scholarly journals Immunotherapy Using Dendritic Cells against Multiple Myeloma: How to Improve?

2012 ◽  
Vol 2012 ◽  
pp. 1-13 ◽  
Author(s):  
Thanh-Nhan Nguyen-Pham ◽  
Yoon-Kyung Lee ◽  
Hyeoung-Joon Kim ◽  
Je-Jung Lee

Multiple myeloma (MM) is a good target disease in which one can apply cellular immunotherapy, which is based on the graft-versus-myeloma effect. This role of immune effector cells provides the framework for the development of immune-based therapeutic options that use antigen-presenting cells (APCs) with increased potency, such as dendritic cells (DCs), in MM. Current isolated idiotype (Id), myeloma cell lysates, myeloma dying cells, DC-myeloma hybrids, or DC transfected with tumor-derived RNA has been used for immunotherapy with DCs. Immunological inhibitory cytokines, such as TGF-β, IL-10, IL-6 and VEGF, which are produced from myeloma cells, can modulate antitumor host immune response, including the abrogation of DC function, by constitutive activation of STAT3. Therefore, even the immune responses have been observed in clinical trials, the clinical response was rarely improved following DC vaccinations in MM patients. We are going to discuss how to improve the efficacy of DC vaccination in MM.

Blood ◽  
1997 ◽  
Vol 90 (11) ◽  
pp. 4266-4270 ◽  
Author(s):  
Terry H. Landowski ◽  
Ning Qu ◽  
Ibrahim Buyuksal ◽  
Jeffrey S. Painter ◽  
William S. Dalton

Programmed cell death, or apoptosis, is well documented as a physiological means of eliminating activated lymphocytes and maintaining immune homeostasis. Apoptosis has also been implicated in the targeting of tumor cells by cytotoxic T lymphocytes and natural killer cells. One of the two primary mechanisms used in cell-mediated cytotoxicity is the Fas/FasLigand system. Activated or transformed cells expressing the Fas antigen on their surface are susceptible to killing by immune effector cells that express the Fas ligand. Many neoplastic cells, including those derived from patients with multiple myeloma, express Fas antigen on their surface, but do not undergo apoptosis in response to antigen crosslinking. One possibility for the lack of Fas-mediated apoptosis includes mutations in the Fas antigen. Loss of function mutations in the Fas antigen have been associated with congenital autoimmune disease in humans, and have been defined as the genetic defect the in lpr mice. Mutations in the Fas antigen have not been previously described in cancer patients. In this study, we show that mutations occur in the Fas antigen which may cause loss of function and contribute to the pathogenesis of the neoplastic disease, multiple myeloma. Using reverse transcriptase-polymerase chain reaction (RT-PCR), single-stranded conformation polymorphism (SSCP) analysis, and DNA sequencing, we examined the cDNA structure of the Fas antigen in 54 bone marrow (BM) specimens obtained from myeloma patients. Six patient specimens (11%) did not express detectable levels of Fas antigen mRNA. Of the 48 BM specimens which did express Fas antigen, 5 (10%) displayed point mutations. All of the mutations identified were located in the cytoplasmic region of the Fas antigen known to be involved in transduction of an apoptotic signal. Two separate individuals demonstrated an identical mutation at a site previously shown to be mutated in the congenital autoimmune syndrome, ALPS. One patient exhibited a point mutation at a site only two amino acids removed from the documented lesion of the lprcg mouse. Although the functional status of these point mutations remains to be determined, we propose that Fas antigen mutations may contribute to the pathogenesis and progression of myeloma in some patients.


