1
|
Kumar D, Kiran FNU, Wheeler A, Dellamano R, Hammer RD. An alternative processing approach to increase CD138 intensity in flow cytometric analysis of plasma cells. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2024; 106:113-116. [PMID: 38010113 DOI: 10.1002/cyto.b.22149] [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: 06/04/2023] [Revised: 10/17/2023] [Accepted: 10/31/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Surface median immunofluorescence intensity (MFI) of plasma cells antigens, particularly CD138, by flow cytometry underestimates plasma cell populations when compared with that estimated by morphological assessment on Wright's-stained slides. CD138 MFI using traditional sample preparation methods for flow cytometric analysis is often dim and difficult to interpret due to multiple factors. This becomes critical when diagnosing and accurately classifying plasma cell dyscrasias. METHODS In this study, we analyzed 280 flow cytometric results collected from 2016 to 2022 for CD38 and CD138 MFI on bone marrow aspirates performed by two different methods of sample processing-traditional method of lyse-wash and the alternative method of lyse-no-wash. RESULTS Visual examination of histograms showed a clear advantage to CD138 expression intensity with the no-wash method. Although no significant difference was observed in CD38 MFI between the two techniques (p = 0.3), considerable improvement was observed in CD138 MFI with the lyse-no-wash technique of sample processing compared with the conventional method (p = 0.003). CONCLUSIONS We concluded that the method of lyse-no-wash is superior to traditional methods especially when it comes to handling bone marrow aspirate samples for plasma cell immunophenotyping. This alternate technique increases the sensitivity of flow cytometry to detect plasma cells resulting in bright and crisp signal intensity for surface CD138. This technique may be particularly advantageous when analyzing low tumor burden such as minimal residual disease.
Collapse
Affiliation(s)
- Deepak Kumar
- Department of Pathology and Anatomical Sciences, University of Missouri Health Care, Columbia, Missouri, USA
| | - F N U Kiran
- Department of Pathology and Anatomical Sciences, University of Missouri Health Care, Columbia, Missouri, USA
| | - Amanda Wheeler
- Department of Pathology and Anatomical Sciences, University of Missouri Health Care, Columbia, Missouri, USA
| | - Rory Dellamano
- Department of Pathology and Anatomical Sciences, University of Missouri Health Care, Columbia, Missouri, USA
| | - Richard D Hammer
- Department of Pathology and Anatomical Sciences, University of Missouri Health Care, Columbia, Missouri, USA
| |
Collapse
|
2
|
Gilligan G, Panico R, Garola F, Jara R, Villarroel-Dorrego M, Martinez B. Unusual clinical presentations of plasma cell mucositis involving oral mucosa. Presentation of 2 cases and review of the literature. Oral Surg Oral Med Oral Pathol Oral Radiol 2023:S2212-4403(23)00436-4. [PMID: 37328328 DOI: 10.1016/j.oooo.2023.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 06/18/2023]
Abstract
OBJECTIVE Plasma cell mucositis (PCM) is a non-neoplastic plasma cell disorder of the upper aerodigestive tract with a high impact on life quality. Less than 70 cases were reported in the literature. The objective of this study was to report 2 cases of PCM. A concise review of the literature is also presented. STUDY DESIGN Two cases of PCM that presented during the COVID-19 quarantine are reported. The inclusion criteria for the literature review were English-indexed case reports of the last 20 years. RESULTS Cases were treated with meprednisone. As mechanical trauma was proposed as a triggering factor, its control was also considered. Patients were followed with no relapses. There were 29 studies included. The mean age was 57 years, with a male predominance, different clinical phenotypes, and intensely erythematous mucosa as a classical finding. The most frequent site was the lip, followed by the buccal mucosa. The final diagnosis is clinicopathologic. CD138 expression is a hallmark of plasma cells, frequently aiding PCM diagnosis. Plasma cell mucositis treatment is mostly symptomatic, and several therapeutic modalities have been mostly unsuccessful. CONCLUSIONS Diagnosing plasma cell mucositis becomes challenging as many lesions may mimic other conditions. Consequently, in these cases, the diagnostic process should gather clinical, histopathologic, and immunohistochemical data.
