1
|
Li Q, Ai L, Zuo L, Li J, Zhao F, Xu A, Zhang B, Cai L, Hu Y, Sun C. Circulating plasma cells as a predictive biomarker in Multiple myeloma: an updated systematic review and meta-analysis. Ann Med 2024; 56:2338604. [PMID: 38599340 PMCID: PMC11008311 DOI: 10.1080/07853890.2024.2338604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/22/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Circulating plasma cells (CPCs) are defined by the presence of peripheral blood clonal plasma cells, which would contribute to the progression and dissemination of multiple myeloma (MM). An increasing number of studies have demonstrated the predictive potential of CPCs in the past few years. Therefore, there is a growing need for an updated meta-analysis to identify the specific relationship between CPCs and the prognosis of MM based on the current research status. METHODS The PubMed, Embase, and Cochrane Library databases were screened to determine eligible studies from inception to November 5, 2023. Publications that reported the prognostic value of CPCs in MM patients were included. Hazard ratios (HRs) with 95% confidence intervals (CIs) of overall survival (OS) and progression-free survival (PFS) were extracted to pool the results. Subgroup analyses were performed based on region, sample size, cut-off value, detection time, initial treatment, and data type. The association between CPCs level and clinicopathological characteristics, including the International Staging System (ISS), Revised-ISS (R-ISS), and cytogenetic abnormalities were also evaluated. Statistical analyses were conducted using STATA 17.0 software. RESULTS Twenty-two studies with a total of 5637 myeloma patients were enrolled in the current meta-analysis. The results indicated that myeloma patients with elevated CPCs were expected to have a poor OS (HR = 2.19, 95% CI: 1.81-2.66, p < 0.001) and PFS (HR = 2.45, 95% CI: 1.93-3.12, p < 0.001). Subgroup analyses did not alter the prognostic role of CPCs, regardless of region, sample size, cut-off value, detection time, initial treatment, or data type. Moreover, the increased CPCs were significantly related to advanced tumour stage (ISS III vs. ISS I-II: pooled OR = 2.89, 95% CI: 2.41-3.46, p < 0.001; R-ISS III vs. R-ISS I-II: pooled OR = 3.65, 95% CI: 2.43-5.50, p < 0.001) and high-risk cytogenetics (high-risk vs. standard-risk: OR = 2.22, 95% CI: 1.60-3.08, p < 0.001). CONCLUSION Our meta-analysis confirmed that the increased number of CPCs had a negative impact on the PFS and OS of MM patients. Therefore, CPCs could be a promising prognostic biomarker that helps with risk stratification and disease monitoring.
Collapse
Affiliation(s)
- Qun Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lisha Ai
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liping Zuo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junying Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Zhao
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Aoshuang Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Cai
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| | - Chunyan Sun
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
Chu B, Wang YT, Gao S, Shi L, Lu MQ, Fang LJ, Xiang QQ, Chen Y, Wang MZ, Wang LF, Sun K, Yang J, Duan F, Bao L. R2-ISS staging combined with circulating plasma cells improves risk stratification for newly diagnosed multiple myeloma: a single-center real-world study. Ann Hematol 2024; 103:3677-3690. [PMID: 38955826 PMCID: PMC11358218 DOI: 10.1007/s00277-024-05806-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/14/2024] [Indexed: 07/04/2024]
Abstract
We aimed to evaluate if circulating plasma cells (CPC) detected by flow cytometry could add prognostic value of R2-ISS staging. We collected the electronic medical records of 336 newly diagnosed MM patients (NDMM) in our hospital from January 2017 to June 2023. The median overall survival (OS) for patients and R2-ISS stage I-IV were not reached (NR), NR, 58 months and 53 months, respectively. There was no significant difference in OS between patients with stage I and patients with stage II (P = 0.309) or between patients with stage III and patients with stage IV (P = 0.391). All the cases were re-classified according to R2-ISS stage and CPC numbers ≥ 0.05% (CPC high) or<0.05% (CPC low) into four new risk groups: Group 1: R2-ISS stage I + R2-ISS stage II and CPC low, Group 2: R2-ISS stage II and CPC high + R2-ISS stage III and CPC low, Group 3: R2-ISS stage III and CPC high + R2-ISS stage IV and CPC low, Group 4: R2-ISS stage IV and CPC high. The median OS were NR, NR, 57 months and 32 months. OS of Group 1 was significantly longer than that of Group 2 (P = 0.033). OS in Group 2 was significantly longer than that of Group 3 (P = 0.007). OS in Group 3 was significantly longer than that of Group 4 (P = 0.041). R2-ISS staging combined with CPC can improve risk stratification for NDMM patients.
Collapse
Affiliation(s)
- Bin Chu
- Department of Hematology, Beijing Jishuitan Hospital, Capital Medical University, No. 31 East Xinjiekou Street, Xicheng District, Beijing, 100035, China
| | - Yu-Tong Wang
- Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Shan Gao
- Department of Hematology, Beijing Jishuitan Hospital, Capital Medical University, No. 31 East Xinjiekou Street, Xicheng District, Beijing, 100035, China
| | - Lei Shi
- Department of Hematology, Beijing Jishuitan Hospital, Capital Medical University, No. 31 East Xinjiekou Street, Xicheng District, Beijing, 100035, China
| | - Min-Qiu Lu
- Department of Hematology, Beijing Jishuitan Hospital, Capital Medical University, No. 31 East Xinjiekou Street, Xicheng District, Beijing, 100035, China
| | - Li-Juan Fang
- Department of Hematology, Beijing Jishuitan Hospital, Capital Medical University, No. 31 East Xinjiekou Street, Xicheng District, Beijing, 100035, China
| | - Qiu-Qing Xiang
- Department of Hematology, Beijing Jishuitan Hospital, Capital Medical University, No. 31 East Xinjiekou Street, Xicheng District, Beijing, 100035, China
| | - Yuan Chen
- Department of Hematology, Beijing Jishuitan Hospital, Capital Medical University, No. 31 East Xinjiekou Street, Xicheng District, Beijing, 100035, China
| | - Meng-Zhen Wang
- Department of Hematology, Beijing Jishuitan Hospital, Capital Medical University, No. 31 East Xinjiekou Street, Xicheng District, Beijing, 100035, China
| | - Li-Fang Wang
- Clinical Epidemiology Research Center, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Kai Sun
- Department of Hematology, Beijing Jishuitan Hospital, Capital Medical University, No. 31 East Xinjiekou Street, Xicheng District, Beijing, 100035, China
| | - Jing Yang
- Houston Methodist Cancer Center, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Fangfang Duan
- Clinical Epidemiology Research Center, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Li Bao
- Department of Hematology, Beijing Jishuitan Hospital, Capital Medical University, No. 31 East Xinjiekou Street, Xicheng District, Beijing, 100035, China.
| |
Collapse
|
3
|
Tessier C, LeBlanc R, Roy J, Trudel S, Côté J, Lalancette M, Boudreault J, Lemieux‐Blanchard É, Kaedbey R, Pavic M. Poor outcome despite modern treatments: A retrospective study of 99 patients with primary and secondary plasma cell leukemia. Cancer Med 2024; 13:e70192. [PMID: 39225552 PMCID: PMC11369989 DOI: 10.1002/cam4.70192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Plasma cell leukemia (PCL) is a rare monoclonal gammopathy, associated with short survival. Because of its very low incidence, only a few cohorts have been reported and thus, information on this disease is scarce. The goal of this study was to better understand the clinical features, prognostic factors, and efficacy of modern treatments in both primary PCL (pPCL) and secondary PCL (sPCL). METHODS We performed a retrospective, multicenter study of patients diagnosed with PCL, defined as circulating plasma cells ≥20% of total leukocytes and/or ≥2 × 109/L. RESULTS We identified 99 eligible PCL patients, of whom 33 were pPCL and 66 were sPCL. The median progression-free survival (PFS) to frontline treatment and overall survival (OS) were, respectively, 4.8 (95% CI, 0.4-9.2) and 18.3 months (95% CI, 0.0-39.0) for pPCL and 0.8 (95% CI, 0.5-1.1) and 1.2 months (95% CI, 0.9-1.5) for sPCL (both p < 0.001). We observed no improvement in OS over time (2005-2012 vs. 2013-2020, p = 0.629 for pPCL and p = 0.329 for sPCL). Finally, our data suggested that sPCL originates from a high-risk multiple myeloma (MM) population with a short OS (median 30.2 months), early relapse after stem cell transplant (median 11.9 months) and a high proportion of patients with multiple cytogenetic abnormalities (36% with ≥2 abnormalities). CONCLUSIONS This study is one of the largest PCL cohorts reported. We are also the first to investigate characteristics of MM before its transformation into sPCL and demonstrate that high-risk biologic features already present at the time of MM diagnosis. Moreover, our data highlights the lack of improvement in PCL survival in recent years and the urgent need for better treatment options.
Collapse
Affiliation(s)
| | | | - Jean Roy
- Hôpital Maisonneuve‐RosemontMontrealQuebecCanada
| | | | - Julie Côté
- Centre Hospitalier Universitaire de Québec (CHUQ), Hôpital de l'Enfant‐JésusQuebecQuebecCanada
| | - Marc Lalancette
- Centre Hospitalier Universitaire de Québec (CHUQ), Hôtel‐Dieu de QuébecQuebecQuebecCanada
| | | | | | | | - Michel Pavic
- Centre Hospitalier Universitaire de Sherbrooke (CHUS)SherbrookeQuebecCanada
- Institut de Recherche sur le Cancer de l'Université de Sherbrooke (IRCUS)SherbrookeQuebecCanada
| |
Collapse
|
4
|
Kogure Y, Handa H, Ito Y, Ri M, Horigome Y, Iino M, Harazaki Y, Kobayashi T, Abe M, Ishida T, Ito S, Iwasaki H, Kuroda J, Shibayama H, Sunami K, Takamatsu H, Tamura H, Hayashi T, Akagi K, Shinozaki T, Yoshida T, Mori I, Iida S, Maeda T, Kataoka K. ctDNA improves prognostic prediction for patients with relapsed/refractory MM receiving ixazomib, lenalidomide, and dexamethasone. Blood 2024; 143:2401-2413. [PMID: 38427753 DOI: 10.1182/blood.2023022540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/03/2024] Open
Abstract
ABSTRACT It remains elusive how driver mutations, including those detected in circulating tumor DNA (ctDNA), affect prognosis in relapsed/refractory multiple myeloma (RRMM). Here, we performed targeted-capture sequencing using bone marrow plasma cells (BMPCs) and ctDNA of 261 RRMM cases uniformly treated with ixazomib, lenalidomide, and dexamethasone in a multicenter, prospective, observational study. We detected 24 and 47 recurrently mutated genes in BMPC and ctDNA, respectively. In addition to clonal hematopoiesis-associated mutations, varying proportion of driver mutations, particularly TP53 mutations (59.2% of mutated cases), were present in only ctDNA, suggesting their subclonal origin. In univariable analyses, ctDNA mutations of KRAS, TP53, DIS3, BRAF, NRAS, and ATM were associated with worse progression-free survival (PFS). BMPC mutations of TP53 and KRAS were associated with inferior PFS, whereas KRAS mutations were prognostically relevant only when detected in both BMPC and ctDNA. A total number of ctDNA mutations in the 6 relevant genes was a strong prognostic predictor (2-year PFS rates: 57.3%, 22.7%, and 0% for 0, 1, and ≥2 mutations, respectively) and independent of clinical factors and plasma DNA concentration. Using the number of ctDNA mutations, plasma DNA concentration, and clinical factors, we developed a prognostic index, classifying patients into 3 categories with 2-year PFS rates of 57.9%, 28.6%, and 0%. Serial analysis of ctDNA mutations in 94 cases revealed that TP53 and KRAS mutations frequently emerge after therapy. Thus, we clarify the genetic characteristics and clonal architecture of ctDNA mutations and demonstrate their superiority over BMPC mutations for prognostic prediction in RRMM. This study is a part of the C16042 study, which is registered at www.clinicaltrials.gov as #NCT03433001.
Collapse
Affiliation(s)
- Yasunori Kogure
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
| | - Hiroshi Handa
- Department of Hematology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yuta Ito
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Masaki Ri
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuichi Horigome
- Department of Hematology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Masaki Iino
- Department of Hematology, Yamanashi Prefectural Central Hospital, Kofu, Japan
| | - Yoriko Harazaki
- Department of Hematology, Miyagi Cancer Center, Natori, Japan
| | - Takahiro Kobayashi
- Department of Hematology, Nephrology and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | - Masahiro Abe
- Department of Hematology, Kawashima Hospital, Tokushima, Japan
| | - Tadao Ishida
- Department of Hematology, Japanese Red Cross Medical Center, Tokyo, Japan
| | - Shigeki Ito
- Department of Hematology and Oncology, Iwate Medical University Hospital, Iwate, Japan
| | - Hiromi Iwasaki
- Department of Hematology, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Junya Kuroda
- Division of Hematology and Oncology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hirohiko Shibayama
- Department of Hematology, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Kazutaka Sunami
- Department of Hematology, National Hospital Organization Okayama Medical Center, Okayama, Japan
| | | | - Hideto Tamura
- Department of Hematology, Nippon Medical School, Tokyo, Japan
| | - Toshiaki Hayashi
- Department of Hematology, Teine Keijinkai Hospital, Sapporo, Japan
| | - Kiwamu Akagi
- Division of Molecular Diagnosis and Cancer Prevention, Saitama Cancer Center, Saitama, Japan
| | - Tomohiro Shinozaki
- Department of Information and Computer Technology, Faculty of Engineering, Tokyo University of Science, Tokyo, Japan
| | | | - Ikuo Mori
- Takeda Pharmaceutical Company Limited, Tokyo, Japan
| | - Shinsuke Iida
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takahiro Maeda
- Division of Precision Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Keisuke Kataoka
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
5
|
Binder M, Szalat RE, Talluri S, Fulciniti M, Avet-Loiseau H, Parmigiani G, Samur MK, Munshi NC. Bone marrow stromal cells induce chromatin remodeling in multiple myeloma cells leading to transcriptional changes. Nat Commun 2024; 15:4139. [PMID: 38755155 PMCID: PMC11098817 DOI: 10.1038/s41467-024-47793-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/12/2024] [Indexed: 05/18/2024] Open
Abstract
The natural history of multiple myeloma is characterized by its localization to the bone marrow and its interaction with bone marrow stromal cells. The bone marrow stromal cells provide growth and survival signals, thereby promoting the development of drug resistance. Here, we show that the interaction between bone marrow stromal cells and myeloma cells (using human cell lines) induces chromatin remodeling of cis-regulatory elements and is associated with changes in the expression of genes involved in the cell migration and cytokine signaling. The expression of genes involved in these stromal interactions are observed in extramedullary disease in patients with myeloma and provides the rationale for survival of myeloma cells outside of the bone marrow microenvironment. Expression of these stromal interaction genes is also observed in a subset of patients with newly diagnosed myeloma and are akin to the transcriptional program of extramedullary disease. The presence of such adverse stromal interactions in newly diagnosed myeloma is associated with accelerated disease dissemination, predicts the early development of therapeutic resistance, and is of independent prognostic significance. These stromal cell induced transcriptomic and epigenomic changes both predict long-term outcomes and identify therapeutic targets in the tumor microenvironment for the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Moritz Binder
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Raphael E Szalat
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
- Department of Data Science, Dana Farber Cancer Institute, Boston, MA, USA
| | - Srikanth Talluri
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | | | - Hervé Avet-Loiseau
- University Cancer Center of Toulouse, Institut National de la Santé, Toulouse, France
| | - Giovanni Parmigiani
- Department of Data Science, Dana Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mehmet K Samur
- Department of Data Science, Dana Farber Cancer Institute, Boston, MA, USA.