2012 ◽  
Vol 2012 ◽  
pp. 1-28 ◽  
Author(s):  
Ivetta Danylesko ◽  
Katia Beider ◽  
Avichai Shimoni ◽  
Arnon Nagler

Multiple myeloma (MM) is a life-threatening haematological malignancy for which standard therapy is inadequate. Autologous stem cell transplantation is a relatively effective treatment, but residual malignant sites may cause relapse. Allogeneic transplantation may result in durable responses due to antitumour immunity mediated by donor lymphocytes. However, morbidity and mortality related to graft-versus-host disease remain a challenge. Recent advances in understanding the interaction between the immune system of the patient and the malignant cells are influencing the design of clinically more efficient study protocols for MM. Cellular immunotherapy using specific antigen-presenting cells (APCs), to overcome aspects of immune incompetence in MM patients, has received great attention, and numerous clinical trials have evaluated the potential for dendritic cell (DC) vaccines as a novel immunotherapeutic approach. This paper will summarize the data investigating aspects of immunity concerning MM, immunotherapy for patients with MM, and strategies, on the way, to target the plasma cell more selectively. We also include the MM antigens and their specific antibodies that are of potential use for MM humoral immunotherapy, because they have demonstrated the most promising preclinical results.


Blood ◽  
1997 ◽  
Vol 90 (11) ◽  
pp. 4266-4270 ◽  
Author(s):  
Terry H. Landowski ◽  
Ning Qu ◽  
Ibrahim Buyuksal ◽  
Jeffrey S. Painter ◽  
William S. Dalton

Abstract Programmed cell death, or apoptosis, is well documented as a physiological means of eliminating activated lymphocytes and maintaining immune homeostasis. Apoptosis has also been implicated in the targeting of tumor cells by cytotoxic T lymphocytes and natural killer cells. One of the two primary mechanisms used in cell-mediated cytotoxicity is the Fas/FasLigand system. Activated or transformed cells expressing the Fas antigen on their surface are susceptible to killing by immune effector cells that express the Fas ligand. Many neoplastic cells, including those derived from patients with multiple myeloma, express Fas antigen on their surface, but do not undergo apoptosis in response to antigen crosslinking. One possibility for the lack of Fas-mediated apoptosis includes mutations in the Fas antigen. Loss of function mutations in the Fas antigen have been associated with congenital autoimmune disease in humans, and have been defined as the genetic defect the in lpr mice. Mutations in the Fas antigen have not been previously described in cancer patients. In this study, we show that mutations occur in the Fas antigen which may cause loss of function and contribute to the pathogenesis of the neoplastic disease, multiple myeloma. Using reverse transcriptase-polymerase chain reaction (RT-PCR), single-stranded conformation polymorphism (SSCP) analysis, and DNA sequencing, we examined the cDNA structure of the Fas antigen in 54 bone marrow (BM) specimens obtained from myeloma patients. Six patient specimens (11%) did not express detectable levels of Fas antigen mRNA. Of the 48 BM specimens which did express Fas antigen, 5 (10%) displayed point mutations. All of the mutations identified were located in the cytoplasmic region of the Fas antigen known to be involved in transduction of an apoptotic signal. Two separate individuals demonstrated an identical mutation at a site previously shown to be mutated in the congenital autoimmune syndrome, ALPS. One patient exhibited a point mutation at a site only two amino acids removed from the documented lesion of the lprcg mouse. Although the functional status of these point mutations remains to be determined, we propose that Fas antigen mutations may contribute to the pathogenesis and progression of myeloma in some patients.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 4891-4891
Author(s):  
Zachary Herse ◽  
Stella Chang ◽  
Denny Liggitt ◽  
Jeffery Fairman ◽  
David F. Claxton

Abstract Cationic Lipid and DNA Complex (CLDC) is a chemically defined preparation of lipids and non-coding bacterial plasmid DNA able to activate antigen presenting and immune effector cells and thereby enhance cellular immune responses. We tested this preparation subcutaneously (SC) in two models of transplantable murine leukemia: 32D -(bcr-abl)p210 (leukemogenic in C3H/HEJ animals) and WEHI-3B cells (leukemogenic in BALB-C mice.) In multiple experiments using the 32Dp210 model a single SC dose of CLDC (200 ul) prevented or significantly delayed death from leukemia when delivered between -1 and 16 days from leukemic challenge. Specifically, control animals died at 28 days (SD=2.73) of leukemia whereas 20/24 similarly leukemia challenged but CLDC treated animals were alive >71 days (p<0.0001 − 4/20 animals died of leukemia). Animals dying of leukemia had swollen spleens and leukemic infiltration of spleen and liver on histopathological evaluation. Similarly Balb-C animals challenged with WEHI-3B cells died at a median of 30 days unless treated with CLDC (1 dose day 6 − 4/5 alive >day 40 p=0.0017). A green fluorescence protein (GFP) expressing subclone of 32Dp210 allowed quantitation of leukemia in peripheral blood (PB). PB leukocytes were 20% GFP positive on day 19. After CLDC on day 19, day 21 PB showed only 2% GFP. Animals receiving 32D-p210-GFP and treated with CLDC and CD8 antibody on day 19 showed higher GFP in followup and died earlier (median day 35) than animals treated with CLDC alone (>42 days) but later than animals receiving no treatment after leukemic challenge (median 23 days, p=0.0002.) See Figure below. We conclude that CLDC shows potent anti-leukemia activity in two murine models of AML. Antibody experiments suggest that CD8 positive effector cells contribute to this effect. Additional experiments are maturing and will be reported. This preparation has promise for activity in human acute leukemias. Figure Figure


