1
|
Guo W, Strouse C, Mery D, Siegel ER, Munshi MN, Ashby TC, Cheng Y, Sun F, Wanchai V, Zhang Z, Bailey C, Alapat DV, Peng H, Al Hadidi S, Thanendrarajan S, Schinke C, Zangari M, van Rhee F, Tricot G, Shaughnessy JD, Zhan F. A Risk Stratification System in Myeloma Patients with Autologous Stem Cell Transplantation. Cancers (Basel) 2024; 16:1116. [PMID: 38539451 PMCID: PMC10969019 DOI: 10.3390/cancers16061116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 06/26/2024] Open
Abstract
Autologous stem cell transplantation (ASCT) has been a mainstay in myeloma treatment for over three decades, but patient prognosis post-ASCT varies significantly. In a retrospective study of 5259 patients with multiple myeloma (MM) at the University of Arkansas for Medical Sciences undergoing ASCT with a median 57-month follow-up, we divided the dataset into training (70%) and validation (30%) subsets. Employing univariable and multivariable Cox analyses, we systematically assessed 29 clinical variables, identifying crucial adverse prognostic factors, such as extended duration between MM diagnosis and ASCT, elevated serum ferritin, and reduced transferrin levels. These factors could enhance existing prognostic models. Additionally, we pinpointed significant poor prognosis markers like high serum calcium and low platelet counts, though they are applicable to a smaller patient population. Utilizing seven easily accessible high-risk variables, we devised a four-stage system (ATM4S) with primary stage borders determined through K-adaptive partitioning. This staging system underwent validation in both the training dataset and an independent cohort of 514 ASCT-treated MM patients from the University of Iowa. We also explored cytogenetic risk factors within this staging system, emphasizing its potential clinical utility for refining prognostic assessments and guiding personalized treatment approaches.
Collapse
Affiliation(s)
- Wancheng Guo
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot# 508, Little Rock, AR 72205, USA; (W.G.); (D.M.); (M.N.M.); (Y.C.); (F.S.); (V.W.); (Z.Z.); (C.B.); (S.A.H.); (S.T.); (C.S.); (M.Z.); (F.v.R.); (G.T.)
- Department of Haematology, Second Xiangya Hospital, Central South University, Changsha 410011, China;
| | | | - David Mery
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot# 508, Little Rock, AR 72205, USA; (W.G.); (D.M.); (M.N.M.); (Y.C.); (F.S.); (V.W.); (Z.Z.); (C.B.); (S.A.H.); (S.T.); (C.S.); (M.Z.); (F.v.R.); (G.T.)
| | - Eric R. Siegel
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Manit N. Munshi
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot# 508, Little Rock, AR 72205, USA; (W.G.); (D.M.); (M.N.M.); (Y.C.); (F.S.); (V.W.); (Z.Z.); (C.B.); (S.A.H.); (S.T.); (C.S.); (M.Z.); (F.v.R.); (G.T.)
| | - Timothy Cody Ashby
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Yan Cheng
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot# 508, Little Rock, AR 72205, USA; (W.G.); (D.M.); (M.N.M.); (Y.C.); (F.S.); (V.W.); (Z.Z.); (C.B.); (S.A.H.); (S.T.); (C.S.); (M.Z.); (F.v.R.); (G.T.)
| | - Fumou Sun
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot# 508, Little Rock, AR 72205, USA; (W.G.); (D.M.); (M.N.M.); (Y.C.); (F.S.); (V.W.); (Z.Z.); (C.B.); (S.A.H.); (S.T.); (C.S.); (M.Z.); (F.v.R.); (G.T.)
| | - Visanu Wanchai
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot# 508, Little Rock, AR 72205, USA; (W.G.); (D.M.); (M.N.M.); (Y.C.); (F.S.); (V.W.); (Z.Z.); (C.B.); (S.A.H.); (S.T.); (C.S.); (M.Z.); (F.v.R.); (G.T.)
| | - Zijun Zhang
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot# 508, Little Rock, AR 72205, USA; (W.G.); (D.M.); (M.N.M.); (Y.C.); (F.S.); (V.W.); (Z.Z.); (C.B.); (S.A.H.); (S.T.); (C.S.); (M.Z.); (F.v.R.); (G.T.)
| | - Clyde Bailey
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot# 508, Little Rock, AR 72205, USA; (W.G.); (D.M.); (M.N.M.); (Y.C.); (F.S.); (V.W.); (Z.Z.); (C.B.); (S.A.H.); (S.T.); (C.S.); (M.Z.); (F.v.R.); (G.T.)
| | - Daisy V. Alapat
- Department of Pathology Clinical, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Hongling Peng
- Department of Haematology, Second Xiangya Hospital, Central South University, Changsha 410011, China;
| | - Samer Al Hadidi
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot# 508, Little Rock, AR 72205, USA; (W.G.); (D.M.); (M.N.M.); (Y.C.); (F.S.); (V.W.); (Z.Z.); (C.B.); (S.A.H.); (S.T.); (C.S.); (M.Z.); (F.v.R.); (G.T.)
| | - Sharmilan Thanendrarajan
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot# 508, Little Rock, AR 72205, USA; (W.G.); (D.M.); (M.N.M.); (Y.C.); (F.S.); (V.W.); (Z.Z.); (C.B.); (S.A.H.); (S.T.); (C.S.); (M.Z.); (F.v.R.); (G.T.)
| | - Carolina Schinke
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot# 508, Little Rock, AR 72205, USA; (W.G.); (D.M.); (M.N.M.); (Y.C.); (F.S.); (V.W.); (Z.Z.); (C.B.); (S.A.H.); (S.T.); (C.S.); (M.Z.); (F.v.R.); (G.T.)
| | - Maurizio Zangari
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot# 508, Little Rock, AR 72205, USA; (W.G.); (D.M.); (M.N.M.); (Y.C.); (F.S.); (V.W.); (Z.Z.); (C.B.); (S.A.H.); (S.T.); (C.S.); (M.Z.); (F.v.R.); (G.T.)
| | - Frits van Rhee
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot# 508, Little Rock, AR 72205, USA; (W.G.); (D.M.); (M.N.M.); (Y.C.); (F.S.); (V.W.); (Z.Z.); (C.B.); (S.A.H.); (S.T.); (C.S.); (M.Z.); (F.v.R.); (G.T.)
| | - Guido Tricot
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot# 508, Little Rock, AR 72205, USA; (W.G.); (D.M.); (M.N.M.); (Y.C.); (F.S.); (V.W.); (Z.Z.); (C.B.); (S.A.H.); (S.T.); (C.S.); (M.Z.); (F.v.R.); (G.T.)
| | - John D. Shaughnessy
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot# 508, Little Rock, AR 72205, USA; (W.G.); (D.M.); (M.N.M.); (Y.C.); (F.S.); (V.W.); (Z.Z.); (C.B.); (S.A.H.); (S.T.); (C.S.); (M.Z.); (F.v.R.); (G.T.)
| | - Fenghuang Zhan
- Myeloma Center, Department of Internal Medicine, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham St. Slot# 508, Little Rock, AR 72205, USA; (W.G.); (D.M.); (M.N.M.); (Y.C.); (F.S.); (V.W.); (Z.Z.); (C.B.); (S.A.H.); (S.T.); (C.S.); (M.Z.); (F.v.R.); (G.T.)
| |
Collapse
|
2
|
Medina-Herrera A, Sarasquete ME, Jiménez C, Puig N, García-Sanz R. Minimal Residual Disease in Multiple Myeloma: Past, Present, and Future. Cancers (Basel) 2023; 15:3687. [PMID: 37509348 PMCID: PMC10377959 DOI: 10.3390/cancers15143687] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Responses to treatment have improved over the last decades for patients with multiple myeloma. This is a consequence of the introduction of new drugs that have been successfully combined in different clinical contexts: newly diagnosed, transplant-eligible or ineligible patients, as well as in the relapsed/refractory setting. However, a great proportion of patients continue to relapse, even those achieving complete response, which underlines the need for updated response criteria. In 2014, the international myeloma working group established new levels of response, prompting the evaluation of minimal residual disease (MRD) for those patients already in complete or stringent complete response as defined by conventional serological assessments: the absence of tumor plasma cells in 100,000 total cells or more define molecular and immunophenotypic responses by next-generation sequencing and flow cytometry, respectively. In this review, we describe all the potential methods that may be used for MRD detection based on the evidence found in the literature, paying special attention to their advantages and pitfalls from a critical perspective.
Collapse
Affiliation(s)
- Alejandro Medina-Herrera
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - María Eugenia Sarasquete
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Cristina Jiménez
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Noemí Puig
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| | - Ramón García-Sanz
- Departament of Hematology, University Hospital of Salamanca (HUSA/IBSAL), CIBERONC, CIC-IBMCC (USAL-CSIC), 37007 Salamanca, Spain
| |
Collapse
|
3
|
Xiong W, Wang Z, Wang T, Yu Y, Huang Y, Sun H, Chen J, Lyu R, Wang H, Yan Y, Wang Q, Liu W, An G, Sui W, Huang W, Zou D, Xiao Z, Wang J, Ouyang G, Qiu L, Yi S. Minimal residual disease status improved the response evaluation in patients with Waldenström's macroglobulinemia. Front Immunol 2023; 14:1171539. [PMID: 37234167 PMCID: PMC10206219 DOI: 10.3389/fimmu.2023.1171539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction Minimal residual disease (MRD) has been recognized as an important prognostic factor of survival in patients with hematological malignancies. However, the prognostic value of MRD in Waldenström macroglobulinemia (WM) remains unexplored. Methods We analyzed 108 newly diagnosed WM patients receiving systematic therapy and assessed for MRD by multiparameter flow cytometry (MFC) using bone marrow samples. Results Of the total patients, 34 (31.5%) achieved undetectable MRD (uMRD). A hemoglobin level of >115 g/L (P=0.03), a serum albumin level of >35 g/L (P=0.01), a β2-MG level of ≤3 mg/L (P=0.03), and a low-risk International Prognostic Scoring System for WM (IPSSWM) stage (P<0.01) were associated with a higher rate of uMRD. Improvements in monoclonal immunoglobulin (P<0.01) and hemoglobin (P=0.03) levels were more evident in uMRD patients compared with that in MRD-positive patients. The 3-year progression-free survival (PFS) was better in uMRD patients compared with that in MRD-positive patients (96.2% vs. 52.8%; P=0.0012). Landmark analysis also showed that uMRD patients had better PFS compared with MRD-positive patients after 6 and 12 months. Patients who achieved partial response (PR) and uMRD had a 3-year PFS of 100%, which was significantly higher than that of patients with MRD-positive PR (62.6%, P=0.029). Multivariate analysis showed that MRD positivity was an independent factor of PFS (HR: 2.55, P=0.03). Moreover, the combination of the 6th International Workshop on WM assessment (IWWM-6 Criteria) and MRD assessment had a higher 3-year AUC compared with the IWWM-6 criteria alone (0.71 vs. 0.67). Discussion MRD status assessed by MFC is an independent prognostic factor for PFS in patients with WM, and its determination could improve the precision of response evaluation, especially in patients who achieved PR.
