99mTc-sestamibi imaging and bone marrow karyotyping in the assessment of multiple myeloma and MGUS

2008 ◽  
Vol 29 (6) ◽  
pp. 535-541 ◽  
Author(s):  
Luca Giovanella ◽  
Monica Taborelli ◽  
Luca Ceriani ◽  
Emanuele Zucca ◽  
Franco Cavalli ◽  
...  
Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3543-3543
Author(s):  
Camila Mosci ◽  
Fernando Vieira Pericole ◽  
Allan de Oliveira Santos ◽  
Mariana Cunha Lima ◽  
Elba Cristina Camargo Etchebehere ◽  
...  

Abstract Introduction: Multiple myeloma (MM) is a plasma cell neoplasm, characterized by plasma cell infiltration inside the bone marrow, secretion of monoclonal immunoglobulin (paraprotein), and end organ damage including lytic lesions in the bones. About 80-90% of myeloma patients suffer from osteolytic lesions during the course of the disease. 18F-FDG PET/CT is an imaging technique capable to detect active disease in patients in multiple myeloma (MM) and can be helpful in staging and prognosis. However, its routine use is still hampered by several factors, including high cost, reimbursement issues, lack of cost-effectiveness studies and limited availability. 99mTc-sestamibi (MIBI) has also been proposed as a potential tracer in MM evaluation and is more accessible with lower costs. The aim of this study was to compare these two imaging modalities at staging disease and their relation with clinical data. Materials and Methods: Sixty-four patients with newly diagnostic MM (30 male; 34 female) were submitted to 18F-FDG PET/CT and 99mTc-Sestamibi SPECT/CT before treatment. Whole body PET/CT images were acquired 60 minutes after FDG administration and anterior and posterior whole-body scans (WBS) plus SPECT/CT of chest and abdomen were obtained 10 minutes after MIBI injection. Number of focal lesions, bone marrow involvement, contiguous soft tissue impairment and extra osseous lesions were recorded. Number of focal lesions was classified in 3 groups: 0 (no lesions); 1 (1-3 lesions); 2 (4-10); 3 (more than 10). A visual degree of uptake was defined for bone marrow involvement: comparison to liver on PET/CT and to myocardium on MIBI. Standardized uptake value (SUVmax) of the hottest lesion of each patient was registered. Potentials factors contributing to progression-free survival (PFS) were assessed with Cox regression model combining baseline clinical data (including renal function, anemia, hypercalcemia, LDH, bone marrow plasma cell percentage and ISS (I, II or III)) along with PET/CT and MIBI scan status. Results: PET/CT was positive in 61 patients (95%) and MIBI in 59 subjects (92%; P = 0.15). WBS was positive in 56 patients while WBS plus SPECT/CT was positive in 59 (p= 0.08). PET/CT detected extra osseous lesions in 4 patients and sestamibi in 1 subject. Contiguous soft tissue involvement was found in 29 and 24 patients on PET/CT and MIBI, respectively (p=0.05). PET/CT detected much more focal lesions than MIBI: 13, 11, 16 and 24 patients were in group 0, 1, 2 and 3 on PET/CT and 30, 18, 6 and 10 were on the same groups respectively on MIBI (p: 0.0001). In the figure below, a comparison between 99mTc-Sestamibi WBS (A) and 18F-FDG PET/CT (B) at staging in a 67 years-old male. SUVmax were statistically different in subjects who presented elevated LDH (p= 0.02). Seventy-five percent and 100% of patients with elevated LDH had contiguous soft tissue involvement on MIBI and PET/CT respectively. More focal lesions on PET/CT were found in patients with hypercalcemia (p=0.02), however this correlation was not observed on MIBI (p=0.45). Renal insufficiency was a negative prognostic factor for PFS (HR: 2.25). The same was observed with advanced ISS staging (HR: 4.29). However, only advanced ISS staging (III) and extramedullary disease detected by MIBI were independent predictors of worse PFS. Conclusion: There was no difference in the detection of active disease when comparing FDG PET/CT and MIBI SPECT/CT in MM staging, although the first one detected more number of lesions. Including SPECT/CT to planar images on MIBI did also not improve the number of positive scans. Elevated LDH and hypercalcemia were the only clinical parameters related to higher number of bone lesions while ISS staging (III) and extramedullary disease detected by MIBI were independent predictors of worse PFS. Our study demonstrated that sestamibi WBI detects less volume of disease compared to PET/CT, however it may substitute PET/CT in centers where it is not available or there is no reimbursement for MM staging. Figure. Figure. Disclosures No relevant conflicts of interest to declare.


