scholarly journals New Xenograft Model of Multiple Myeloma and Efficacy of a Humanized Antibody Against Human Interleukin-6 Receptor

Blood ◽  
1997 ◽  
Vol 90 (6) ◽  
pp. 2437-2444 ◽  
Author(s):  
Toshiaki Tsunenari ◽  
Yasuo Koishihara ◽  
Akito Nakamura ◽  
Miki Moriya ◽  
Hiroyuki Ohkawa ◽  
...  

Abstract A new xenograft model of multiple myeloma (MM), where growth is strongly regulated by interleukin-6 (IL-6), was established in severe combined immunodeficiency (SCID) mice. In this model, endogenous IL-6 from SCID mice was ineffective at eliciting growth of the established human MM cell line KPMM2; these cells achieved autonomous growth through their autocrine secretion of IL-6. The etiopathology in this disease model is consistent with that of human MM. When greater than 3 × 106 KPMM2 cells were injected intravenously (IV), tumors developed in all mice and were predominantly localized in their bone marrow. Tumors were also apparent in the lymph nodes, but absent from other organs. Immunostaining of cell surface antigen (CD38) showed that more than 40% of bone marrow cells in femur were of myeloma origin in the advanced stage of tumor progression (day 37). Histologic analysis of these mice show that bone marrow was largely occupied by plasmablastic cells and bones had developed osteolytic lesions at multiple sites. Concurrently, there was a decrease in bone density throughout the body and a significant increase in ionized plasma calcium. M-protein was detected in the serum within 10 days after transplantation, which correlated with the tumor progression. Between 30 and 40 days after the transplantation, mice presented with a rapid and severe loss of body weight, hind leg paralysis, and fatigue. Subsequently, the mice died within a week. A single IV injection of 0.2 mg humanized anti–IL-6 receptor antibody (hPM1) into mice on the day after tumor transplantation substantially suppressed the elevation of serum M-protein and development of the tumor-associated abnormalities and significantly increased in the life span of tumor-bearing mice. Our data show the usefulness of this model to analyze the pathologic role of IL-6 in MM and the efficacy of targeting the IL-6 receptor in IL-6–dependent KPMM2 cells.

Blood ◽  
1997 ◽  
Vol 90 (6) ◽  
pp. 2437-2444 ◽  
Author(s):  
Toshiaki Tsunenari ◽  
Yasuo Koishihara ◽  
Akito Nakamura ◽  
Miki Moriya ◽  
Hiroyuki Ohkawa ◽  
...  

A new xenograft model of multiple myeloma (MM), where growth is strongly regulated by interleukin-6 (IL-6), was established in severe combined immunodeficiency (SCID) mice. In this model, endogenous IL-6 from SCID mice was ineffective at eliciting growth of the established human MM cell line KPMM2; these cells achieved autonomous growth through their autocrine secretion of IL-6. The etiopathology in this disease model is consistent with that of human MM. When greater than 3 × 106 KPMM2 cells were injected intravenously (IV), tumors developed in all mice and were predominantly localized in their bone marrow. Tumors were also apparent in the lymph nodes, but absent from other organs. Immunostaining of cell surface antigen (CD38) showed that more than 40% of bone marrow cells in femur were of myeloma origin in the advanced stage of tumor progression (day 37). Histologic analysis of these mice show that bone marrow was largely occupied by plasmablastic cells and bones had developed osteolytic lesions at multiple sites. Concurrently, there was a decrease in bone density throughout the body and a significant increase in ionized plasma calcium. M-protein was detected in the serum within 10 days after transplantation, which correlated with the tumor progression. Between 30 and 40 days after the transplantation, mice presented with a rapid and severe loss of body weight, hind leg paralysis, and fatigue. Subsequently, the mice died within a week. A single IV injection of 0.2 mg humanized anti–IL-6 receptor antibody (hPM1) into mice on the day after tumor transplantation substantially suppressed the elevation of serum M-protein and development of the tumor-associated abnormalities and significantly increased in the life span of tumor-bearing mice. Our data show the usefulness of this model to analyze the pathologic role of IL-6 in MM and the efficacy of targeting the IL-6 receptor in IL-6–dependent KPMM2 cells.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1565-1565
Author(s):  
Sunil Iyengar ◽  
Linda Ariza-McNaughton ◽  
Andrew James Clear ◽  
Amy Roe ◽  
Debra Lillington ◽  
...  