Collapse
Affiliation(s)
- Wenjie Xiong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Zanzan Wang
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Tingyu Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ying Yu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yanshan Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Hao Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jiawen Chen
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Rui Lyu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Huijun Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yuting Yan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Qi Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wei Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Gang An
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Weiwei Sui
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Wenyang Huang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Dehui Zou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Zhijian Xiao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Jianxiang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Guifang Ouyang
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Shuhua Yi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Department of Lymphma & Myeloma, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
4
|
Suzuki K, Yano S. Treatment Strategy for Ultra-High-Risk Multiple Myelomas with Chromosomal Aberrations Considering Minimal Residual Disease Status and Bone Marrow Microenvironment. Cancers (Basel) 2023; 15:cancers15092418. [PMID: 37173885 PMCID: PMC10177433 DOI: 10.3390/cancers15092418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/12/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Despite the development of anti-myeloma therapeutics, such as proteasome inhibitors, immunomodulatory drugs, anti-CD38 monoclonal antibodies, and autologous stem cell transplantation (ASCT), multiple myeloma remains incurable. A trial treatment combining four drugs-daratumumab, carfilzomib, lenalidomide, and dexamethasone-followed by ASCT frequently results in minimal residual disease (MRD) negativity and prevents progressive disease in patients with standard- and high-risk cytogenetics; however, it is insufficient to overcome the poor outcomes in patients with ultra-high-risk chromosomal aberration (UHRCA). In fact, MRD status in autografts can predict clinical outcomes after ASCT. Therefore, the current treatment strategy might be insufficient to overcome the negative impact of UHRCA in patients with MRD positivity after the four-drug induction therapy. High-risk myeloma cells lead to poor clinical outcomes not only by aggressive myeloma behavior but also via the generation of a poor bone marrow microenvironment. Meanwhile, the immune microenvironment effectively suppresses myeloma cells with a low frequency of high-risk cytogenetic abnormalities in early-stage myeloma compared to late-stage myeloma. Therefore, early intervention might be key to improving clinical outcomes in myeloma patients. The purpose of this review is to improve clinical outcomes in patients with UHRCA by considering MRD assessment results and improvement of the microenvironment.
Collapse
Affiliation(s)
- Kazuhito Suzuki
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, 3-19-18 Nishi-Shimbashi, Minato-ku, Tokyo 105-0003, Japan
| | - Shingo Yano
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, 3-19-18 Nishi-Shimbashi, Minato-ku, Tokyo 105-0003, Japan
| |
Collapse
|
5
|
Cédile O, Hansen MH, Dahlmann SK, Kristensen TK, Abildgaard N, Nyvold CG. Reproducibility of low-level residual myeloma immunoglobulin detection using ultra-deep sequencing. Exp Hematol 2023; 119-120:14-20. [PMID: 36708872 DOI: 10.1016/j.exphem.2023.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/20/2022] [Accepted: 01/16/2023] [Indexed: 01/26/2023]
Abstract
Multiple myeloma, a mature B-cell neoplasm, is the second most common hematologic malignancy. Despite advancements in treatment, the disease remains incurable, with more than 100,000 annual deaths worldwide. As recommended by the International Myeloma Working Group, measurable residual disease (MRD) should be addressed at a 10-5 sensitivity level or beyond for practical purposes. Next-generation sequencing (NGS) has provided new opportunities with deep sequencing of clonal rearrangements of the immunoglobulin heavy chain (IGH) locus in B-cell malignancies. Although the ability to resolve one cancerous cell in a million other B cells is becoming attractive as a prognostic indicator in sustained patients who are MRD-negative, reaching consistent sensitivity levels is challenging because of sample stochasticity and the substantial amount of deoxyribonucleic acid (DNA) required for library preparation. Thus, in the presented study, we implemented ultra-deep sequencing of rearranged IGH to investigate the reproducibility and consistency aimed at the 10-5 sensitivity level. In this controlled setup, our data provided stable MRD detection of 1.2 clonal cells per 100,000 analyzed cells and longitudinal reproducibility. We also demonstrated a low false-negative rate using 4-5 replicates and 700-800 ng DNA per sequencing replicate. In conclusion, adding an internal control to the replicates enabled clonal cell normalization for MRD evaluation as a stable reference. These findings may guide MRD-level reporting and comparisons between laboratories.
Collapse
Affiliation(s)
- Oriane Cédile
- Haematology-Pathology Research Laboratory, Research Unit for Haematology and Research Unit for Pathology, University of Southern Denmark and Odense University Hospital, Odense, Denmark; OPEN, Odense Patient Data Explorative Network, Odense University Hospital, Odense, Denmark
| | - Marcus Høy Hansen
- Haematology-Pathology Research Laboratory, Research Unit for Haematology and Research Unit for Pathology, University of Southern Denmark and Odense University Hospital, Odense, Denmark.
| | - Sara Kamuk Dahlmann
- Haematology-Pathology Research Laboratory, Research Unit for Haematology and Research Unit for Pathology, University of Southern Denmark and Odense University Hospital, Odense, Denmark
| | - Thomas Kielsgaard Kristensen
- Haematology-Pathology Research Laboratory, Research Unit for Haematology and Research Unit for Pathology, University of Southern Denmark and Odense University Hospital, Odense, Denmark; Department of Clinical Development, Odense University Hospital, Odense, Denmark
| | - Niels Abildgaard
- Haematology-Pathology Research Laboratory, Research Unit for Haematology and Research Unit for Pathology, University of Southern Denmark and Odense University Hospital, Odense, Denmark
| | - Charlotte Guldborg Nyvold
- Haematology-Pathology Research Laboratory, Research Unit for Haematology and Research Unit for Pathology, University of Southern Denmark and Odense University Hospital, Odense, Denmark; OPEN, Odense Patient Data Explorative Network, Odense University Hospital, Odense, Denmark
| |
Collapse
|
6
|
Urushihara R, Takezako N, Yoroidaka T, Yamashita T, Murata R, Satou K, Nakao S, Takamatsu H. Eight-color multiparameter flow cytometry (EuroFlow-NGF) is as sensitive as next-generation sequencing in detecting minimal/measurable residual disease in autografts of patients with multiple myeloma. EJHAEM 2023; 4:184-191. [PMID: 36819171 PMCID: PMC9928796 DOI: 10.1002/jha2.633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 12/07/2022] [Indexed: 01/25/2023]
Abstract
The prognostic value of minimal/measurable residual disease (MRD) detection in autografts of patients with multiple myeloma (MM) in an autologous stem-cell transplantation setting has been reported. Next-generation flow (NGF) cytometry has lower sensitivity (2 × 10-6) to detect MRD than next-generation sequencing (NGS) (<10-6). We compared the clinical value of high-sensitivity NGF (cutoff: <10-6) and NGS (cutoff: 10-6) for the detection of MRD in the cryopreserved autografts of 49 patients with newly diagnosed MM. The sensitivity test using frozen/thawed autografts revealed a strong correlation among MRD levels of 5 × 10-7 and 1 × 10-4 (r = 0.9997, p < 0.0001) when an adequate number of cells were analyzed. Autograft MRD levels determined using NGF and NGS were highly correlated (r = 0.811, p < 0.0001). MRD-negative patients identified with NGF (cutoff: <10-6) showed significantly longer progression-free survival (PFS) than MRD-positive patients (p = 0.026). The PFS of MRD-negative patients determined by NGS (cutoff: 10-6) was similar to that determined by NGF. These results show that the high-sensitivity NGF method can assess MRD in frozen/thawed autografts, and its prognostic value is comparable to that of NGS.
Collapse
Affiliation(s)
- Ryota Urushihara
- Department of Hematology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health SciencesKanazawa UniversityKanazawaJapan
| | - Naoki Takezako
- Department of HematologyDisaster Medical Center of JapanTachikawaJapan
| | - Takeshi Yoroidaka
- Department of Hematology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health SciencesKanazawa UniversityKanazawaJapan
| | | | - Ryoichi Murata
- Division of Internal MedicineKeiju Kanazawa HospitalKanazawaJapan
| | - Kenji Satou
- Faculty of Transdisciplinary Sciences for InnovationInstitute of Transdisciplinary Sciences for InnovationKanazawa UniversityKanazawaJapan
| | - Shinji Nakao
- Department of Hematology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health SciencesKanazawa UniversityKanazawaJapan
| | - Hiroyuki Takamatsu
- Department of Hematology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health SciencesKanazawa UniversityKanazawaJapan
- Faculty of Transdisciplinary Sciences for InnovationInstitute of Transdisciplinary Sciences for InnovationKanazawa UniversityKanazawaJapan
| |
Collapse
|
7
|
MRD in multiple myeloma: does CR really matter? Blood 2022; 140:2423-2428. [PMID: 35560160 DOI: 10.1182/blood.2022016170] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/11/2022] [Accepted: 04/26/2022] [Indexed: 01/01/2023] Open
Abstract
Multiple myeloma embodies the paradigm of the deeper the response, the longer the survival. However, results are conflicting regarding achievement of complete remission (CR) and minimal residual disease (MRD) negativity; some patients with persistent M protein have undetectable MRD. We reviewed the frequency of this discordance and outcomes of these patients. We spotlight possible explanations for and consequences of conflicting response criteria and suggest that MRD be assessed in patients achieving very good partial response or better in clinical trials.
Collapse
|
8
|
Deng X, Zhang M, Zhou J, Xiao M. Next-generation sequencing for MRD monitoring in B-lineage malignancies: from bench to bedside. Exp Hematol Oncol 2022; 11:50. [PMID: 36057673 PMCID: PMC9440501 DOI: 10.1186/s40164-022-00300-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 08/21/2022] [Indexed: 12/02/2022] Open
Abstract
Minimal residual disease (MRD) is considered the strongest relevant predictor of prognosis and an effective decision-making factor during the treatment of hematological malignancies. Remarkable breakthroughs brought about by new strategies, such as epigenetic therapy and chimeric antigen receptor-T (CAR-T) therapy, have led to considerably deeper responses in patients than ever, which presents difficulties with the widely applied gold-standard techniques of MRD monitoring. Urgent demands for novel approaches that are ultrasensitive and provide sufficient information have put a spotlight on high-throughput technologies. Recently, advances in methodology, represented by next-generation sequencing (NGS)-based clonality assays, have proven robust and suggestive in numerous high-quality studies and have been recommended by some international expert groups as disease-monitoring modalities. This review demonstrates the applicability of NGS-based clonality assessment for MRD monitoring of B-cell malignancies by summarizing the oncogenesis of neoplasms and the corresponding status of immunoglobulin (IG) rearrangements. Furthermore, we focused on the performance of NGS-based assays compared with conventional approaches and the interpretation of results, revealing directions for improvement and prospects in clinical practice.