Collapse
Affiliation(s)
- Gerardo Gilligan
- Oral Medicine Department, Facultad de Odontología, Universidad Nacional de Córdoba, Córdoba, Argentina.
| | - René Panico
- Oral Medicine Department, Facultad de Odontología, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Federico Garola
- Oral Medicine Department, Facultad de Odontología, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - René Jara
- Oral Pathology Service, Universidad Mayor de Chile, Santiago de Chile, Chile
| | | | - Benjamín Martinez
- Oral Pathology Service, Universidad Mayor de Chile, Santiago de Chile, Chile
| |
Collapse
|
3
|
Development of an Independent Prognostic Signature Based on Three Hypoxia-Related Genes for Breast Cancer. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:2974126. [DOI: 10.1155/2022/2974126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/29/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022]
Abstract
Background. Hypoxia was considered to be a prognostic indicator in a variety of solid tumors. This study aims at identifying the hypoxia-related genes (HRGs) in breast cancer (BC) and the feasibility of HRGs as a prognostic indicator. Methods. We downloaded the mRNA expression data of BC patients from TCGA and GEO databases. The LASSO Cox regression analysis was applied to screen the hub HRGs to establish a prognostic Risk Score. The independence of Risk Score was assessed by multivariate Cox regression analysis. And the immune checkpoint analysis was also performed. In addition, we also detected the expression level of hub HRGs in MCF-10A cells, MCF-7 cells, and SK-BR-3 cells by RT-qPCR. Results. Three HRGs were identified as hub genes with prognostic value in BC, including CA9, PGK1, and SDC1. The Risk Score constructed by these three genes could efficiently distinguish the prognosis of different BC patients and has been shown to be an independent prognostic indicator. In the high-risk group, patients had lower overall survival and poorer prognosis. In addition, the expression levels of five immune checkpoints (PD1, CTLA4, TIGIT, LAG3, and TIM3) in the high-risk group were significantly higher than those in the low-risk group. Moreover, the expression levels of PGK1 and SDC1 in BC cells were significantly increased. Conclusion. In this study, we established an efficiently model based on three optimal HRGs (CA9, PGK1, and SDC1) could clearly distinguish the prognosis of different BC patients.
Collapse
|
4
|
Junctional Adhesion Molecule-C expression specifies a CD138low/neg multiple myeloma cell population in mice and humans. Blood Adv 2021; 6:2195-2206. [PMID: 34861679 PMCID: PMC9006287 DOI: 10.1182/bloodadvances.2021004354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 10/04/2021] [Indexed: 11/20/2022] Open
Abstract
JAM-C identifies a distinct MM cell population in bone marrow of patients and mice. Targeting JAM-C ameliorates MM progression and offers potential therapeutic options that might complement standard treatment regimens.
Deregulation such as overexpression of adhesion molecules influences cancer progression and survival. Metastasis of malignant cells from their primary tumor site to distant organs is the most common reason for cancer-related deaths. Junctional adhesion molecule-C (JAM-C), a member of the immunoglobulin-like JAM family, can homodimerize and aid cancer cell migration and metastasis. Here we show that this molecule is dynamically expressed on multiple myeloma (MM) cells in the bone marrow and co-localizes with blood vessels within the bone marrow of patients and mice. In addition, upregulation of JAM-C inversely correlates with the downregulation of the canonical plasma cell marker CD138 (syndecan-1), whose surface expression has recently been found to dynamically regulate a switch between MM growth in situ and MM dissemination. Moreover, targeting JAM-C in a syngeneic in vivo MM model ameliorates MM progression and improves outcome. Overall, our data demonstrate that JAM-C might serve not only as an additional novel diagnostic biomarker but also as a therapeutic target in MM disease.
Collapse
|
5
|
Alhallak K, Jeske A, de la Puente P, Sun J, Fiala M, Azab F, Muz B, Sahin I, Vij R, DiPersio JF, Azab AK. A pilot study of 3D tissue-engineered bone marrow culture as a tool to predict patient response to therapy in multiple myeloma. Sci Rep 2021; 11:19343. [PMID: 34588522 PMCID: PMC8481555 DOI: 10.1038/s41598-021-98760-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/07/2021] [Indexed: 12/28/2022] Open
Abstract
Cancer patients undergo detrimental toxicities and ineffective treatments especially in the relapsed setting, due to failed treatment attempts. The development of a tool that predicts the clinical response of individual patients to therapy is greatly desired. We have developed a novel patient-derived 3D tissue engineered bone marrow (3DTEBM) technology that closely recapitulate the pathophysiological conditions in the bone marrow and allows ex vivo proliferation of tumor cells of hematologic malignancies. In this study, we used the 3DTEBM to predict the clinical response of individual multiple myeloma (MM) patients to different therapeutic regimens. We found that while no correlation was observed between in vitro efficacy in classic 2D culture systems of drugs used for MM with their clinical efficacious concentration, the efficacious concentration in the 3DTEBM were directly correlated. Furthermore, the 3DTEBM model retrospectively predicted the clinical response to different treatment regimens in 89% of the MM patient cohort. These results demonstrated that the 3DTEBM is a feasible platform which can predict MM clinical responses with high accuracy and within a clinically actionable time frame. Utilization of this technology to predict drug efficacy and the likelihood of treatment failure could significantly improve patient care and treatment in many ways, particularly in the relapsed and refractory setting. Future studies are needed to validate the 3DTEBM model as a tool for predicting clinical efficacy.