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Nikhil C Munshi
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
6
|
Liu X, Wu F, Ye W, Deng J, Zhang M, Zhang C, Yu Q, Cao L, Gan S, Ma J. Prognostic value of circulating plasma cells detected by flow cytometry in newly diagnosed multiple myeloma patients: a systematic review and meta-analysis. BMJ Open 2024; 14:e071548. [PMID: 38216195 PMCID: PMC10810043 DOI: 10.1136/bmjopen-2022-071548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 12/21/2023] [Indexed: 01/14/2024] Open
Abstract
OBJECTIVES Multiple myeloma (MM) is a malignant plasma cell disorder. The most widely accepted staging system for MM is the revised International Staging System based on cytogenetic and clinical biomarkers. The circulating clonal plasma cells (CPCs) were reported to have potential prognostic impact on MM. Among various diagnostic approaches, multiparametric flow cytometry (FCM) offers heightened sensitivity, minimal invasiveness and reproducibility. We conducted a meta-analysis to evaluate the prognostic value of quantifying CPCs via FCM in newly diagnosed symptomatic MM (NDMM) patients. DESIGN Systematic review and meta-analysis. DATA SOURCE PubMed, Web of Science, Embase and references of included studies. ELIGIBILITY CRITERIA FOR SELECTING STUDIES We included observational studies that evaluated the prognostic value of CPCs detected by FCM in NDMM. DATA EXTRACTION AND SYNTHESIS Data were screened and extracted independently by two investigators. The pooled results originated from random effects models. The primary endpoint was overall survival (OS). The secondary endpoint was progression-free survival (PFS). To evaluate the prognostic value of CPCs in NDMM, HRs and their 95% CI for both OS and PFS were derived using COX multivariable models. These values were then used to compute the pooled estimated effect. RESULTS Our meta-analysis encompassed a total of 2704 NDMM patients from 11 studies up to 27 August 2022. The pooled HR for OS and PFS in CPC-positive (CPCs+) group and CPC-negative group were 1.95 (95% CI 1.24 to 3.07) and 2.07 (95% CI 1.79 to 2.39), respectively. The autologous stem cell transplantation (ASCT) failed to eliminate the adverse impact on OS and PFS. The heterogeneity may stem from the use of novel agents or traditional chemotherapy as initial treatment. CONCLUSION This meta-analysis indicates CPCs+ had an adverse impact on the prognosis of NDMM patients in the total population, and the adverse impact could not be eliminated by ASCT. PROSPERO REGISTRATION NUMBER CRD42021272381.
Collapse
Affiliation(s)
- Xiaoyan Liu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feifei Wu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wu Ye
- Department of Hematology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Jili Deng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital and Institute, Beijing, China
| | - Mengmeng Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Congli Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qingfeng Yu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li Cao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Silin Gan
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jie Ma
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
7
|
Jerez J, Sanchez F, Flores F, Guajardo L, Briones JL, Selman C. Early Detection and Diagnostic Approach Through Automated Hematological Analysis for Plasma Cell Leukemia. J Med Cases 2024; 15:31-36. [PMID: 38328809 PMCID: PMC10846498 DOI: 10.14740/jmc4188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 01/16/2024] [Indexed: 02/09/2024] Open
Abstract
Plasma cell leukemia (PCL) is a clinically aggressive variant of multiple myeloma, characterized by a high burden of circulating plasma cells, necessitating swift and accurate diagnosis due to its poor prognosis. The conventional diagnostic criteria, including the recent recommendation by the International Myeloma Working Group (IMWG) of > 5% circulating plasma cells as positive, have evolved over time. In this context, we present a detailed case report that underscores the pivotal role of the ADVIA 2120 automated hematology counter in detecting plasma cells through cytogram analysis, along with the significance of routine peripheral blood smear analysis and the utility of a large unstained cells (LUCs) threshold of > 4.5% as an indicator for PCL. The case involves a 64-year-old patient with relapsed multiple myeloma and stable paraprotein levels who experienced sudden renal impairment. In this case report, we highlight how ADVIA analysis and cytochemistry assisted in the diagnosis, and further explore ADVIA's utility in this challenging leukemia.
Collapse
Affiliation(s)
- Joaquin Jerez
- Department of Haematology, Fundacion Arturo Lopez Perez, Providencia, Chile
| | - Francisca Sanchez
- Department of Diagnostics Units, Fundacion Arturo Lopez Perez, Providencia, Chile
| | - Francisco Flores
- Department of Diagnostics Units, Fundacion Arturo Lopez Perez, Providencia, Chile
| | - Lissette Guajardo
- Department of Diagnostics Units, Fundacion Arturo Lopez Perez, Providencia, Chile
| | - Jose Luis Briones
- Department of Haematology, Fundacion Arturo Lopez Perez, Providencia, Chile
- Department of Diagnostics Units, Fundacion Arturo Lopez Perez, Providencia, Chile
| | - Carolina Selman
- Department of Diagnostics Units, Fundacion Arturo Lopez Perez, Providencia, Chile
| |
Collapse
|
8
|
Pérez-Escurza O, Flores-Montero J, Óskarsson JÞ, Sanoja-Flores L, Del Pozo J, Lecrevisse Q, Martín S, Reed ER, Hákonardóttir GK, Harding S, Þorsteinsdóttir S, Rögnvaldsson S, Love TJ, Durie B, Kristinsson SY, Orfao A. Immunophenotypic assessment of clonal plasma cells and B-cells in bone marrow and blood in the diagnostic classification of early stage monoclonal gammopathies: an iSTOPMM study. Blood Cancer J 2023; 13:182. [PMID: 38072838 PMCID: PMC10711003 DOI: 10.1038/s41408-023-00944-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/26/2023] [Accepted: 11/03/2023] [Indexed: 12/18/2023] Open
Abstract
Monoclonal gammopathy of undetermined significance (MGUS) is the earliest discernible stage of multiple myeloma (MM) and Waldenström's macroglobulinemia (WM). Early diagnosis of MG may be compromised by the low-level infiltration, undetectable to low-sensitive methodologies. Here, we investigated the prevalence and immunophenotypic profile of clonal (c) plasma cells (PC) and/or cB-lymphocytes in bone marrow (BM) and blood of subjects with a serum M-component from the iSTOPMM program, using high-sensitive next-generation flow cytometry (NGF), and its utility in the diagnostic classification of early-stage MG. We studied 164 paired BM and blood samples from 82 subjects, focusing the analysis on: 55 MGUS, 12 smoldering MM (SMM) and 8 smoldering WM (SWM). cPC were detected in 84% of the BM samples and cB-lymphocytes in 45%, coexisting in 39% of cases. In 29% of patients, the phenotypic features of cPC and/or cB-lymphocytes allowed a more accurate disease classification, including: 19/55 (35%) MGUS, 1/12 (8%) SMM and 2/8 (25%) SWM. Blood samples were informative in 49% of the BM-positive cases. We demonstrated the utility of NGF for a more accurate diagnostic classification of early-stage MG.
Collapse
Affiliation(s)
- Oihane Pérez-Escurza
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
| | - Juan Flores-Montero
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Department of Hematology, University Hospital of Salamanca, Salamanca, Spain
| | | | - Luzalba Sanoja-Flores
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Institute of Biomedicine of Seville, Department of Hematology, University Hospital Virgen del Rocío of the Consejo Superior de Investigaciones Científicas (CSIC), University of Seville, Seville, Spain
| | - Julio Del Pozo
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
| | - Quentin Lecrevisse
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Silvia Martín
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain
| | - Elín Ruth Reed
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | | | | | - Sigrún Þorsteinsdóttir
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Rigshospitalet, Copenhagen, Denmark
| | - Sæmundur Rögnvaldsson
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Department of Science, Landspitali University Hospital, Reykjavík, Iceland
| | - Thorvardur Jon Love
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Department of Science, Landspitali University Hospital, Reykjavík, Iceland
| | - Brian Durie
- Department of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sigurður Yngvi Kristinsson
- Faculty of Medicine, University of Iceland, Reykjavík, Iceland
- Department of Science, Landspitali University Hospital, Reykjavík, Iceland
| | - Alberto Orfao
- Translational and Clinical Research Program, Cancer Research Center (IBMCC, CSIC-University of Salamanca); Cytometry Service, NUCLEUS; Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain.
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.
- Department of Medicine, University of Salamanca (Universidad de Salamanca), Salamanca, Spain.
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
9
|
Sathya P, Kayal S, Srinivas BH, Hamide A, Kar R. Quantification of circulating clonal plasma cells by multiparametric flow cytometry as a prognostic marker in patients with newly diagnosed multiple myeloma. Int J Lab Hematol 2023; 45:917-926. [PMID: 37632156 DOI: 10.1111/ijlh.14156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 08/10/2023] [Indexed: 08/27/2023]
Abstract
BACKGROUND Studies have shown that the quantification of circulating clonal plasma cells (cCPCs) in peripheral blood using flow cytometry could be used as a prognostic predictor of poor outcome in multiple myeloma (MM). METHODS In 66 newly diagnosed MM, cCPCs were quantified (cCPC%) and analysed for association with outcome and survival. Single-tube combined surface (CD45/CD19/CD138/CD38/CD56/CD27/CD81 as per availability) and cytoplasmic (kappa/lambda) staining was done using pre-titrated volumes of antibodies. In 26 patients, repeat cCPC% was assessed post-induction therapy. For association studies, treatment response has been taken as good (VGPR and above) and poor (PR and below). All statistical analyses were performed with SPSS software version 16.0. RESULTS There was no significant association between cCPC% at baseline with staging (p = 0.43), β2 -microglobulin (p = 0.27) and albumin (p = 0.08). There was a significant difference between the pre-induction and post-induction cCPC% (p = 0.0001). The patients were segregated using a cut-off of ≥0.197 and <0.197 based on the median values of baseline cCPC%. The post-induction outcome was available for 47 patients among whom 33 (70%) had VGPR and above. There was a significant association between higher cCPC% at baseline with poor treatment response (p = 0.008). The median OS in the study patients was 42 (CI 28.14-43.03) months and the median PFS was 39 (CI 28.49-49.04) months. Higher cCPC% showed a lower median PFS (30 vs. 39 months) and OS (35 vs. 41 months) compared to lower cCPC% though it was not statistically significant. CONCLUSION Flow cytometric baseline cCPC% in newly diagnosed MM was associated with poor treatment response and survival.
Collapse
Affiliation(s)
- Pandurangan Sathya
- Department of Pathology, Jawaharlal Institute of Post-Graduate Medical Education and Research, Pondicherry, India
| | - Smita Kayal
- Department of Medical Oncology, Jawaharlal Institute of Post-Graduate Medical Education and Research, Pondicherry, India
| | - B H Srinivas
- Department of Pathology, Jawaharlal Institute of Post-Graduate Medical Education and Research, Pondicherry, India
| | - Abdoul Hamide
- Department of Medicine, Jawaharlal Institute of Post-Graduate Medical Education and Research, Pondicherry, India
| | - Rakhee Kar
- Department of Pathology, Jawaharlal Institute of Post-Graduate Medical Education and Research, Pondicherry, India
| |
Collapse
|
10
|
Banerjee R, Cicero KI, Lee SS, Cowan AJ. Definers and drivers of functional high-risk multiple myeloma: insights from genomic, transcriptomic, and immune profiling. Front Oncol 2023; 13:1240966. [PMID: 37849816 PMCID: PMC10577204 DOI: 10.3389/fonc.2023.1240966] [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: 06/15/2023] [Accepted: 09/18/2023] [Indexed: 10/19/2023] Open
Abstract
Traditional prognostic models for newly diagnosed patients with multiple myeloma (MM), including International Staging System criteria and number of high-risk chromosomal abnormalities, are based on disease characteristics at diagnosis. However, the identification of patients at risk of more rapidly progressive MM is inherently a dynamic assessment. In a subset of patients with MM, adverse disease biology only becomes evident after the failure of first-line therapy. We define this entity as functional high-risk MM (FHRMM), encompassing relapse within 18 months of treatment initiation and/or within 12 months of frontline autologous stem cell transplantation. FHRMM is not adequately captured by traditional prognostic models, and there is a need for better understanding of mechanisms or risk factors for early relapse or progression. In this review, we explore potential definitions of FHRMM before delving into its underlying drivers based on genetic, transcriptomic, and immune cell profiling studies. Emerging data suggest that specific features of both myeloma cells and immune cells can enable the FHRMM phenotype. We conclude our review by discussing ongoing and future studies that seek to identify and intervene upon patients with FHRMM preemptively.
Collapse
Affiliation(s)
- Rahul Banerjee
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Kara I. Cicero
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Sarah S. Lee
- Division of Myeloma, Department of Hematology & Hematopoietic Cell Transplantation, City of Hope, CA, United States
| | - Andrew J. Cowan
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| |
Collapse
|
11
|
Du J, Gu XR, Yu XX, Cao YJ, Hou J. Essential procedures of single-cell RNA sequencing in multiple myeloma and its translational value. BLOOD SCIENCE 2023; 5:221-236. [PMID: 37941914 PMCID: PMC10629747 DOI: 10.1097/bs9.0000000000000172] [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: 05/17/2023] [Accepted: 09/18/2023] [Indexed: 11/10/2023] Open
Abstract
Multiple myeloma (MM) is a malignant neoplasm characterized by clonal proliferation of abnormal plasma cells. In many countries, it ranks as the second most prevalent malignant neoplasm of the hematopoietic system. Although treatment methods for MM have been continuously improved and the survival of patients has been dramatically prolonged, MM remains an incurable disease with a high probability of recurrence. As such, there are still many challenges to be addressed. One promising approach is single-cell RNA sequencing (scRNA-seq), which can elucidate the transcriptome heterogeneity of individual cells and reveal previously unknown cell types or states in complex tissues. In this review, we outlined the experimental workflow of scRNA-seq in MM, listed some commonly used scRNA-seq platforms and analytical tools. In addition, with the advent of scRNA-seq, many studies have made new progress in the key molecular mechanisms during MM clonal evolution, cell interactions and molecular regulation in the microenvironment, and drug resistance mechanisms in target therapy. We summarized the main findings and sequencing platforms for applying scRNA-seq to MM research and proposed broad directions for targeted therapies based on these findings.
Collapse
Affiliation(s)
- Jun Du
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Xiao-Ran Gu
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Xiao-Xiao Yu
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Yang-Jia Cao
- Department of Hematology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shanxi 710000, China
| | - Jian Hou
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
12
|
Abdelmonem ME, Nooh HA, El Ashry MS. Clinical Relevance of Interferon Regulatory Family-4 (IRF4) Expression in Newly Diagnosed Patients with Multiple Myeloma. Indian J Hematol Blood Transfus 2023; 39:525-536. [PMID: 37786826 PMCID: PMC10542031 DOI: 10.1007/s12288-023-01628-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 01/06/2023] [Indexed: 01/19/2023] Open
Abstract
Multiple myeloma (MM) is a malignant plasma cell neoplasm with complex biology and heterogenous course. Interferon regulatory factor 4 (IRF4) transcription factor, important key developmental stages of hematopoiesis, represents an excellent potential therapeutic target. The present work aimed to investigate the expression status of IRF4 in the diagnostic bone marrow biopsy (BMB) cores of MM patients. This prospective study included 62 newly diagnosed MM patients. The expression of IRF4 was assessed in the BMB by immunohistochemistry (IHC). The data were correlated to the patients' clinico-pathological features, response to treatment and survival rates. IRF4 expression was observed in 50% of MM patients (31/62). IRF-4 positive patients were more frequently male patients (P = 0.018), have immunoglobulin heavy chain (IgH) translocations (P = 0.05) and tended to present with a higher platelets count (P = 0.07). Multiple myeloma patients presenting with urine M-protein had worse overall survival (OS) than negative cases (P = 0.012). Normocellular BM aspirate (BMA) was associated with better OS than hypercellular and hypocellular BMA (P = 0.006). Patchy distribution of plasma cells in BMB was associated with better disease-free survival (DFS) while diffuse infiltration had the worst (P = 0.019). Of note, after treatment, MM patients had significantly lower percentage of BMA plasma cells, platelet count, β2 microglobulin and creatinine levels (P = 0.037, < 0.001, 0.022 and 0.026, respectively). Had higher albumin level (P = 0.007), compared to initial investigations. No significant association was found between IRF4 expression and the patients'clinical outcomes. Patterns of plasma cells distribution in BMB, BMA cellularity and urine M-protein are prognostically relevant in MM. Supplementary Information The online version contains supplementary material available at 10.1007/s12288-023-01628-3.