Collapse
Affiliation(s)
- Xinyue Deng
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, 430030, China
| | - Meilan Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, 430030, China
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, 430030, China
| | - Min Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, 430030, China.
| |
Collapse
|
9
|
Ferla V, Antonini E, Perini T, Farina F, Masottini S, Malato S, Marktel S, Lupo Stanghellini MT, Tresoldi C, Ciceri F, Marcatti M. Minimal residual disease detection by next-generation sequencing in multiple myeloma: Promise and challenges for response-adapted therapy. Front Oncol 2022; 12:932852. [PMID: 36052251 PMCID: PMC9426755 DOI: 10.3389/fonc.2022.932852] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/06/2022] [Indexed: 11/18/2022] Open
Abstract
Assessment of minimal residual disease (MRD) is becoming a standard diagnostic tool for curable hematological malignancies such as chronic and acute myeloid leukemia. Multiple myeloma (MM) remains an incurable disease, as a major portion of patients even in complete response eventually relapse, suggesting that residual disease remains. Over the past decade, the treatment landscape of MM has radically changed with the introduction of new effective drugs and the availability of immunotherapy, including targeted antibodies and adoptive cell therapy. Therefore, conventional serological and morphological techniques have become suboptimal for the evaluation of depth of response. Recently, the International Myeloma Working Group (IMWG) introduced the definition of MRD negativity as the absence of clonal Plasma cells (PC) with a minimum sensitivity of <10−5 either by next-generation sequencing (NGS) using the LymphoSIGHT platform (Sequenta/Adaptative) or by next-generation flow cytometry (NGF) using EuroFlow approaches as the reference methods. While the definition of the LymphoSIGHT platform (Sequenta/Adaptive) as the standard method derives from its large use and validation in clinical studies on the prognostic value of NGS-based MRD, other commercially available options exist. Recently, the LymphoTrack assay has been evaluated in MM, demonstrating a sensitivity level of 10−5, hence qualifying as an alternative effective tool for MRD monitoring in MM. Here, we will review state-of-the-art methods for MRD assessment by NGS. We will summarize how MRD testing supports clinical trials as a useful tool in dynamic risk-adapted therapy. Finally, we will also discuss future promise and challenges of NGS-based MRD determination for clinical decision-making. In addition, we will present our real-life single-center experience with the commercially available NGS strategy LymphoTrack-MiSeq. Even with the limitation of a limited number of patients, our results confirm the LymphoTrack-MiSeq platform as a cost-effective, readily available, and standardized workflow with a sensitivity of 10−5. Our real-life data also confirm that achieving MRD negativity is an important prognostic factor in MM.
Collapse
Affiliation(s)
- Valeria Ferla
- Hematology and Bone Marrow Transplantation, San Raffaele Scientific Institute, Milan, Italy
- *Correspondence: Valeria Ferla,
| | - Elena Antonini
- Molecular Hematology Laboratory, San Raffaele Scientific Institute, Milan, Italy
| | - Tommaso Perini
- Hematology and Bone Marrow Transplantation, San Raffaele Scientific Institute, Milan, Italy
- Age Related Diseases Laboratory, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Farina
- Hematology and Bone Marrow Transplantation, San Raffaele Scientific Institute, Milan, Italy
| | - Serena Masottini
- Molecular Hematology Laboratory, San Raffaele Scientific Institute, Milan, Italy
| | - Simona Malato
- Hematology and Bone Marrow Transplantation, San Raffaele Scientific Institute, Milan, Italy
| | - Sarah Marktel
- Hematology and Bone Marrow Transplantation, San Raffaele Scientific Institute, Milan, Italy
| | | | - Cristina Tresoldi
- Molecular Hematology Laboratory, San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Ciceri
- Hematology and Bone Marrow Transplantation, San Raffaele Scientific Institute, Milan, Italy
- University Vita-Salute San Raffaele, Milan, Italy
| | - Magda Marcatti
- Hematology and Bone Marrow Transplantation, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
10
|
Takamatsu H. Guest Editorial: Innovation will be a bridge to cure in patients with multiple myeloma? Int J Hematol 2022; 115:760-761. [PMID: 35543898 DOI: 10.1007/s12185-022-03371-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 10/18/2022]
Affiliation(s)
- Hiroyuki Takamatsu
- Department of Hematology, Kanazawa University Hospital/School of Entrepreneurial and Innovation Studies, Institute of Transdisciplinary Sciences for Innovation, Kanazawa University, 13-1 Takaramachi, Kanazawa, 920-8641, Japan.
| |
Collapse
|
11
|
Cho H, Shin S, Chung H, Jang JE, Kim YR, Cheong JW, Min YH, Lee ST, Choi JR, Kim JS. Real-world data on prognostic value of measurable residual disease assessment by fragment analysis or next-generation sequencing in multiple myeloma. Br J Haematol 2022; 198:503-514. [PMID: 35505579 DOI: 10.1111/bjh.18211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/07/2022] [Accepted: 04/09/2022] [Indexed: 01/12/2023]
Abstract
Measurable residual disease (MRD) negativity is a strong prognostic indicator in multiple myeloma (MM). However, the optimal use of MRD in daily clinical practice has been hampered by the limited feasibility of MRD testing. Therefore, we examined the clinical relevance of commercially available MRD modalities based on clonality assays by fragment analysis with IdentiClone® (n = 73 patients) and next-generation sequencing (NGS) with LymphoTrack® (n = 116 patients) in newly diagnosed patients with MM who received autologous stem cell transplantation (ASCT). MRD was assessed at the end of induction (pre-ASCT) and/or at 100 days after ASCT (post-ASCT). MRD could not predict survival when assessed by fragment analysis. However, NGS-based MRD negativity at pre- or post-ASCT was beneficial in terms of progression-free and overall survival. Moreover, NGS-based MRD negativity was independently associated with improved progression-free and overall survival, and MRD-positive patients both pre- and post-ASCT had worst outcome. Indeed, initial adverse prognostic features by high-risk cytogenetics could be mitigated upon achieving MRD negativity by NGS. We demonstrate the feasibility and clinical benefit of achieving MRD negativity by commercially available clonality-based MRD assays in MM and support incorporating NGS, but not fragment analysis, to tailor therapeutic strategies in real-world practice.
Collapse
Affiliation(s)
- Hyunsoo Cho
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Saeam Shin
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Haerim Chung
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji Eun Jang
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yu Ri Kim
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - June-Won Cheong
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoo Hong Min
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Rak Choi
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin Seok Kim
- Division of Hematology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
12
|
Elkjær LAL, Cédile O, Hansen MH, Nielsen C, Møller MB, Abildgaard N, Haaber J, Nyvold CG. Exploration of residual disease in stem cell products from mantle cell lymphoma using next-generation sequencing. Leuk Res Rep 2022; 18:100341. [PMID: 36039182 PMCID: PMC9418493 DOI: 10.1016/j.lrr.2022.100341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 07/01/2022] [Accepted: 08/07/2022] [Indexed: 11/05/2022] Open
Abstract
High-dose chemotherapy followed by autologous stem cell transplantation (ASCT) has become a treatment option for fit patients with mantle cell lymphoma (MCL). However, these patients often relapse within few years, potentially caused by contaminating lymphoma cells within the reinfused stem cell product (SCP). Studies have shown that measurable residual disease, also termed minimal residual disease (MRD), following ASCT predicts shorter survival. Using next-generation sequencing, we explore whether the diagnostic MCL clonotype is present within the infused SCP. MRD was detected in 4/17 of the SCPs, ranging 4–568 clonal cells/100,000 cells. With a median survival of 17 months, 3/4 of patients with MRD+ graft succumbed from MCL relapse versus 2/13 in the MRD– fraction. Patients receiving MRD+ grafts had increased risk of mortality, and thus screening of SCPs may be important for clinical decision-making.
Collapse
|
13
|
Yoshihara S, Yoshihara K, Shimizu Y, Imado T, Takatsuka H, Kawamoto H, Misawa M, Ifuku H, Ohe Y, Okada M, Fujimori Y. Feasibility of six cycles of lenalidomide-based triplet induction before stem cell collection for newly diagnosed transplant-eligible multiple myeloma. HEMATOLOGY (AMSTERDAM, NETHERLANDS) 2021; 26:388-392. [PMID: 34000225 DOI: 10.1080/16078454.2021.1926101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVES Achieving a deep response with induction therapy has a major impact on outcomes following autologous stem cell transplantation. Although longer and intensified induction therapy may provide better disease control, longer exposure to lenalidomide negatively affects stem cell yield. We examined the feasibility of 6 cycles of lenalidomide-based triplet induction therapy before stem cell collection in transplant-eligible multiple myeloma patients. METHODS In this prospective study, patients received a combination of bortezomib, lenalidomide, and dexamethasone for 6 cycles. For patients who did not achieve a deep response after 3 cycles, bortezomib was substituted with carfilzomib for the last 2 cycles (5th and 6th courses). RESULTS Although only half of the patients achieved a deep response after 3 cycles, all but 1 patient achieved a very good partial response (n = 4) or complete response (n = 5) after completing 6 cycles. Among 9 patients who received cyclophosphamide-based stem cell mobilization, 1 patient required a second mobilization that was successfully performed using plerixafor. After autologous transplantation, 7 patients showed complete response, including 5 with minimal residual disease-negative status. CONCLUSION This study demonstrates that 6 cycles of lenalidomide-based induction therapy before stem cell collection are a feasible and promising approach for transplant-eligible newly diagnosed multiple myeloma patients.The study is registered at UMIN Clinical Trials Registry as UMIN000026936.Trial registration: UMIN Japan identifier: UMIN000026936.
Collapse
Affiliation(s)
- Satoshi Yoshihara
- Department of Hematology, Hyogo College of Medicine Hospital, Hyogo, Japan.,Department of Transfusion Medicine and Cellular Therapy, Hyogo College of Medicine Hospital, Hyogo, Japan
| | - Kyoko Yoshihara
- Department of Hematology, Hyogo College of Medicine Hospital, Hyogo, Japan
| | | | - Takehito Imado
- Department of Hematology, Takarazuka City Hospital, Hyogo, Japan
| | | | | | - Mahito Misawa
- Department of Hematology, Ako Central Hospital, Hyogo, Japan
| | - Hideki Ifuku
- Department of Hematology, Amagasaki Chuo Hospital, Hyogo, Japan
| | - Yokiko Ohe
- Department of Hematology, Uegahara Hospital, Hyogo, Japan
| | - Masaya Okada
- Department of Hematology, Hyogo College of Medicine Hospital, Hyogo, Japan
| | - Yoshihiro Fujimori
- Department of Hematology, Hyogo College of Medicine Hospital, Hyogo, Japan.,Department of Transfusion Medicine and Cellular Therapy, Hyogo College of Medicine Hospital, Hyogo, Japan.,Department of Hematology, Uegahara Hospital, Hyogo, Japan
| |
Collapse
|
14
|
Yoroidaka T, Narita K, Takamatsu H, Fujisawa M, Nakao S, Matsue K. Comparison of minimal residual disease detection in multiple myeloma between the DuraClone and EuroFlow methods. Sci Rep 2021; 11:11218. [PMID: 34045494 PMCID: PMC8160149 DOI: 10.1038/s41598-021-89761-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 04/23/2021] [Indexed: 11/09/2022] Open
Abstract
In this study, the minimal residual disease (MRD) levels in patients with multiple myeloma (MM) were assessed by comparing the new 8-color single-tube multiparameter flow cytometry method (DuraClone), which reduces the cost of antibodies and labor burden of laboratories, with the EuroFlow next-generation flow (NGF) method. A total of 96 samples derived from 69 patients with MM were assessed to determine the total cell acquisition number (tCAN), percentages of total and normal plasma cells (PCs), and MRD levels using two methods. We found that the tCAN was significantly higher with EuroFlow-NGF than with DuraClone (median 8.6 × 106 vs. 5.7 × 106; p < 0.0001). In addition, a significant correlation in the MRD levels between the two methods was noted (r = 0.92, p < 0.0001). However, in the qualitative analysis, 5.2% (5/96) of the samples showed discrepancies in the MRD levels. In conclusion, the DuraClone is a good option to evaluate MRD in multiple myeloma but it should be used with caution.
