Targeting Myeloma Cells and Their Progenitors By Pim Kinase Inhibition

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5724-5724
Author(s):  
Shiro Fujii ◽  
Hirokazu Miki ◽  
Asuka Oda ◽  
Keiichiro Watanabe ◽  
Ryota Amachi ◽  
...  

Abstract Myeloma (MM) cells grow and expand almost exclusively in the bone marrow while creating a cellular microenvironment suitable for MM cell growth and survival (MM niche). In pursuing the molecular mechanisms whereby MM cells gain drug resistance in the “MM niche”, we have found that the serine/threonine kinase Pim-2 is constitutively over-expressed in MM cells, and further up-regulated by co-cultures with bone marrow stromal cells (BMSCs) as well as osteoclasts (Leukemia, 2011), and that Pim-2 is an important therapeutic target in MM for the progression of MM tumor and bone disease (Leukemia, 2014). The ABC transporter BCRP is preferentially expressed in drug resistant MM cells as well as in MM progenitors or stem cells. BCRP has been demonstrated to be phosphorylated by Pim kinases to trigger its dimerization and function; Pim inhibition may suppress the BCRP function to sensitize BCRP-expressing MM cells to chemotherapeutic agents. In the present study we therefore explored whether Pim inhibition is able to target and impair BCRP-expressing drug-resistant MM cells and MM progenitors. We analyzed an ABC transporter activity in BCRP-expressing RPMI8226 and KMS11 cells by intracellular accumulation and retention of BCRP substrates with auto-fluorescence emission, mitoxantrone and doxorubicin, in flow cytometry. Treatment with Pim inhibitors, SMI-16a or SMI-4a, increased the incorporation of these drugs into the MM cells and enhanced their subsequent intracellular retention after 6-hour incubation without these drugs, although BCRP expression on their surface was only marginally affected by the Pim inhibition. Interestingly, acidic conditions up-regulated Pim-2 expression while reducing the accumulation and retention of these drugs in BCRP-expressing RPMI8226 and KMS11 cells. However, the Pim inhibitors efficaciously restored the drug accumulation and retention reduced by extracellular acidification, and enhanced the cytotoxic activity of the BCRP substrate doxorubicin against RPMI8226 cells rather preferentially in acidic conditions. Furthermore, the Pim inhibition minimized the sizes of “side populations”, highly drug-resistant fractions with enhanced BCRP activity, and the ability of colony formation in RPMI8226 and KMS11 cells, which was more marked in acidic conditions. We previously demonstrated the in vivo effects of the Pim inhibitors in human INA-6 cell-bearing SCID-rab MM models and syngeneic mouse MM models with an intra-tibial inoculation of 5TGM1 MM cells (Leukemia, 2014). To further examine the acid-tropism of anti-tumorigenic activity of Pim inhibition, we pretreated murine 5TGM1 MM cells in vitro with or without SMI16a at pH6.8 for 24 hours, and transplanted to the tibiae in mice the same numbers of viable MM cells remaining in each treatment group. Treatment with SMI16a at pH6.8 almost completely abrogated in vivo tumorigenic capacity of 5TGM1 cells, while MM cells without the treatment rapidly grew and expanded in and outside of the tibiae, suggesting targeting clonogenic MM cells by Pim inhibition preferentially in acidic conditions. Taken together, Pim-2 may become an important therapeutic target of drug-resistant BCRP-expressing MM cells and their progenitors which appear to gain more drug resistance in acidic bone lesions. Combinatory treatment with Pim inhibitors warrants further study to overcome drug resistance in MM cells, including their tumorigenic cancer stem cells. Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 547-547
Author(s):  
Julia Kirshner ◽  
Kyle J. Thulien ◽  
Lorri D. Martin ◽  
Carina Debes Marun ◽  
Tony Reiman ◽  
...  