Abstract Abstract 1565 Background: The relatively low incidence of MCL (0.55 per 100,000) poses a challenge for effective evaluation of novel therapies in patients affected by this aggressive and incurable lymphoma. A xenograft model of human MCL provides a useful model for pre-clinical evaluation of novel drugs, rational drug combinations and biomarkers. The only mouse xenograft model of primary human MCL reported was established by injecting CD19 selected PBMCs in a subcutaneously implanted human embryonic bone graft in SCID mice and SCID mice without subcutaneous bone grafts did not show engraftment. NOD/SCID/IL2Rγ chain null (NSG) mice, which lack mature T or B cells and are also deficient in NK cells, permit engraftment of a wider range of primary human cells compared to SCID mice. In view of recent reports of successful engraftment of human CLL in NSG mice, we hypothesised that primary human MCL can be established in these mice. Methods: We initially introduced luciferase transduced Jeko-1 cells at 2 concentrations – 0.5 and 2 million cells by tail vein injection of 8 to 12 week old γ-irradiated (3.75 Gy) NSG mice in an attempt to track the kinetics and distribution of MCL cells. Bioluminescent imaging (BLI) following injection of luciferin was performed weekly for 4 weeks. We then injected NSG mice (4 replicates) with 107, T-cell depleted, previously cyropreserved human MCL cells from 3 patients. Mice were bled at 3, 6 and 12 weeks and flow cytometry was performed on PBMCs for human CD45, CD5 and CD20. Mice were sacrificed at 20 weeks and immunohistochemistry (IHC) for human CD20 and cyclin D1 was performed on formalin fixed paraffin embedded spleen, ileo-caecal junction and liver while FACS for human CD45, CD5 and CD20 was performed on bone marrow cells flushed from the femur. Tissues harbouring CD20 and cyclin D1 positive cells were stained with Ki-67 to assess proliferation and FISH for t(11;14) was performed on fresh cells isolated from the spleen to further confirm engraftment. Results: NSG mice injected with Jeko-1 cells showed rapid engraftment at both concentrations (0.5 and 2 million) on assessment with BLI, with a more rapid progression after 3 weeks in mice injected with 2 million cells. Mice had to be sacrificed at 4 weeks because of illness. Bioluminescence was seen primarily in the bone marrow, spleen and along the spine. Consistent engraftment was also seen in all mice injected with sample 1 - PBMCs from a patient in 1st relapse with blastoid morphology, classic immunophenotypic features and the IgH:CCND1 translocation. CD5 and CD20 double positive cells were consistently detected at 6 and 12 weeks in the peripheral blood of all 4 mice examined. Mice were not visibly ill at 20 weeks but had gross splenomegaly at sacrifice. No lymph node or abdominal masses were found. A clear human CD5/CD20 population was found on flow cytometry of bone marrow cells. The splenic architecture was disrupted in mice that engrafted, compared to those that did not and a heavy infiltration of CD20 and cyclin D1 positive cells was found with proliferation estimated at 35–40% by Ki-67 staining. Interphase FISH on fresh cells derived from the spleen showed the IGH/CCND1 [t(11;14)] rearrangement in all cells examined. Scattered CD20 positive cells were observed in the liver but no polyps or submucosal infiltration was found in the ileo-caecal regions of these mice. NSG mice injected with samples 2 and 3 had no evidence of engraftment in peripheral blood, bone marrow or other tissues. Conclusion: Our studies demonstrate that a mouse model of human MCL can be established in NSG mice and are encouraging for developing this model for pre-clinical evaluation of novel drugs. The lymphoma cells that engrafted were from a patient with relapsed, blastoid MCL suggesting that a more aggressive phenotype may favor engraftment as seen with AML xenografts in NSG mice. Ongoing studies are examining additional patient samples and the need for T cell depletion. This model will also provide an opportunity to investigate the role of tumor-initiating side populations in this disease. Disclosures: Gribben: Celgene: Honoraria; Roche: Honoraria; Pharmacyclics: Honoraria; GSK: Honoraria; Mundipharma: Honoraria; Gilead: Honoraria.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5720-5720 ◽  
Author(s):  
Eric Sanchez ◽  
Mingjie Li ◽  
Abigail Gillespie ◽  
Nika M Harutyunyan ◽  
Gigi Garzio ◽  
...  