Collapse
Affiliation(s)
- Takeshi Yoroidaka
- Department of Hematology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Kentaro Narita
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, 929 Higashi-chou, Kamogawa-shi, Chiba, 296-8602, Japan
| | - Hiroyuki Takamatsu
- Department of Hematology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8641, Japan.
| | - Momoko Fujisawa
- Department of Hematology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Shinji Nakao
- Department of Hematology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8641, Japan
| | - Kosei Matsue
- Division of Hematology/Oncology, Department of Medicine, Kameda Medical Center, 929 Higashi-chou, Kamogawa-shi, Chiba, 296-8602, Japan.
| |
Collapse
|
15
|
Hansen MH, Cédile O, Larsen TS, Abildgaard N, Nyvold CG. Perspective: sensitive detection of residual lymphoproliferative disease by NGS and clonal rearrangements-how low can you go? Exp Hematol 2021; 98:14-24. [PMID: 33823225 DOI: 10.1016/j.exphem.2021.03.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/22/2021] [Accepted: 03/30/2021] [Indexed: 01/12/2023]
Abstract
Malignant lymphoproliferative disorders collectively constitute a large fraction of the hematological cancers, ranging from indolent to highly aggressive neoplasms. Being a diagnostically important hallmark, clonal gene rearrangements of the immunoglobulins enable the detection of residual disease in the clinical course of patients down to a minute fraction of malignant cells. The introduction of next-generation sequencing (NGS) has provided unprecedented assay specificity, with a sensitivity matching that of polymerase chain reaction-based measurable residual disease (MRD) detection down to the 10-6 level. Although reaching 10-6 to 10-7 is theoretically feasible, employing a sufficient amount of DNA and sequencing coverage is placed in the perspective of the practical challenges when relying on clinical samples in contrast to controlled serial dilutions. As we discuss, the randomness of subsampling must be taken into account to accommodate the sensitivity threshold-in terms of both the required number of cells and sequencing coverage. As a substantial part of the reviewed studies do not state the depth of coverage or even amount of DNA in some cases, we call for increased transparency to enable critical assessment of the MRD assays for clinical implementation and feasibility.
Collapse
Affiliation(s)
- Marcus H Hansen
- Hematology-Pathology Research Laboratory, Research Unit for Hematology and Research Unit for Pathology, University of Southern Denmark and Odense University Hospital, Odense, Denmark; Department of Hematology, Odense University Hospital, Odense, Denmark.
| | - Oriane Cédile
- Hematology-Pathology Research Laboratory, Research Unit for Hematology and Research Unit for Pathology, University of Southern Denmark and Odense University Hospital, Odense, Denmark; Department of Hematology, Odense University Hospital, Odense, Denmark
| | - Thomas S Larsen
- Department of Hematology, Odense University Hospital, Odense, Denmark
| | - Niels Abildgaard
- Hematology-Pathology Research Laboratory, Research Unit for Hematology and Research Unit for Pathology, University of Southern Denmark and Odense University Hospital, Odense, Denmark; Department of Hematology, Odense University Hospital, Odense, Denmark
| | - Charlotte G Nyvold
- Hematology-Pathology Research Laboratory, Research Unit for Hematology and Research Unit for Pathology, University of Southern Denmark and Odense University Hospital, Odense, Denmark; Department of Hematology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
16
|
Van der Straeten J, De Brouwer W, Kabongo E, Dresse MF, Fostier K, Schots R, Van Riet I, Bakkus M. Validation of a PCR-Based Next-Generation Sequencing Approach for the Detection and Quantification of Minimal Residual Disease in Acute Lymphoblastic Leukemia and Multiple Myeloma Using gBlocks as Calibrators. J Mol Diagn 2021; 23:599-611. [PMID: 33549860 DOI: 10.1016/j.jmoldx.2021.01.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 10/22/2022] Open
Abstract
Detection of minimal residual disease (MRD) to guide therapy has been a standard practice in treatment of childhood acute lymphoblastic leukemia (ALL) for decades. In multiple myeloma (MM), a clear correlation is found between absence of MRD and longer survival. Quantitative allele-specific oligonucleotide (qASO)-PCR is the standard molecular method for MRD detection in these hematologic malignant tumors. However, this technique has some drawbacks that can be overcome by next-generation sequencing (NGS). In this study, NGS is validated as an alternative method for qASO-PCR for MRD detection in both ALL and MM. MRD results obtained by NGS and qASO-PCR were compared in 59 and 39 bone marrow samples of 33 and 14 patients with ALL and MM, respectively. Our results indicate that the use of gBlocks as calibrators makes the NGS approach a powerful tool to quantify MRD. With an input of 400 ng of DNA (corresponding to approximately 7 × 104 cells), a limit of detection of 0.01% can be achieved. The specificity of the NGS-MRD technique was 100%, and a correlation with qASO-PCR for quantifiable MRD results of 0.93 and 0.91 was found in ALL and MM, respectively. Especially for MM, the higher applicability (100%) of the NGS-MRD protocol, compared with qASO-PCR (57%), was clearly demonstrated. These results demonstrate that NGS is an even better alternative to qASO-PCR.
Collapse
Affiliation(s)
- Jona Van der Straeten
- Molecular Hematology Laboratory, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Wouter De Brouwer
- Department of Hematology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Emmanuelle Kabongo
- Molecular Hematology Laboratory, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | | | - Karel Fostier
- Department of Hematology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Rik Schots
- Department of Hematology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Ivan Van Riet
- Department of Hematology, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Marleen Bakkus
- Molecular Hematology Laboratory, Vrije Universiteit Brussel, Universitair Ziekenhuis Brussel, Brussels, Belgium.
| |
Collapse
|
17
|
Legarda MA, Cejalvo MJ, de la Rubia J. Recent Advances in the Treatment of Patients with Multiple Myeloma. Cancers (Basel) 2020; 12:E3576. [PMID: 33265952 PMCID: PMC7761116 DOI: 10.3390/cancers12123576] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/17/2020] [Accepted: 11/25/2020] [Indexed: 12/22/2022] Open
Abstract
In the past 20 years, few diseases have seen as great progress in their treatment as multiple myeloma. With the approval of many new drugs and the limited availability of clinical trials comparing head-to-head the different possible combinations, the choice of the best treatments at each stage of the disease becomes complex as well as crucial since multiple myeloma remains incurable. This article presents a general description of the novelties of the whole treatment of multiple myeloma, from induction in the newly diagnosed patient through the role of hematopoietic stem cell transplantation and maintenance treatment until early and late relapses, including a section on recently approved drugs as well as novel drugs and immunotherapy in advanced stages of research, and that will surely play a relevant role in the treatment of this devastating disease in the coming years.
Collapse
Affiliation(s)
- Mario A. Legarda
- Hematology Department, University Hospital Doctor Peset, 46017 Valencia, Spain; (M.A.L.); (M.J.C.)
| | - María J. Cejalvo
- Hematology Department, University Hospital Doctor Peset, 46017 Valencia, Spain; (M.A.L.); (M.J.C.)
| | - Javier de la Rubia
- Hematology Department, University Hospital Doctor Peset, 46017 Valencia, Spain; (M.A.L.); (M.J.C.)
- Hematology Department, Internal Medicine, School of Medicine and Dentistry, Catholic University of Valencia, 46017 Valencia, Spain
| |
Collapse
|
18
|
Ho C, Syed M, Roshal M, Petrova-Drus K, Moung C, Yao J, Quesada AE, Benhamida J, Vanderbilt C, Liu Y, Zhu M, Yu W, Maciag L, Wang M, Ma Y, Gao Q, Rustad EH, Hultcrantz M, Diamond BT, Zheng-Lin B, Huang Y, Hutt K, Miller JE, Dogan A, Nafa K, Landgren O, Arcila ME. Routine Evaluation of Minimal Residual Disease in Myeloma Using Next-Generation Sequencing Clonality Testing: Feasibility, Challenges, and Direct Comparison with High-Sensitivity Flow Cytometry. J Mol Diagn 2020; 23:181-199. [PMID: 33217553 PMCID: PMC7874334 DOI: 10.1016/j.jmoldx.2020.10.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/13/2020] [Accepted: 10/22/2020] [Indexed: 01/12/2023] Open
Abstract
The 2016 International Myeloma Working Group consensus recommendations emphasize high-sensitivity methods for minimal residual disease (MRD) detection, treatment response assessment, and prognostication. Next-generation sequencing (NGS) of IGH gene rearrangements is highly specific and sensitive, but its description in routine clinical practice and performance comparison with high-sensitivity flow cytometry (hsFC) remain limited. In this large, single-institution study including 438 samples from 251 patients, the use of NGS targeting the IGH and IGK genes for clonal characterization and monitoring, with comparison to hsFC, is described. The index clone characterization success rate was 93.6% (235/251), which depended on plasma cell (PC) cellularity, reaching 98% when PC ≥10% and below 80% when PC <5%. A total of 85% of cases were successfully characterized using leader and FR1 primer sets, and most clones showed high somatic hypermutation rates (median, 8.1%). Among monitoring samples from 124 patients, 78.6% (147/187) had detectable disease by NGS. Concordance with hsFC was 92.9% (170/183). Discordant cases encompassed 8 of 124 hsFC MRD+/NGS MRD− patients (6.5%) and 4 of 124 hsFC MRD−/NGS MRD+ patients (3.2%), all with low-level disease near detection limits for both assays. Among concordant hsFC MRD−/NGS MRD− cases, only 5 of 24 patients (20.8%) showed subsequent overt relapse at 3-year follow-up. HsFC and NGS showed similar operational sensitivity, and the choice of test may depend on practical, rather than test performance, considerations.