Abstract Bone marrow (BM), a site of hematopoiesis, is a multicellular tissue with a complex architecture. Multiple myeloma (MM) is an incurable plasma cell malignancy where even patients in remission succumb to an inevitable relapse. While considerable progress has been made towards understanding and treating MM, to date, there is no culture system which can recapitulate the complex interactions within the BM microenvironment. Current failure to grow the MM clone within the context of human microenvironment hampers progress into the understanding of the biology of MM and design of biologically relevant therapies. Here we present an in vitro three-dimensional (3-D) tissue culture model which recapitulates the human BM microenvironment allowing for the growth and expansion of the MM clone. Cells from the BM aspirates are grown in a fibronectin, laminin and collagen rich ECM designed to reconstruct in vitro endosteum and central marrow, mimicking the in vivo microenvironment of the BM. Proliferation and redistribution of cells within reconstructed ECM results in stratification of the culture, mimicking the in vivo condition where cells occupy individual niches. Cellular composition of the culture is maintained in accordance with the proliferation properties of the BM where osteoblasts, osteoclasts, adipocytes and stromal cells differentiate along with the full complement of the hematopoietic cells. BM cultures from normal donors are well-organized with osteoclasts and hematopoietic cells occupying distinct positions in the ECM. In contrast, reconstructed BM from MM patients is disorganized in 3-D where osteoclasts intermingle with the hematopoietic compartment. The MM malignant clone is expanded in 3-D cultures as measured by real-time quantitative PCR (rqPCR) for genomic clonotypic VDJ sequences. Malignant B and plasma cells proliferate in these cultures and FISH analysis reveals that their progeny harbor chromosomal abnormalities identical to those that mark the malignant clone prior to culture. Preclinical testing of emerging therapeutics targeted for multiple myeloma is hindered by the failure of the current models to sustain growth of the myeloma clone. In the 3-D culture, myeloma clone expands within its native environment providing an ideal preclinical model where conventional (Melphalan) and novel (Velcade) therapeutics efficiently and selectively kill their target cells. In the 3-D BM culture model, non-proliferating, label retaining cells (LRC) concentrate at a putative endosteum-marrow junction, where hematopoietic stem cells have been shown to localize in vivo, suggesting that the drug-resistant myeloma stem cells localize to the endosteal niche. In a colony-forming assay, drug-resistant LRC purified from the 3-D cultures form clonal colonies composed of malignant cells with patient specific clonotypic VDJ sequences. Recapitulation of the BM architecture in vitro is a first step towards the identification and therapeutic targeting of the elusive myeloma stem cell.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 602-602
Author(s):  
Erbey Ziya Özdemir ◽  
Sarah Ebinger ◽  
Christoph Ziegenhain ◽  
Wolfgang Enard ◽  
Olivier Gires ◽  
...  

Abstract Introduction Drug resistant cells represent a major threat for tumor patients as they might induce relapse and severely decrease disease outcome. Relapse represents a major drawback in patients with acute lymphoblastic leukemia (ALL), the single most frequent malignancy in children. Novel treatment options are intensively desired to remove drug resistant cells, which often additionally display dormancy. Aim We aimed at unraveling basic mechanisms determining drug resistance and dormancy, as basis for developing novel treatment strategies to prevent relapse. Methods Using cutting edge in vivo technology, we performed genetic engineering in the individualized xenograft mouse model of ALL. Primary patients' ALL cells were amplified in mice to generate patient-derived xenograft (PDX) cells. ALL PDX cells were lentivirally transduced to express transgenes. Recombinant luciferase allowed highly sensitive and reliable follow-up of leukemia growth and treatment. Recombinant surface markers enabled an unbiased approach to reliably and effectively enriching minute numbers of PDX cells from mouse bone marrow. Two independent, complementary innovative preclinical in vivo mouse models were established.In the first model, proliferation sensitive dyes allowed identifying and enriching in vivo long-term dormant PDX ALL cells.In the second model, the clinically highly relevant and challenging situation of MRD was mimicked in mice. PDX ALL cells were grown to advanced leukemia stages of above 30 % human blasts in bone marrow, when systemic chemotherapy with conventional cytotoxic drugs was initiated for prolonged periods of time, similar as applied in ALL patients. Chemotherapy reduced advanced leukemia down to 0,1 % or 10-3 leukemia cells in bone marrow, resembling not only complete morphologic remission, but even molecular remission. This novel preclinical model allows for the first time to characterize patients' dormant and MRD cells in detail including functional in vivo assays. Results Using our innovative preclinical model of dormancy, we identified a novel, distinct, rare subpopulation of PDX ALL cells that displayed long term dormancy in vivo. Long-term dormant cells showed significant resistance against drug treatment in vivo, as therapy nearly exclusively targeted proliferating cells. Dormant cells showed stem cell behavior as they initiated leukemia upon re-transplantation into further recipient mice. Long-term dormant cells thus combined the three challenging characteristics of relapse-inducing cells dormancy, drug resistance and stemness with re-growth upon withdrawal of treatment pressure. Using our second novel preclinical model, we isolated a pure, vivid fraction of rare MRD cells. These cells showed drug resistance in vivo and stemness features. We used single cell RNA sequencing to compare the transcriptomes of dormant and MRD populations and found that they were highly similar. Both populations had further similarities with primary high-risk ALL cells and dormant sub-fractions in patients' leukemia samples. Of high relevance for future treatment strategies, both, dormancy and drug resistance revealed transient characteristics in PDX ALL cells. When PDX long-term dormant ALL cells were distracted from their in vivo environment, they started proliferating similarly as their previously highly proliferative counterparts. When in vivo drug resistant PDX ALL cells were retrieved from murine bone marrow, they showed similar drug sensitivity in vitro as their sensitive counterparts. Summary/Conclusion Thus, both in vivo dormancy and drug resistance represent reversible characteristics in ALL cells which might result from the localization of ALL cells in the bone marrow niche. Dissolving ALL cells from their in vivo environment might sensitize them towards treatment. Addressing and inhibiting the interaction between ALL cells and their bone marrow niche might represent an attractive future therapeutic strategy to prevent ALL relapse. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 5011-5011
Author(s):  
Shiro Fujii ◽  
Masami Iwasa ◽  
Takeshi Harada ◽  
Shingen Nakamura ◽  
Hirokazu Miki ◽  
...  

Abstract Abstract 5011 Myeloma (MM) cells and osteoclasts (OCs) mutually interact in MM bone lesions to confer aggressiveness and drug resistance in MM cells along with the progression of bone destruction. The MM-OC interaction appears to create a highly acidic milieu in bone lesions by proton produced from activated OCs and lactate from proliferating MM cells (the Warburg effect). Tumor acidity is known to cause drug resistance in cancers. As suggested, acidic conditions or cocultures with OCs blunt the cytotoxic effects of anti-MM agents such as doxorubicin. Therefore, the development of innovative modalities is strongly needed to overcome the drug resistance in an acidic microenvironment in MM. We have found that the serine/threonine kinase Pim-2 is over-expressed in MM cells as an anti-apoptotic mediator, and further up-regulated to cause their aggressiveness and drug resistance when cocultured with bone marrow stromal cells or OCs (Leukemia, 2011). In the present study, we aim to clarify the role of Pim-2 in MM cell growth and survival in acidic conditions and the therapeutic impact of Pim inhibition on drug-resistant MM cells. The phosphorylation of 4E-BP1, a substrate of Pim-2, as well as Pim-2 expression were up-regulated in MM cells in media acidified by lactic acid or HCl; the Pim inhibitor SMI-16a preferentially induced MM cell death at pH 6. 8 or 6. 4 rather than at pH7. 4, suggesting Pim-2-dependent MM cell survival in an acidic milieu. In contrast to their quick death at pH 7. 4, substantial numbers of MM cells remained intact at pH6. 8 or lower upon treatment with doxorubicin. Interestingly, Pim inhibition enhanced the cytotoxic activity of doxorubicin against MM cells, which was more marked in acidic conditions. The ABC transporter BCRP is aberrantly over-expressed in drug-resistant MM cells. The intracellular levels of auto-fluorescence emitting doxorubicin and mitoxantron were reduced over time after their passive incorporation into BCRP-expressing RPMI8226 cells. However, the Pim inhibitor SMI-16a at 50 microM substantially restored the intracellular levels of these drugs; the intracellular retention of these drugs by the Pim inhibition was more prominent in acidic conditions, suggesting the correlation of BCRP function with Pim-2 up-regulation by acid. BCRP phosphorylation has been demonstrated to be essential for its functionality. Treatment with SMI-16a reduced BCRP phosphorylation in BCRP-expressing INA6 cells, suggesting Pim-dependent activation of BCRP. The reduction was more obvious at pH6. 8 than at pH7. 4. “Side population (SP)” is regarded as a highly drug-resistant fraction with enhanced BCRP activity, which is considered to contain clonogenic or tumor-initiating cells. The Pim inhibitor SMI-16a minimized the size of SP fractions in RPMI8226 and KMS11 cells; the reduction of SP fractions by the Pim inhibition was also more marked in acidic conditions. Collectively, these results suggest that Pim-2 plays an important role in MM cell survival and drug resistance in an acidic milieu, and that Pim-2 may become an important therapeutic target of MM cells which preferentially gain drug resistance in acidic bone lesions. Disclosures: Nakamura: Janssen Pharmaceutical K. K.: Honoraria. Abe:Janssen Pharmaceutical K. K.: Honoraria, Research Funding.


2020 ◽  
Vol 2020 ◽  
pp. 1-13
Author(s):  
Xiaoyan Zhang ◽  
Yazhi Yang ◽  
Yang Yang ◽  
Huijun Chen ◽  
Huaijun Tu ◽  
...  

Although major advances have been achieved in the treatment of chronic myeloid leukemia (CML) by using tyrosine kinase inhibitors, patients relapse after withdrawal and need long-term medication. This reflects the CML clones have not been eliminated completely. The precise mechanisms for the maintenance of CML cells are not yet fully understood. The bone marrow microenvironment constitutes the sanctuary for leukemic cells. Mesenchymal stem cells (MSC) are an important component of the bone marrow microenvironment (BM). It plays an important role in the development and drug resistance of CML. Accumulating evidence indicates that exosomes play a vital role in cell-to-cell communication. We successfully isolated and purified exosomes from human bone marrow microenvironment-derived mesenchymal stem cells (hBMMSC-Exo) by serial centrifugation. In the present study, we investigated the effect of hBMMSC-Exo on the proliferation, apoptosis, and drug resistance of CML cells. The results demonstrated that hBMMSC-Exo had the ability to inhibit the proliferation of CML cells in vitro via miR-15a and arrest cell cycle in the G0/G1 phase. However, the results obtained from BALB/c nu/nu mice studies apparently contradicted the in vitro results. In fact, hBMMSC-Exo increased tumor incidence and promoted tumor growth in vivo. Further study showed the antiapoptotic protein Bcl-2 expression increased, whereas the Caspase3 expression decreased. Moreover, the in vivo study in the xenograft tumor model showed that hBMMSC-Exo promoted the proliferation and decreased the sensitivity of CML cells to tyrosine kinase inhibitors, resulting in drug resistance. These results demonstrated that hBMMSC-Exo supported the maintenance of CML cells and drug resistance in BM by cell-extrinsic protective mechanisms. They also suggested that hBMMSC-Exo might be a potential target to overcome the microenvironment-mediated drug resistance.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 708-708
Author(s):  
Ehsan Bahrami ◽  
Jan Philipp Schmid ◽  
Martin Becker ◽  
Anna-Katharina Wirth ◽  
Rupert Öllinger ◽  
...  

Abstract Acute leukemias require more accurate and effective treatments, especially upon disease relapse. In search for novel therapeutic targets for acute leukemias, we established a pipeline for CRISPR-Cas9 mediated functional genomic screens which harbor the ability to elegantly increase our knowledge about vulnerabilities and gene dependencies. For a highly patient-related setting, we performed CRISPR knockout (KO) dropout screens in patient-derived xenograft (PDX) models in vivo, combining the advantages of studying an individual patient´s tumor cell in the physiologic in vivo bone marrow microenvironment. Serially transplantable PDX models were lentivirally transduced to stably express Cas9. A customized CRISPR-Cas9 library targeting about 100 genes addressing surface molecules was designed, cloned and transduced into two PDX models of acute lymphoblastic leukemia (ALL). Enriched PDX ALL cells were transplanted into NSG mice and grown until advanced disease stage. Input versus end stage cells were subjected to next generation sequencing, followed by data analysis using MAGeCK algorithm. Data analysis revealed commonly depleted as well as sample-specific depleted genes between the two PDX models tested, and CXCR4 and ITGB1 were the top commonly depleted genes from the screens. For target validation, single sgRNA were cloned into the knockout vector; concomitant expression of recombinant fluorochromes allowed competitive growth assays in PDX models in vivo, comparing cells with KO of interest versus control KO in the same animal. In vivo competitive assay showed that both PDX models clearly depended on both CXCR4 and ITGB1, validating an essential function for both genes in the two PDX ALL models. Of note, disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) was among the list of dropout genes in the screens. ADAM10 is known for its role in the central nervous system and considered as a therapeutic target in Alzheimer disease, but poorly studied in the context of leukemia. In competitive validation assays in vivo, ADAM10 KO population showed a clear growth disadvantage compared to control KO cells in a number of PDX models of ALL, but also acute myeloid leukemia (AML); in some PDX models, ADAM10 KO cells were completely abrogated, indicating that ADAM10 plays an essential role for different types of acute leukemias and represents a yet unknown vulnerability. To better characterize the role of ADAM10 for the clinical situation, we performed further in vivo assays with PDX models. Re-expression of ADAM10 in ADAM10 KO PDX cells could partially rescue the phenotype in an in vivo competitive reconstitution assay, unequivocally proving ADAM10 essentiality in ALL cells. Interestingly, a similar rescue assay expressing a ADAM10 variant lacking the disintegrin domain resulted in the same phenotypical compensation, highlighting essentiality of the enzymatic but not adhesion domain of ADAM10 in tumor engraftment and growth in BM. Important for translating the molecular insights into clinical use, PDX cells treated with an ADAM10 chemical inhibitor ex vivo, showed reduced tumor engraftment capacity compared to the vehicle treated cells, suggesting a role for ADAM10 in tumor-niche interactions and homing to the bone marrow. Further, we performed limiting dilution transplantation assays to determine stem cell frequencies; ADAM10 loss resulted in reduced stemness and a reduced number of leukemia-initiating cells compared to control KO cells, indicating that ADAM10 is essential also in leukemia stem cells. Of clinical relevance, ADAM10 KO significantly sensitized leukemic cells towards treatment of mice with the routine chemotherapeutic drug Cyclophosphamide in vivo, suggesting a putative synergistic effect when addressing ADAM10 as a therapeutic target. In summary, our data revealed ADAM10 as attractive novel vulnerability in acute leukemias with essential function for the tumor-niche interaction, leukemia stem cells and anti-leukemia treatment. ADAM10 might be addressed as therapeutic target to treat acute leukemias in the future. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 9 (1) ◽  
Author(s):  
Jian Wu ◽  
Min Zhang ◽  
Omar Faruq ◽  
Eldad Zacksenhaus ◽  
Wenming Chen ◽  
...  