Collapse
Affiliation(s)
- Kinan Alhallak
- Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park Ave, St. Louis, MO, 63108, USA.,Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Amanda Jeske
- Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park Ave, St. Louis, MO, 63108, USA.,Department of Biomedical Engineering, Washington University, St. Louis, MO, USA.,Cellatrix LLC, St. Louis, MO, USA
| | - Pilar de la Puente
- Cellatrix LLC, St. Louis, MO, USA.,Cancer Biology and Immunotherapies Group, Sanford Research, Sioux Falls, SD, USA
| | - Jennifer Sun
- Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park Ave, St. Louis, MO, 63108, USA.,Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Mark Fiala
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Barbara Muz
- Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park Ave, St. Louis, MO, 63108, USA
| | - Ilyas Sahin
- Division of Hematology/Oncology, The Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Ravi Vij
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - John F DiPersio
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park Ave, St. Louis, MO, 63108, USA. .,Department of Biomedical Engineering, Washington University, St. Louis, MO, USA. .,Cellatrix LLC, St. Louis, MO, USA.
| |
Collapse
|
6
|
Alhallak K, Sun J, Jeske A, Park C, Yavner J, Bash H, Lubben B, Adebayo O, Khaskiah A, Azab AK. Bispecific T Cell Engagers for the Treatment of Multiple Myeloma: Achievements and Challenges. Cancers (Basel) 2021; 13:2853. [PMID: 34201007 PMCID: PMC8228067 DOI: 10.3390/cancers13122853] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/26/2021] [Accepted: 06/02/2021] [Indexed: 12/15/2022] Open
Abstract
MM is the second most common hematological malignancy and represents approximately 20% of deaths from hematopoietic cancers. The advent of novel agents has changed the therapeutic landscape of MM treatment; however, MM remains incurable. T cell-based immunotherapy such as BTCEs is a promising modality for the treatment of MM. This review article discusses the advancements and future directions of BTCE treatments for MM.
Collapse
Affiliation(s)
- Kinan Alhallak
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, MO 63108, USA; (K.A.); (J.S.); (A.J.); (C.P.); (J.Y.); (H.B.); (B.L.); (O.A.)
- Department of Biomedical Engineering, Washington University in St. Louis McKelvey School of Engineering, St. Louis, MO 63130, USA
| | - Jennifer Sun
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, MO 63108, USA; (K.A.); (J.S.); (A.J.); (C.P.); (J.Y.); (H.B.); (B.L.); (O.A.)
- Department of Biomedical Engineering, Washington University in St. Louis McKelvey School of Engineering, St. Louis, MO 63130, USA
| | - Amanda Jeske
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, MO 63108, USA; (K.A.); (J.S.); (A.J.); (C.P.); (J.Y.); (H.B.); (B.L.); (O.A.)
- Department of Biomedical Engineering, Washington University in St. Louis McKelvey School of Engineering, St. Louis, MO 63130, USA
| | - Chaelee Park
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, MO 63108, USA; (K.A.); (J.S.); (A.J.); (C.P.); (J.Y.); (H.B.); (B.L.); (O.A.)
| | - Jessica Yavner
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, MO 63108, USA; (K.A.); (J.S.); (A.J.); (C.P.); (J.Y.); (H.B.); (B.L.); (O.A.)
| | - Hannah Bash
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, MO 63108, USA; (K.A.); (J.S.); (A.J.); (C.P.); (J.Y.); (H.B.); (B.L.); (O.A.)
| | - Berit Lubben
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, MO 63108, USA; (K.A.); (J.S.); (A.J.); (C.P.); (J.Y.); (H.B.); (B.L.); (O.A.)
| | - Ola Adebayo
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, MO 63108, USA; (K.A.); (J.S.); (A.J.); (C.P.); (J.Y.); (H.B.); (B.L.); (O.A.)
| | - Ayah Khaskiah
- Faculty of Pharmacy, Nursing and Health Professions, Birzeit University, Birzeit 627, West Bank, Palestine;
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Washington University in St. Louis School of Medicine, St. Louis, MO 63108, USA; (K.A.); (J.S.); (A.J.); (C.P.); (J.Y.); (H.B.); (B.L.); (O.A.)