Collapse
Affiliation(s)
- Caleb Ho
- Hematopathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York; Diagnostic Molecular Pathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York.
| | - Mustafa Syed
- Diagnostic Molecular Pathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Mikhail Roshal
- Hematopathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Kseniya Petrova-Drus
- Hematopathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York; Diagnostic Molecular Pathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Christine Moung
- Diagnostic Molecular Pathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Jinjuan Yao
- Diagnostic Molecular Pathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Andres E Quesada
- Hematopathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York; Diagnostic Molecular Pathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Jamal Benhamida
- Diagnostic Molecular Pathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Chad Vanderbilt
- Diagnostic Molecular Pathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Ying Liu
- Hematopathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Menglei Zhu
- Hematopathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Wayne Yu
- Diagnostic Molecular Pathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Lidia Maciag
- Diagnostic Molecular Pathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Meiyi Wang
- Diagnostic Molecular Pathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Yuanyuan Ma
- Diagnostic Molecular Pathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Qi Gao
- Hematopathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Even H Rustad
- Myeloma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Malin Hultcrantz
- Myeloma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Benjamin T Diamond
- Myeloma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Binbin Zheng-Lin
- Myeloma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Ying Huang
- Invivoscribe, Inc., San Diego, California
| | - Kasey Hutt
- Invivoscribe, Inc., San Diego, California
| | | | - Ahmet Dogan
- Hematopathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Khedoudja Nafa
- Diagnostic Molecular Pathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Ola Landgren
- Myeloma Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Maria E Arcila
- Hematopathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York; Diagnostic Molecular Pathology Service, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York.
| |
Collapse
|
19
|
Flach J, Shumilov E, Joncourt R, Porret N, Novak U, Pabst T, Bacher U. Current concepts and future directions for hemato-oncologic diagnostics. Crit Rev Oncol Hematol 2020; 151:102977. [DOI: 10.1016/j.critrevonc.2020.102977] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 03/22/2020] [Accepted: 04/28/2020] [Indexed: 02/07/2023] Open
|
20
|
Thakral D, Das N, Basnal A, Gupta R. Cell-free DNA for genomic profiling and minimal residual disease monitoring in Myeloma- are we there yet? AMERICAN JOURNAL OF BLOOD RESEARCH 2020; 10:26-45. [PMID: 32685257 PMCID: PMC7364270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/11/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVE Multiple myeloma (MM), a plasma cell neoplasm, afflicts elder individuals accounting for 10% of hematologic malignancies. The MM plasma cells largely reside within the bone marrow niche and are accessible through an invasive bone marrow biopsy, which is challenging during serial monitoring of patients. In this setting, cell free DNA (cfDNA) may have a role to ascertain the molecular aberrations at diagnosis and in assessment of residual disease during therapy. The aim of this review was to explore the utility and current status of cfDNA in MM. METHOD PubMed was searched with terms including cell-free DNA, circulating-tumor DNA, Multiple Myeloma, diagnosis, genomic profiling, Minimal Residual Disease individually or in combination to shortlist the relevant studies. RESULT cfDNA serves as a non-invasive source of tumor-specific molecular biomarker, ctDNA that has immense potential in facilitating management of cancer patients. The mutation detection platforms for ctDNA include hybrid capture and ultra-deep sequencing. Hybrid capture allows full length gene sequencing for mutation and CNV detection. The disease progression can be monitored by profiling prognostic somatic copy number alterations by ultra-low pass whole genome sequencing of ctDNA cost-effectively. Evolution of both the laboratory protocols and bioinformatics tools may further improve the sensitivity of ctDNA detection for better disease management. Only a limited number of studies were available in MM exploring the potential utility of cfDNA. CONCLUSION In this review, we discuss the nuances and challenges associated with molecular evaluation of cfDNA and its potential role in diagnosis and monitoring of treatment response in MM.
Collapse
Affiliation(s)
- Deepshi Thakral
- Laboratory Oncology Unit, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences New Delhi, India
| | - Nupur Das
- Laboratory Oncology Unit, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences New Delhi, India
| | - Atul Basnal
- Laboratory Oncology Unit, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences New Delhi, India
| | - Ritu Gupta
- Laboratory Oncology Unit, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences New Delhi, India
| |
Collapse
|
21
|
Bal S, Landau HJ, Shah GL, Scordo M, Dahi P, Lahoud OB, Hassoun H, Hultcrantz M, Korde N, Lendvai N, Lesokhin AM, Mailankody S, Shah UA, Smith E, Devlin SM, Avecilla S, Dogan A, Roshal M, Landgren O, Giralt SA, Chung DJ. Stem Cell Mobilization and Autograft Minimal Residual Disease Negativity with Novel Induction Regimens in Multiple Myeloma. Biol Blood Marrow Transplant 2020; 26:1394-1401. [PMID: 32442725 DOI: 10.1016/j.bbmt.2020.04.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/10/2020] [Accepted: 04/11/2020] [Indexed: 12/22/2022]
Abstract
Autologous stem cell transplantation (ASCT) remains the standard of care for transplantation-eligible patients with multiple myeloma (MM). Bortezomib with lenalidomide and dexamethasone (VRD) is the most common triplet regimen for newly diagnosed MM in the United States. Carfilzomib with lenalidomide and dexamethasone (KRD) has shown promising efficacy and may supplant VRD. We compared stem cell yields and autograft minimal residual disease (MRD)-negativity after VRD and KRD induction. Deeper responses (ie, very good partial response or better) were more common with KRD. Precollection bone marrow (BM) cellularity, interval from the end of induction therapy to start of stem cell collection, and method of stem cell mobilization were similar for the 2 cohorts. Days to complete collection was greater with KRD (2.2 days, versus 1.81 days with VRD), which more often required ≥3 days of apheresis. Precollection viable CD34+ cell content was greater with VRD, as was collection yield (11.11 × 106, versus 9.19 × 106 with KRD). Collection failure (defined as <2 × 106 CD34+ cells/kg) was more frequent with KRD (5.4% versus .7% with VRD). The difference in stem cell yield between VRD and KRD is associated with the degree of lenalidomide exposure. Age ≥70 years predicted poorer collection for both cohorts. Stem cell autograft purity/MRD-negativity was higher with KRD (81.4%, versus 57.1% with VRD). For both cohorts, MRD-negativity was attained in a larger fraction of autografts than in precollection BM. For patients proceeding to ASCT, the time to neutrophil/platelet engraftment was comparable in the 2 study arms. In summary, our data demonstrate that KRD induces deeper clinical responses and greater autograft purity than VRD without compromising stem cell yield or post-transplantation engraftment kinetics.
Collapse
Affiliation(s)
- Susan Bal
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Heather J Landau
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Gunjan L Shah
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Michael Scordo
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Parastoo Dahi
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Oscar B Lahoud
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Hani Hassoun
- Department of Medicine, Weill Cornell Medical College, New York, New York; Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Malin Hultcrantz
- Department of Medicine, Weill Cornell Medical College, New York, New York; Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Neha Korde
- Department of Medicine, Weill Cornell Medical College, New York, New York; Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nikoletta Lendvai
- Department of Medicine, Weill Cornell Medical College, New York, New York; Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alexander M Lesokhin
- Department of Medicine, Weill Cornell Medical College, New York, New York; Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sham Mailankody
- Department of Medicine, Weill Cornell Medical College, New York, New York; Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Urvi A Shah
- Department of Medicine, Weill Cornell Medical College, New York, New York; Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eric Smith
- Department of Medicine, Weill Cornell Medical College, New York, New York; Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sean M Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Scott Avecilla
- Cell Therapy Laboratory, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ahmet Dogan
- Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mikhail Roshal
- Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ola Landgren
- Department of Medicine, Weill Cornell Medical College, New York, New York; Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sergio A Giralt
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - David J Chung
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York; The Rockefeller University, New York, New York.
| |
Collapse
|
22
|
Paiva B, Puig N, Cedena MT, Rosiñol L, Cordón L, Vidriales MB, Burgos L, Flores-Montero J, Sanoja-Flores L, Lopez-Anglada L, Maldonado R, de la Cruz J, Gutierrez NC, Calasanz MJ, Martin-Ramos ML, Garcia-Sanz R, Martinez-Lopez J, Oriol A, Blanchard MJ, Rios R, Martin J, Martinez-Martinez R, Sureda A, Hernandez MT, de la Rubia J, Krsnik I, Moraleda JM, Palomera L, Bargay J, Van Dongen JJ, Orfao A, Mateos MV, Blade J, San-Miguel JF, Lahuerta JJ. Measurable Residual Disease by Next-Generation Flow Cytometry in Multiple Myeloma. J Clin Oncol 2020; 38:784-792. [DOI: 10.1200/jco.19.01231] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Assessing measurable residual disease (MRD) has become standard with many tumors, but the clinical meaning of MRD in multiple myeloma (MM) remains uncertain, particularly when assessed by next-generation flow (NGF) cytometry. Thus, we aimed to determine the applicability and sensitivity of the flow MRD-negative criterion defined by the International Myeloma Working Group (IMWG). PATIENTS AND METHODS In the PETHEMA/GEM2012MENOS65 trial, 458 patients with newly diagnosed MM had longitudinal assessment of MRD after six induction cycles with bortezomib, lenalidomide, and dexamethasone (VRD), autologous transplantation, and two consolidation courses with VRD. MRD was assessed in 1,100 bone marrow samples from 397 patients; the 61 patients without MRD data discontinued treatment during induction and were considered MRD positive for intent-to-treat analysis. The median limit of detection achieved by NGF was 2.9 × 10−6. Patients received maintenance (lenalidomide ± ixazomib) according to the companion PETHEMA/GEM2014MAIN trial. RESULTS Overall, 205 (45%) of 458 patients had undetectable MRD after consolidation, and only 14 of them (7%) have experienced progression thus far; seven of these 14 displayed extraosseous plasmacytomas at diagnosis and/or relapse. Using time-dependent analysis, patients with undetectable MRD had an 82% reduction in the risk of progression or death (hazard ratio, 0.18; 95% CI, 0.11 to 0.30; P < .001) and an 88% reduction in the risk of death (hazard ratio, 0.12; 95% CI, 0.05 to 0.29; P < .001). Timing of undetectable MRD (after induction v intensification) had no impact on patient survival. Attaining undetectable MRD overcame poor prognostic features at diagnosis, including high-risk cytogenetics. By contrast, patients with Revised International Staging System III status and positive MRD had dismal progression-free and overall survivals (median, 14 and 17 months, respectively). Maintenance increased the rate of undetectable MRD by 17%. CONCLUSION The IMWG flow MRD-negative response criterion is highly applicable and sensitive to evaluate treatment efficacy in MM.