Abstract Background SMAD1, a central mediator in TGF-β signaling, is involved in a broad range of biological activities including cell growth, apoptosis, development and immune response, and is implicated in diverse type of malignancies. Whether SMAD1 plays an important role in multiple myeloma (MM) pathogenesis and can serve as a therapeutic target are largely unknown. Methods Myeloma cell lines and primary MM samples were used. Cell culture, cytotoxicity and apoptosis assay, siRNA transfection, Western blot, RT-PCR, Soft-agar colony formation, and migration assay, Chromatin immunoprecipitation (Chip), animal xenograft model studies and statistical analysis were applied in this study. Results We demonstrate that SMAD1 is highly expressed in myeloma cells of MM patients with advanced stages or relapsed disease, and is associated with significantly shorter progression-free and overall survivals. Mechanistically, we show that SMAD1 is required for TGFβ-mediated proliferation in MM via an ID1/p21/p27 pathway. TGF-β also enhanced TNFα-Induced protein 8 (TNFAIP8) expression and inhibited apoptosis through SMAD1-mediated induction of NF-κB1. Accordingly, depletion of SMAD1 led to downregulation of NF-κB1 and TNFAIP8, resulting in caspase-8-induced apoptosis. In turn, inhibition of NF-κB1 suppressed SMAD1 and ID1 expression uncovering an autoregulatory loop. Dorsomorphin (DM), a SMAD1 inhibitor, exerted a dose-dependent cytotoxic effect on drug-resistant MM cells with minimal cytotoxicity to normal hematopoietic cells, and further synergized with the proteasomal-inhibitor bortezomib to effectively kill drug-resistant MM cells in vitro and in a myeloma xenograft model. Conclusions This study identifies SMAD1 regulation of NF-κB1/TNFAIP8 and ID1-p21/p27 as critical axes of MM drug resistance and provides a potentially new therapeutic strategy to treat drug resistance MM through targeted inhibition of SMAD1.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Pegah Nammian ◽  
Seyedeh-Leili Asadi-Yousefabad ◽  
Sajad Daneshi ◽  
Mohammad Hasan Sheikhha ◽  
Seyed Mohammad Bagher Tabei ◽  
...  

Abstract Introduction Critical limb ischemia (CLI) is the most advanced form of peripheral arterial disease (PAD) characterized by ischemic rest pain and non-healing ulcers. Currently, the standard therapy for CLI is the surgical reconstruction and endovascular therapy or limb amputation for patients with no treatment options. Neovasculogenesis induced by mesenchymal stem cells (MSCs) therapy is a promising approach to improve CLI. Owing to their angiogenic and immunomodulatory potential, MSCs are perfect candidates for the treatment of CLI. The purpose of this study was to determine and compare the in vitro and in vivo effects of allogeneic bone marrow mesenchymal stem cells (BM-MSCs) and adipose tissue mesenchymal stem cells (AT-MSCs) on CLI treatment. Methods For the first step, BM-MSCs and AT-MSCs were isolated and characterized for the characteristic MSC phenotypes. Then, femoral artery ligation and total excision of the femoral artery were performed on C57BL/6 mice to create a CLI model. The cells were evaluated for their in vitro and in vivo biological characteristics for CLI cell therapy. In order to determine these characteristics, the following tests were performed: morphology, flow cytometry, differentiation to osteocyte and adipocyte, wound healing assay, and behavioral tests including Tarlov, Ischemia, Modified ischemia, Function and the grade of limb necrosis scores, donor cell survival assay, and histological analysis. Results Our cellular and functional tests indicated that during 28 days after cell transplantation, BM-MSCs had a great effect on endothelial cell migration, muscle restructure, functional improvements, and neovascularization in ischemic tissues compared with AT-MSCs and control groups. Conclusions Allogeneic BM-MSC transplantation resulted in a more effective recovery from critical limb ischemia compared to AT-MSCs transplantation. In fact, BM-MSC transplantation could be considered as a promising therapy for diseases with insufficient angiogenesis including hindlimb ischemia.


2018 ◽  
Vol 106 ◽  
pp. 1126-1134 ◽  
Author(s):  
Wenbo Zhang ◽  
Chao Huang ◽  
Aijun Sun ◽  
Liang Qiao ◽  
Xi Zhang ◽  
...  

2001 ◽  
Vol 169 (1) ◽  
pp. 12-20 ◽  
Author(s):  
Jizong Gao ◽  
James E. Dennis ◽  
Raymond F. Muzic ◽  
Magnus Lundberg ◽  
Arnold I. Caplan

Sign in / Sign up

Export Citation Format

Share Document