- Department of Biomedical Engineering, Washington University in St. Louis McKelvey School of Engineering, St. Louis, MO 63130, USA
| |
Collapse
|
7
|
Co-evolution of tumor and immune cells during progression of multiple myeloma. Nat Commun 2021; 12:2559. [PMID: 33963182 PMCID: PMC8105337 DOI: 10.1038/s41467-021-22804-x] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 03/10/2021] [Indexed: 12/17/2022] Open
Abstract
Multiple myeloma (MM) is characterized by the uncontrolled proliferation of plasma cells. Despite recent treatment advances, it is still incurable as disease progression is not fully understood. To investigate MM and its immune environment, we apply single cell RNA and linked-read whole genome sequencing to profile 29 longitudinal samples at different disease stages from 14 patients. Here, we collect 17,267 plasma cells and 57,719 immune cells, discovering patient-specific plasma cell profiles and immune cell expression changes. Patients with the same genetic alterations tend to have both plasma cells and immune cells clustered together. By integrating bulk genomics and single cell mapping, we track plasma cell subpopulations across disease stages and find three patterns: stability (from precancer to diagnosis), and gain or loss (from diagnosis to relapse). In multiple patients, we detect “B cell-featured” plasma cell subpopulations that cluster closely with B cells, implicating their cell of origin. We validate AP-1 complex differential expression (JUN and FOS) in plasma cell subpopulations using CyTOF-based protein assays, and integrated analysis of single-cell RNA and CyTOF data reveals AP-1 downstream targets (IL6 and IL1B) potentially leading to inflammation regulation. Our work represents a longitudinal investigation for tumor and microenvironment during MM progression and paves the way for expanding treatment options. Clonal evolution in multiple myeloma (MM) needs to be understood in both the tumor and its microenvironment. Here the authors perform single-cell multi-omics profiling of samples from MM patients at different stages, finding transitions in the immune cell composition throughout progression.
Collapse
|
8
|
Martino M, Paviglianiti A. An update on B-cell maturation antigen-targeted therapies in Multiple Myeloma. Expert Opin Biol Ther 2021; 21:1025-1034. [PMID: 33412948 DOI: 10.1080/14712598.2021.1872540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction: B-cell maturation antigen (BCMA) targeted therapy (BCMA-TT) has emerged as a promising treatment for Multiple Myeloma (MM). the three most common treatment modalities for targeting BCMA are antibody-drug conjugates (ADCs), bispecific antibody constructs, including BiTE (bispecific T-cell engager) immuno-oncology therapies, and chimeric antigen receptor (CAR)-modified T-cell therapy.Areas covered: The review provides an overview of the main published studies on clinical and pre-clinical data from trials using BCMA-TT.Expert opinion: Despite progresses in survival outcomes and the availability of new drugs, MM remains an incurable disease. ADC is a promising antibody-based treatment and Belantamab mafodotin showed an anti-myeloma effect alone or in combination with other drugs. The major issue of ADC is the occurrence of events interfering with the efficacy and the off-target cytotoxicity. Bispecific antibody constructs are off-the-shelf therapies characterized by a potential rapid availability. The most critical limitation of bispecific antibody constructs is their short half-life necessitating prolonged intravenous infusion. CAR-T cells produced unprecedented results in heavily pretreated RRMM. The most common toxicities include neurologic toxicity and cytokine release syndrome, B-cell aplasia, cytopenias, and hypogammaglobulinemia. Further studies are needed to detect which are the eligible patients who could benefit from one treatment more than another.
Collapse
Affiliation(s)
- Massimo Martino
- Stem Cell Transplant and Cellular Therapies Unit, Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Reggio Calabria, Italy
| | - Annalisa Paviglianiti
- Stem Cell Transplant and Cellular Therapies Unit, Department of Hemato-Oncology and Radiotherapy, Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Reggio Calabria, Italy
| |
Collapse
|
9
|
Nanoparticle T-cell engagers as a modular platform for cancer immunotherapy. Leukemia 2021; 35:2346-2357. [PMID: 33479469 PMCID: PMC8292428 DOI: 10.1038/s41375-021-01127-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 12/01/2020] [Accepted: 01/07/2021] [Indexed: 01/29/2023]
Abstract
T-cell-based immunotherapy, such as CAR-T cells and bispecific T-cell engagers (BiTEs), has shown promising clinical outcomes in many cancers; however, these therapies have significant limitations, such as poor pharmacokinetics and the ability to target only one antigen on the cancer cells. In multiclonal diseases, these therapies confer the development of antigen-less clones, causing tumor escape and relapse. In this study, we developed nanoparticle-based bispecific T-cell engagers (nanoBiTEs), which are liposomes decorated with anti-CD3 monoclonal antibodies (mAbs) targeting T cells, and mAbs targeting the cancer antigen. We also developed a nanoparticle that targets multiple cancer antigens by conjugating multiple mAbs against multiple cancer antigens for T-cell engagement (nanoMuTEs). NanoBiTEs and nanoMuTEs have a long half-life of about 60 h, which enables once-a-week administration instead of continuous infusion, while maintaining efficacy in vitro and in vivo. NanoMuTEs targeting multiple cancer antigens showed greater efficacy in myeloma cells in vitro and in vivo, compared to nanoBiTEs targeting only one cancer antigen. Unlike nanoBiTEs, treatment with nanoMuTEs did not cause downregulation (or loss) of a single antigen, and prevented the development of antigen-less tumor escape. Our nanoparticle-based immuno-engaging technology provides a solution for the major limitations of current immunotherapy technologies.