Collapse
Affiliation(s)
| | - Hend A. Nooh
- Clinical Pathology Department, National Cancer Institute, Cairo University, Kasr Al Eini Street, Fom El Khalig, P.O Box 11796, Cairo, Egypt
| | - Mona S. El Ashry
- Clinical Pathology Department, National Cancer Institute, Cairo University, Kasr Al Eini Street, Fom El Khalig, P.O Box 11796, Cairo, Egypt
| |
Collapse
|
13
|
Kreiniz N, Gertz MA. Understanding high-risk smoldering multiple myeloma. Leuk Lymphoma 2023; 64:1361-1372. [PMID: 37229535 DOI: 10.1080/10428194.2023.2216818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/27/2023]
Abstract
Smoldering multiple myeloma (SMM) is an asymptomatic condition with heterogeneous biology and various risks of progression to symptomatic disease. The best-known risk stratification models are Mayo-2018, and IWWG based on tumor burden. Recently, the personalized risk assessment tool PANGEA was introduced. New markers of SMM progression, including genomic and immune characteristics of plasma cells (PCs) and tumor microenvironment, are under investigation, and some have been incorporated into traditional scoring systems. Only one phase 3 clinical trial demonstrated an overall survival benefit of lenalidomide for high-risk SMM patients. The study has limitations, and most guidelines recommend observation or participation in clinical trials for high-risk SMM. High-intensity time-limited treatment strategies for high-risk SMM demonstrated deep responses in single-arm studies. But these treatments can cause adverse effects in asymptomatic patients.This review aims to understand better the risk of SMM progression from a clinical and biological point of view.
Collapse
Affiliation(s)
- Natalia Kreiniz
- Division of Hematology, Bnai Zion Medical Centre, Haifa, Israel
- The Ruth and Bruce Rappaport, Technion, Haifa, Israel
| | - Morie A Gertz
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
14
|
Li S, Zhang E, Cai Z. Liquid biopsy by analysis of circulating myeloma cells and cell-free nucleic acids: a novel noninvasive approach of disease evaluation in multiple myeloma. Biomark Res 2023; 11:27. [PMID: 36890597 PMCID: PMC9997021 DOI: 10.1186/s40364-023-00469-6] [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: 12/20/2022] [Accepted: 02/26/2023] [Indexed: 03/10/2023] Open
Abstract
Multiple myeloma (MM) is an incurable hematological cancer with high spatial- and temporal-heterogeneity. Invasive single-point bone marrow sampling cannot capture the tumor heterogeneity and is difficult to repeat for serial assessments. Liquid biopsy is a technique for identifying and analyzing circulating MM cells and cell products produced by tumors and released into the circulation, allowing for the minimally invasive and comprehensive detection of disease burden and molecular alterations in MM and monitoring treatment response and disease progression. Furthermore, liquid biopsy can provide complementary information to conventional detection approaches and improve their prognostic values. This article reviewed the technologies and applications of liquid biopsy in MM.
Collapse
Affiliation(s)
- Shuchan Li
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79, Qingchun Road, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Enfan Zhang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79, Qingchun Road, Hangzhou, Zhejiang, China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhen Cai
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No. 79, Qingchun Road, Hangzhou, Zhejiang, China. .,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
15
|
Jurczyszyn A, Olszewska-Szopa M, Vesole DH. The Current State of Knowledge About Evolution of Multiple Myeloma to Plasma Cell Leukemia. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:188-193. [PMID: 36593169 DOI: 10.1016/j.clml.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 11/20/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
Plasma cell leukemia is a rare form of multiple myeloma (MM). In contrast to de novo primary plasma cell leukemia (pPCL), which is very uncommon presentation of MM, there is increasing frequency of transformation to secondary plasma cell leukemia (sPCL) with increasing survival of patients (MM). The molecular basis of sPCL remains poorly understood sPCL is particularly aggressive and is associated with an extremely poor prognosis, constituting a major unmet medical need. High-quality data in sPCL regarding presentation, treatment and outcomes is limited. Herein we review the current state of knowledge on sPCL diagnostics, molecular biology, clinical characteristics, prognosis and reported treatment outcomes and the emergence of the new therapeutic strategies.
Collapse
Affiliation(s)
- Artur Jurczyszyn
- Hematology Department, Jagiellonian University Medical College, Cracow, Poland.
| | - Magdalena Olszewska-Szopa
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - David H Vesole
- Department of Medicine, John Theurer Cancer Center at Hackensack Meridian School of Medicine, Hackensack, NJ
| |
Collapse
|
16
|
Kostopoulos IV, Ntanasis-Stathopoulos I, Rousakis P, Eleutherakis-Papaiakovou E, Panteli C, Malandrakis P, Angelis N, Kanellias N, Orologas-Stavrou N, Papanota A, Fotiou D, Migkou M, Gavriatopoulou M, Kastritis E, Tsitsilonis O, Terpos E, Dimopoulos MA. Circulating Plasma Cells in Newly Diagnosed Multiple Myeloma: Prognostic and More. J Clin Oncol 2023; 41:708-710. [PMID: 36179274 DOI: 10.1200/jco.22.01606] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Affiliation(s)
- Ioannis V Kostopoulos
- Ioannis V. Kostopoulos, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Ioannis Ntanasis-Stathopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Pantelis Rousakis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Evangelos Eleutherakis-Papaiakovou, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Chrysanthi Panteli, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Panagiotis Malandrakis, MD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Angelis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Kanellias, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Orologas-Stavrou, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Aristina Papanota, MD, Despina Fotiou, MD, PhD, Magdalini Migkou, MD, PhD, Maria Gavriatopoulou, MD, PhD, and Efstathios Kastritis, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Ourania Tsitsilonis, MD, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; and Evangelos Terpos, MD, PhD, and Meletios-Athanasios Dimopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Ntanasis-Stathopoulos
- Ioannis V. Kostopoulos, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Ioannis Ntanasis-Stathopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Pantelis Rousakis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Evangelos Eleutherakis-Papaiakovou, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Chrysanthi Panteli, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Panagiotis Malandrakis, MD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Angelis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Kanellias, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Orologas-Stavrou, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Aristina Papanota, MD, Despina Fotiou, MD, PhD, Magdalini Migkou, MD, PhD, Maria Gavriatopoulou, MD, PhD, and Efstathios Kastritis, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Ourania Tsitsilonis, MD, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; and Evangelos Terpos, MD, PhD, and Meletios-Athanasios Dimopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Pantelis Rousakis
- Ioannis V. Kostopoulos, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Ioannis Ntanasis-Stathopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Pantelis Rousakis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Evangelos Eleutherakis-Papaiakovou, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Chrysanthi Panteli, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Panagiotis Malandrakis, MD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Angelis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Kanellias, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Orologas-Stavrou, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Aristina Papanota, MD, Despina Fotiou, MD, PhD, Magdalini Migkou, MD, PhD, Maria Gavriatopoulou, MD, PhD, and Efstathios Kastritis, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Ourania Tsitsilonis, MD, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; and Evangelos Terpos, MD, PhD, and Meletios-Athanasios Dimopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Eleutherakis-Papaiakovou
- Ioannis V. Kostopoulos, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Ioannis Ntanasis-Stathopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Pantelis Rousakis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Evangelos Eleutherakis-Papaiakovou, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Chrysanthi Panteli, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Panagiotis Malandrakis, MD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Angelis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Kanellias, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Orologas-Stavrou, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Aristina Papanota, MD, Despina Fotiou, MD, PhD, Magdalini Migkou, MD, PhD, Maria Gavriatopoulou, MD, PhD, and Efstathios Kastritis, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Ourania Tsitsilonis, MD, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; and Evangelos Terpos, MD, PhD, and Meletios-Athanasios Dimopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Chrysanthi Panteli
- Ioannis V. Kostopoulos, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Ioannis Ntanasis-Stathopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Pantelis Rousakis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Evangelos Eleutherakis-Papaiakovou, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Chrysanthi Panteli, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Panagiotis Malandrakis, MD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Angelis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Kanellias, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Orologas-Stavrou, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Aristina Papanota, MD, Despina Fotiou, MD, PhD, Magdalini Migkou, MD, PhD, Maria Gavriatopoulou, MD, PhD, and Efstathios Kastritis, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Ourania Tsitsilonis, MD, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; and Evangelos Terpos, MD, PhD, and Meletios-Athanasios Dimopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Malandrakis
- Ioannis V. Kostopoulos, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Ioannis Ntanasis-Stathopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Pantelis Rousakis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Evangelos Eleutherakis-Papaiakovou, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Chrysanthi Panteli, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Panagiotis Malandrakis, MD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Angelis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Kanellias, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Orologas-Stavrou, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Aristina Papanota, MD, Despina Fotiou, MD, PhD, Magdalini Migkou, MD, PhD, Maria Gavriatopoulou, MD, PhD, and Efstathios Kastritis, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Ourania Tsitsilonis, MD, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; and Evangelos Terpos, MD, PhD, and Meletios-Athanasios Dimopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Angelis
- Ioannis V. Kostopoulos, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Ioannis Ntanasis-Stathopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Pantelis Rousakis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Evangelos Eleutherakis-Papaiakovou, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Chrysanthi Panteli, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Panagiotis Malandrakis, MD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Angelis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Kanellias, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Orologas-Stavrou, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Aristina Papanota, MD, Despina Fotiou, MD, PhD, Magdalini Migkou, MD, PhD, Maria Gavriatopoulou, MD, PhD, and Efstathios Kastritis, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Ourania Tsitsilonis, MD, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; and Evangelos Terpos, MD, PhD, and Meletios-Athanasios Dimopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Kanellias
- Ioannis V. Kostopoulos, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Ioannis Ntanasis-Stathopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Pantelis Rousakis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Evangelos Eleutherakis-Papaiakovou, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Chrysanthi Panteli, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Panagiotis Malandrakis, MD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Angelis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Kanellias, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Orologas-Stavrou, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Aristina Papanota, MD, Despina Fotiou, MD, PhD, Magdalini Migkou, MD, PhD, Maria Gavriatopoulou, MD, PhD, and Efstathios Kastritis, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Ourania Tsitsilonis, MD, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; and Evangelos Terpos, MD, PhD, and Meletios-Athanasios Dimopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Orologas-Stavrou
- Ioannis V. Kostopoulos, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Ioannis Ntanasis-Stathopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Pantelis Rousakis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Evangelos Eleutherakis-Papaiakovou, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Chrysanthi Panteli, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Panagiotis Malandrakis, MD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Angelis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Kanellias, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Orologas-Stavrou, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Aristina Papanota, MD, Despina Fotiou, MD, PhD, Magdalini Migkou, MD, PhD, Maria Gavriatopoulou, MD, PhD, and Efstathios Kastritis, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Ourania Tsitsilonis, MD, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; and Evangelos Terpos, MD, PhD, and Meletios-Athanasios Dimopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Aristina Papanota
- Ioannis V. Kostopoulos, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Ioannis Ntanasis-Stathopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Pantelis Rousakis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Evangelos Eleutherakis-Papaiakovou, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Chrysanthi Panteli, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Panagiotis Malandrakis, MD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Angelis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Kanellias, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Orologas-Stavrou, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Aristina Papanota, MD, Despina Fotiou, MD, PhD, Magdalini Migkou, MD, PhD, Maria Gavriatopoulou, MD, PhD, and Efstathios Kastritis, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Ourania Tsitsilonis, MD, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; and Evangelos Terpos, MD, PhD, and Meletios-Athanasios Dimopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Despina Fotiou
- Ioannis V. Kostopoulos, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Ioannis Ntanasis-Stathopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Pantelis Rousakis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Evangelos Eleutherakis-Papaiakovou, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Chrysanthi Panteli, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Panagiotis Malandrakis, MD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Angelis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Kanellias, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Orologas-Stavrou, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Aristina Papanota, MD, Despina Fotiou, MD, PhD, Magdalini Migkou, MD, PhD, Maria Gavriatopoulou, MD, PhD, and Efstathios Kastritis, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Ourania Tsitsilonis, MD, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; and Evangelos Terpos, MD, PhD, and Meletios-Athanasios Dimopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Magdalini Migkou
- Ioannis V. Kostopoulos, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Ioannis Ntanasis-Stathopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Pantelis Rousakis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Evangelos Eleutherakis-Papaiakovou, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Chrysanthi Panteli, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Panagiotis Malandrakis, MD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Angelis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Kanellias, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Orologas-Stavrou, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Aristina Papanota, MD, Despina Fotiou, MD, PhD, Magdalini Migkou, MD, PhD, Maria Gavriatopoulou, MD, PhD, and Efstathios Kastritis, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Ourania Tsitsilonis, MD, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; and Evangelos Terpos, MD, PhD, and Meletios-Athanasios Dimopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Gavriatopoulou
- Ioannis V. Kostopoulos, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Ioannis Ntanasis-Stathopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Pantelis Rousakis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Evangelos Eleutherakis-Papaiakovou, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Chrysanthi Panteli, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Panagiotis Malandrakis, MD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Angelis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Kanellias, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Orologas-Stavrou, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Aristina Papanota, MD, Despina Fotiou, MD, PhD, Magdalini Migkou, MD, PhD, Maria Gavriatopoulou, MD, PhD, and Efstathios Kastritis, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Ourania Tsitsilonis, MD, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; and Evangelos Terpos, MD, PhD, and Meletios-Athanasios Dimopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios Kastritis
- Ioannis V. Kostopoulos, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Ioannis Ntanasis-Stathopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Pantelis Rousakis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Evangelos Eleutherakis-Papaiakovou, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Chrysanthi Panteli, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Panagiotis Malandrakis, MD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Angelis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Kanellias, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Orologas-Stavrou, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Aristina Papanota, MD, Despina Fotiou, MD, PhD, Magdalini Migkou, MD, PhD, Maria Gavriatopoulou, MD, PhD, and Efstathios Kastritis, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Ourania Tsitsilonis, MD, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; and Evangelos Terpos, MD, PhD, and Meletios-Athanasios Dimopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ourania Tsitsilonis
- Ioannis V. Kostopoulos, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Ioannis Ntanasis-Stathopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Pantelis Rousakis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Evangelos Eleutherakis-Papaiakovou, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Chrysanthi Panteli, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Panagiotis Malandrakis, MD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Angelis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Kanellias, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Orologas-Stavrou, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Aristina Papanota, MD, Despina Fotiou, MD, PhD, Magdalini Migkou, MD, PhD, Maria Gavriatopoulou, MD, PhD, and Efstathios Kastritis, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Ourania Tsitsilonis, MD, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; and Evangelos Terpos, MD, PhD, and Meletios-Athanasios Dimopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Terpos
- Ioannis V. Kostopoulos, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Ioannis Ntanasis-Stathopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Pantelis Rousakis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Evangelos Eleutherakis-Papaiakovou, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Chrysanthi Panteli, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Panagiotis Malandrakis, MD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Angelis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Kanellias, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Orologas-Stavrou, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Aristina Papanota, MD, Despina Fotiou, MD, PhD, Magdalini Migkou, MD, PhD, Maria Gavriatopoulou, MD, PhD, and Efstathios Kastritis, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Ourania Tsitsilonis, MD, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; and Evangelos Terpos, MD, PhD, and Meletios-Athanasios Dimopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Meletios-Athanasios Dimopoulos
- Ioannis V. Kostopoulos, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Ioannis Ntanasis-Stathopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Pantelis Rousakis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Evangelos Eleutherakis-Papaiakovou, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Chrysanthi Panteli, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Panagiotis Malandrakis, MD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Angelis, MSc, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Kanellias, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Nikolaos Orologas-Stavrou, MSc, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; Aristina Papanota, MD, Despina Fotiou, MD, PhD, Magdalini Migkou, MD, PhD, Maria Gavriatopoulou, MD, PhD, and Efstathios Kastritis, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Ourania Tsitsilonis, MD, PhD, Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece; and Evangelos Terpos, MD, PhD, and Meletios-Athanasios Dimopoulos, MD, PhD, Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
17
|
Mejia Saldarriaga M, Darwiche W, Jayabalan D, Monge J, Rosenbaum C, Pearse RN, Niesvizky R, Bustoros M. Advances in the molecular characterization of multiple myeloma and mechanism of therapeutic resistance. Front Oncol 2022; 12:1020011. [PMID: 36387095 PMCID: PMC9646612 DOI: 10.3389/fonc.2022.1020011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/07/2022] [Indexed: 11/25/2022] Open
Abstract
Recent insight in the genomic landscape of newly diagnosed multiple myeloma (NDMM) and its precursor conditions, monoclonal gammopathy of uncertain significance (MGUS), and smoldering myeloma have allowed the identification of patients with precursor conditions with a high risk of progression. These cases with "progressor" MGUS/SMM have a higher average mutation burden, have higher rates of mutations in specific genes such as MAPK, DNA repair, MYC, DIS3, and are enriched for specific mutational signatures when compared to non-progressors and are comparable to those found in NDMM. The highly preserved clonal heterogeneity seen upon progression of SMM, combined with the importance of these early variables, suggests that the identification of progressors based on these findings could complement and enhance the currently available clinical models based on tumor burden. Mechanisms leading to relapse/refractory multiple myeloma (RRMM) are of clinical interest given worse overall survival in this population. An Increased mutational burden is seen in patients with RRMM when compared to NDMM, however, there is evidence of branching evolution with many of these mutations being present at the subclonal level. Likewise, alterations in proteins associated with proteosome inhibitor and immunomodulatory drugs activity could partially explain clinical resistance to these agents. Evidence of chromosomal events leading to copy number changes is seen, with the presence of TP53 deletion, mutation, or a combination of both being present in many cases. Additional chromosomal events such as 1q gain and amplification may also interact and lead to resistance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mark Bustoros
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
18
|
Hofste op Bruinink D, Kuiper R, van Duin M, Cupedo T, van der Velden VH, Hoogenboezem R, van der Holt B, Beverloo HB, Valent ET, Vermeulen M, Gay F, Broijl A, Avet-Loiseau H, Munshi NC, Musto P, Moreau P, Zweegman S, van de Donk NW, Sonneveld P. Identification of High-Risk Multiple Myeloma With a Plasma Cell Leukemia-Like Transcriptomic Profile. J Clin Oncol 2022; 40:3132-3150. [PMID: 35357885 PMCID: PMC9509081 DOI: 10.1200/jco.21.01217] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 01/15/2022] [Accepted: 02/11/2022] [Indexed: 12/24/2022] Open
Abstract
PURPOSE Primary plasma cell leukemia (pPCL) is an aggressive subtype of multiple myeloma, which is distinguished from newly diagnosed multiple myeloma (NDMM) on the basis of the presence of ≥ 20% circulating tumor cells (CTCs). A molecular marker for pPCL is currently lacking, which could help identify NDMM patients with high-risk PCL-like disease, despite not having been recognized as such clinically. METHODS A transcriptomic classifier for PCL-like disease was bioinformatically constructed and validated by leveraging information on baseline CTC levels, tumor burden, and tumor transcriptomics from 154 patients with NDMM included in the Cassiopeia or HO143 trials and 29 patients with pPCL from the EMN12/HO129 trial. Its prognostic value was assessed in an independent cohort of 2,139 patients with NDMM from the HOVON-65/GMMG-HD4, HOVON-87/NMSG-18, EMN02/HO95, MRC-IX, Total Therapy 2, Total Therapy 3, and MMRF CoMMpass studies. RESULTS High CTC levels were associated with the expression of 1,700 genes, independent of tumor burden (false discovery rate < 0.05). Of these, 54 genes were selected by leave-one-out cross-validation to construct a transcriptomic classifier representing PCL-like disease. This not only demonstrated a sensitivity of 93% to identify pPCL in the validation cohort but also classified 10% of NDMM tumors as PCL-like. PCL-like MM transcriptionally and cytogenetically resembled pPCL, but presented with significantly lower CTC levels and tumor burden. Multivariate analyses in NDMM confirmed the significant prognostic value of PCL-like status in the context of Revised International Staging System stage, age, and treatment, regarding both progression-free (hazard ratio, 1.64; 95% CI, 1.30 to 2.07) and overall survival (hazard ratio, 1.89; 95% CI, 1.42 to 2.50). CONCLUSION pPCL was identified on the basis of a specific tumor transcriptome, which was also present in patients with high-risk NDMM, despite not being clinically leukemic. Incorporating PCL-like status into current risk models in NDMM may improve prognostic accuracy.