Blood ◽  
2000 ◽  
Vol 96 (13) ◽  
pp. 4300-4306 ◽  
Author(s):  
Patrizia Rovere ◽  
Giuseppe Peri ◽  
Fausto Fazzini ◽  
Barbara Bottazzi ◽  
Andrea Doni ◽  
...  

Pentraxins are acute-phase proteins produced in vivo during inflammatory reactions. Classical short pentraxins, C-reactive protein, and serum amyloid P component are generated in the liver in response to interleukin (IL)–6. The long pentraxin PTX3 is produced in tissues under the control of primary proinflammatory signals, such as lipopolysaccharide, IL-1β, and tumor necrosis factor-α, which also promote maturation of dendritic cells (DCs). Cell death commonly occurs during inflammatory reactions. In this study, it is shown that PTX3 specifically binds to dying cells. The binding was dose dependent and saturable. Recognition was restricted to extranuclear membrane domains and to a chronological window after UV irradiation or after CD95 cross-linking–induced or spontaneous cell death in vitro. PTX3 bound to necrotic cells to a lesser extent. Human DCs failed to internalize dying cells in the presence of PTX3, while they took up normally soluble or inert particulate substrates. These results suggest that PTX3 sequesters cell remnants from antigen-presenting cells, possibly contributing to preventing the onset of autoimmune reactions in inflamed tissues.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 5617-5617 ◽  
Author(s):  
Tengteng Yu ◽  
Lijie Xing ◽  
Liang Lin ◽  
Jiye Liu ◽  
Kenneth Wen ◽  
...  