2020 ◽  
Vol 92 (7) ◽  
pp. 85-89
Author(s):  
L. P. Mendeleeva ◽  
I. G. Rekhtina ◽  
A. M. Kovrigina ◽  
I. E. Kostina ◽  
V. A. Khyshova ◽  
...  

Our case demonstrates severe bone disease in primary AL-amyloidosis without concomitant multiple myeloma. A 30-year-old man had spontaneous vertebral fracture Th8. A computed tomography scan suggested multiple foci of lesions in all the bones. In bone marrow and resected rib werent detected any tumor cells. After 15 years from the beginning of the disease, nephrotic syndrome developed. Based on the kidney biopsy, AL-amyloidosis was confirmed. Amyloid was also detected in the bowel and bone marrow. On the indirect signs (thickening of the interventricular septum 16 mm and increased NT-proBNP 2200 pg/ml), a cardial involvement was confirmed. In the bone marrow (from three sites) was found 2.85% clonal plasma cells with immunophenotype СD138+, СD38dim, СD19-, СD117+, СD81-, СD27-, СD56-. FISH method revealed polysomy 5,9,15 in 3% of the nuclei. Serum free light chain Kappa 575 mg/l (/44.9) was detected. Multiple foci of destruction with increased metabolic activity (SUVmax 3.6) were visualized on PET-CT, and an surgical intervention biopsy was performed from two foci. The number of plasma cells from the destruction foci was 2.5%, and massive amyloid deposition was detected. On CT scan foci of lesions differed from bone lesions at multiple myeloma. Bone fragments of point and linear type (button sequestration) were visualized in most of the destruction foci. The content of the lesion was low density. There was no extraossal spread from large zones of destruction. There was also spontaneous scarring of the some lesions (without therapy). Thus, the diagnosis of multiple myeloma was excluded on the basis based on x-ray signs, of the duration of osteodestructive syndrome (15 years), the absence of plasma infiltration in the bone marrow, including from foci of bone destruction by open biopsy. This observation proves the possibility of damage to the skeleton due to amyloid deposition and justifies the need to include AL-amyloidosis in the spectrum of differential diagnosis of diseases that occur with osteodestructive syndrome.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A168-A168
Author(s):  
Eric Lutz ◽  
Lakshmi Rudraraju ◽  
Elizabeth DeOliveira ◽  
Amanda Seiz ◽  
Monil Shah ◽  
...  