Collapse
|
10
|
Targeting CD47 as a Novel Immunotherapy for Multiple Myeloma. Cancers (Basel) 2020; 12:cancers12020305. [PMID: 32012878 PMCID: PMC7072283 DOI: 10.3390/cancers12020305] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/17/2020] [Accepted: 01/22/2020] [Indexed: 12/25/2022] Open
Abstract
Multiple myeloma (MM) remains to be incurable despite recent therapeutic advances. CD47, an immune checkpoint known as the "don't eat me" signal, is highly expressed on the surface of various cancers, allowing cancer cells to send inhibitory signals to macrophages and impede phagocytosis and immune response. In this study, we hypothesized that blocking the "don't eat me" signaling using an anti-CD47 monoclonal antibody will induce killing of MM cells. We report that CD47 expression was directly correlated with stage of the disease, from normal to MGUS to MM. Moreover, MM cells had remarkably higher CD47 expression than other cell populations in the bone marrow. These findings indicate that CD47 is specifically expressed on MM and can be used as a potential therapeutic target. Further, blocking of CD47 using an anti-CD47 antibody induced immediate activation of macrophages, which resulted in induction of phagocytosis and killing of MM cells in the 3D-tissue engineered bone marrow model, as early as 4 hours. These results suggest that macrophage checkpoint immunotherapy by blocking the CD47 "don't eat me" signal is a novel and promising strategy for the treatment of MM, providing a basis for additional studies to validate these effects in vivo and in patients.
Collapse
|
11
|
Wang N, Tesfaluul N, Li J, Gao X, Liu S, Yue B. Enrichment of circulating myeloma cells by immunomagnetic beads combined with flow cytometry for monitoring minimal residual disease and relapse in patients with multiple myeloma. Ann Hematol 2019; 98:2769-2780. [PMID: 31748925 DOI: 10.1007/s00277-019-03833-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 10/28/2019] [Indexed: 12/14/2022]
Abstract
Difficulty in regularly analyzing marrow myeloma cells (MMCs) and low frequency of circulating myeloma cells (CMCs) in blood presents challenges for monitoring minimal residual disease (MRD) in multiple myeloma (MM). We have developed a set of method for enrichment of CMCs by immunomagetic beads (IMB) combined with flow cytometry (IMB-FCM) based on CD38-APC/CD138-APC antibodies in U266-spiked samples and in 122 patient samples. U266 cell capture efficiency of CD38/CD138-IMB-FCM (6.960, 2.574) was 6- and 2-fold higher than that of FCM (1.032), and the sensitivity of FCM and IMB-FCM was 0.01% and 0.001%, respectively. In MM cohort, the positive rate of CMCs by IMB-FCM increased from 60.5~70.0 to 85~87.2% in newly diagnosed/relapsed and partial remission (PR) patients compared with by FCM (P < 0.05). Two complete remission (CR) patients contain certain amounts of CMCs by IMB-FCM while no CMCs and MMCs were detectable by FCM. Patients exhibiting PR and CR upon therapy had much lower CMC and MMC counts than newly diagnosed/relapsed patients (P < 0.005). Based on MRD measurement in BM and PB samples, all FCM-negative BM samples were also paired with FCM/IMB-FCM-negative PB samples among newly diagnosed, relapsed, and PR patients, and FCM-positive BM samples were accompanied by IMB-FCM-positive results in 88% of corresponding PB samples. CMCs strongly associated with other clinical biomarkers of disease burden, including elevated MMCs, β2-MG, sCrea, and DS and ISS stages, and more serious anemia, bone destruction, and renal impairment (P < 0.05). Logistic regression analysis revealed that elevated β2-MG and moderate-to-more anemia were significant risk factors for the presence of CMCs (P < 0.05). As a noninvasive "liquid biopsy" of monitoring MRD, the potential of IMB-FCM for CMC detection may complement or minimize bone marrow aspiration in future treatment of MM patients.
Collapse
Affiliation(s)
- Ningning Wang
- Department of Laboratory Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Department of Laboratory Medicine, the First People's Hospital of Pingdingshan, Pingdingshan, Henan, China
| | - Nahom Tesfaluul
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jia Li
- Department of Laboratory Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaojuan Gao
- Department of Laboratory Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shuai Liu
- Department of Laboratory Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Faculty of Laboratory Medicine, Zhengzhou University, Zhengzhou, Henan, China.,Key Laboratory Medicine of Henan Province, Faculty of Laboratory Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Baohong Yue
- Department of Laboratory Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China. .,Faculty of Laboratory Medicine, Zhengzhou University, Zhengzhou, Henan, China. .,Key Laboratory Medicine of Henan Province, Faculty of Laboratory Medicine, Zhengzhou University, Zhengzhou, Henan, China. .,Open Laboratory, Henan Province Key Subject of Clinical Medicine, Zhengzhou, Henan, China.