Collapse
Affiliation(s)
- Bruno Paiva
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada Instituto de Investigacion Sanitaria de Navarra, Pamplona, Spain
| | - Noemi Puig
- Hospital Universitario de Salamanca, Instituto de Investigacion Biomedica de Salamanca, Salamanca, Spain
| | | | - Laura Rosiñol
- Hospital Clínic Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lourdes Cordón
- Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - María-Belén Vidriales
- Hospital Universitario de Salamanca, Instituto de Investigacion Biomedica de Salamanca, Salamanca, Spain
| | - Leire Burgos
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada Instituto de Investigacion Sanitaria de Navarra, Pamplona, Spain
| | - Juan Flores-Montero
- Centro de Investigación Biomédica en Red de Cáncer, Instituto Carlos III, Madrid, Spain
- University of Salamanca, Salamanca, Spain
| | - Luzalba Sanoja-Flores
- Centro de Investigación Biomédica en Red de Cáncer, Instituto Carlos III, Madrid, Spain
- University of Salamanca, Salamanca, Spain
| | | | | | | | - Norma C. Gutierrez
- Hospital Universitario de Salamanca, Instituto de Investigacion Biomedica de Salamanca, Salamanca, Spain
| | - Maria-Jose Calasanz
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada Instituto de Investigacion Sanitaria de Navarra, Pamplona, Spain
| | | | - Ramón Garcia-Sanz
- Hospital Universitario de Salamanca, Instituto de Investigacion Biomedica de Salamanca, Salamanca, Spain
| | | | - Albert Oriol
- Institut Català d’Oncologia i Institut Josep Carreras, Badalona, Spain
| | | | - Rafael Rios
- Hospital Virgen de las Nieves, Granada, Spain
| | - Jesus Martin
- Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | | | - Anna Sureda
- Institut Català d'Oncologia L’Hospitalet, Barcelona, Spain
| | | | - Javier de la Rubia
- Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Catholic University of Valencia, Valencia, Spain
| | | | | | | | - Joan Bargay
- Hospital Son Llatzer, Palma de Mallorca, Spain
| | | | - Alberto Orfao
- Centro de Investigación Biomédica en Red de Cáncer, Instituto Carlos III, Madrid, Spain
- University of Salamanca, Salamanca, Spain
| | - Maria-Victoria Mateos
- Hospital Universitario de Salamanca, Instituto de Investigacion Biomedica de Salamanca, Salamanca, Spain
| | - Joan Blade
- Hospital Clínic Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jesús F. San-Miguel
- Clinica Universidad de Navarra, Centro de Investigacion Medica Aplicada Instituto de Investigacion Sanitaria de Navarra, Pamplona, Spain
| | | | | |
Collapse
|
23
|
Takamatsu H. Clinical value of measurable residual disease testing for multiple myeloma and implementation in Japan. Int J Hematol 2020; 111:519-529. [DOI: 10.1007/s12185-020-02828-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 01/15/2020] [Indexed: 11/24/2022]
|
24
|
Oliva S, D'Agostino M, Boccadoro M, Larocca A. Clinical Applications and Future Directions of Minimal Residual Disease Testing in Multiple Myeloma. Front Oncol 2020; 10:1. [PMID: 32076595 PMCID: PMC7006453 DOI: 10.3389/fonc.2020.00001] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/02/2020] [Indexed: 12/20/2022] Open
Abstract
In the last years, the life expectancy of multiple myeloma (MM) patients has substantially improved thanks to the availability of many new drugs. Our ability to induce deep responses has improved as well, and the treatment goal in patients tolerating treatment moved from the delay of progression to the induction of the deepest possible response. As a result of these advances, a great scientific effort has been made to redefine response monitoring, resulting in the development and validation of high-sensitivity techniques to detect minimal residual disease (MRD). In 2016, the International Myeloma Working Group (IMWG) updated MM response categories defining MRD-negative responses both in the bone marrow (assessed by next-generation flow cytometry or next-generation sequencing) and outside the bone marrow. MRD is an important factor independently predicting prognosis during MM treatment. Moreover, using novel combination therapies, MRD-negative status can be achieved in a fairly high percentage of patients. However, many questions regarding the clinical use of MRD status remain unanswered. MRD monitoring can guide treatment intensity, although well-designed clinical trials are needed to demonstrate this potential. This mini-review will focus on currently available techniques and data on MRD testing and their potential future applications.
Collapse
Affiliation(s)
- Stefania Oliva
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Cittá Della Salute e Della Scienza di Torino, Turin, Italy
| | - Mattia D'Agostino
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Cittá Della Salute e Della Scienza di Torino, Turin, Italy
| | - Mario Boccadoro
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Cittá Della Salute e Della Scienza di Torino, Turin, Italy
| | - Alessandra Larocca
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Cittá Della Salute e Della Scienza di Torino, Turin, Italy
| |
Collapse
|
25
|
Kunacheewa C, Lee HC, Patel K, Thomas S, Amini B, Srour S, Bashir Q, Nieto Y, Qazilbash MH, Weber DM, Feng L, Orlowski RZ, Lin P, Manasanch EE. Minimal Residual Disease Negativity Does Not Overcome Poor Prognosis in High-Risk Multiple Myeloma: A Single-Center Retrospective Study. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e221-e238. [PMID: 32037287 DOI: 10.1016/j.clml.2020.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 12/28/2019] [Accepted: 01/02/2020] [Indexed: 01/14/2023]
Abstract
BACKGROUND Minimal residual disease (MRD) is a standard measurement for response assessment in multiple myeloma (MM). Despite new treatments, high-risk MM patients continue to have poor prognosis. We evaluated the effect of MRD negativity in high-risk versus standard-risk patients. PATIENTS AND METHODS We retrospectively evaluated all consecutive MM patients who underwent routine MRD testing by 1-tube 8-color advanced flow cytometry with 2,000,000 events and sensitivity level 10-5 at our center from 2015 to 2018 after initial therapy. Kaplan-Meier and log-rank test were used to assess survival estimates and differences between study groups. RESULTS One hundred thirty-six patients with MRD testing after initial therapy or autologous stem-cell transplantation were identified. At a median follow-up of 14 months (range, 1-36 months), progression-free survival and overall survival were significantly worse in high-risk versus standard-risk patients. During the study period, 50% of high-risk group had experienced disease progression (relapse and/or death) versus 20% in the standard-risk group (P = .0006). No patients with standard-risk died, but 4 (14%) in the high-risk group did (P = .0007). Regardless of MRD status, high-risk patients had statistically significant worse progression-free survival than standard-risk patients. At median follow-up, those with disease 10% standard-risk/MRD negative; 20% standard-risk/MRD positive; 40% high-risk/MRD negative; and 45% high-risk/MRD positive had either experienced relapse or died (P = .0041). MRD status did not significantly affect overall survival in either group (P = .0914); however, longer follow-up is needed to assess survival. CONCLUSION Genetic abnormalities remain a powerful prognostic indicator for MM, regardless of MRD status. For newly diagnosed MM patients treated with novel triple-drug initial therapy and frontline autologous stem-cell transplantation, MRD-negative status did not mitigate the poor-prognosis outcomes of high-risk MM patients.
Collapse
Affiliation(s)
- Chutima Kunacheewa
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hans C Lee
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Krina Patel
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sheeba Thomas
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Behrang Amini
- Division of Diagnostic Imaging, Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Samer Srour
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Qaiser Bashir
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yago Nieto
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Muzzaffar H Qazilbash
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Donna M Weber
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lei Feng
- Department of Statistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Robert Z Orlowski
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Pei Lin
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Elisabet E Manasanch
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
26
|
Wang G, Ning FY, Wang JH, Yan HM, Kong HW, Zhang YT, Shen Q. Expression of interleukin-32 in bone marrow of patients with myeloma and its prognostic significance. World J Clin Cases 2019; 7:4234-4244. [PMID: 31911904 PMCID: PMC6940335 DOI: 10.12998/wjcc.v7.i24.4234] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 11/14/2019] [Accepted: 11/26/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The guiding effect of prognostic stratification in multiple myeloma (MM) for treatment has been increasingly emphasized in recent years. The stratification of risk factors based on the International Staging System (ISS), Durie-Salmon (DS) staging and related indicators is affected by the renal function of patients, resulting in poor performance. This study assesses the relationship between interleukin-32 (IL-32) and related risk factors in 67 patients with MM and their clinical outcomes.
AIM To investigate the feasibility of IL-32 in evaluating prognosis in patients with MM and the factors influencing prognosis.
METHODS This was a pragmatic, prospective observational study of patients with MM at a single center. According to IL-32 level, patients were divided into two groups. The variables under consideration included age, blood β2-microglobulin, albumin, C-reactive protein, serum calcium, serum creatinine, lactate dehydrogenase, M protein type, ISS stage, DS stage, and IL-32 levels and minimal residual disease (MRD) after induction treatment. The main outcomes were progression-free survival (PFS) and overall survival (OS).
RESULTS IL-32 was an important factor affecting PFS and OS in patients with MM. Compared with patients with IL-32 levels ≥ 856.4 pg/mL, patients with IL-32 levels < 856.4 pg/mL had longer PFS (P = 0.0387) and OS (P = 0.0379); Univariate analysis showed that IL-32 level and MRD were significantly associated with OS and PFS (P < 0.05). Multivariate analysis showed that IL-32 levels ≥ 856.4 pg/mL and MRD positive were still independent risk factors for OS and PFS (P < 0.05).
CONCLUSION IL-32 is valuable for assessing the prognosis of MM patients. IL-32 level combined with MRD may be a useful routine evaluation index for MM patients after treatment.
Collapse
Affiliation(s)
- Gang Wang
- Department of Hematology, Quzhou People’s Hospital, Quzhou 324000, Zhejiang Province, China
| | - Fang-Ying Ning
- Department of Hematology, People’s Hospital of Hangzhou Medical College, Zhejiang Provincial People’s Hospital, Hangzhou 310000, Zhejiang Province, China
| | - Jia-Heng Wang
- Department of Hematology, Quzhou People’s Hospital, Quzhou 324000, Zhejiang Province, China
| | - Hai-Meng Yan
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| | - Hong-Wei Kong
- Department of Hematology, Quzhou People’s Hospital, Quzhou 324000, Zhejiang Province, China
| | - Yu-Ting Zhang
- Adicon Clinical Laboratories Inc., Hangzhou 310023, Zhejiang Province, China
| | - Qiang Shen
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
27
|
D’Agostino M, Bertamini L, Oliva S, Boccadoro M, Gay F. Pursuing a Curative Approach in Multiple Myeloma: A Review of New Therapeutic Strategies. Cancers (Basel) 2019; 11:E2015. [PMID: 31847174 PMCID: PMC6966449 DOI: 10.3390/cancers11122015] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/05/2019] [Accepted: 12/09/2019] [Indexed: 12/28/2022] Open
Abstract
Multiple myeloma (MM) is still considered an incurable hematologic cancer and, in the last decades, the treatment goal has been to obtain a long-lasting disease control. However, the recent availability of new effective drugs has led to unprecedented high-quality responses and prolonged progression-free survival and overall survival. The improvement of response rates has prompted the development of new, very sensitive methods to measure residual disease, even when monoclonal components become undetectable in patients' serum and urine. Several scientific efforts have been made to develop reliable and validated techniques to measure minimal residual disease (MRD), both within and outside the bone marrow. With the newest multidrug combinations, a good proportion of MM patients can achieve MRD negativity. Long-lasting MRD negativity may prove to be a marker of "operational cure", although the follow-up of the currently ongoing studies is still too short to draw conclusions. In this article, we focus on results obtained with new-generation multidrug combinations in the treatment of high-risk smoldering MM and newly diagnosed MM, including the potential role of MRD and MRD-driven treatment strategies in clinical trials, in order to optimize and individualize treatment.