BackgroundMarrow infiltrating lymphocytes (MILsTM) are the product of activating and expanding bone marrow T cells.1 The bone marrow is a specialized niche in the immune system enriched for antigen-experienced, memory T cells. In patients with multiple myeloma and other hematological malignancies that relapse post-transplant, MILs have been shown to contain tumor antigen-specific T cells and adoptive cell therapy (ACT) using MILs has demonstrated antitumor activity.2 3 The bone marrow has been shown to harbor tumor-antigen specific T cells in patients with melanoma,4 5 glioblastoma,6 breast,7 non-small-cell lung8 and pancreatic cancers.9 Here, we sought to determine if tumor-specific MILs could be expanded from the bone marrow of patients with a range of different solid tumors.MethodsBone marrow and blood samples were collected from patients with advanced and metastatic cancers. To date, samples have been collected from a minimum of four patients with non-small cell lung cancer (NSCLC), prostate cancer, head and neck cancer, glioblastoma, and breast cancer. Samples from patients with multiple myeloma were used as a reference control. Utilizing a 10-day proprietary process, MILs and peripheral blood lymphocytes (PBLs) were activated and expanded from patient bone marrow and blood samples, respectively. T cell lineage-specific markers (CD3, CD4 and CD8) were characterized by flow cytometry pre- and post-expansion.Tumor-specific T cells were quantitated in expanded MILs and PBLs using a previously described cytokine-secretion assay [2]. Briefly, autologous antigen-presenting cells (APCs) were pulsed with lysates from allogeneic cancer cell lines and co-cultured with activated MILs or PBLs. APCs pulsed with irrelevant mis-matched cancer cell line lysates or media alone were used as negative controls. Tumor-specific T cells were defined as the IFNgamma-producing population by flow cytometry.ResultsMILs were successfully expanded from all patient bone marrow samples tested, regardless of tumor type. Cytokine-producing tumor-specific CD4+ and CD8+ T cells were detected in each of the expanded MILs. In contrast, tumor-specific T cells were not detected in any of the matched activated and expanded PBLs.ConclusionsMILs have been successfully grown for all solid tumor types evaluated, including NSCLC, prostate, head and neck, glioblastoma and breast cancer. Clinical studies have been completed in patients with multiple myeloma and other hematological cancers. 2 3 A phase IIa trial to evaluate MILs in combination with a checkpoint inhibitor is underway in patients with anti-PD1/PDL1-refractory NSCLC (ClinicalTrials.gov Identifier: NCT04069936). The preclinical data presented herein demonstrate that expanding MILs is feasible. MILs-based therapies hold therapeutic promise across a wide range of tumor indications.Ethics ApprovalThis study was approved by each participating instituion’s IRB.ReferencesBorrello I and Noonan KA. Marrow-Infiltrating Lymphocytes - Role in Biology and Cancer Therapy. Front Immunol 2016 March 30; 7(112)Noonan KA, Huff CA, Davis J, et al. Adoptive transfer of activated marrow-infiltrating lymphocytes induces measurable antitumor immunity in the bone marrow in multiple myeloma. Sci. Transl. Med 2015;7:288ra78.Biavati L, Noonan K, Luznik L, Borrello I. Activated allogeneic donor-derived marrow-infiltrating lymphocytes display measurable in vitro antitumor activity. J Immunother 2019 Apr;42(3):73–80.Müller-Berghaus J, Ehlert K, Ugurel S, et al. Melanoma-reactive T cells in the bone marrow of melanoma patients: association with disease stage and disease duration. Cancer Res 2006;66(12):5997–6001.Letsch A, Keilholz U, Assfalg G, et al., Bone marrow contains melanoma-reactive CD8+ effector T Cells and, compared with peripheral blood, enriched numbers of melanoma-reactive CD8+ memory T cells. Cancer Res 2003 Sep 1;63(17):5582–5586.Chongsathidkiet P, Jackson C, Koyama S, et al., Sequestration of T cells in bone marrow in the setting of glioblastoma and other intracranial tumors. Nature Medicine 2018 Aug 13; 24:1459–1468.Feuerer M, Rocha M, Bai L, et al. Enrichment of memory T cells and other profound immunological changes in the bone marrow from untreated breast cancer patients. Int J Cancer 2001; 92(1):96–105.Safi S, Yamauchi Y, Stamova S, et al. Bone marrow expands the repertoire of functional T cells targeting tumor-associated antigens in patients with resectable non-small-cell lung cancer. Oncoimmunology 2019;8(12):e1671762.Schmitz-Winnenthal FH, Volk C, Z’Graggen K, et al. High frequencies of functional tumor-reactive T cells in bone marrow and blood of pancreatic cancer patients. Cancer Res 2005;65(21):10079–87.


Cancers ◽  
2021 ◽  
Vol 13 (2) ◽  
pp. 164
Author(s):  
Federica Costa ◽  
Valentina Marchica ◽  
Paola Storti ◽  
Fabio Malavasi ◽  
Nicola Giuliani

The emerging role of the PD-1/PD-L1 axis in MM immune-microenvironment has been highlighted by several studies. However, discordant data have been reported on PD-1/PD-L1 distribution within the bone marrow (BM) microenvironment of patients with monoclonal gammopathies. In addition, the efficacy of PD-1/PD-L1 blockade as a therapeutic strategy to reverse myeloma immune suppression and inhibit myeloma cell survival still remains unknown. Recent data suggest that, among the potential mechanisms behind the lack of responsiveness or resistance to anti-PD-L1/PD-1 antibodies, the CD38 metabolic pathways involving the immune-suppressive factor, adenosine, could play an important role. This review summarizes the available data on PD-1/PD-L1 expression in patients with MM, reporting the main mechanisms of regulation of PD-1/PD-L1 axis. The possible link between the CD38 and PD-1/PD-L1 pathways is also reported, highlighting the rationale for the potential use of a combined therapeutic approach with CD38 blocking agents and anti-PD-1/PD-L1 antibodies in order to improve their anti-tumoral effect in MM patients.


Sign in / Sign up

Export Citation Format

Share Document