Collapse
Affiliation(s)
- Davine Hofste op Bruinink
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
- Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Rowan Kuiper
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
- SkylineDx, Rotterdam, the Netherlands
| | - Mark van Duin
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Tom Cupedo
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | - Remco Hoogenboezem
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Bronno van der Holt
- HOVON Data Center, Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - H. Berna Beverloo
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Michael Vermeulen
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Francesca Gay
- Myeloma Unit, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Annemiek Broijl
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | - Nikhil C. Munshi
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Pellegrino Musto
- “Aldo Moro” University School of Medicine, Unit of Hematology and Stem Cell Transplantation, AOUC Policlinico, Bari, Italy
| | - Philippe Moreau
- Hematology Department, University Hospital Hôtel-Dieu, Nantes, France
| | - Sonja Zweegman
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Niels W.C.J. van de Donk
- Department of Hematology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Pieter Sonneveld
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| |
Collapse
|
19
|
Allegra A, Cancemi G, Mirabile G, Tonacci A, Musolino C, Gangemi S. Circulating Tumour Cells, Cell Free DNA and Tumour-Educated Platelets as Reliable Prognostic and Management Biomarkers for the Liquid Biopsy in Multiple Myeloma. Cancers (Basel) 2022; 14:cancers14174136. [PMID: 36077672 PMCID: PMC9454477 DOI: 10.3390/cancers14174136] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Even though the presently employed biomarkers in the detection and management of multiple myeloma are demonstrating encouraging results, the mortality percentage of the malignancy is still elevated. Thus, searching for new diagnostic or prognostic markers is pivotal. Liquid biopsy allows the examination of circulating tumour DNA, cell-free DNA, extracellular RNA, and cell free proteins, which are released into the bloodstream due to the breakdown of tumour cells or exosome delivery. Liquid biopsy can now be applied in clinical practice to diagnose, and monitor multiple myeloma, probably allowing a personalized treatment of the disease. Abstract Liquid biopsy is one of the fastest emerging fields in cancer evaluation. Circulating tumour cells and tumour-originated DNA in plasma have become the new targets for their possible employ in tumour diagnosis, and liquid biopsy can define tumour burden without invasive procedures. Multiple Myeloma, one of the most frequent hematologic tumors, has been the target of therapeutic progresses in the last few years. Bone marrow aspirate is the traditional tool for diagnosis, prognosis, and genetic evaluation in multiple myeloma patients. However, this painful procedure presents a relevant drawback for regular disease examination as it requires an invasive practice. Moreover, new data demonstrated that a sole bone marrow aspirate is incapable of expressing the multifaceted multiple myeloma genetic heterogeneity. In this review, we report the emerging usefulness of the assessment of circulating tumour cells, cell-free DNA, extracellular RNA, cell-free proteins, extracellular vesicles, and tumour-educated platelets to evaluate the changing mutational profile of multiple myeloma, as early markers of disease, reliable predictors of prognosis, and as useful tools to perform less invasive monitoring in multiple myeloma.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy
- Correspondence:
| | - Gabriella Cancemi
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy
| | - Giuseppe Mirabile
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy
| | - Alessandro Tonacci
- Clinical Physiology Institute, National Research Council of Italy (IFC-CNR), 56124 Pisa, Italy
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| |
Collapse
|
20
|
Forster S, Radpour R. Molecular Impact of the Tumor Microenvironment on Multiple Myeloma Dissemination and Extramedullary Disease. Front Oncol 2022; 12:941437. [PMID: 35847862 PMCID: PMC9284036 DOI: 10.3389/fonc.2022.941437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/06/2022] [Indexed: 12/25/2022] Open
Abstract
Multiple myeloma (MM) is the most common malignant monoclonal disease of plasma cells. Aside from classical chemotherapy and glucocorticoids, proteasome inhibitors, immunomodulatory agents and monoclonal antibodies are used in the current treatment scheme of MM. The tumor microenvironment (TME) plays a fundamental role in the development and progression of numerous solid and non-solid cancer entities. In MM, the survival and expansion of malignant plasma cell clones heavily depends on various direct and indirect signaling pathways provided by the surrounding bone marrow (BM) niche. In a number of MM patients, single plasma cell clones lose their BM dependency and are capable to engraft at distant body sites or organs. The resulting condition is defined as an extramedullary myeloma (EMM). EMMs are highly aggressive disease stages linked to a dismal prognosis. Emerging literature demonstrates that the dynamic interactions between the TME and malignant plasma cells affect myeloma dissemination. In this review, we aim to summarize how the cellular and non-cellular BM compartments can promote plasma cells to exit their BM niche and metastasize to distant intra-or extramedullary locations. In addition, we list selected therapy concepts that directly target the TME with the potential to prevent myeloma spread.
Collapse
Affiliation(s)
- Stefan Forster
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ramin Radpour
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- *Correspondence: Ramin Radpour,
| |
Collapse
|
21
|
Chakraborty R, Lentzsch S. Circulating Tumor Cell Burden as a Component of Staging in Multiple Myeloma: Ready for Prime Time? J Clin Oncol 2022; 40:3099-3102. [PMID: 35759726 DOI: 10.1200/jco.22.01040] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Rajshekhar Chakraborty
- Multiple Myeloma and Amyloidosis Program, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
| | - Suzanne Lentzsch
- Multiple Myeloma and Amyloidosis Program, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
22
|
Bertamini L, Oliva S, Rota-Scalabrini D, Paris L, Morè S, Corradini P, Ledda A, Gentile M, De Sabbata G, Pietrantuono G, Pascarella A, Tosi P, Curci P, Gilestro M, Capra A, Galieni P, Pisani F, Annibali O, Monaco F, Liberati AM, Palmieri S, Luppi M, Zambello R, Fazio F, Belotti A, Tacchetti P, Musto P, Boccadoro M, Gay F. High Levels of Circulating Tumor Plasma Cells as a Key Hallmark of Aggressive Disease in Transplant-Eligible Patients With Newly Diagnosed Multiple Myeloma. J Clin Oncol 2022; 40:3120-3131. [PMID: 35666982 DOI: 10.1200/jco.21.01393] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE High levels of circulating tumor plasma cells (CTC-high) in patients with multiple myeloma are a marker of aggressive disease. We aimed to confirm the prognostic impact and identify a possible cutoff value of CTC-high for the prediction of progression-free survival (PFS) and overall survival (OS), in the context of concomitant risk features and minimal residual disease (MRD) achievement. METHODS CTC were analyzed at diagnosis with two-tube single-platform flow cytometry (sensitivity 4 × 10-5) in patients enrolled in the multicenter randomized FORTE clinical trial (ClinicalTrials.gov identifier: NCT02203643). MRD was assessed by second-generation multiparameter flow cytometry (sensitivity 10-5). We tested different cutoff values in series of multivariate (MV) Cox proportional hazards regression analyses on PFS outcome and selected the value that maximized the Harrell's C-statistic. We analyzed the impact of CTC on PFS and OS in a MV analysis including baseline features and MRD negativity. RESULTS CTC analysis was performed in 401 patients; the median follow-up was 50 months (interquartile range, 45-54 months). There was a modest correlation between the percentage of CTC and bone marrow plasma cells (r = 0.38). We identified an optimal CTC cutoff of 0.07% (approximately 5 cells/µL, C-index 0.64). In MV analysis, CTC-high versus CTC-low patients had significantly shorter PFS (hazard ratio, 2.61; 95% CI, 1.49 to 2.97, P < .001; 4-year PFS 38% v 69%) and OS (hazard ratio, 2.61; 95% CI, 1.49 to 4.56; P < .001; 4-year OS 68% v 92%). The CTC levels, but not the bone marrow plasma cell levels, affected the outcome. The only factor that reduced the negative impact of CTC-high was the achievement of MRD negativity (interaction P = .039). CONCLUSION In multiple myeloma, increasing levels of CTC above an optimal cutoff represent an easy-to-assess, robust, and independent high-risk factor. The achievement of MRD negativity is the most important factor that modulates their negative prognostic impact.
Collapse
Affiliation(s)
- Luca Bertamini
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Stefania Oliva
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Delia Rota-Scalabrini
- Multidisciplinary Oncology Outpatient Clinic, Candiolo Cancer Institute, FPO - IRCCS, Torino, Italy
| | - Laura Paris
- Division of Hematology, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Sonia Morè
- Clinica di Ematologia, AOU Ospedali Riuniti di Ancona, Ancona, Italy
| | - Paolo Corradini
- Hematology and Bone Marrow Transplant Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, University of Milan, Milan, Italy
| | - Antonio Ledda
- Ematologia/CTMO, Ospedale "A. Businco," Cagliari, Italy
| | | | - Giovanni De Sabbata
- Ematologia, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Giuseppe Pietrantuono
- Hematology and Stem Cell Transplantation Unit, IRCCS Centro di Riferimento Oncologico della Basilicata, Rionero in Vulture, Italy
| | | | | | - Paola Curci
- Unit of Hematology and Stem Cell Transplantation, AOUC Policlinico, Bari, Italy
| | - Milena Gilestro
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Andrea Capra
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Piero Galieni
- UOC Ematologia e Terapia cellulare, Ospedale C. e G. Mazzoni, Ascoli Piceno, Italy
| | - Francesco Pisani
- Hematology and Stem Cell Transplant Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Ombretta Annibali
- Unit of Hematology, Stem Cell Transplantation, University Campus Bio-Medico, Rome, Italy
| | - Federico Monaco
- SC Ematologia, Azienda Ospedaliera SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | - Anna Marina Liberati
- Università degli Studi di Perugia, Azienda Ospedaliera Santa Maria, Terni, Italy
| | | | - Mario Luppi
- Dipartimento di Scienze Mediche e Chirurgiche Materno Infantili e dell'Adulto, UNIMORE, UOC Ematologia, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Renato Zambello
- Department of Medicine (DIMED), Hematology and Clinical Immunology Section, Padova University School of Medicine, Padova, Italy
| | - Francesca Fazio
- Hematology, Department of Translational and Precision Medicine, Azienda Ospedaliera Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Angelo Belotti
- Department of Hematology, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Paola Tacchetti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli," Bologna, Italy
| | - Pellegrino Musto
- Unit of Hematology and Stem Cell Transplantation, AOUC Policlinico, Bari, Italy.,Department of Emergency and Organ Transplantation, "Aldo Moro" University School of Medicine, Bari, Italy
| | - Mario Boccadoro
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Francesca Gay
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| |
Collapse
|
23
|
Ndacayisaba LJ, Rappard KE, Shishido SN, Ruiz Velasco C, Matsumoto N, Navarez R, Tang G, Lin P, Setayesh SM, Naghdloo A, Hsu CJ, Maney C, Symer D, Bethel K, Kelly K, Merchant A, Orlowski R, Hicks J, Mason J, Manasanch EE, Kuhn P. Enrichment-Free Single-Cell Detection and Morphogenomic Profiling of Myeloma Patient Samples to Delineate Circulating Rare Plasma Cell Clones. Curr Oncol 2022; 29:2954-2972. [PMID: 35621632 PMCID: PMC9139906 DOI: 10.3390/curroncol29050242] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/07/2022] [Accepted: 04/18/2022] [Indexed: 01/27/2023] Open
Abstract
Multiple myeloma is an incurable malignancy that initiates from a bone marrow resident clonal plasma cell and acquires successive mutational changes and genomic alterations, eventually resulting in tumor burden accumulation and end-organ damage. It has been recently recognized that myeloma secondary genomic events result in extensive sub-clonal heterogeneity both in localized bone marrow areas and circulating peripheral blood plasma cells. Rare genomic subclones, including myeloma initiating cells, could be the drivers of disease progression and recurrence. Additionally, evaluation of rare myeloma cells in blood for disease monitoring has numerous advantages over invasive bone marrow biopsies. To this end, an unbiased method for detecting rare cells and delineating their genomic makeup enables disease detection and monitoring in conditions with low abundant cancer cells. In this study, we applied an enrichment-free four-plex (CD138, CD56, CD45, DAPI) immunofluorescence assay and single-cell DNA sequencing for morphogenomic characterization of plasma cells to detect and delineate common and rare plasma cells and discriminate between normal and malignant plasma cells in paired blood and bone marrow aspirates from five patients with newly diagnosed myeloma (N = 4) and monoclonal gammopathy of undetermined significance (n = 1). Morphological analysis confirms CD138+CD56+ cells in the peripheral blood carry genomic alterations that are clonally identical to those in the bone marrow. A subset of altered CD138+CD56- cells are also found in the peripheral blood consistent with the known variability in CD56 expression as a marker of plasma cell malignancy. Bone marrow tumor clinical cytogenetics is highly correlated with the single-cell copy number alterations of the liquid biopsy rare cells. A subset of rare cells harbors genetic alterations not detected by standard clinical diagnostic methods of random localized bone marrow biopsies. This enrichment-free morphogenomic approach detects and characterizes rare cell populations derived from the liquid biopsies that are consistent with clinical diagnosis and have the potential to extend our understanding of subclonality at the single-cell level in this disease. Assay validation in larger patient cohorts has the potential to offer liquid biopsy for disease monitoring with similar or improved disease detection as traditional blind bone marrow biopsies.
Collapse
Affiliation(s)
- Libere J. Ndacayisaba
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA; (L.J.N.); (K.E.R.); (S.N.S.); (C.R.V.); (N.M.); (R.N.); (S.M.S.); (A.N.); (C.-J.H.); (C.M.); (J.H.); (J.M.)
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
| | - Kate E. Rappard
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA; (L.J.N.); (K.E.R.); (S.N.S.); (C.R.V.); (N.M.); (R.N.); (S.M.S.); (A.N.); (C.-J.H.); (C.M.); (J.H.); (J.M.)
| | - Stephanie N. Shishido
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA; (L.J.N.); (K.E.R.); (S.N.S.); (C.R.V.); (N.M.); (R.N.); (S.M.S.); (A.N.); (C.-J.H.); (C.M.); (J.H.); (J.M.)
| | - Carmen Ruiz Velasco
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA; (L.J.N.); (K.E.R.); (S.N.S.); (C.R.V.); (N.M.); (R.N.); (S.M.S.); (A.N.); (C.-J.H.); (C.M.); (J.H.); (J.M.)
| | - Nicholas Matsumoto
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA; (L.J.N.); (K.E.R.); (S.N.S.); (C.R.V.); (N.M.); (R.N.); (S.M.S.); (A.N.); (C.-J.H.); (C.M.); (J.H.); (J.M.)
| | - Rafael Navarez
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA; (L.J.N.); (K.E.R.); (S.N.S.); (C.R.V.); (N.M.); (R.N.); (S.M.S.); (A.N.); (C.-J.H.); (C.M.); (J.H.); (J.M.)
| | - Guilin Tang
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (G.T.); (P.L.)
| | - Pei Lin
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (G.T.); (P.L.)
| | - Sonia M. Setayesh
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA; (L.J.N.); (K.E.R.); (S.N.S.); (C.R.V.); (N.M.); (R.N.); (S.M.S.); (A.N.); (C.-J.H.); (C.M.); (J.H.); (J.M.)
| | - Amin Naghdloo
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA; (L.J.N.); (K.E.R.); (S.N.S.); (C.R.V.); (N.M.); (R.N.); (S.M.S.); (A.N.); (C.-J.H.); (C.M.); (J.H.); (J.M.)
| | - Ching-Ju Hsu
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA; (L.J.N.); (K.E.R.); (S.N.S.); (C.R.V.); (N.M.); (R.N.); (S.M.S.); (A.N.); (C.-J.H.); (C.M.); (J.H.); (J.M.)
| | - Carlisle Maney
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA; (L.J.N.); (K.E.R.); (S.N.S.); (C.R.V.); (N.M.); (R.N.); (S.M.S.); (A.N.); (C.-J.H.); (C.M.); (J.H.); (J.M.)
| | - David Symer
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.S.); (R.O.); (E.E.M.)
| | - Kelly Bethel
- Department of Pathology, Scripps Clinic Medical Group, La Jolla, CA 92037, USA;
| | - Kevin Kelly
- Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA;
| | - Akil Merchant
- Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Robert Orlowski
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.S.); (R.O.); (E.E.M.)
| | - James Hicks
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA; (L.J.N.); (K.E.R.); (S.N.S.); (C.R.V.); (N.M.); (R.N.); (S.M.S.); (A.N.); (C.-J.H.); (C.M.); (J.H.); (J.M.)
- Department of Pathology, Scripps Clinic Medical Group, La Jolla, CA 92037, USA;
| | - Jeremy Mason
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA; (L.J.N.); (K.E.R.); (S.N.S.); (C.R.V.); (N.M.); (R.N.); (S.M.S.); (A.N.); (C.-J.H.); (C.M.); (J.H.); (J.M.)
- Institute of Urology, Catherine & Joseph Aresty Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Elisabeth E. Manasanch
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (D.S.); (R.O.); (E.E.M.)
| | - Peter Kuhn
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA 90089, USA; (L.J.N.); (K.E.R.); (S.N.S.); (C.R.V.); (N.M.); (R.N.); (S.M.S.); (A.N.); (C.-J.H.); (C.M.); (J.H.); (J.M.)
- Institute of Urology, Catherine & Joseph Aresty Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Department of Aerospace and Mechanical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA 90089, USA
- Correspondence: ; Tel.: +1-213-821-3980
| |
Collapse
|
24
|
Jia S, Bi L, Chu Y, Liu X, Feng J, Xu L, Zhang T, Gu H, Yang L, Bai Q, Liang R, Tian B, Gao Y, Tang H, Gao G. Development and Validation of a Novel Prognostic Model for Overall Survival in Newly Diagnosed Multiple Myeloma Integrating Tumor Burden and Comorbidities. Front Oncol 2022; 12:805702. [PMID: 35372057 PMCID: PMC8968003 DOI: 10.3389/fonc.2022.805702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/21/2022] [Indexed: 11/24/2022] Open
Abstract
Background Multiple myeloma (MM) is a highly heterogeneous disease with enormously variable outcomes. It remains to be a major challenge to conduct a more precise estimation of the survival of MM patients. The existing stratifications attached less importance to the prognostic significance of comorbidities. In the present study, we aimed to develop and validate a novel and simple prognostic stratification integrating tumor burden and comorbidities measured by HCT-CI. Method We retrospectively enrolled 385 consecutive newly diagnosed multiple myeloma (NDMM) patients in Xijing Hospital from January 2013 to December 2020. The cohort between January 2016 and December 2020 was selected as development cohort (N = 233), and the cohort between January 2013 and December 2015 was determined as validation cohort (N = 152). By using LASSO analysis and univariate and multivariable Cox regression analyses, we developed the MM-BHAP model in the way of nomogram composed of β2-MG, HCT-CI, ALB, and PBPC. We internally and externally validated the MM-BHAP model and compared it with ISS stage and R-ISS stage. Results The MM-BHAP model was superior to the ISS stage and partially better than the R-ISS stage according to time-dependent AUC, time-dependent C-index, DCA, IDI, and continuous NRI analyses. In predicting OS, only the MM-BHAP stratification clearly divided patients into three groups while both the ISS stage and R-ISS stage had poor classifications in patients with stage I and stage II. Moreover, the MM-BHAP stratification and the R-ISS stage performed well in predicting PFS, but not for the ISS stage. Besides, the MM-BHAP model was also applied to the patients with age ≤65 or age >65 and with or without HRCA and could enhance R-ISS or ISS classifications. Conclusions Our study offered a novel simple MM-BHAP stratification containing tumor burden and comorbidities to predict outcomes in the real-world unselected NDMM population.
Collapse
Affiliation(s)
- Shuangshuang Jia
- Department of Hematology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Lei Bi
- Department of Hematology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yuping Chu
- Department of Hematology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Xiao Liu
- Department of Hematology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Juan Feng
- Department of Hematology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Li Xu
- Department of Hematology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Tao Zhang
- Department of Hematology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Hongtao Gu
- Department of Hematology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Lan Yang
- Department of Hematology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Qingxian Bai
- Department of Hematology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Rong Liang
- Department of Hematology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Biao Tian
- Department of Hematology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Yaya Gao
- Department of Hematology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Hailong Tang
- Department of Hematology, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Guangxun Gao
- Department of Hematology, Xijing Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
25
|
Bonello F, Cani L, D'Agostino M. Risk Stratification Before and During Treatment in Newly Diagnosed Multiple Myeloma: From Clinical Trials to the Real-World Setting. Front Oncol 2022; 12:830922. [PMID: 35356221 PMCID: PMC8959380 DOI: 10.3389/fonc.2022.830922] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/08/2022] [Indexed: 12/22/2022] Open
Abstract
Multiple Myeloma (MM) is a hematologic malignancy characterized by a wide clinical and biological heterogeneity leading to different patient outcomes. Various prognostic tools to stratify newly diagnosed (ND)MM patients into different risk groups have been proposed. At baseline, the standard-of-care prognostic score is the Revised International Staging System (R-ISS), which stratifies patients according to widely available serum markers (i.e., albumin, β 2-microglobulin, lactate dehydrogenase) and high-risk cytogenetic abnormalities detected by fluorescence in situ hybridization. Though this score clearly identifies a low-risk and a high-risk population, the majority of patients are categorized as at “intermediate risk”. Although new prognostic factors identified through molecular assays (e.g., gene expression profiling, next-generation sequencing) are now available and may improve risk stratification, the majority of them need specialized centers and bioinformatic expertise that may preclude their broad application in the real-world setting. In the last years, new tools to monitor response and measurable residual disease (MRD) with very high sensitivity after the start of treatment have been developed. MRD analyses both inside and outside the bone marrow have a strong prognostic impact, and the achievement of MRD negativity may counterbalance the high-risk behavior identified at baseline. All these techniques have been developed in clinical trials. However, their efficient application in real-world clinical practice and their potential role to guide treatment-decision making are still open issues. This mini review will cover currently known prognostic factors identified before and during first-line treatment, with a particular focus on their potential applications in real-world clinical practice.
Collapse
Affiliation(s)
- Francesca Bonello
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Lorenzo Cani
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| | - Mattia D'Agostino
- SSD Clinical Trial in Oncoematologia e Mieloma Multiplo, Division of Hematology, University of Torino, Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Torino, Italy
| |
Collapse
|
26
|
Cengiz Seval G, Beksac M. Is Quantification of Measurable Clonal Plasma Cells in Stem Cell Grafts (gMRD) Clinically Meaningful? Front Oncol 2022; 12:800711. [PMID: 35280810 PMCID: PMC8904734 DOI: 10.3389/fonc.2022.800711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 01/20/2022] [Indexed: 11/20/2022] Open
Abstract
With the introduction of more effective novel therapies, the prognosis of multiple myeloma (MM) has improved significantly over the past decade, resulting with a significant proportion of patients achieving durable remissions that may reach even more than 10 years. Several studies demonstrated that the real prognostic value of complete remission (CR) relies on sustained undetectable minimal residual disease (MRD). Additionally, advances in MRD detection methods used for the detection of clonal plasma cells (cPC) inside or outside the bone marrow have also improved the value of MRD. The use of peripheral blood for MRD detection could be an effective method that overcomes the spatial heterogeneity and invasive intervention with recurrent bone marrow aspirations. During the last two decades, many groups have investigated the role of circulating plasma cells (CPCs) at diagnosis. As also presented by multiple groups during the recent ASH 2021 annual meeting, CPCs are becoming recognized as an independent prognostic factor. In addition, measurement of post-induction residual plasma cells in the stem cell graft is identified as another option for MRD assessment. Earlier studies in the era of less intensive induction regimens attempts to analyze the level of CPC contamination in the graft was shown to contribute to myeloma relapse and progression. According to these recent results, higher graft purity has been found to be in concordance with deeper responses. As expected, graft minimal residual disease (gMRD) may reflect the efficacy of induction as an additional response assessment tool. Although gMRD is a non-invasive approach, it has not gained sufficient support for routine use. In view of the hurdles related to monoclonal protein assessments, high-sensitivity cellular component measurement continues to possess its value as an end point for therapeutic efficacy. In this review, we will present a structural framework for MRD testing in peripheral blood stem cell autografts in MM and review the clinical integration into MM management.
Collapse
Affiliation(s)
| | - Meral Beksac
- Department of Hematology, School of Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
27
|
Garcés JJ, San-Miguel J, Paiva B. Biological Characterization and Clinical Relevance of Circulating Tumor Cells: Opening the Pandora's Box of Multiple Myeloma. Cancers (Basel) 2022; 14:cancers14061430. [PMID: 35326579 PMCID: PMC8946760 DOI: 10.3390/cancers14061430] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 02/28/2022] [Accepted: 03/09/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Bone marrow (BM) aspirates are mandatory for diagnosis and follow-up of patients with multiple myeloma (MM). However, they present two important caveats: Their invasiveness and limited scope to capture the broad tumor heterogeneity. Conversely, circulating tumor cells (CTCs) are detectable in the peripheral blood of patients with precursor and malignant disease states and have strong prognostic value. Moreover, the high genetic and transcriptomic overlap between both plasma cell compartments suggests that CTCs might reflect with notable precision the medullar clone. Furthermore, the study of CTCs could be used as a model to identify mechanisms favoring BM egression and disease spreading. Here, we summarize the state of the art on MM CTCs and provide insights on what they may offer in research and clinical scenarios. Abstract Bone marrow (BM) aspirates are the gold standard for patient prognostication and genetic characterization in multiple myeloma (MM). However, they represent an important limitation for periodic disease monitoring because they entail an aggressive procedure. Moreover, recent findings show that a single BM aspirate is unable to reflect the complex MM heterogeneity. Recent advances in flow cytometry, microfluidics, and “omics” technologies have opened Pandora’s box of MM: The detection and isolation of circulating tumor cells (CTCs) offer a promising and minimally invasive alternative for tumor assessment and metastasis study. CTCs are detectable in premalignant and active MM states, and their enumeration has strong prognostic value, to the extent that it is challenging current stratification systems. In addition, CTCs reflect with high precision both intra- and extra-medullary disease at the phenotypic, genomic, and transcriptomic levels. Despite this high resemblance between tumor clones in distinct locations, some subtle (not random) differences might shed some light on the metastatic process. Thus, it has been suggested that a hypoxic and pro-inflammatory microenvironment could induce an arrest in proliferation forcing tumor cells to recirculate. Herein, we summarize data on the characterization of MM CTCs as well as their clinical and research potential.
Collapse
|
28
|
Alonso R, Lahuerta JJ. Tumor Reduction in Multiple Myeloma: New Concepts for New Therapeutics. Front Oncol 2022; 11:800309. [PMID: 35096603 PMCID: PMC8794792 DOI: 10.3389/fonc.2021.800309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/23/2021] [Indexed: 12/22/2022] Open
Abstract
The development of new resources for a more accurate diagnosis and response assessment in multiple myeloma has been a long process for decades, mainly since the middle of the 20th century. During this time, the succession of technical advances has run parallel to the better knowledge of disease biology and the availability of novel therapeutic strategies. The cornerstone of standardized criteria to uniformly evaluate the disease response in myeloma dates back to the 1990s when the key role of complete remission was established. Since then, different updates have been implemented according to available scientific evidences not always without certain controversies. The progressive improvements in survival results of myeloma patients and the growing quality of responses due to the novel therapies have led to the need of developing new tools for better monitoring of tumor burden. In this way, the concept of minimal residual disease and its key value based on the prognostic significance and the clinical relevance has been consolidated during the last years, overcoming the value of conventional response criteria or classical adverse prognosis markers. Nevertheless, its precise role in the clinical management of myeloma patients to detect early treatment failure and trigger early rescue strategies is still pending to be defined. In this review, we revisit the major milestones in the understanding of tumor reduction in multiple myeloma until the most recent imaging techniques or liquid biopsy approaches, including a critical view of conventional response criteria, whose backbone has remained unchanged during the last 20 years.
Collapse
Affiliation(s)
- Rafael Alonso
- Hematology Department, Hospital Universitario 12 de Octubre, CIBERONC CB16/12/00369, Madrid, Spain
| | - Juan José Lahuerta
- Instituto de Investigación Sanitaria, Hospital Universitario 12 de Octubre (imas12) CIBERONC CB16/12/00369, Madrid, Spain
| |
Collapse
|
29
|
Mack EKM, Hartmann S, Ross P, Wollmer E, Mann C, Neubauer A, Brendel C, Hoffmann J. Monitoring multiple myeloma in the peripheral blood based on cell-free DNA and circulating plasma cells. Ann Hematol 2022; 101:811-824. [PMID: 35106639 PMCID: PMC8913458 DOI: 10.1007/s00277-022-04771-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/16/2022] [Indexed: 11/25/2022]
Abstract
With the advent of novel, highly effective therapies for multiple myeloma (MM), classical serologic monitoring appears insufficient for response assessment and prediction of relapse. Moreover, serologic studies in MM are hampered by interference of therapeutic antibodies. The detection of malignant plasma cell clones by next generation sequencing (NGS) or multiparameter flow cytometry (MFC) circumvents these difficulties and can be performed in the peripheral blood (pB) by targeting circulating cell-free DNA (cfDNA) or circulating plasma cells (CPCs), thus also avoiding an invasive sampling procedure. Here, we applied NGS of VJ light chain (LC) rearrangements in cfDNA and MFC of magnetically-enriched CD138-positive CPCs (me-MFC) to investigate disease burden in unselected MM patients. Sequencing was successful for 114/130 (87.7%) cfDNA samples and me-MFC results were analyzable for 196/205 (95.6%) samples. MM clones were detectable in 38.9% of samples taken at initial diagnosis or relapse (ID/RD), but only in 11.8% of samples taken during complete remission (CR). Circulating MM plasma cells were present in 83.3% of ID/RD samples and 9.9% of CR samples. Residual disease assessment by NGS or me-MFC in samples taken during very good partial remission or CR was 80% concordant. Notably, 4/4 (NGS) and 5/8 (me-MFC) positive CR samples were from patients with oligo- or non-secretory myeloma. The time to progression was shorter if there was evidence of residual myeloma in the pB. Together, our findings indicate that our two novel analytical approaches accurately indicate the course of MM and may be particularly valuable for monitoring patients with serologically non-trackable disease.