Abstract CD138 (Syndecan-1), a member of integral membrane family of heparan sulfate proteoglycans (HSPGS), is highly expressed on differentiated plasma cells (PC) and is both a primary diagnostic biomarker of multiple myeloma (MM) as well as an indicator of poor clinical prognosis. This surface antigen is an attractive candidate for targeted immunotherapy for MM, given its constitutive expression during disease progression, including smoldering myeloma, a relatively early asymptomatic phase of disease that is potentially amenable to early treatment. We here investigated the targeted use of chimeric anti-CD138 monoclonal antibody (mAb) 1610 and confirm its in vitro anti-tumor potency based on an immune directed cellular cytotoxicity against a diverse panel of CD138 positive MM cell lines, both resistant or sensitive to conventional and current MM therapies and varying levels of CD138 expression as measured by cell immunostaining and quantitative RT-PCR. Antibody-dependent cellular cytotoxicity (ADCC) was evaluated using a calcein-AM based release assay in the presence of human natural killer (NK) effector cells purified from four different healthy donors. MAb 1610 lysed CD138-expressing MM cell lines in a dose dependent manner. This ADCC activity was mAb 1610 specific (in comparison to isotype control), CD138 target dependent, and mediated in the presence of human NK effector cells (co-cultured at an effector:target cell ratio of 20:1). MAb 1610 dependent-cytotoxicity was observed at concentrations as low as 0.01 µg/ml with maximal lysis occurring at approximately 1 µg/ml and extrapolated sub-nanomolar ED50 potencies (Table 1) based on these data. All MM cell lines were subject to mAb 1610-mediated lysis, albeit with slightly different sensitivities that modestly correlated with their relative CD138 cell surface expression levels. This anti CD138 mAb-dependent cellular toxicity included MM1SR and H929R cell lines, both of which are resistant to lenalidomide. MAb 1610 induced specific cell lysis of JJN3 cells, but not of CD138 knock out JJN3 cells or CD138-negative B lymphocytes, further confirming that mAb 1610 specifically induced ADCC against-CD138 expressing MM cells in a target specific manner. Using an orthogonal cytometric based assay, the ability of mAb 1610, in a dose-dependent manner, to activate NK cells was also shown in the presence of CD138 target cells, as evidenced by increased expression of CD107 (a marker for NK cell degranulation) and cytokine production in NK cells. Importantly, the CD138 targeting cytotoxic activities of mAb 1610 translationally extend to MM cells autologously derived directly from MM patients with newly diagnosed and relapsed/refractory diseases. The concomitant use of autologously derived effector cells from these patients to mediate antibody dependent myeloma cell killing further suggests the relevance of anti-CD138 directed immune-based therapeutic strategy in humans. In further replication of human disease, we also co-cultured MM1.S or MM1.R cells with human bone marrow stromal cells (BMSCs) which support myeloma cell growth by promoting an immunosuppressive microenvironment within the BM. Importantly, mAb 1610-dependent cytotoxicity against MM1.S or MM1.R cells was not attenuated by the co-presence of BMSCs. Similarly, IL-6 (10 ng/ml) did not significantly affect mAb 1610-induced ADCC activity, indicating a mechanism of action that can overcome growth promotion, immune suppression, and drug resistance conferred by the tumor promoting BM microenvironment. Taken together, these in vitro studies further demonstrate as a proof-of-concept the use of an antibody CD138 targeting strategy mediated through an immune based mechanism of myeloma plasma cell killing. Based on these results, optimization and further biological characterization of chimeric mAb 1610 in advance of pre-clinical studies is anticipated. Disclosures Myette: Visterra Inc.: Employment. Chaganty:Visterra Inc.: Employment. Adari:Visterra Inc.: Employment. Tissire:Visterra Inc.: Employment. Deotale:Visterra Inc.: Employment. Shriver:Visterra Inc.: Employment. Munshi:OncoPep: Other: Board of director. Anderson:Millennium Takeda: Consultancy; Gilead: Membership on an entity's Board of Directors or advisory committees; OncoPep: Equity Ownership, Other: Scientific founder; C4 Therapeutics: Equity Ownership, Other: Scientific founder; Celgene: Consultancy; Bristol Myers Squibb: Consultancy.


Blood ◽  
1998 ◽  
Vol 91 (1) ◽  
pp. 196-206 ◽  
Author(s):  
Anja B. Geldhof ◽  
Muriel Moser ◽  
Laurence Lespagnard ◽  
Kris Thielemans ◽  
Patrick De Baetselier

Activation of natural killer (NK) cells in the presence of interleukin-12 (IL-12) augments the capacity of these effector cells to recognize B7-1– and B7-2–expressing target cells. These effector cells also efficiently lyse autologous B7-positive progenitor or organ-derived dendritic cells, suggesting a physiologic regulatory pathway between IL-12, NK cells, and B7-expressing antigen-presenting cells. Although IL-12–activated NK cells secreted higher levels of interferon-γ, this cytokine did not play a role in synergistic effects of IL-12 and B7 on NK activation. The B7-counterreceptor was found to be selectively upregulated on IL-2/IL-12 as compared with IL-2–activated NK cells. CD28 is functionally involved in the recognition of B7 on target cells since IL-2/IL-12–activated NK cells derived from CD28 knockout mice were strongly reduced in their capacity to lyse syngeneic B7-positive tumor cells as well as antigen-presenting cells. However, recognition of B7 on allogeneic targets did not require the expression of CD28 on the IL-2/IL-12–activated NK cells. Hence, IL-12 triggers the expression of both CD28-dependent and CD28-independent mechanisms that allow NK cells to eliminate B7-positive target cells including autologous dendritic cells.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1487-1487 ◽  
Author(s):  
Ali Jalili ◽  
Shuji Ozaki ◽  
Tomoko Hara ◽  
Etsuko Sekimoto ