PTH and Bortezomib Suppress Growth of Primary Human Myeloma through Increased Bone Formation In Vivo.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 509-509 ◽  
Author(s):  
Angela Pennisi ◽  
Wen Ling ◽  
Paul Perkins ◽  
Rinku Saha ◽  
Xin Li ◽  
...  

Abstract We have recently demonstrated the inhibitory effect of osteoblasts on myeloma (MM) ex vivo and in vivo (Yaccoby et al., Haematologica 2006) and that anti-MM response of bortezomib is associated with osteoblast activation in patients with MM (Zangari et al., BJH 2005). The aims of this study were to investigate the effect of intermittent PTH and bortezomib on bone remodeling and tumor growth, and the consequences of PTH pretreatment on MM progression in our SCID-rab model for primary MM (Yata & Yaccoby, Leukemia 2004). In nonmyelomatous hosts, both PTH and bortezomib significantly increased bone mineral density (BMD) of the implanted bone. SCID-rab mice were engrafted with MM cells from 13 patients. Following establishment of MM growth, as monitored by bi-weekly measurement of human monoclonal immunoglobulins (hIg) in mice sera and by x-rays, mice were injected subcutaneously with bortezomib (0.5 mg/kg twice a week, n=10), PTH (0.3 mg/kg/day, n=5) or PBS for 4–8 weeks. Whereas all PBS-treated mice had increased hIg levels during the experimental period, bortezomib treatment resulted in marked reduction of hIg in 5/10 experiments by 73±10% from pretreatment levels (responding hosts) and stabilized or delayed growth in additional 5 experiments. Overall, tumor burden in control- and bortezomib-treated mice was increased by 447±118% and 157±97% from pretreatment levels, respectively (p<0.02). Whereas in control mice the BMD of the implanted bone was reduced by 17±5% from pretreatment, it increased in bortezomib-treated hosts by 4±10% from pretreatment (p<0.03). While in bortezomib-responding hosts BMD increased by 20±14% (n=5), it decreased in partial/non-responding hosts (n=5) by 13±12% (n=5) from pretreatment (p<0.047). This bone anabolic effect was unique to bortezomib and was not observed in hosts responding to dexamethasone. Histological examination revealed that myelomatous bones from bortezomib-treated hosts had increased numbers of osteocalcin-expressing osteoblasts (34±7 vs. 13±3 per mm bone in control mice, p<0.03) and reduced numbers of multinucleated TRAP-expressing osteoclasts (10±3 vs. 28±7 per mm bone in control mice, p<0.02). We further demonstrated that bortezomib suppresses osteoclastogenesis through downregulation of NF-κB activity in osteoclast precursors. Since bortezomib also directly inhibits MM cell growth we further studied the association between increased bone formation and MM growth by treating hosts engrafted with MM cells from 5 patients with PTH, a bone anabolic agent with no known direct apoptotic effect on MM cells. Whereas PTH treatment resulted in increased BMD of the implanted bone by 19±5%, BMD in control hosts was reduced by 5±8% from pre-treatment (p<0.05). The bone anabolic effect of PTH was associated with inhibition of MM progression in 4/5 experiments. Overall, hIg in PBS- and PTH-treated mice was increased by 947±247% and 391±217% from pretreatment levels, respectively (p<0.04). In additional set of experiments hosts received PTH or PBS, 4 weeks prior to inoculation of MM cells from 3 patients and thereafter. In all experiments, PTH pretreatment, which increased implanted BMD by 48±11%, resulted in slower growth of MM cells as compared to control hosts. We conclude that increased bone formation by PTH and bortezomib contributes to controlling MM growth and that pretreatment with PTH, in addition to improving skeletal complications, may be a promising approach to prevent MM progression.

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3450-3450
Author(s):  
Shmuel Yaccoby ◽  
Wen Ling ◽  
Rinku Saha ◽  
Bart Barlogie ◽  
Guido Tricot ◽  
...  

Abstract Recent studies have indicated that certain proteasome inhibitors have bone anabolic activity in vivo. Our clinical study have shown a significant association between anti-myeloma (MM) response of bortezomib (Millennium Pharmaceuticals) and increased serum bone alkaline phosphatase in patients with MM (Zangari et al., BJH 2005, In press). The aim of this study was to investigate the effect of bortezomib on bone remodeling and the association between anti-myeloma response and increased bone formation in our established SCID-rab model for primary MM. In this system rabbit bones were implanted S.C. in unconditioned SCID mice. After 6 weeks, primary MM cells were injected directly into the implanted bone. MM cells from >85% of patients (n>70) were successfully engrafted, grew exclusively in the implanted bone and produced typical disease manifestations including increased osteoclast activity, reduced osteoblast numbers and induction of osteolytic bone disease (Yata & Yaccoby, Leukemia 2004). In the present study, SCID-rab mice were engrafted with MM cells from 5 patients. Following establishment of MM growth, as monitored by weekly measurement of human monoclonal immunoglobulins (hIg) in mice sera (587±276 μg/ml) and by x-rays, mice were injected subcutaneously twice a week with 0.5 mg/kg bortezomib or PBS for 4–8 weeks. Whereas all PBS-treated mice had increased hIg levels during the experimental period, bortezomib treatment resulted in marked reduction of hIg in 2 experiments by 95% and 73% from pretreatment levels, respectively, retardation of myeloma growth in an additional experiment and no response in 2 experiments. Overall, tumor burden in control PBS- and bortezomib-treated mice was increased by 298%±51 and 110%±88 from pretreatment levels, respectively (p<0.03). In control mice the implanted rabbit bone mineral density (BMD) and bone mineral content (BMC) were reduced by 23%±6% and 27%±10% from pretreatment levels, respectively, whereas in bortezomib-responsive mice BMD and BMC were increased by 57%±23% and 79%±60% from pretreatment levels, respectively (p<0.05, bortezomib vs. PBS). In the 2 non-responsive mice BMD and BMC were reduced by 36%±7% and 40%±3% from pretreatment levels, respectively, similar to the matched control mice. The bone anabolic effect of bortezomib could also be visualized on x-rays. Histological examination of a bone from a responsive myeloma revealed increased numbers of osteocalcin-expressing osteoblasts (25±4 vs. 8±3 per mm bone in control mice, p<0.03) and reduced number of multinucleated TRAP-expressing multinucleated osteoclasts (59±6 vs. 20±4 per mm bone in control mice, p<0.008). We conclude that anti-myeloma response to bortezomib is associated with increased osteoblast activity and bone formation in myelomatous bone. Since we previously demonstrated the ability of osteoblasts to inhibit growth of primary MM cells ex vivo, our study suggests an additional mechanism by which bortezomib inhibits myeloma.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 255-255
Author(s):  
Claire M. Edwards ◽  
Robert L. Caldwell ◽  
Andreia L. Bates ◽  
Jessica A. Fowler ◽  
James R. Edwards ◽  
...  

Abstract The proteasome inhibitor bortezomib (VELCADE®) represents an important advance in the treatment of multiple myeloma. Despite its very significant beneficial effects, its precise mechanism of action remains unclear. There is currently evidence to suggest that bortezomib has the potential to directly affect both myeloma cells and osteoblasts. However, it is not possible to discern the precise mechanism of action of bortezomib in patients with myeloma. To address this we used a well-characterized murine model of myeloma combined with MALDI mass spectrometry (MALDI MS) to study the effect of bortezomib on bone formation and tumor burden in vivo and protein profiles of myeloma cells. To more closely reflect the clinical setting, we used an established treatment protocol, in which bortezomib treatment was initiated upon development of myeloma. 5TGM1-GFP myeloma cells were inoculated into C57BlKaLwRij mice, resulting in tumor growth within the bone marrow and the development of an osteolytic bone disease. Tumor burden was monitored by measurement of serum IgG2bκ, and a significant increase was detected 14 days following tumor inoculation. Mice were then randomized to receive either bortezomib (0.5mg/kg, 3x week) or vehicle control for the remainder of the experiment. Treatment with bortezomib resulted in a significant reduction in tumor burden, as determined by serum IgG2bκ concentrations and by flow cytometric analysis of GFP-positive cells in the bone marrow. The tibia was analyzed by microCT, and bortezomib was found to significantly increase trabecular bone volume and reduce the number of osteolytic bone lesions. In order to study bone formation, mice were treated with 2 doses of calcein at a 7 day interval. Myeloma-bearing mice were associated with a significant reduction in rates of bone formation, which was prevented by treatment with bortezomib. GFP-positive myeloma cells were isolated from bone marrow of control- and bortezomib-treated mice and purified by fluorescence-activated cell sorting. Protein profiling by MALDI MS identified a number of proteins, both known and unknown, which were regulated by bortezomib treatment in vivo. Free ubiquitin (m/z 8565) was down-regulated, which is a known effect of proteasome inhibition resulting from the inhibition of protein degradation and release of free ubiquitin. Regulated proteins also included the up-regulation of thymosin beta-10 (m/z 4936) following bortezomib treatment. Thymosin beta 10 was undetectable in myeloma cells from control-treated mice. In addition, the calcium binding protein calgranulin A (S100A8) (m/z 10163) was up-regulated in myeloma cells isolated from bortezomib-treated mice. Neither thymosin-beta 10 nor calgranulin A have been previously linked to the activity of the proteasome, and thus may indicate novel molecular mechanisms involved in the anti-myeloma effect of bortezomib. Our data demonstrate that treatment with bortezomib from the time of established myeloma not only reduces myeloma tumor burden but also significantly increases bone formation in vivo. In addition, proteomic analysis of myeloma cells following in vivo treatment with bortezomib reveals a number of regulated proteins, providing novel insights into the molecular mechanism of action of proteasome inhibition in multiple myeloma in vivo.


2018 ◽  
Vol 15 (6) ◽  
pp. 531-543 ◽  
Author(s):  
Dominik Szwajgier ◽  
Ewa Baranowska-Wojcik ◽  
Kamila Borowiec

Numerous authors have provided evidence regarding the beneficial effects of phenolic acids and their derivatives against Alzheimer's disease (AD). In this review, the role of phenolic acids as inhibitors of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) is discussed, including the structure-activity relationship. In addition, the inhibitory effect of phenolic acids on the formation of amyloid β-peptide (Aβ) fibrils is presented. We also cover the in vitro, ex vivo, and in vivo studies concerning the prevention and treatment of the cognitive enhancement.


Biomolecules ◽  
2021 ◽  
Vol 11 (2) ◽  
pp. 131
Author(s):  
Aira Matsugaki ◽  
Yumi Kimura ◽  
Ryota Watanabe ◽  
Fumihito Nakamura ◽  
Ryo Takehana ◽  
...  

Malignant melanoma favors spreading to bone, resulting in a weakened bone with a high fracture risk. Here, we revealed the disorganized alignment of apatite crystals in the bone matrix associated with the homing of cancer cells by developing an artificially controlled ex vivo melanoma bone metastasis model. The ex vivo metastasis model reflects the progressive melanoma cell activation in vivo, resulting in decreased bone mineral density and expression of MMP1-positive cells. Moreover, less organized intercellular connections were observed in the neighboring osteoblasts in metastasized bone, indicating the abnormal and randomized organization of bone matrix secreted by disconnected osteoblasts. Our study revealed that the deteriorated microstructure associated with disorganized osteoblast arrangement was a determinant of malignant melanoma-related bone dysfunction.


1982 ◽  
Vol 48 (01) ◽  
pp. 087-090 ◽  
Author(s):  
Carlos O Esquivel ◽  
David Bergqvist ◽  
Claes-Göran Björck ◽  
Stan N Carson ◽  
Bodil Nilsson

SummaryThe effect of sodium ibuprofen on platelet activity in vivo and the lysability of ex vivo thrombi was investigated. The formation of a hemostatic platelet plug in the rabbit mesentery and platelet embolism as a response to a laser-induced injury in the ear chamber of rabbits were used as models for determining platelet activity. Ibuprofen at a dose of 25 mg/kg i.v. was found to increase the primary (PHT) and the total hemostatic plug formation time (THT). The same dose decreased the number of cumulative emboli over a 10 min period after a laser injury to arterioles. A dose of 10 mg/kg i.v. did not affect the formation of the hemostatic platelet plug. In dogs, doses of 10, 25 und 50 mg/kg did not enhance the release of 125I-FDP from the thrombi after incubation in plasmin, but the largest dose which is approximately five times the recommended dose in humans, did significantly decrease the thrombus weight 90 and 180 min after the drug administration. In conclusion, sodium ibuprofen was shown to have an inhibitory effect on platelet function in vivo and in large doses was also found to diminish the thrombus weight.


Cancers ◽  
2022 ◽  
Vol 14 (2) ◽  
pp. 337
Author(s):  
John D. Christie ◽  
Nicole Appel ◽  
Liqiang Zhang ◽  
Kenneth Lowe ◽  
Jacquelyn Kilbourne ◽  
...  

Cancers that metastasize to the lungs represent a major challenge in both basic and clinical cancer research. Oncolytic viruses are newly emerging options but successful delivery and choice of appropriate therapeutic armings are two critical issues. Using an immunocompetent murine K7M2-luc lung metastases model, the efficacy of MYXV armed with murine LIGHT (TNFSF14/CD258) expressed under virus-specific early/late promoter was tested in an advanced later-stage disease K7M2-luc model. Results in this model show that mLIGHT-armed MYXV, delivered systemically using ex vivo pre-loaded PBMCs as carrier cells, reduced tumor burden and increased median survival time. In vitro, when comparing direct infection of K7M2-luc cancer cells with free MYXV vs. PBMC-loaded virus, vMyx-mLIGHT/PBMCs also demonstrated greater cytotoxic capacity against the K7M2 cancer cell targets. In vivo, systemically delivered vMyx-mLIGHT/PBMCs increased viral reporter transgene expression levels both in the periphery and in lung tumors compared to unarmed MYXV, in a tumor- and transgene-dependent fashion. We conclude that vMyx-mLIGHT, especially when delivered using PBMC carrier cells, represents a new potential therapeutic strategy for solid cancers that metastasize to the lung.


1998 ◽  
Vol 72 (6) ◽  
pp. 5121-5127 ◽  
Author(s):  
Prasad S. Koka ◽  
John K. Fraser ◽  
Yvonne Bryson ◽  
Gregory C. Bristol ◽  
Grace M. Aldrovandi ◽  
...  

ABSTRACT Human immunodeficiency virus type 1 (HIV-1)-infected individuals often exhibit multiple hematopoietic abnormalities reaching far beyond loss of CD4+ lymphocytes. We used the SCID-hu (Thy/Liv) mouse (severe combined immunodeficient mouse transplanted with human fetal thymus and liver tissues), which provides an in vivo system whereby human pluripotent hematopoietic progenitor cells can be maintained and undergo T-lymphoid differentiation and wherein HIV-1 infection causes severe depletion of CD4-bearing human thymocytes. Herein we show that HIV-1 infection rapidly and severely decreases the ex vivo recovery of human progenitor cells capable of differentiation into both erythroid and myeloid lineages. However, the total CD34+ cell population is not depleted. Combination antiretroviral therapy administered well after loss of multilineage progenitor activity reverses this inhibitory effect, establishing a causal role of viral replication. Taken together, our results suggest that pluripotent stem cells are not killed by HIV-1; rather, a later stage important in both myeloid and erythroid differentiation is affected. In addition, a primary virus isolated from a patient exhibiting multiple hematopoietic abnormalities preferentially depleted myeloid and erythroid colony-forming activity rather than CD4-bearing thymocytes in this system. Thus, HIV-1 infection perturbs multiple hematopoietic lineages in vivo, which may explain the many hematopoietic defects found in infected patients.


2001 ◽  
Vol 281 (2) ◽  
pp. E283-E288 ◽  
Author(s):  
Dennis L. Andress

Insulin-like growth factor binding protein-5 (IGFBP-5) is an osteoblast secretory protein that becomes incorporated into the mineralized bone matrix. In osteoblast cultures, IGFBP-5 stimulates cell proliferation by an IGF-independent mechanism. To evaluate whether IGFBP-5 can stimulate osteoblast activity and enhance bone accretion in a mouse model of osteoblast insufficiency, daily subcutaneous injections of either intact [IGFBP-5 (intact)] or carboxy-truncated IGFBP-5 [IGFBP-5-(1–169)] were given to ovariectomized (OVX) mice for 8 wk. Femur and spine bone mineral density (BMD), measured every 2 wk, showed early and sustained increases in response to IGFBP-5. Bone histomorphometry of cancellous bone showed significant elevations in the bone formation rate in both the femur metaphysis [IGFBP-5- (1)] only) and spine compared with OVX controls. IGFBP-5 also stimulated osteoblast number in the femur IGFBP-5-(1–169) only) and spine. These data indicate that IGFBP-5 effectively enhances bone formation and bone accretion in OVX mice by stimulating osteoblast activity. The finding that IGFBP-5-(1–169) is bioactive in vivo indicates that the carboxy-terminal portion is not required for this bone anabolic effect.


BioTechniques ◽  
2020 ◽  
Vol 69 (1) ◽  
pp. 37-45
Author(s):  
Murali K Ravoori ◽  
Sheela Singh ◽  
Peiying Yang ◽  
Wei Wei ◽  
Huiqin Chen ◽  
...  

Methods for imaging orthotopic prostate tumors within the prostate or small tumors with extension outside the prostate are needed to more closely model human prostate tumors, which are most commonly located within the gland or may extend just through the gland. By comparing MR sequences, we found that the T2-based Dixon ‘water only’ sequence best visualized tumors within the prostate of mouse models in both young and old mice and that tumor weight derived from this sequence correlated highly with ex vivo tumor weight (r2 = 0.98, p < 0.001, n = 12). This should aid tumor detection, margin delineation and evaluation of tumor burden to enable studies including, but not limited to, evaluating the natural history of the disease, the mechanisms of action and the efficacy of therapeutic interventions.


Sign in / Sign up

Export Citation Format

Share Document