Collapse
Affiliation(s)
| | | | | | | | - Francesca Gay
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, 10126 Torino, Italy
| |
Collapse
|
28
|
Monitoring the cytogenetic architecture of minimal residual plasma cells indicates therapy-induced clonal selection in multiple myeloma. Leukemia 2019; 34:578-588. [PMID: 31591469 DOI: 10.1038/s41375-019-0590-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/19/2019] [Indexed: 11/09/2022]
Abstract
Recent attempts have focused on identifying fewer magnitude of minimal residual disease (MRD) rather than exploring the biological and genetic features of the residual plasma cells (PCs). Here, a cohort of 193 patients with at least one cytogenetic abnormalities (CA) at diagnosis were analyzed, and interphase fluorescence in situ hybridization (iFISH) analyses were performed in patient-paired diagnostic and posttherapy samples. Persistent CA in residual PCs were observed for the majority of patients (63%), even detectable in 28/63 (44%) patients with MRD negativity (<10-4). The absence of CA in residual PCs was associated with prolonged survival regardless of MRD status. According to the change of the clonal size of specific CA, patients were clustered into five groups, reflecting different patterns of clone selection under therapy pressure. Therapy-induced clonal selection exerted a significant impact on survival (HR = 4.0; P < 0.001). According to the longitudinal cytogenetic studies at relapse, sequential cytogenetic dynamics were observed in most patients, and cytogenetic architecture of residual PCs could to some extent predict the evolutional pattern at relapse. Collectively, the repeat cytogenetic evaluation in residual PCs could not only serves as a good complementary tool for MRD detection, but also provides a better understanding of clinical response and clonal evolution.
Collapse
|
29
|
Romano A, Palumbo GA, Parrinello NL, Conticello C, Martello M, Terragna C. Minimal Residual Disease Assessment Within the Bone Marrow of Multiple Myeloma: A Review of Caveats, Clinical Significance and Future Perspectives. Front Oncol 2019; 9:699. [PMID: 31482061 PMCID: PMC6710454 DOI: 10.3389/fonc.2019.00699] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022] Open
Abstract
There is an increasing clinical interest in the measure and achievement of minimal residual disease (MRD) negativity in the bone marrow of Multiple Myeloma (MM) patients, as defined equally either by Multicolor Flow Cytometry (MFC) or by Next Generation Sequencing (NGS) technologies. At present, modern technologies allow to detect up to one on 104 or on 105 or even on 106 cells, depending on their throughput. MFC approaches, which have been progressively improved up to the so-called Next Generation Flow (NGF), and NGS, which proved clear advantages over ASO-PCR, can detect very low levels of residual disease in the BM. These methods are actually almost superimposable, in terms of MRD detection power, supporting the lack of unanimous preference for either technique on basis of local availability. However, some technical issues are still open: the optimal assay to use to detect either phenotype (e.g., next generation multidimensional flow cytometry, imaging) or genotype aberrations (e.g., ASO-RQ PCR, digital droplet PCR, NGS) and their standardization, the sample source (BM or peripheral blood, PB) and its pre-processing (red-cell lysis vs. Ficoll, fresh vs. frozen samples, requirement of CD138+ cells enrichment). Overall, MRD negativity is considered as the most powerful predictor of favorable long-term outcomes in MM and is likely to represent the major driver of treatment strategies in the near future. In this manuscript, we reviewed the main pitfalls and caveats of MRD detection within bone marrow in MM patients after front-line therapy, highlighting the improving of the currently employed technology and describing alternative methods for MRD testing in MM, such as liquid biopsy.
Collapse
Affiliation(s)
- Alessandra Romano
- Department of Surgery and Medical Specialties, University of Catania, Catania, Italy
| | - Giuseppe Alberto Palumbo
- Division of Hematology, Azienda Ospedaliero-Universitaria Policlinico Vittorio Emanuele di Catania, Catania, Italy
- Dipartimento di Scienze Mediche, Chirurgiche e Tecnologie avanzate “G.F. Ingrassia,” University of Catania, Catania, Italy
| | - Nunziatina Laura Parrinello
- Division of Hematology, Azienda Ospedaliero-Universitaria Policlinico Vittorio Emanuele di Catania, Catania, Italy
- Dipartimento di Scienze Mediche, Chirurgiche e Tecnologie avanzate “G.F. Ingrassia,” University of Catania, Catania, Italy
| | - Concetta Conticello
- Division of Hematology, Azienda Ospedaliero-Universitaria Policlinico Vittorio Emanuele di Catania, Catania, Italy
| | - Marina Martello
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale (DIMES), Università degli Studi di Bologna, Bologna, Italy
| | - Carolina Terragna
- Istituto di Ematologia “L.A.Seràgnoli,” Azienda Ospedaliera Sant'Orsola-Malpighi, Bologna, Italy
| |
Collapse
|
30
|
Bal S, Weaver A, Cornell RF, Costa LJ. Challenges and opportunities in the assessment of measurable residual disease in multiple myeloma. Br J Haematol 2019; 186:807-819. [PMID: 31364160 DOI: 10.1111/bjh.16130] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Treatment response assessment in multiple myeloma (MM) relies on the detection of paraprotein in serum and/or urine, bone marrow morphology and immunohistochemistry. With remarkable advances in therapy, particularly in the newly diagnosed setting, achievement of complete remission became frequent, creating the need to identify smaller amounts of residual disease and understand their prognostic and therapeutic implications. Measurable residual disease (MRD) can be assessed primarily by flow cytometry and next generation sequencing and state-of-the-art assays have sensitivity approaching 1 in 106 cells. This review discusses the existing challenges in utilizing MRD to inform management of MM and highlights open research questions and opportunities as MRD is more routinely incorporated into clinical practice for patients with MM.
Collapse
Affiliation(s)
- Susan Bal
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Allison Weaver
- Division of Hematology and Oncology, Vanderbilt University, Nashville, TN, USA
| | - Robert F Cornell
- Division of Hematology and Oncology, Vanderbilt University, Nashville, TN, USA
| | - Luciano J Costa
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
31
|
|
32
|
Mizuta S, Kawata T, Kawabata H, Yamane N, Mononobe S, Komai T, Koba Y, Ukyo N, Tamekane A, Watanabe M. VS38 as a promising CD38 substitute antibody for flow cytometric detection of plasma cells in the daratumumab era. Int J Hematol 2019; 110:322-330. [DOI: 10.1007/s12185-019-02685-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 05/30/2019] [Accepted: 05/31/2019] [Indexed: 12/15/2022]
|
33
|
Yao Q, Bai Y, Orfao A, Chim CS. Standardized Minimal Residual Disease Detection by Next-Generation Sequencing in Multiple Myeloma. Front Oncol 2019; 9:449. [PMID: 31245284 PMCID: PMC6563351 DOI: 10.3389/fonc.2019.00449] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 05/13/2019] [Indexed: 11/13/2022] Open
Abstract
Next-generation sequencing (NGS) has been applied to monitor minimal residual disease (MRD) in multiple myeloma (MM). Standardized DNA input and sequencing depth is essential for achieving a uniform sensitivity in NGS-based MRD study. Herein, the sensitivity of 10−5 was verified by a standardized experimental design based on triplicate measurements of 1 μg DNA input and 1 million sequencing reads using the LymphoTrack-MiSeq platform. MRD level was defined as the mean MRD burden of the triplicates. Two spike-in controls at concentrations of 0.001% tumor plasma cells (PC) for verifying the sensitivity of 10−5 and 0.01% (or 0.005%) for MRD normalization were systematically analyzed. The spike-in control of 0.001% MRD was consistently detected in all samples, confirming a sensitivity of 10−5. Moreover, this standardized NGS approach yielded MRD measurements concordant with serological response and comparable to allele-specific oligonucleotide (ASO) real-time quantitative (RQ)-PCR. Moreover, NGS showed an improved sensitivity and provided quantification of MRD for cases assigned “positive but not quantifiable” (PNQ) by ASO RQ-PCR, even without the use of patient-specific probes/primers. Issues regarding the specificity of myeloma-specific sequences as MRD target, optimal input for spike-in normalization, and interpretation of MRD from triplicates are discussed. Herein, the standardized LymphoTrack-MiSeq-based method is verified to carry a sensitivity of 10−5, hence an effective tool for MRD monitoring in MM. As only a small number of samples are tested here, further study with a larger number of patients is warranted.
Collapse
Affiliation(s)
- Qiumei Yao
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| | - Yinlei Bai
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Alberto Orfao
- Department of Medicine and Cytometry Service (Nucleus), Cancer Research Centre (IBMCC, USAL-CSIC) and CIBERONC, Institute for Biomedical Research of Salamanca (IBSAL), University of Salamanca (USAL), Salamanca, Spain
| | - Chor Sang Chim
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Pokfulam, Hong Kong
| |
Collapse
|
34
|
A Phase I Study of the Safety and Feasibility of Bortezomib in Combination With G-CSF for Stem Cell Mobilization in Patients With Multiple Myeloma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 19:e588-e593. [PMID: 31358485 DOI: 10.1016/j.clml.2019.04.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/24/2019] [Accepted: 04/29/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND We previously reported that administration of bortezomib (BTZ) after 4 days of granulocyte colony-stimulating factor (G-CSF) significantly augments mobilization in mice. We hypothesized that administration of BTZ at peak G-CSF mobilization in patients with multiple myeloma (MM) would be safe, augment mobilization, and have an in vivo purging effect on circulating myeloma cells. PATIENTS AND METHODS This was a phase I study using 3 dose levels of BTZ. G-CSF was administered for 5 days. On the evening of the fourth day, a single dose of BTZ was administered. Peripheral blood was drawn 1 to 2 hours before and 15 to 18 hours after BTZ administration (before day 5 G-CSF administration) to analyze the mobilization effect of BTZ. Standard apheresis was then performed starting on day 5. After mobilization, patients underwent autologous stem cell transplantation (ASCT) per institutional guidelines. RESULTS Ten patients were enrolled. There were no dose-limiting toxicities. Median peripheral blood CD34+ cells at day 4 before BTZ administration was 16 per microliter and 15 hours later was 32 per microliter suggesting that administration of BTZ at peak G-CSF mobilization augments the mobilization effect of G-CSF. The effect of BTZ on circulating MM cells was unclear. All patients had successful engraftment after ASCT. CONCLUSION Administration of 1 dose of BTZ at peak G-CSF mobilization was safe and well tolerated, enhanced stem cell mobilization, and did not affect graft viability. The mobilization effect of BTZ at peak G-CSF mobilization shown in this phase I study needs to be confirmed in a larger randomized trial.