| |
Collapse
|
12
|
Muz B, Azab F, Fiala M, King J, Kohnen D, Fogler WE, Smith T, Magnani JL, Vij R, Azab AK. Inhibition of E-Selectin (GMI-1271) or E-selectin together with CXCR4 (GMI-1359) re-sensitizes multiple myeloma to therapy. Blood Cancer J 2019; 9:68. [PMID: 31431613 PMCID: PMC6702213 DOI: 10.1038/s41408-019-0227-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/07/2019] [Accepted: 05/14/2019] [Indexed: 12/26/2022] Open
Affiliation(s)
- Barbara Muz
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Feda Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Mark Fiala
- Department of Medicine, Division of Medical Oncology, Washington University in St. Louis School of Medicine, Saint Louis, MO, USA
| | - Justin King
- Department of Medicine, Division of Medical Oncology, Washington University in St. Louis School of Medicine, Saint Louis, MO, USA
| | - Daniel Kohnen
- Department of Medicine, Division of Medical Oncology, Washington University in St. Louis School of Medicine, Saint Louis, MO, USA
| | | | - Ted Smith
- GlycoMimetics Inc., Rockville, MD, USA
| | | | - Ravi Vij
- Department of Medicine, Division of Medical Oncology, Washington University in St. Louis School of Medicine, Saint Louis, MO, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
13
|
Kamande JW, Lindell MAM, Witek MA, Voorhees PM, Soper SA. Isolation of circulating plasma cells from blood of patients diagnosed with clonal plasma cell disorders using cell selection microfluidics. Integr Biol (Camb) 2018; 10:82-91. [PMID: 29372735 PMCID: PMC5877822 DOI: 10.1039/c7ib00183e] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Blood samples from patients with plasma cell disorders were analysed for the presence of circulating plasma cells (CPCs) using a microfluidic device modified with monoclonal anti-CD138 antibodies. CPCs were immuno-phenotyped using a CD38/CD56/CD45 panel and identified in 78% of patients with monoclonal gammopathy of undetermined significance (MGUS), all patients with smouldering and symptomatic multiple myeloma (MM), and none in the controls. The burden of CPCs was higher in patients with symptomatic MM compared with MGUS and smouldering MM (p < 0.05). FISH analysis revealed the presence of chromosome 13 deletions in CPCs that correlated with bone marrow results. Point mutations in KRAS were identified, including different mutations from sub-clones derived from the same patient. The microfluidic assay represents a highly sensitive method for enumerating CPCs and allows for the cytogenetic and molecular characterization of CPCs.
Collapse
Affiliation(s)
- Joyce W Kamande
- Department of Biomedical Engineering, The University of North Carolina at Chapel Hill, NC 27599, USA
| | | | | | | | | |
Collapse
|
14
|
de la Puente P, Luderer MJ, Federico C, Jin A, Gilson RC, Egbulefu C, Alhallak K, Shah S, Muz B, Sun J, King J, Kohnen D, Salama NN, Achilefu S, Vij R, Azab AK. Enhancing proteasome-inhibitory activity and specificity of bortezomib by CD38 targeted nanoparticles in multiple myeloma. J Control Release 2017; 270:158-176. [PMID: 29196043 DOI: 10.1016/j.jconrel.2017.11.045] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/29/2017] [Accepted: 11/27/2017] [Indexed: 01/03/2023]
Abstract
The establishment of more effective treatments that can circumvent chemoresistance in Multiple Myeloma (MM) is a priority. Although bortezomib (BTZ) is one of the most potent proteasome inhibitors available, still possesses limitations related to dose limiting side effects. Several strategies have been developed to improve the delivery of chemotherapies to MM by targeting different moieties expressed on MM cells to nanoparticle delivery systems (NPs), which have failed mainly due to their heterogeneous expression on these cells. Our goal was to test CD38 targeted chitosan NPs as novel targeting moiety for MM to improve the potency and efficacy of BTZ in MM cells and reduce the side effects in healthy tissue. We have showed preferential BTZ release in tumor-microenvironment, specific binding to MM cells, and an improved drug cellular uptake through BTZ diffusion from the surface and endocytosed NPs, which translated in enhanced proteasome inhibition and robust cytotoxic effect on MM cells when BTZ was administered through anti-CD38 chitosan NPs. Furthermore, the anti-CD38 chitosan NPs specifically delivered therapeutic agents to MM cells improving therapeutic efficacy and reducing side effects in vivo. The anti-CD38 chitosan NPs showed low toxicity profile allowing enhancement of proteasome-inhibitory activity and specificity of BTZ by endocytosis-mediated uptake of CD38 representing a promising therapy in MM.
Collapse
Affiliation(s)
- Pilar de la Puente
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Micah J Luderer
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Cinzia Federico
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Abbey Jin
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA; Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, MO, USA
| | - Rebecca C Gilson
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, MO, USA
| | - Christopher Egbulefu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, MO, USA
| | - Kinan Alhallak
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Shruti Shah
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Barbara Muz
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Jennifer Sun
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA
| | - Justin King
- Section of Stem Cell Transplant and Leukemia, Division of Medical Oncology, USA
| | - Daniel Kohnen
- Section of Stem Cell Transplant and Leukemia, Division of Medical Oncology, USA
| | - Noha Nabil Salama
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, MO, USA; Department of Pharmaceutics and Industrial Pharmacy, Cairo University Faculty of Pharmacy, Cairo, Egypt
| | - Samuel Achilefu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, MO, USA
| | - Ravi Vij
- Section of Stem Cell Transplant and Leukemia, Division of Medical Oncology, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, MO, USA.
| |
Collapse
|
15
|
Foulk B, Schaffer M, Gross S, Rao C, Smirnov D, Connelly MC, Chaturvedi S, Reddy M, Brittingham G, Mata M, Repollet M, Rojas C, Auclair D, DeRome M, Weiss B, Sasser AK. Enumeration and characterization of circulating multiple myeloma cells in patients with plasma cell disorders. Br J Haematol 2017; 180:71-81. [DOI: 10.1111/bjh.15003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/01/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Brad Foulk
- Janssen Research & Development, LLC; Spring House PA USA
| | - Mike Schaffer
- Janssen Research & Development, LLC; Spring House PA USA
| | - Steve Gross
- Menarini Silicon Biosystems; Huntingdon Valley PA USA
| | - Chandra Rao
- Janssen Research & Development, LLC; Spring House PA USA
| | - Denis Smirnov
- Janssen Research & Development, LLC; Spring House PA USA
| | | | | | - Manjula Reddy
- Janssen Research & Development, LLC; Spring House PA USA
| | | | - Marielena Mata
- Janssen Research & Development, LLC; Spring House PA USA
| | | | - Claudia Rojas
- Menarini Silicon Biosystems; Huntingdon Valley PA USA
| | | | - Mary DeRome
- Multiple Myeloma Research Foundation; Norwalk CT USA
| | - Brendan Weiss
- Abramson Cancer Center and Perelman School of Medicine; University of Pennsylvania; Philadelphia PA USA
| | - Amy K. Sasser
- Janssen Research & Development, LLC; Spring House PA USA
| | | |
Collapse
|
16
|
Mykytiv V, Alwaheed A, Mohd Hashim NA. Double CD38 -/CD138 - negative multiple myeloma. Hematol Oncol Stem Cell Ther 2017; 12:64-66. [PMID: 29079129 DOI: 10.1016/j.hemonc.2017.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/23/2017] [Accepted: 08/07/2017] [Indexed: 10/18/2022] Open
Abstract
The standard diagnosis of multiple myeloma by flow cytometry is based on selection of population of CD38+/CD138+ positives cells. As the result treatment with proteasome inhibitors, CD138 may be underexpressed on atypical plasma cells. Thus, in order to improve this strategy, recently new CD138-independent method, based on CD38 positivity of plasma cells was developed. We present an unusual case of CD138- negative multiple myeloma which had become double CD138-/CD38- negative after treatment with daratumumab by which we would like to illustrate potential pitfalls of both strategies.
Collapse
Affiliation(s)
- Vitaliy Mykytiv
- Haematology Department, Cork University Hospital, Cork, Ireland.
| | - Abrar Alwaheed
- Haematology Department, Cork University Hospital, Cork, Ireland.
| | | |
Collapse
|
17
|
Muz B, Azab F, de la Puente P, Landesman Y, Azab AK. Selinexor Overcomes Hypoxia-Induced Drug Resistance in Multiple Myeloma. Transl Oncol 2017; 10:632-640. [PMID: 28668761 PMCID: PMC5496204 DOI: 10.1016/j.tranon.2017.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/20/2017] [Accepted: 04/24/2017] [Indexed: 01/07/2023] Open
Abstract
Increased levels of the nuclear export protein, exportin 1 (XPO1), were demonstrated in multiple myeloma (MM) patients. Targeting XPO1 with selinexor (the selective inhibitor of nuclear export; SINE compound KPT-330) demonstrates broad antitumor activity also in patient cells resistant to bortezomib; hence, it is a promising target in MM patients. Hypoxia is known to mediate tumor progression and drug resistance (including bortezomib resistance) in MM cells. In this study, we tested the effects of selinexor alone or in combination with bortezomib in normoxia and hypoxia on MM cell survival and apoptosis in vitro and in vivo. In vitro, selinexor alone decreased survival and increased apoptosis, resensitizing MM cells to bortezomib. In vivo, we examined the effects of selinexor alone on tumor initiation and tumor progression, as well as selinexor in combination with bortezomib, on tumor growth in a bortezomib-resistant MM xenograft mouse model. Selinexor, used as a single agent, delayed tumor initiation and tumor progression, prolonging mice survival. In bortezomib-resistant xenografts, selinexor overcame drug resistance, significantly decreasing tumor burden and extending mice survival when combined with bortezomib.