Abstract Introduction: We previously demonstrated that when carfilzomib (CFZ) was administered first to SCID mice bearing our human multiple myeloma (MM) xenografts, followed by the addition of melphalan (MEL) at the time of disease progression, a modest further reduction in tumor size was observed compared with continuing CFZ or adding MEL alone. In addition, substitution of single agent bortezomib (BOR) for CFZ showed no anti-MM effects. However, when MEL was administered first and CFZ was added after disease progression, mice treated with the combination showed a reduction in tumor volume compared with mice that discontinued MEL. Mice treated with CFZ alone showed no reduction in tumor size. Throughout the study, there was a trend toward smaller tumors among mice in which MEL was followed by the addition of CFZ when compared to mice in which MEL was continued or discontinued and treated with single-agent CFZ or vehicle alone. In the current study, we evaluated the anti-MM effects of CFZ, MEL and BOR for severe combined immunodeficient (SCID) mice progressing from BOR+MEL treatment using our human MM xenograft model LAGk-1A. Methods: Each SCID mouse was surgically implanted with a 20 – 40 mm3 LAGk-1A tumor piece into the left hind limb muscle. Seven days post-implantation mice were randomized into treatment groups based on human immunoglobulin (Ig) G levels. Carfilzomib stock solution (2 mg/ml) was diluted to 3 mg/kg using 5% dextrose and administered twice weekly on two consecutive days (Sundays and Mondays) via intravenous (i.v.) injection.Bortezomib stock solution (1 mg/ml) was diluted to 0.25 mg/kg using NaCl and administered twice weekly (Sundays and Tuesdays) via i.v. injection.MEL stock solution (3 mg/ml) was diluted to 1 mg/kg using PBS and administered once weekly (Fridays) via intraperitoneal injection. Mice (n = 10/group) were initially treated with the combination of BOR + MEL until tumor progression. Progression was defined as an increase in paraprotein > 25% confirmed on one consecutive assessment. After tumor progression, mice initially treated with BOR + MEL were randomized to continue to receive BOR + MEL, receive MEL only (discontinue BOR), receive BOR only (discontinue MEL), substitute BOR with CFZ, discontinue BOR + MEL and add in CFZ alone, or discontinue treatment altogether. Tumor size was measured using standard calipers and human IgG levels with an ELISA (Bethyl Laboratories, Montgomery, TX). This study was conducted according to protocols approved by the Institutional Animal Care and Use Committee. Results: Following progression from the combination of BOR + MEL among mice bearing LAGk-1A, substitution of these drugs with single agent CFZ alone did not produce a reduction in tumor volume when compared with mice continuing to receive BOR + MEL. However, significantly smaller tumors were observed when mice progressing from BOR + MEL were switched to CFZ + MEL compared with mice continued on BOR alone (P = 0.0044) or animals switched to CFZ alone (P = 0.05). There was a trend throughout the study toward smaller tumors in mice receiving CFZ + MEL when compared to mice continuing on BOR + MEL, receiving BOR, CFZ or vehicle alone or discontinuing BOR + MEL. Following progression from BOR + MEL, similar anti-MM effects were observed on human IgG (paraprotein) levels among animals treated with CFZ + MEL when compared to mice continuing BOR + MEL, receiving BOR alone switching to CFZ alone or discontinuing both BOR and MEL. Conclusions: These in vivo studies using our human MM xenograft model, LAGk-1A, show that SCID mice progressing from initial BOR + MEL treatment show a reduction in MM tumor burden when BOR is replaced with CFZ at the time of disease progression, and these tumors are significantly smaller than among mice continued on BOR + MEL. These studies demonstrate that once tumors progress from BOR + MEL treatment, CFZ can replace BOR and produce anti-tumor effects. A recent clinical trial confirms our preclinical findings and shows that most MM patients refractory to BOR in combination with other agents, including MEL, will respond to the substitution of CFZ for BOR (Berenson et al., Leukemia 2014). Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1992 ◽  
Vol 79 (7) ◽  
pp. 1775-1780 ◽  
Author(s):  
RJ Kreitman ◽  
CB Siegall ◽  
DJ FitzGerald ◽  
J Epstein ◽  
B Barlogie ◽  
...  

Abstract IL-6-PE4E is a recombinant protein consisting of interleukin-6 (IL-6) fused to a mutant form of Pseudomonas exotoxin in which four basic amino acids are changed to glutamate (PE4E). The chimeric toxin has been previously shown to specifically kill malignant hepatic, prostatic, epidermoid, and myeloma cell lines in vitro. To explore the possible clinical utility of IL-6-PE4E, particularly as an agent for ex vivo purging of marrow for autologous bone marrow transplantation (ABMT), we tested malignant cells from patients with multiple myeloma for sensitivity to this chimeric toxin. Ficoll-purified bone marrow cells were incubated with and without IL-6-toxin for 2 to 3 days. Eight of the 15 myeloma patients had cells that were sensitive to IL-6-toxin as measured by a decrease in the level of protein synthesis. Cells from five patients were very sensitive to IL-6-PE4E, with 50% inhibition of protein synthesis (ID50) achieved at or below 6 ng/mL (7 x 10(-11) mol/L). Cells from three additional patients showed moderate sensitivity, with ID50s between 30 and 140 ng/mL. The remaining seven samples showed little or no sensitivity, with ID50s greater than or equal to 400 ng/mL. Normal bone marrow cells or normal BFU-E and CFU-GM were resistant to the IL-6-toxin even at 1,000 ng/mL. Neither IL-6, IL- 2-PE4E, nor an enzymatically deficient mutant of IL-6-PE4E was cytotoxic toward the myeloma cells, indicating that the cytotoxic effect of IL-6-PE4E required the adenosine diphosphate-ribosylation function as well as the specific ligand. Our data suggest that IL-6- toxin could be effective in ex vivo marrow purging in selected multiple myeloma patients who are candidates for ABMT, and that this toxin should also be investigated further for in vivo therapy.


Blood ◽  
1992 ◽  
Vol 79 (7) ◽  
pp. 1775-1780 ◽  
Author(s):  
RJ Kreitman ◽  
CB Siegall ◽  
DJ FitzGerald ◽  
J Epstein ◽  
B Barlogie ◽  
...  

IL-6-PE4E is a recombinant protein consisting of interleukin-6 (IL-6) fused to a mutant form of Pseudomonas exotoxin in which four basic amino acids are changed to glutamate (PE4E). The chimeric toxin has been previously shown to specifically kill malignant hepatic, prostatic, epidermoid, and myeloma cell lines in vitro. To explore the possible clinical utility of IL-6-PE4E, particularly as an agent for ex vivo purging of marrow for autologous bone marrow transplantation (ABMT), we tested malignant cells from patients with multiple myeloma for sensitivity to this chimeric toxin. Ficoll-purified bone marrow cells were incubated with and without IL-6-toxin for 2 to 3 days. Eight of the 15 myeloma patients had cells that were sensitive to IL-6-toxin as measured by a decrease in the level of protein synthesis. Cells from five patients were very sensitive to IL-6-PE4E, with 50% inhibition of protein synthesis (ID50) achieved at or below 6 ng/mL (7 x 10(-11) mol/L). Cells from three additional patients showed moderate sensitivity, with ID50s between 30 and 140 ng/mL. The remaining seven samples showed little or no sensitivity, with ID50s greater than or equal to 400 ng/mL. Normal bone marrow cells or normal BFU-E and CFU-GM were resistant to the IL-6-toxin even at 1,000 ng/mL. Neither IL-6, IL- 2-PE4E, nor an enzymatically deficient mutant of IL-6-PE4E was cytotoxic toward the myeloma cells, indicating that the cytotoxic effect of IL-6-PE4E required the adenosine diphosphate-ribosylation function as well as the specific ligand. Our data suggest that IL-6- toxin could be effective in ex vivo marrow purging in selected multiple myeloma patients who are candidates for ABMT, and that this toxin should also be investigated further for in vivo therapy.


Blood ◽  
2005 ◽  
Vol 106 (2) ◽  
pp. 713-716 ◽  
Author(s):  
Pierfrancesco Tassone ◽  
Paola Neri ◽  
Daniel R. Carrasco ◽  
Renate Burger ◽  
Victor S. Goldmacher ◽  
...  

Abstract We developed a novel in vivo multiple myeloma (MM) model by engrafting the interleukin 6 (IL-6)-dependent human MM cell line INA-6 into severe combined immunodeficiency (SCID) mice previously given implants of a human fetal bone chip (SCID-hu mice). INA-6 cells require either exogenous human IL-6 (huIL-6) or bone marrow stromal cells (BMSCs) to proliferate in vitro. In this model, we monitored the in vivo growth of INA-6 cells stably transduced with a green fluorescent protein (GFP) gene (INA-6GFP+ cells). INA-6 MM cells engrafted in SCID-hu mice but not in SCID mice that had not been given implants of human fetal bone. The level of soluble human IL-6 receptor (shuIL-6R) in murine serum and fluorescence imaging of host animals were sensitive indicators of tumor growth. Dexamethasone as well as experimental drugs, such as Atiprimod and B-B4-DM1, were used to confirm the utility of the model for evaluation of anti-MM agents. We report that this model is highly reproducible and allows for evaluation of investigational drugs targeting IL-6-dependent MM cells in the human bone marrow (huBM) milieu. (Blood. 2005;106:713-716)


