z.; Lengyel, L.; Torbagyi, E.; Barta, A.; et al. Early Versus Delayed Autologous Stem Cell Transplantation and Interferon Maintenance in Multiple Myeloma: Single-Center Experience of 18 Years. Transpl. Proc. 2016, 48, 177–184.
  45. Lemieux, C.; Muffly, L.S.; Iberri, D.J.; Craig, J.K.; Johnston, L.J.; Lowsky, R.; Shiraz, P.; Rezvani, A.R.; Frank, M.J.; Weng, W.K.; et al. Outcomes after Delayed and Second Autologous Stem Cell Transplant in Patients with Relapsed Multiple Myeloma. Bone Marrow Transpl. 2021, 56, 2664–2671.
  46. Van Oekelen, O.; Nath, K.; Mouhieddine, T.H.; Farzana, T.; Aleman, A.; Melnekoff, D.T.; Ghodke-Puranik, Y.; Shah, G.L.; Lesokhin, A.; Giralt, S.; et al. Interventions and Outcomes of Patients with Multiple Myeloma Receiving Salvage Therapy after Bcma-Directed Car T Therapy. Blood 2023, 141, 756–765.
More
Upload a video for this entry
Information
Subjects: Oncology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , Monique A. Hartley-Brown , , Paul G. Richardson
View Times: 528
Revisions: 2 times (View History)
Update Date: 21 Dec 2023
Notice
You are not a member of the advisory board for this topic. If you want to update advisory board member profile, please contact office@encyclopedia.pub.
OK
Confirm
Only members of the Encyclopedia advisory board for this topic are allowed to note entries. Would you like to become an advisory board member of the Encyclopedia?
Yes
No
${ textCharacter }/${ maxCharacter }
Submit
Cancel
There is no comment~
${ textCharacter }/${ maxCharacter }
Submit
Cancel
${ selectedItem.replyTextCharacter }/${ selectedItem.replyMaxCharacter }
Submit
Cancel
Confirm
Are you sure to Delete?
Yes No
Academic Video Service