Collapse
Affiliation(s)
- Barbara Muz
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, Saint Louis, MO 63108, USA
| | - Feda Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, Saint Louis, MO 63108, USA
| | - Pilar de la Puente
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, Saint Louis, MO 63108, USA
| | | | - Abdel Kareem Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in Saint Louis School of Medicine, Saint Louis, MO 63108, USA.
| |
Collapse
|
18
|
Chakraborty R, Muchtar E, Kumar SK, Jevremovic D, Buadi FK, Dingli D, Dispenzieri A, Hayman SR, Hogan WJ, Kapoor P, Lacy MQ, Leung N, Gertz MA. Serial measurements of circulating plasma cells before and after induction therapy have an independent prognostic impact in patients with multiple myeloma undergoing upfront autologous transplantation. Haematologica 2017; 102:1439-1445. [PMID: 28473618 PMCID: PMC5541877 DOI: 10.3324/haematol.2017.166629] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 04/28/2017] [Indexed: 12/14/2022] Open
Abstract
Circulating plasma cells at diagnosis, prior to auto-transplant and at relapse have a negative impact on survival in multiple myeloma. However, the impact of kinetics of circulating plasma cells along the course of illness has not been defined. We have analyzed 247 newly diagnosed multiple myeloma patients undergoing early auto-transplant who had paired evaluation of circulating plasma cells at diagnosis and pre-transplant by 6-color flow cytometry. A total of 117 patients had no detectable circulating plasma cells at both time points (CPC−/−), 82 had circulating plasma cells at diagnosis followed by complete eradication after induction (CPC+/−) and 48 had circulating plasma cells at transplant, including persistence of cells (CPC+/+; n=45) or emergence of new cells (CPC−/+; n=3) after induction. The rate of post-transplant stringent complete response was 32% in the CPC−/−, 30% in CPC+/− and 12% in CPC+/+ or −/+ groups (P=0.018). At a median follow up of 58 months from transplantation, the median progression-free survival in the 3 respective groups were 30, 24 and 14 months, and the 5-year overall survival rates were 83%, 70% and 43% (P<0.001 for both comparisons). On a multivariate analysis for overall survival, the risk of mortality was higher in CPC +/− (hazard ratio 2.7, 95%CI: 1.3–5.8; P=0.009) and CPC+/+ or −/+ (hazard ratio 5.7, 95%CI: 2.5–13.1; P<0.001) groups compared to the CPC−/− group. Monitoring for circulating plasma cells before induction therapy and before transplant by 6-color flow cytometry is predictive of survival in newly diagnosed myeloma and should be incorporated into clinical trials.
Collapse
Affiliation(s)
- Rajshekhar Chakraborty
- Division of Hematology, Mayo Clinic, Rochester, MN, USA.,Hospitalist Services, Essentia Health-St. Joseph's Medical Center, Brainerd, MN, USA
| | - Eli Muchtar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Shaji K Kumar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Dragan Jevremovic
- Department of Laboratory Medicine and Pathology, Division of Hematopathology, Mayo Clinic, Rochester, MN, USA
| | | | - David Dingli
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Martha Q Lacy
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Nelson Leung
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Morie A Gertz
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
19
|
de la Puente P, Azab AK. Nanoparticle delivery systems, general approaches, and their implementation in multiple myeloma. Eur J Haematol 2017; 98:529-541. [PMID: 28208215 DOI: 10.1111/ejh.12870] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2017] [Indexed: 12/25/2022]
Abstract
Multiple myeloma (MM) is a hematological malignancy that remains incurable, with relapse rates >90%. The main limiting factor for the effective use of chemotherapies in MM is the serious side effects caused by these drugs. The emphasis in cancer treatment has shifted from cytotoxic, non-specific chemotherapies to molecularly targeted and rationally designed therapies showing greater efficacy and fewer side effects. Traditional chemotherapy has shown several disadvantages such as lack of targeting capabilities, systemic toxicity, and side effects; low therapeutic index, as well as most anticancer drugs, has poor water solubility. Nanoparticle delivery systems (NPs) are capable of targeting large doses of chemotherapies into the target area while sparing healthy tissues, overcoming the limitations of traditional chemotherapy. Here, we review the current state of the art in nanoparticle-based strategies designed to treat MM. Many nanoparticle delivery systems have been studied for myeloma using non-targeted NPs (liposomes, polymeric NPs, and inorganic NPs), triggered NPs, as well as targeted NPs (VLA-4, ABC drug transporters, bone microenvironment targeting). The results in preclinical and clinical studies are promising; however, there remains much to be learned in the emerging field of nanomedicine in myeloma.
Collapse
Affiliation(s)
- Pilar de la Puente
- Cancer Biology Division, Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, St. Louis, MO, USA
| | - Abdel Kareem Azab
- Cancer Biology Division, Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|