Collapse
Affiliation(s)
- Elisabeth K M Mack
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany.
| | - Sören Hartmann
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Petra Ross
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Ellen Wollmer
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Christoph Mann
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Andreas Neubauer
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Cornelia Brendel
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany
| | - Jörg Hoffmann
- Department of Hematology, Oncology and Immunology, Philipps-University Marburg and University Hospital Gießen and Marburg, 35032, Baldingerstraße, Marburg, Germany.
| |
Collapse
|
30
|
Genomic characterization of functional high-risk multiple myeloma patients. Blood Cancer J 2022; 12:24. [PMID: 35102139 PMCID: PMC8803925 DOI: 10.1038/s41408-021-00576-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 12/18/2022] Open
Abstract
Multiple myeloma (MM) patients with suboptimal response to induction therapy or early relapse, classified as the functional high-risk (FHR) patients, have been shown to have poor outcomes. We evaluated newly-diagnosed MM patients in the CoMMpass dataset and divided them into three groups: genomic high-risk (GHR) group for patients with t(4;14) or t(14;16) or complete loss of functional TP53 (bi-allelic deletion of TP53 or mono-allelic deletion of 17p13 (del17p13) and TP53 mutation) or 1q21 gain and International Staging System (ISS) stage 3; FHR group for patients who had no markers of GHR group but were refractory to induction therapy or had early relapse within 12 months; and standard-risk (SR) group for patients who did not fulfill any of the criteria for GHR or FHR. FHR patients had the worst survival. FHR patients are characterized by increased mutations affecting the IL-6/JAK/STAT3 pathway, and a gene expression profile associated with aberrant mitosis and DNA damage response. This is also corroborated by the association with the mutational signature associated with abnormal DNA damage response. We have also developed a machine learning based classifier that can identify most of these patients at diagnosis.
Collapse
|
31
|
Single-cell profiling of tumour evolution in multiple myeloma - opportunities for precision medicine. Nat Rev Clin Oncol 2022; 19:223-236. [PMID: 35017721 DOI: 10.1038/s41571-021-00593-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2021] [Indexed: 11/08/2022]
Abstract
Multiple myeloma (MM) is a haematological malignancy of plasma cells characterized by substantial intraclonal genetic heterogeneity. Although therapeutic advances made in the past few years have led to improved outcomes and longer survival, MM remains largely incurable. Over the past decade, genomic analyses of patient samples have demonstrated that MM is not a single disease but rather a spectrum of haematological entities that all share similar clinical symptoms. Moreover, analyses of samples from monoclonal gammopathy of undetermined significance and smouldering MM have also shown the existence of genetic heterogeneity in precursor stages, in some cases remarkably similar to that of MM. This heterogeneity highlights the need for a greater dissection of underlying disease biology, especially the clonal diversity and molecular events underpinning MM at each stage to enable the stratification of individuals with a high risk of progression. Emerging single-cell sequencing technologies present a superlative solution to delineate the complexity of monoclonal gammopathy of undetermined significance, smouldering MM and MM. In this Review, we discuss how genomics has revealed novel insights into clonal evolution patterns of MM and provide examples from single-cell studies that are beginning to unravel the mutational and phenotypic characteristics of individual cells within the bone marrow tumour, immune microenvironment and peripheral blood. We also address future perspectives on clinical application, proposing that multi-omics single-cell profiling can guide early patient diagnosis, risk stratification and treatment strategies.
Collapse
|
32
|
Russo GI, Musso N, Romano A, Caruso G, Petralia S, Lanzanò L, Broggi G, Camarda M. The Role of Dielectrophoresis for Cancer Diagnosis and Prognosis. Cancers (Basel) 2021; 14:198. [PMID: 35008359 PMCID: PMC8750463 DOI: 10.3390/cancers14010198] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/27/2021] [Accepted: 12/30/2021] [Indexed: 12/17/2022] Open
Abstract
Liquid biopsy is emerging as a potential diagnostic tool for prostate cancer (PC) prognosis and diagnosis. Unfortunately, most circulating tumor cells (CTC) technologies, such as AdnaTest or Cellsearch®, critically rely on the epithelial cell adhesion molecule (EpCAM) marker, limiting the possibility of detecting cancer stem-like cells (CSCs) and mesenchymal-like cells (EMT-CTCs) that are present during PC progression. In this context, dielectrophoresis (DEP) is an epCAM independent, label-free enrichment system that separates rare cells simply on the basis of their specific electrical properties. As compared to other technologies, DEP may represent a superior technique in terms of running costs, cell yield and specificity. However, because of its higher complexity, it still requires further technical as well as clinical development. DEP can be improved by the use of microfluid, nanostructured materials and fluoro-imaging to increase its potential applications. In the context of cancer, the usefulness of DEP lies in its capacity to detect CTCs in the bloodstream in their epithelial, mesenchymal, or epithelial-mesenchymal phenotype forms, which should be taken into account when choosing CTC enrichment and analysis methods for PC prognosis and diagnosis.
Collapse
Affiliation(s)
| | - Nicolò Musso
- Department of Biomedical and Biotechnological Science (BIOMETEC), University of Catania, 95123 Catania, Italy
- STLab s.r.l., Via Anapo 53, 95126 Catania, Italy;
| | - Alessandra Romano
- Haematological Section, University of Catania, 95125 Catania, Italy;
| | - Giuseppe Caruso
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (G.C.); (S.P.)
| | - Salvatore Petralia
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (G.C.); (S.P.)
| | - Luca Lanzanò
- Department of Physics and Astronomy “Ettore Majorana”, University of Catania, 95123 Catania, Italy;
| | - Giuseppe Broggi
- Pathology Section, Department of Medical, Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, University of Catania, 95123 Catania, Italy;
| | | |
Collapse
|
33
|
Primary plasma cell leukemia: consensus definition by the International Myeloma Working Group according to peripheral blood plasma cell percentage. Blood Cancer J 2021; 11:192. [PMID: 34857730 PMCID: PMC8640034 DOI: 10.1038/s41408-021-00587-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 11/08/2022] Open
Abstract
Primary plasma cell leukemia (PCL) has a consistently ominous prognosis, even after progress in the last decades. PCL deserves a prompt identification to start the most effective treatment for this ultra-high-risk disease. The aim of this position paper is to revisit the diagnosis of PCL according to the presence of circulating plasma cells in patients otherwise meeting diagnostic criteria of multiple myeloma. We could identify two retrospective series where the question about what number of circulating plasma cells in peripheral blood should be used for defining PCL. The presence of ≥5% circulating plasma cells in patients with MM had a similar adverse prognostic impact as the previously defined PCL. Therefore, PCL should be defined by the presence of 5% or more circulating plasma cells in peripheral blood smears in patients otherwise diagnosed with symptomatic multiple myeloma.
Collapse
|
34
|
Fouquet G, Wartski M, Dechmi A, Willems L, Deau-Fischer B, Franchi P, Descroocq J, Deschamps P, Blanc-Autran E, Clerc J, Bouscary D, Barreau S, Chapuis N, Vignon M, Cottereau AS. Prognostic Value of FDG-PET/CT Parameters in Patients with Relapse/Refractory Multiple Myeloma before Anti-CD38 Based Therapy. Cancers (Basel) 2021; 13:4323. [PMID: 34503133 PMCID: PMC8431719 DOI: 10.3390/cancers13174323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 11/16/2022] Open
Abstract
Although anti-CD38 monoclonal antibodies have improved the prognosis of relapsed/refractory multiple myeloma (RRMM), some patients still experience early relapses with poor outcomes. This present study evaluated the predictive value of FDG PET/CT parameters for RRMM prior to initiating anti-CD38 treatment. We included 38 consecutive RRMM patients who underwent a PET/CT scan treated at our institution at relapse. The median PFS was 12.5 months and the median OS was not reached. 42% of the patients had an initial ISS score of 1, 37% of 2, and 21% of 3. The presence of >3 focal lesions (FLs, n = 19) and the ISS score were associated with inferior PFS (p = 0.0036 and p = 0.0026) and OS (p = 0.025 and p = 0.0098). Patients with >3 FLs had a higher initial ISS score (p = 0.028). In multivariable analysis, the ISS score and >3 FLs were independent prognostic factors for PFS (p = 0.010 and p = 0.025 respectively), and combined they individualized a high-risk group with a median PFS and OS of 3.1 months and 8.5 months respectively vs. not reached for the other patients. The presence of >3 FLs on PET was predictive of survival outcomes in patients with RRMM treated using CD38 targeted therapy. Combined with the initial ISS, an ultra-high-risk RRMM population can thus be identified.
Collapse
Affiliation(s)
- Guillemette Fouquet
- Assistance Publique-Hôpitaux de Paris Centre, Université de Paris, Service d’Hématologie Clinique, Hôpital Cochin, 75014 Paris, France; (G.F.); (L.W.); (B.D.-F.); (P.F.); (J.D.); (P.D.); (D.B.); (M.V.)
| | - Myriam Wartski
- Assistance Publique-Hôpitaux de Paris Centre, Université de Paris, Service de Médecine Nucléaire, Hôpital Cochin, 75014 Paris, France; (M.W.); (A.D.); (J.C.)
| | - Amina Dechmi
- Assistance Publique-Hôpitaux de Paris Centre, Université de Paris, Service de Médecine Nucléaire, Hôpital Cochin, 75014 Paris, France; (M.W.); (A.D.); (J.C.)
| | - Lise Willems
- Assistance Publique-Hôpitaux de Paris Centre, Université de Paris, Service d’Hématologie Clinique, Hôpital Cochin, 75014 Paris, France; (G.F.); (L.W.); (B.D.-F.); (P.F.); (J.D.); (P.D.); (D.B.); (M.V.)
| | - Bénédicte Deau-Fischer
- Assistance Publique-Hôpitaux de Paris Centre, Université de Paris, Service d’Hématologie Clinique, Hôpital Cochin, 75014 Paris, France; (G.F.); (L.W.); (B.D.-F.); (P.F.); (J.D.); (P.D.); (D.B.); (M.V.)
| | - Patricia Franchi
- Assistance Publique-Hôpitaux de Paris Centre, Université de Paris, Service d’Hématologie Clinique, Hôpital Cochin, 75014 Paris, France; (G.F.); (L.W.); (B.D.-F.); (P.F.); (J.D.); (P.D.); (D.B.); (M.V.)
| | - Justine Descroocq
- Assistance Publique-Hôpitaux de Paris Centre, Université de Paris, Service d’Hématologie Clinique, Hôpital Cochin, 75014 Paris, France; (G.F.); (L.W.); (B.D.-F.); (P.F.); (J.D.); (P.D.); (D.B.); (M.V.)
| | - Paul Deschamps
- Assistance Publique-Hôpitaux de Paris Centre, Université de Paris, Service d’Hématologie Clinique, Hôpital Cochin, 75014 Paris, France; (G.F.); (L.W.); (B.D.-F.); (P.F.); (J.D.); (P.D.); (D.B.); (M.V.)
| | - Estelle Blanc-Autran
- Hôpital Marie Lannelongue, Service de Médecine Nucléaire, 92350 Le Plessis-Robinson, France;
| | - Jérôme Clerc
- Assistance Publique-Hôpitaux de Paris Centre, Université de Paris, Service de Médecine Nucléaire, Hôpital Cochin, 75014 Paris, France; (M.W.); (A.D.); (J.C.)
| | - Didier Bouscary
- Assistance Publique-Hôpitaux de Paris Centre, Université de Paris, Service d’Hématologie Clinique, Hôpital Cochin, 75014 Paris, France; (G.F.); (L.W.); (B.D.-F.); (P.F.); (J.D.); (P.D.); (D.B.); (M.V.)
| | - Sylvain Barreau
- Assistance Publique-Hôpitaux de Paris Centre, Université de Paris, Service D’hémato-Biologie, Hôpital Cochin, 75014 Paris, France; (S.B.); (N.C.)
| | - Nicolas Chapuis
- Assistance Publique-Hôpitaux de Paris Centre, Université de Paris, Service D’hémato-Biologie, Hôpital Cochin, 75014 Paris, France; (S.B.); (N.C.)
| | - Marguerite Vignon
- Assistance Publique-Hôpitaux de Paris Centre, Université de Paris, Service d’Hématologie Clinique, Hôpital Cochin, 75014 Paris, France; (G.F.); (L.W.); (B.D.-F.); (P.F.); (J.D.); (P.D.); (D.B.); (M.V.)
| | - Anne-Ségolène Cottereau
- Assistance Publique-Hôpitaux de Paris Centre, Université de Paris, Service de Médecine Nucléaire, Hôpital Cochin, 75014 Paris, France; (M.W.); (A.D.); (J.C.)
| |
Collapse
|
35
|
PIM Kinases in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13174304. [PMID: 34503111 PMCID: PMC8428354 DOI: 10.3390/cancers13174304] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) remains an incurable disease and novel therapeutic agents/approaches are urgently needed. The PIM (Proviral insertion in murine malignancies) serine/threonine kinases have 3 isoforms: PIM1, PIM2, and PIM3. PIM kinases are engaged with an expansive scope of biological activities including cell growth, apoptosis, drug resistance, and immune response. An assortment of molecules and pathways that are critical to myeloma tumorigenesis has been recognized as the downstream targets of PIM kinases. The inhibition of PIM kinases has become an emerging scientific interest for the treatment of multiple myeloma and several PIM kinase inhibitors, such as SGI-1776, AZD1208, and PIM447 (formerly LGH447), have been developed and are under different phases of clinical trials. Current research has been focused on the development of a new generation of potent PIM kinase inhibitors with appropriate pharmacological profiles reasonable for human malignancy treatment. Combination therapy of PIM kinase inhibitors with chemotherapeutic appears to create an additive cytotoxic impact in cancer cells. Notwithstanding, the mechanisms by which PIM kinases modulate the immune microenvironment and synergize with the immunomodulatory agents such as lenalidomide have not been deliberately depicted. This review provides a comprehensive overview of the PIM kinase pathways and the current research status of the development of PIM kinase inhibitors for the treatment of MM. Additionally, the combinatorial effects of the PIM kinase inhibitors with other targeted agents and the promising strategies to exploit PIM as a therapeutic target in malignancy are highlighted.
Collapse
|
36
|
Han W, Jin Y, Xu M, Zhao SS, Shi Q, Qu X, Zhang R, Li J, Wu Y, Chen L. Prognostic value of circulating clonal plasma cells in newly diagnosed multiple myeloma. ACTA ACUST UNITED AC 2021; 26:510-517. [PMID: 34314294 DOI: 10.1080/16078454.2021.1948208] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVES Multiple myeloma (MM) involves a clinically and biologically heterogeneous malignancy of plasma cells. It is difficult to predict the prognosis of MM. The presence of circulating clonal plasma cells (CPC) has been associated with a worse prognosis in patients with MM. METHODS This study retrospectively analysed CPC in 108 newly diagnosed MM patients by 8-colour flow cytometry to investigate their value for predicting the outcome and combined the level of CPC with the revised International Staging System (R-ISS) to stratify the MM patients according to risk. RESULTS CPC were detected in 58/108 patients (53.7%). The optimum cut-off for the prediction of overall survival was determined to be 0.105%. Patients with higher R-ISS stages seemed to harbour more CPC. A level of CPC≥0.105% was an independent risk factor for adverse outcomes (P<0.001). The combination of the R-ISS staging system and level of CPC was used to stratify MM patients according to risk, and the combination of R-ISS stage III and a level of CPC≥0.105% defined the ultra-high-risk group. CONCLUSION This study suggests that a high proportion of CPC is associated with aggressive disease and that the use of the current R-ISS system in conjunction with assessment of the level of CPC may facilitate the stratification of newly diagnosed MM patients into clinically relevant prognostic subgroups.
Collapse
Affiliation(s)
- Wenmin Han
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, People's Republic of China.,Department of Hematology, Changzhou Second Affiliated Hospital of Nanjing Medical University, Changzhou, People's Republic of China
| | - Yuanyuan Jin
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, People's Republic of China
| | - Min Xu
- Department of Hematology, Zhangjiagang First Affiliated Hospital of Soochow University, Zhangjiagang, People's Republic of China
| | - Si-Shu Zhao
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, People's Republic of China
| | - Qinglin Shi
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, People's Republic of China
| | - Xiaoyan Qu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, People's Republic of China
| | - Run Zhang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, People's Republic of China
| | - Jianyong Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, People's Republic of China
| | - Yujie Wu
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, People's Republic of China
| | - Lijuan Chen
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, People's Republic of China
| |
Collapse
|
37
|
O'Brien A, O'Halloran F, Mykytiv V. Minimal Residual Disease in Multiple Myeloma: Potential for Blood-Based Methods to Monitor Disease. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 22:e34-e40. [PMID: 34470720 DOI: 10.1016/j.clml.2021.07.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/25/2021] [Accepted: 07/29/2021] [Indexed: 10/20/2022]
Abstract
In recent years, the life expectancy of Multiple Myeloma (MM) patients has substantially improved, but this cancer remains incurable with increasing incidence in the developed world. Most MM patients will eventually relapse due to residual drug-resistant cancerous cells that survive treatment, commonly referred to as minimal residual disease (MRD). Methods to improve MRD detection in MM patients are generating considerable interest as a means of monitoring patients' response to treatment. In clinical laboratories, these methods currently require bone marrow aspirates which are invasive and frequently miss detection of localised disease due to the spatial heterogeneity of disease infiltration. By simplifying serial sampling and allowing for the detection of extramedullary disease, a blood-based method could significantly impact treatment duration and intensity and minimise chemotherapy-induced toxicity. This review will describe the current blood-based techniques available to detect MRD in MM and compare their potential to evaluate patient prognosis and drive therapeutic decisions.
Collapse
Affiliation(s)
- Aisling O'Brien
- Dept. of Biological Sciences, Munster Technological University, Cork, Ireland; Dept. of Immunology, Cork University Hospital, Cork, Ireland.
| | - Fiona O'Halloran
- Dept. of Biological Sciences, Munster Technological University, Cork, Ireland
| | - Vitaliy Mykytiv
- Dept. of Haematology, Cork University Hospital, Cork, Ireland; Dept. of Medicine, University College Cork, Cork, Ireland
| |
Collapse
|
38
|
Ebert LM, Vandyke K, Johan MZ, DeNichilo M, Tan LY, Myo Min KK, Weimann BM, Ebert BW, Pitson SM, Zannettino ACW, Wallington-Beddoe CT, Bonder CS. Desmoglein-2 expression is an independent predictor of poor prognosis patients with multiple myeloma. Mol Oncol 2021; 16:1221-1240. [PMID: 34245117 PMCID: PMC8936512 DOI: 10.1002/1878-0261.13055] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/09/2021] [Indexed: 12/20/2022] Open
Abstract
Multiple myeloma (MM) is the second most common haematological malignancy and is an incurable disease of neoplastic plasma cells (PC). Newly diagnosed MM patients currently undergo lengthy genetic testing to match chromosomal mutations with the most potent drug/s to decelerate disease progression. With only 17% of MM patients surviving 10‐years postdiagnosis, faster detection and earlier intervention would unequivocally improve outcomes. Here, we show that the cell surface protein desmoglein‐2 (DSG2) is overexpressed in ~ 20% of bone marrow biopsies from newly diagnosed MM patients. Importantly, DSG2 expression was strongly predictive of poor clinical outcome, with patients expressing DSG2 above the 70th percentile exhibiting an almost 3‐fold increased risk of death. As a prognostic factor, DSG2 is independent of genetic subtype as well as the routinely measured biomarkers of MM activity (e.g. paraprotein). Functional studies revealed a nonredundant role for DSG2 in adhesion of MM PC to endothelial cells. Together, our studies suggest DSG2 to be a potential cell surface biomarker that can be readily detected by flow cytometry to rapidly predict disease trajectory at the time of diagnosis.
Collapse
Affiliation(s)
- Lisa M Ebert
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Kate Vandyke
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia.,Myeloma Research Laboratory, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - M Zahied Johan
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Mark DeNichilo
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Lih Y Tan
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Kay K Myo Min
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Benjamin M Weimann
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia.,College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Brenton W Ebert
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Andrew C W Zannettino
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia.,Myeloma Research Laboratory, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Craig T Wallington-Beddoe
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia.,College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia.,Flinders Medical Centre, Bedford Park, SA, Australia
| | - Claudine S Bonder
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
39
|
Goldman‐Mazur S, Kumar SK. Current approaches to management of high-risk multiple myeloma. Am J Hematol 2021; 96:854-871. [PMID: 33725367 DOI: 10.1002/ajh.26161] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 12/15/2022]
Abstract
The median overall survival in multiple myeloma is rapidly approaching 10 years; however, in nearly a fifth of patients the prognosis remains poor. Therefore, the modern-day management of myeloma patients should be individualized, with a more intense and continuous approach in these high-risk patients. This includes first-line treatment based on multi-drug combinations employing the most effective drug combinations, upfront autologous stem cell transplantation (in eligible patients with tandem transplantation being a consideration), and maintenance based on proteasome inhibitor-based combinations. This paper reviews the results of recent retrospective analyses and clinical trials, but also gives a glance into the future by presenting the ongoing trials.
Collapse
Affiliation(s)
| | - Shaji K. Kumar
- Division of Hematology Mayo Clinic Rochester Minnesota USA
| |
Collapse
|
40
|
Vasco-Mogorrón MA, Campillo JA, Periago A, Cabañas V, Berenguer M, García-Garay MC, Gimeno L, Soto-Ramírez MF, Martínez-Hernández MD, Muro M, Minguela A. Blood-based risk stratification for pre-malignant and symptomatic plasma cell neoplasms to improve patient management. Am J Cancer Res 2021; 11:2736-2753. [PMID: 34249425 PMCID: PMC8263680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 05/21/2021] [Indexed: 06/13/2023] Open
Abstract
Standard risk stratification (sRisk) guides clinical management in monoclonal gammopathy of undetermined significance (MGUS), smoldering multiple myeloma (SMM) and multiple myeloma (MM). Nonetheless, clinical results are considerably heterogeneous among patients with similar risk status. Blood and bone marrow samples from 276 MGUS, 56 SMM and 242 MM in regular clinical practice were analyzed at diagnosis by flow cytometry. Higher levels of aberrant circulating plasma cells (cPC) (> 0.0035% of leukocytes), combined with albumin, beta2-microglobuline and lactate-dehydrogenase levels, offered minimally-invasive risk stratification (RcPC) with results comparable to sRisk. RcPC and sRisk 10-year progression-free-survival (10y-PFS) rates were: 93.8% vs. 95.1% for low-risk, 78.4% vs. 81.7% for intermediate-risk and 50.0% vs. 47.8% for high-risk MGUS; 58.3% vs. 57.8% low-risk, 44.4% vs. 45.8% intermediate-risk and 8.9% vs. 15.0% high-risk SMM; and 44.4% vs. 44.4% low-risk, 36.1% vs. 36.8% intermediate-risk, and 13.3% vs. 16.2% high-risk MM. Circulating-PC > 0.0035% vs. cPC<0.0035% was an independent prognostic factor for PFS (HR=4.389, P=1.2×10-15, Harrell C-statistic =0.7705±0.0190) and over-all survival (OS, HR=4.286, 2.3×10-9, Harrell C-statistic =0.8225±0.0197) that complemented sRisk in patients with low-sRisk (10y-PFS rates 48.1% vs. 87.3%, P=1.2×10-8) and intermediate-sRisk (10y-PFS rates 28.9% vs. 74.1%, P=8.6×10-12). Patients with high cPCs values are associated with higher proliferation and lower apoptosis rates of PC. Circulating-PC > 0.0035% identified MGUS, SMM and MM patients at higher risk of progression or death and predicted a cohort of patients that after relapse from stringent complete response showed shorter OS. These patients could benefit from early consolidation therapy, tandem ASCT or intensive maintenance.
Collapse
Affiliation(s)
- María A Vasco-Mogorrón
- Immunology Service, Clinic University Hospital Virgen de la Arrrixaca (HCUVA) and Biomedical Research Institute of Murcia (IMIB)Murcia, Spain
| | - José A Campillo
- Immunology Service, Clinic University Hospital Virgen de la Arrrixaca (HCUVA) and Biomedical Research Institute of Murcia (IMIB)Murcia, Spain
| | - Adela Periago
- Hematology Service, General University Hospital Rafael Méndez, Lorca, and Biomedical Research Institute of Murcia (IMIB)Murcia, Spain
| | - Valentin Cabañas
- Hematology Service, Clinic University Hospital Virgen de la Arrrixaca (HCUVA) and Biomedical Research Institute of Murcia (IMIB)Murcia, Spain
| | - Mercedes Berenguer
- Hematology Service, General University Hospital Santa Lucía, Cartagena, and Biomedical Research Institute of Murcia (IMIB)Murcia, Spain
| | - María C García-Garay
- Hematology Service, Clinic University Hospital Virgen de la Arrrixaca (HCUVA) and Biomedical Research Institute of Murcia (IMIB)Murcia, Spain
| | - Lourdes Gimeno
- Immunology Service, Clinic University Hospital Virgen de la Arrrixaca (HCUVA) and Biomedical Research Institute of Murcia (IMIB)Murcia, Spain
- Human Anatomy Department, Medicine Faculty, Murcia UniversityMurcia, Spain
| | - María F Soto-Ramírez
- Immunology Service, Clinic University Hospital Virgen de la Arrrixaca (HCUVA) and Biomedical Research Institute of Murcia (IMIB)Murcia, Spain
| | - María D Martínez-Hernández
- Immunology Service, Clinic University Hospital Virgen de la Arrrixaca (HCUVA) and Biomedical Research Institute of Murcia (IMIB)Murcia, Spain
| | - Manuel Muro
- Immunology Service, Clinic University Hospital Virgen de la Arrrixaca (HCUVA) and Biomedical Research Institute of Murcia (IMIB)Murcia, Spain
| | - Alfredo Minguela
- Immunology Service, Clinic University Hospital Virgen de la Arrrixaca (HCUVA) and Biomedical Research Institute of Murcia (IMIB)Murcia, Spain
| |
Collapse
|
41
|
Maiso P, Mogollón P, Ocio EM, Garayoa M. Bone Marrow Mesenchymal Stromal Cells in Multiple Myeloma: Their Role as Active Contributors to Myeloma Progression. Cancers (Basel) 2021; 13:2542. [PMID: 34067236 PMCID: PMC8196907 DOI: 10.3390/cancers13112542] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/16/2021] [Accepted: 05/19/2021] [Indexed: 01/01/2023] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy of plasma cells that proliferate and accumulate within the bone marrow (BM). Work from many groups has made evident that the complex microenvironment of the BM plays a crucial role in myeloma progression and response to therapeutic agents. Within the cellular components of the BM, we will specifically focus on mesenchymal stromal cells (MSCs), which are known to interact with myeloma cells and the other components of the BM through cell to cell, soluble factors and, as more recently evidenced, through extracellular vesicles. Multiple structural and functional abnormalities have been found when characterizing MSCs derived from myeloma patients (MM-MSCs) and comparing them to those from healthy donors (HD-MSCs). Other studies have identified differences in genomic, mRNA, microRNA, histone modification, and DNA methylation profiles. We discuss these distinctive features shaping MM-MSCs and propose a model for the transition from HD-MSCs to MM-MSCs as a consequence of the interaction with myeloma cells. Finally, we review the contribution of MM-MSCs to several aspects of myeloma pathology, specifically to myeloma growth and survival, drug resistance, dissemination and homing, myeloma bone disease, and the induction of a pro-inflammatory and immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Patricia Maiso
- University Hospital Marqués de Valdecilla (IDIVAL), University of Cantabria, 39008 Santander, Spain
| | - Pedro Mogollón
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (P.M.); (M.G.)
| | - Enrique M. Ocio
- University Hospital Marqués de Valdecilla (IDIVAL), University of Cantabria, 39008 Santander, Spain
| | - Mercedes Garayoa
- Cancer Research Center (IBMCC-CSIC-USAL), University Hospital of Salamanca (IBSAL), 37007 Salamanca, Spain; (P.M.); (M.G.)
| |
Collapse
|
42
|
Galieni P, Travaglini F, Vagnoni D, Ruggieri M, Caraffa P, Bigazzi C, Falcioni S, Picardi P, Mazzotta S, Troiani E, Dalsass A, Mestichelli F, Angelini M, Camaioni E, Maravalle D, Angelini S, Pezzoni V. The detection of circulating plasma cells may improve the Revised International Staging System (R-ISS) risk stratification of patients with newly diagnosed multiple myeloma. Br J Haematol 2021; 193:542-550. [PMID: 33792026 DOI: 10.1111/bjh.17118] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/02/2020] [Indexed: 11/25/2022]
Abstract
The Revised International Staging System (R-ISS) stratifies patients affected by Multiple Myeloma (MM) into three distinct risk groups: R-ISS I [ISS Stage I, Standard-Risk cytogenetics and normal Lactase DeHydrogenase (LDH)], R-ISS III (ISS stage III and either high-risk cytogenetics or high LDH) and R-ISS II (any other characteristics). With the aim to verify whether the three R-ISS groups could be divided into subgroups with different prognostic factors based on the detection of Circulating Plasma Cells (CPCs) at diagnosis, in this retrospective analysis, we evaluated 161 patients with MM treated at our centre between 2005 and 2017. In all, 57 patients (33·9%) were staged as R-ISS III, 98 (58·3%) as R-ISS II and six (3·6%) as R-ISS I. CPCs were detected in 125 patients (74·4%), while in 43 patients (25·6%) no CPCs were seen. Our analysis revealed that Overall Survival (OS) and progression-free survival (PFS) rates in R-ISS II patients were higher in the subgroup without CPCs compared to the subgroup with ≥1 CPCs (OS: 44·7% vs. 16·3%, P = 0·0089; PFS: 27·8% vs. 8·1%, P = 0·0118). Our present findings suggest that the detection of CPCs at diagnosis may be used as a further prognostic biomarker to improve the risk stratification of patients with MM staged as R-ISS II.
Collapse
Affiliation(s)
- Piero Galieni
- Department of Haematology and Stem Cell Transplantation Unit, C. e G. Mazzoni Hospital, Ascoli Piceno, Italy
| | - Fosco Travaglini
- Department of Haematology and Stem Cell Transplantation Unit, C. e G. Mazzoni Hospital, Ascoli Piceno, Italy
| | - Davide Vagnoni
- UOC Clinical Pathology, ASUR Marche, Civitanova Marche Hospital, Costamartina, Italy
| | - Miriana Ruggieri
- Department of Haematology and Stem Cell Transplantation Unit, C. e G. Mazzoni Hospital, Ascoli Piceno, Italy
| | - Patrizia Caraffa
- Department of Haematology and Stem Cell Transplantation Unit, C. e G. Mazzoni Hospital, Ascoli Piceno, Italy
| | - Catia Bigazzi
- Department of Haematology and Stem Cell Transplantation Unit, C. e G. Mazzoni Hospital, Ascoli Piceno, Italy
| | - Sadia Falcioni
- Department of Haematology and Stem Cell Transplantation Unit, C. e G. Mazzoni Hospital, Ascoli Piceno, Italy
| | - Paola Picardi
- Department of Haematology and Stem Cell Transplantation Unit, C. e G. Mazzoni Hospital, Ascoli Piceno, Italy
| | - Serena Mazzotta
- Department of Haematology and Stem Cell Transplantation Unit, C. e G. Mazzoni Hospital, Ascoli Piceno, Italy
| | - Emanuela Troiani
- Department of Haematology and Stem Cell Transplantation Unit, C. e G. Mazzoni Hospital, Ascoli Piceno, Italy
| | - Alessia Dalsass
- Department of Haematology and Stem Cell Transplantation Unit, C. e G. Mazzoni Hospital, Ascoli Piceno, Italy
| | - Francesca Mestichelli
- Department of Haematology and Stem Cell Transplantation Unit, C. e G. Mazzoni Hospital, Ascoli Piceno, Italy
| | - Mario Angelini
- Department of Haematology and Stem Cell Transplantation Unit, C. e G. Mazzoni Hospital, Ascoli Piceno, Italy
| | - Elisa Camaioni
- Department of Haematology and Stem Cell Transplantation Unit, C. e G. Mazzoni Hospital, Ascoli Piceno, Italy
| | - Denise Maravalle
- Department of Haematology and Stem Cell Transplantation Unit, C. e G. Mazzoni Hospital, Ascoli Piceno, Italy
| | - Stefano Angelini
- Department of Haematology and Stem Cell Transplantation Unit, C. e G. Mazzoni Hospital, Ascoli Piceno, Italy
| | - Valerio Pezzoni
- Department of Haematology and Stem Cell Transplantation Unit, C. e G. Mazzoni Hospital, Ascoli Piceno, Italy
| |
Collapse
|
43
|
Immunological Prognostic Factors in Multiple Myeloma. Int J Mol Sci 2021; 22:ijms22073587. [PMID: 33808304 PMCID: PMC8036885 DOI: 10.3390/ijms22073587] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/25/2021] [Accepted: 03/29/2021] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) is a plasma cell neoplasm characterized by an abnormal proliferation of clonal, terminally differentiated B lymphocytes. Current approaches for the treatment of MM focus on developing new diagnostic techniques; however, the search for prognostic markers is also crucial. This enables the classification of patients into risk groups and, thus, the selection of the most optimal treatment method. Particular attention should be paid to the possible use of immune factors, as the immune system plays a key role in the formation and course of MM. In this review, we focus on characterizing the components of the immune system that are of prognostic value in MM patients, in order to facilitate the development of new diagnostic and therapeutic directions.
Collapse
|
44
|
Cheng Q, Cai L, Zhang Y, Chen L, Hu Y, Sun C. Circulating Plasma Cells as a Biomarker to Predict Newly Diagnosed Multiple Myeloma Prognosis: Developing Nomogram Prognostic Models. Front Oncol 2021; 11:639528. [PMID: 33747963 PMCID: PMC7973368 DOI: 10.3389/fonc.2021.639528] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/10/2021] [Indexed: 12/11/2022] Open
Abstract
Background: To investigate the prognostic value of circulating plasma cells (CPC) and establish novel nomograms to predict individual progression-free survival (PFS) as well as overall survival (OS) of patients with newly diagnosed multiple myeloma (NDMM). Methods: One hundred ninetyone NDMM patients in Wuhan Union Hospital from 2017.10 to 2020.8 were included in the study. The entire cohort was randomly divided into a training (n = 130) and a validation cohort (n = 61). Univariate and multivariate analyses were performed on the training cohort to establish nomograms for the prediction of survival outcomes, and the nomograms were validated by calibration curves. Results: When the cut-off value was 0.038%, CPC could well distinguish patients with higher tumor burden and lower response rates (P < 0.05), and could be used as an independent predictor of PFS and OS. Nomograms predicting PFS and OS were developed according to CPC, lactate dehydrogenase (LDH) and creatinine. The C-index and the area under receiver operating characteristic curves (AUC) of the nomograms showed excellent individually predictive effects in training cohort, validation cohort or entire cohort. Patients with total points of the nomograms ≤ 60.7 for PFS and 75.8 for OS could be defined as low-risk group and the remaining as high-risk group. The 2-year PFS and OS rates of patients in low-risk group was significantly higher than those in high-risk group (p < 0.001). Conclusions: CPC is an independent prognostic factor for NDMM patients. The proposed nomograms could provide individualized PFS and OS prediction and risk stratification.
Collapse
Affiliation(s)
- Qianwen Cheng
- Institute of Hematology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Cai
- Institute of Hematology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuyang Zhang
- Institute of Hematology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Chen
- Institute of Hematology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| | - Chunyan Sun
- Institute of Hematology, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
45
|
Maclachlan KH, Came N, Diamond B, Roshal M, Ho C, Thoren K, Mayerhoefer ME, Landgren O, Harrison S. Minimal residual disease in multiple myeloma: defining the role of next generation sequencing and flow cytometry in routine diagnostic use. Pathology 2021; 53:385-399. [PMID: 33674146 DOI: 10.1016/j.pathol.2021.02.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022]
Abstract
For patients diagnosed with multiple myeloma (MM) there have been significant treatment advances over the past decade, reflected in an increasing proportion of patients achieving durable remissions. Clinical trials repeatedly demonstrate that achieving a deep response to therapy, with a bone marrow assessment proving negative for minimal residual disease (MRD), confers a significant survival advantage. To accurately assess for minute quantities of residual cancer requires highly sensitive methods; either multiparameter flow cytometry or next generation sequencing are currently recommended for MM response assessment. Under optimal conditions, these methods can detect one aberrant cell amongst 1,000,000 normal cells (a sensitivity of 10-6). Here, we will review the practical use of MRD assays in MM, including challenges in implementation for the routine diagnostic laboratory, standardisation across laboratories and clinical trials, the clinical integration of MRD status assessment into MM management and future directions for ongoing research.
Collapse
Affiliation(s)
- Kylee H Maclachlan
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Haematology Service, Peter MacCallum Cancer Centre, East Melbourne, Vic, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic, Australia.
| | - Neil Came
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic, Australia; Pathology Department, Peter MacCallum Cancer Centre, East Melbourne, Vic, Australia
| | - Benjamin Diamond
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mikhail Roshal
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Caleb Ho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katie Thoren
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marius E Mayerhoefer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ola Landgren
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Myeloma Program, Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Simon Harrison
- Haematology Service, Peter MacCallum Cancer Centre, East Melbourne, Vic, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Vic, Australia
| |
Collapse
|
46
|
Diamond BT, Rustad E, Maclachlan K, Thoren K, Ho C, Roshal M, Ulaner GA, Landgren CO. Defining the undetectable: The current landscape of minimal residual disease assessment in multiple myeloma and goals for future clarity. Blood Rev 2021; 46:100732. [PMID: 32771227 PMCID: PMC9928431 DOI: 10.1016/j.blre.2020.100732] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/05/2020] [Accepted: 07/06/2020] [Indexed: 01/19/2023]
Abstract
Multiple Myeloma, the second most prevalent hematologic malignancy, yet lacks an established curative therapy. However, overall response rate to modern four-drug regimens approaches 100%. Major efforts have thus focused on the measurement of minute quantities of residual disease (minimal residual disease or MRD) for prognostic metrics and therapeutic response evaluation. Currently, MRD is assessed by flow cytometry or by next generation sequencing to track tumor-specific immunoglobulin V(D)J rearrangements. These bone marrow-based methods can reach sensitivity thresholds of the identification of one neoplastic cell in 1,000,000 (10-6). New technologies are being developed to be used alone or in conjunction with established methods, including peripheral blood-based assays, mass spectrometry, and targeted imaging. Data is also building for MRD as a surrogate endpoint for overall survival. Here, we will address the currently utilized MRD assays, challenges in validation across labs and clinical trials, techniques in development, and future directions for successful clinical application of MRD in multiple myeloma.
Collapse
Affiliation(s)
| | | | | | | | - Caleb Ho
- Memorial Sloan Kettering Cancer Center, USA
| | | | | | | |
Collapse
|
47
|
Caro J, Al Hadidi S, Usmani S, Yee AJ, Raje N, Davies FE. How to Treat High-Risk Myeloma at Diagnosis and Relapse. Am Soc Clin Oncol Educ Book 2021; 41:291-309. [PMID: 34010042 DOI: 10.1200/edbk_320105] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Survival in multiple myeloma has improved greatly during the past 2 decades, but this change has primarily benefited patients who have standard-risk disease. Patients with high-risk disease remain a challenge to diagnose and treat. To improve their clinical outcomes, it is imperative to develop tools to readily identify them and to provide them with the most effective available treatments. The most widely used stratification system, the revised International Staging System, incorporates serum β-2 microglobulin, albumin, lactate dehydrogenase, and high-risk chromosomal abnormalities [del(17p), t(4;14), and t(14;16)]. Recent updates have included mutational status and chromosome 1q abnormalities. Plasma cell leukemia, extramedullary disease, circulating plasma cells, renal failure, and frailty are also associated with poor outcome. The treatment approach for a newly diagnosed patient with high-risk multiple myeloma should include induction therapy, autologous stem cell transplantation if appropriate, and maintenance therapy. Triplet therapy with a proteasome inhibitor, immunomodulatory drug, and steroid, with or without an anti-CD38 antibody, should be considered for induction, along with a proteasome inhibitor and/or immunomodulatory drug for maintenance. Aiming for a deep and sustained response is important. Similar principles apply at relapse, with close monitoring of response, especially extramedullary disease and active management of side effects, so that patients can continue therapy and benefit from treatment. Immune-based therapies, including autologous CAR T-cell-based therapies and bispecific antibodies, show promising activity in relapsed disease and are being actively explored in earlier disease settings. As the prognosis for high-risk disease remains poor, the future goal for this patient group is to develop specific clinical trials to explore novel approaches and therapies efficiently.
Collapse
Affiliation(s)
- Jessica Caro
- Perlmutter Cancer Center, NYU Langone Health, New York, NY
| | - Samer Al Hadidi
- Department of Hematology and Oncology, Baylor College of Medicine, Houston, TX
| | - Saad Usmani
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute/Atrium Health, Charlotte, NC
| | - Andrew J Yee
- Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA
| | - Noopur Raje
- Center for Multiple Myeloma, Massachusetts General Hospital, Boston, MA
| | - Faith E Davies
- Perlmutter Cancer Center, NYU Langone Health, New York, NY
| |
Collapse
|
48
|
Zeissig MN, Zannettino ACW, Vandyke K. Tumour Dissemination in Multiple Myeloma Disease Progression and Relapse: A Potential Therapeutic Target in High-Risk Myeloma. Cancers (Basel) 2020; 12:cancers12123643. [PMID: 33291672 PMCID: PMC7761917 DOI: 10.3390/cancers12123643] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/02/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Like in solid cancers, the process of dissemination is a critical feature of disease progression in the blood cancer multiple myeloma. At diagnosis, myeloma patients have cancer that has spread throughout the bone marrow, with patients with more disseminatory myeloma having worse outcomes for their disease. In this review, we discuss the current understanding of the mechanisms that underpin the dissemination process in multiple myeloma. Furthermore, we discuss the potential for the use of therapies that target the dissemination process as a novel means of improving outcomes for multiple myeloma patients. Abstract Multiple myeloma (MM) is a plasma cell (PC) malignancy characterised by the presence of MM PCs at multiple sites throughout the bone marrow. Increased numbers of peripheral blood MM PCs are associated with rapid disease progression, shorter time to relapse and are a feature of advanced disease. In this review, the current understanding of the process of MM PC dissemination and the extrinsic and intrinsic factors potentially driving it are addressed through analysis of patient-derived MM PCs and MM cell lines as well as mouse models of homing and dissemination. In addition, we discuss how patient cytogenetic subgroups that present with highly disseminated disease, such as t(4;14), t(14;16) and t(14;20), suggest that intrinsic properties of MM PC influence their ability to disseminate. Finally, we discuss the possibility of using therapeutic targeting of tumour dissemination to slow disease progression and prevent overt relapse.
Collapse
Affiliation(s)
- Mara N. Zeissig
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Australia, Adelaide 5005, Australia; (M.N.Z.); (A.C.W.Z.)
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia
| | - Andrew C. W. Zannettino
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Australia, Adelaide 5005, Australia; (M.N.Z.); (A.C.W.Z.)
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia
- Central Adelaide Local Health Network, Adelaide 5000, Australia
- Centre for Cancer Biology, University of South Australia, Adelaide 5000, Australia
| | - Kate Vandyke
- Myeloma Research Laboratory, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Australia, Adelaide 5005, Australia; (M.N.Z.); (A.C.W.Z.)
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia
- Correspondence: ; Tel.: +61-8-8128-4694
| |
Collapse
|
49
|
Biological properties of bone marrow plasma cells influence their recovery in aspirate specimens: impact on classification of plasma cell disorders and potential bias to evaluation of treatment response. Ann Hematol 2020; 99:2599-2609. [PMID: 32935190 PMCID: PMC7536141 DOI: 10.1007/s00277-020-04249-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 09/01/2020] [Indexed: 01/22/2023]
Abstract
Methods to estimate bone marrow plasma cells (BMPC) basically include histopathology, cytomorphology, and flow cytometry. The present study compares the outcomes of these methods with special focus on the impact of BMPC-specific characteristics on their recovery by either method. Laboratory reports of diagnostic samples from 238 consecutive patients with suspected or known plasma cell disease were retrospectively analyzed. The median (IQR) proportion of BMPC was 30.0% (15.0–70.0%) by histological review (hBMPC), 7.0% (2.0–16.0%) by smear review (sBMPC), and 3.0% (0.8–10.0%) by flow cytometry (fBMPC). The disparity of results between core biopsy and aspirate smear was enhanced in case of poor quality of the smear, increased BM fiber content, higher grade cell atypia, expression of CD56 (all P < 0.0001), the number of cytogenetic aberrations (P = 0.0002), and abnormalities of the MYC gene (P = 0.0002). Conversely, expression of CD19 and a non-clonal plasma cell phenotype were associated with a lower difference between hBMPC and sBMPC (both P < 0.0001). The disparity between the percentages of sBMPC and fBMPC was associated with the quality of the smear (P = 0.0007) and expression of CD56 (P < 0.0001). Our results suggest that the recovery of BMPC in aspirate specimens not only is a matter of sampling quality but also depends on biological cell properties. Aspiration failure due to malignant type features of BMPC may lead to misclassification of plasma cell disorders and represent a bias for the detection of minimal residual disease after therapy.
Collapse
|
50
|
Kapoor R, Kumar R, Dubey AP. Risk Stratification in Multiple Myeloma in Indian Settings. Indian J Hematol Blood Transfus 2020; 36:464-472. [PMID: 32647419 DOI: 10.1007/s12288-019-01240-4] [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: 09/19/2019] [Accepted: 12/06/2019] [Indexed: 01/03/2023] Open
Abstract
Multiple myeloma (MM) constitutes 10% of all hematological malignancies. The last one decade has seen a phenomenal progress in the therapeutic options available for the management. Although it still remains incurable, with the advent of newer therapies, the median survival in many risk groups is now around 10 years. Conventional karyotyping of bone marrow samples has a positivity rate of 20-30% at diagnosis in patients of Multiple Myeloma. However, array Comparative Genomic Hybridisation (aCGH) has revealed that almost all MM patients have cytogenetic abnormalities which may affect the pathophysiology, selection of therapy and outcomes of the disease. The progress in the field of exploring the genetic landscape of multiple myeloma with multiple tools like Fluorescent in-situ hybridization, aCGH, Next Generation Sequencing, Flow cytometry, etc., combined with the traditional risk stratification markers like albumin, β2 microglobulin and LDH, is gradually leading towards a risk-adapted therapy. The recent R-ISS risk stratification has combined these two group of information to validate a prognostic score which is an improvement over the past tools like DSS and ISS. In view of the plethora of information available on the multitude of cytogenetic markers there is a tendency to evaluate for all of them at diagnosis, especially in research centers. This leads to a significant increase in the cost of therapy of Multiple Myeloma in day-to-day clinical practice and an increased out-of-pocket spending to the patient, especially in resource-limited settings like India. Also, there is a variable approach to pre-therapy cytogenetic evaluation and risk stratification at different Hematology centres in the country, often dictated by financial constraints and availability of specialized tests. This review discusses the risk stratification markers and tools available in MM in 2019 and how it can be adapted in the resource constraint settings so as to derive the maximum prognostic information from a minimal prognostic panel, as well as lead to standardization of the prognostic protocols in resource limited settings across various Hematology centres in India.
Collapse
Affiliation(s)
- Rajan Kapoor
- Medicine and Clinical Hematology, Command Hospital (EC), Kolkata, India
| | - Rajiv Kumar
- Department of Medicine and Clinical Hematology, INHS Asvini, R C Church, Colaba, Mumbai, 400005 India
| | - A P Dubey
- Medical Oncology, Clear Medi Hospitals and Cancer Centre, Ghaziabad, UP India
| |
Collapse
|