Collapse
|
35
|
Terpos E, Ntanasis-Stathopoulos I. Multiple Myeloma: Clinical Updates From the American Society of Hematology Annual Meeting 2018. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 19:e324-e336. [PMID: 31076371 DOI: 10.1016/j.clml.2019.03.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 03/08/2019] [Indexed: 12/28/2022]
Abstract
Herein, we summarize the novel clinical data for multiple myeloma (MM) that were presented in the 2019 Annual Meeting of the American Society of Hematology. Triplet regimens including lenalidomide-dexamethasone for high-risk smoldering MM are effective but longer follow-up data are needed. Among transplant-eligible, newly diagnosed MM (NDMM) patients, carfilzomib- and daratumumab-based combinations are promising as effective and safe induction regimens and do not impair stem cell collection. Maintenance with ixazomib results in prolonged progression-free survival (PFS) compared with placebo. Regarding transplant-ineligible NDMM patients, large phase III studies showed that the additional use of daratumumab in backbone first-line regimens provides deep responses and PFS prolongation, whereas dose-/schedule-adjusted lenalidomide-dexamethasone has similar efficacy and is more tolerable than continuous lenalidomide-dexamethasone. In the relapsed/refractory setting carfilzomib- and daratumumab-based regimens remain highly effective and safe treatments, whereas the introduction of venetoclax, isatuximab, atezolizumab, and oprozomib broadens the therapeutic options. Among heavily pretreated MM patients, selinexor and melflufen showed particularly encouraging results. Novel immunotherapeutic approaches including chimeric antigen receptor T cells against B-cell maturation antigen and bispecific antibodies constitute a promising alternative that remains to be evaluated in later-phase studies.
Collapse
Affiliation(s)
- Evangelos Terpos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | -
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
36
|
Rustad EH, Hultcrantz M, Yellapantula VD, Akhlaghi T, Ho C, Arcila ME, Roshal M, Patel A, Chen D, Devlin SM, Jacobsen A, Huang Y, Miller JE, Papaemmanuil E, Landgren O. Baseline identification of clonal V(D)J sequences for DNA-based minimal residual disease detection in multiple myeloma. PLoS One 2019; 14:e0211600. [PMID: 30901326 PMCID: PMC6430394 DOI: 10.1371/journal.pone.0211600] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 01/16/2019] [Indexed: 12/21/2022] Open
Abstract
Tracking of clonal immunoglobulin V(D)J rearrangement sequences by next generation sequencing is highly sensitive for minimal residual disease in multiple myeloma. However, previous studies have found variable rates of V(D)J sequence identification at baseline, which could limit tracking. Here, we aimed to define the factors influencing the identification of clonal V(D)J sequences. Bone marrow mononuclear cells from 177 myeloma patients underwent V(D)J sequencing by the LymphoTrack assays (Invivoscribe). As a molecular control for tumor cell content, we sequenced the samples using our in-house myeloma panel myTYPE. V(D)J sequence clonality was identified in 81% of samples overall, as compared with 95% in samples where tumor-derived DNA was detectable by myTYPE. Clonality was detected more frequently in patients with lambda-restricted disease, mainly because of increased detection of kappa gene rearrangements. Finally, we describe how the tumor cell content of bone marrow aspirates decrease gradually in sequential pulls because of hemodilution: From the initial pull used for aspirate smear, to the final pull that is commonly used for research. In conclusion, baseline clonality detection rates of 95% or higher are feasible in multiple myeloma. Optimal performance depends on the use of good quality aspirates and/or subsequent tumor cell enrichment.
Collapse
Affiliation(s)
- Even H. Rustad
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, NTNU, Trondheim, Norway
| | - Malin Hultcrantz
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Venkata D. Yellapantula
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Theresia Akhlaghi
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Caleb Ho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Maria E. Arcila
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Mikhail Roshal
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Akshar Patel
- Center for Hematological Malignancies, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Denise Chen
- Marie-Josee and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Sean M. Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | | | - Ying Huang
- Invivoscribe, Inc, San Diego, CA, United States of America
| | | | - Elli Papaemmanuil
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Ola Landgren
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
- * E-mail:
| |
Collapse
|
37
|
Measurable residual disease detection by high-throughput sequencing improves risk stratification for pediatric B-ALL. Blood 2017; 131:1350-1359. [PMID: 29284596 DOI: 10.1182/blood-2017-09-806521] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 12/12/2017] [Indexed: 12/13/2022] Open
Abstract
Early response to induction chemotherapy is an important prognostic factor in B-lymphoblastic leukemia (B-ALL). Here, we compare high-throughput sequencing (HTS) of IGH and TRG genes vs flow cytometry (FC) for measurable residual disease (MRD) detection at the end of induction chemotherapy in pediatric patients with newly diagnosed B-ALL. Six hundred nineteen paired pretreatment and end-of-induction bone marrow samples from Children's Oncology Group studies AALL0331 (clinicaltrials.gov #NCT00103285) (standard risk [SR]; with MRD by FC at any level) and AALL0232 (clinicaltrials.gov #NCT00075725) (high risk; with day 29 MRD <0.1% by FC) were evaluated by HTS and FC for event-free (EFS) and overall survival (OS). HTS and FC showed similar 5-year EFS and OS for MRD-positive and -negative patients using an MRD threshold of 0.01%. However, there was a high discordant rate with HTS identifying 55 (38.7%) more patients MRD positive at this threshold. These discrepant patients have worse outcomes than FC MRD-negative patients. In addition, the increased analytic sensitivity of HTS permitted identification of 19.9% of SR patients without MRD at any detectable level who had excellent 5-year EFS (98.1%) and OS (100%). The higher analytic sensitivity and lower false-negative rate of HTS improves upon FC for MRD detection in pediatric B-ALL by identifying a novel subset of patients at end of induction who are essentially cured using current chemotherapy and identifying MRD at 0.01% in up to one-third of patients who are missed at the same threshold by FC.
Collapse
|
38
|
Takamatsu H, Wee RK, Zaimoku Y, Murata R, Zheng J, Moorhead M, Carlton VEH, Kong KA, Takezako N, Ito S, Miyamoto T, Yokoyama K, Matsue K, Sato T, Kurokawa T, Yagi H, Terasaki Y, Ohata K, Matsumoto M, Yoshida T, Faham M, Nakao S. A comparison of minimal residual disease detection in autografts among ASO-qPCR, droplet digital PCR, and next-generation sequencing in patients with multiple myeloma who underwent autologous stem cell transplantation. Br J Haematol 2017; 183:664-668. [PMID: 29270982 DOI: 10.1111/bjh.15002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Hiroyuki Takamatsu
- Haematology/Respiratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Rachel K Wee
- Haematology/Respiratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Yoshitaka Zaimoku
- Haematology/Respiratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Ryoichi Murata
- Division of Internal Medicine, Keiju Kanazawa Hospital, Kanazawa, Japan
| | | | | | | | | | - Naoki Takezako
- Department of Haematology, National Hospital Organization Disaster Medical Centre of Japan, Tachikawa, Japan
| | - Shigeki Ito
- Department of Clinical Oncology, Iwate Medical University School of Medicine, Morioka, Japan
| | - Toshihiro Miyamoto
- Medicine and Biosystemic Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenji Yokoyama
- Department of Haematology/Oncology, Tokai University Hachioji Hospital, Hachioji, Japan
| | - Kosei Matsue
- Division of Haematology/Oncology, Department of Internal Medicine, Kameda Medical Centre, Kamogawa, Japan
| | - Tsutomu Sato
- Department of Medical Oncology and Haematology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshiro Kurokawa
- Department of Haematology, Toyama Red Cross Hospital, Toyama, Japan
| | - Hideo Yagi
- Department of Haematology, Kinki University School of Medicine Nara Hospital, Ikoma, Japan
| | - Yasushi Terasaki
- Division of Internal Medicine, Toyama City Hospital, Toyama, Japan
| | - Kinya Ohata
- Haematology/Respiratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Morio Matsumoto
- Department of Haematology, National Hospital Organization Shibukawa Medical Center, Shibukawa, Japan
| | - Takashi Yoshida
- Department of Haematology, Shizuoka City Shimizu Hospital, Shizuoka, Japan
| | - Malek Faham
- Adaptive Biotechnologies Corp, Seattle, WA, USA
| | - Shinji Nakao
- Haematology/Respiratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
39
|
Takamatsu H. Comparison of Minimal Residual Disease Detection by Multiparameter Flow Cytometry, ASO-qPCR, Droplet Digital PCR, and Deep Sequencing in Patients with Multiple Myeloma Who Underwent Autologous Stem Cell Transplantation. J Clin Med 2017; 6:jcm6100091. [PMID: 28946710 PMCID: PMC5664006 DOI: 10.3390/jcm6100091] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 09/17/2017] [Accepted: 09/20/2017] [Indexed: 12/28/2022] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy with a poor prognosis, characterized by clonal proliferation of plasma cells in the bone marrow (BM). Relapse due to undetected minimal residual disease (MRD) is the leading cause of death among patients with MM. This review summarizes the methods and prognostic value of MRD assessment in BM and autografts from MM patients who underwent autologous stem cell transplantation (ASCT) by multiparameter flow cytometry (MFC), allele-specific oligonucleotide real-time quantitative PCR (ASO-qPCR), droplet digital PCR (ddPCR), and next-generation sequencing (NGS)-based detection methods. MRD assessment using NGS-based approaches has clear prognostic value and better sensitivity compared to traditional methods.
Collapse
Affiliation(s)
- Hiroyuki Takamatsu
- Hematology/Respiratory Medicine, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8641, Japan.
| |
Collapse
|
40
|
High-throughput sequencing for noninvasive disease detection in hematologic malignancies. Blood 2017; 130:440-452. [PMID: 28600337 DOI: 10.1182/blood-2017-03-735639] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/25/2017] [Indexed: 12/20/2022] Open
Abstract
Noninvasive monitoring of minimal residual disease (MRD) has led to significant advances in personalized management of patients with hematologic malignancies. Improved therapeutic options and prolonged survival have further increased the need for sensitive tumor assessment that can inform treatment decisions and patient outcomes. At diagnosis or relapse of most hematologic neoplasms, malignant cells are often easily accessible in the blood as circulating tumor cells (CTCs), making them ideal targets to noninvasively profile the molecular features of each patient. In other cancer types, CTCs are generally rare and noninvasive molecular detection relies on circulating tumor DNA (ctDNA) shed from tumor deposits into circulation. The ability to precisely detect and quantify CTCs and ctDNA could minimize invasive procedures and improve prediction of clinical outcomes. Technical advances in MRD detection methods in recent years have led to reduced costs and increased sensitivity, specificity, and applicability. Among currently available tests, high-throughput sequencing (HTS)-based approaches are increasingly attractive for noninvasive molecular testing. HTS-based methods can simultaneously identify multiple genetic markers with high sensitivity and specificity without individual optimization. In this review, we present an overview of techniques used for noninvasive molecular disease detection in selected myeloid and lymphoid neoplasms, with a focus on the current and future role of HTS-based assays.
Collapse
|