Blood ◽  
2010 ◽  
Vol 116 (6) ◽  
pp. 900-908 ◽  
Author(s):  
Sheng Zhou ◽  
Disha Mody ◽  
Suk See DeRavin ◽  
Julia Hauer ◽  
Taihe Lu ◽  
...  

Abstract To develop safer and more effective vectors for gene therapy of X-linked severe combined immunodeficiency (SCID-X1), we have evaluated new self-inactivating lentiviral vectors based on the HIV virus. The CL20i4-hγc-Revgen vector contains the entire human common γ chain (γc) genomic sequence driven by the γc promoter. The CL20i4-EF1α-hγcOPT vector uses a promoter fragment from the eukaryotic elongation factor alpha (EF1α) gene to express a codon-optimized human γc cDNA. Both vectors contain a 400-bp insulator fragment from the chicken β-globin locus within the self-inactivating long-terminal repeat. Transduction of bone marrow cells using either of these vectors restored T, B, and natural killer lymphocyte development and function in a mouse SCID-X1 transplantation model. Transduction of human CD34+ bone marrow cells from SCID-X1 patients with either vector restored T-cell development in an in vitro assay. In safety studies using a Jurkat LMO2 activation assay, only the CL20i4-EF1α-hγcOPT vector lacked the ability to transactivate LMO2 protein expression, whereas the CL20i4-hγc-Revgen vector significantly activated LMO2 protein expression. In addition, the CL20i4-EF1α-hγcOPT vector has not caused any tumors in transplanted mice. We conclude that the CL20i4-EF1α-hγcOPT vector may be suitable for testing in a clinical trial based on these preclinical demonstrations of efficacy and safety.


Blood ◽  
2013 ◽  
Vol 121 (12) ◽  
pp. e90-e97 ◽  
Author(s):  
Mark Wunderlich ◽  
Benjamin Mizukawa ◽  
Fu-Sheng Chou ◽  
Christina Sexton ◽  
Mahesh Shrestha ◽  
...  

Key Points A relevant xenograft chemotherapy model was developed by using standard AML induction therapy drugs and primary human AML patient samples. Human AML cells show significantly increased sensitivity to in vivo chemotherapy treatment compared with murine LSK and total bone marrow cells.


2019 ◽  
Vol 508 (4) ◽  
pp. 1175-1181 ◽  
Author(s):  
Jessica L. Berlier ◽  
Malini Rethnam ◽  
A'Qilah Banu Binte Abdul Majeed ◽  
Toshio Suda

Blood ◽  
1990 ◽  
Vol 75 (12) ◽  
pp. 2286-2291 ◽  
Author(s):  
K Koike ◽  
T Nakahata ◽  
T Kubo ◽  
T Kikuchi ◽  
M Takagi ◽  
...  

We investigated the effect of interleukin-6 (IL-6) on murine megakaryocytopoiesis in a serum-free culture system. The addition of IL- 6 to a culture containing interleukin-3 (IL-3) resulted in a significant increase in the number of megakaryocyte colonies by bone marrow cells of normal mice. The megakaryocytic progenitors that survive exposure to 5-fluorouracil (5-FU) exhibited a more significant response to IL-6 and IL-3. Polyclonal anti-IL-6 antibody neutralized the stimulatory effect of IL-6 on megakaryocyte colony growth supported by IL-3. Delayed addition experiments and replating experiments of blast cell colonies showed that megakaryocytic progenitors are supported by IL-3 in the early stage of the development but require IL- 6 for their subsequent proliferation and differentiation. In addition, IL-6 increased the size of megakaryocytes in granulocyte-macrophage- megakaryocyte colonies. The combination of granulocyte colony- stimulating factor or granulocyte-macrophage colony stimulating factor with IL-3 resulted in an increase in the granulocyte-macrophage colony growth of bone marrow cells of 5-FU-treated mice or normal mice, respectively, but had little effect on the enhancement of pure and mixed megakaryocyte colony growth. These results suggest that IL-6 plays an important role in murine megakaryocytopoiesis.


Sign in / Sign up

Export Citation Format

Share Document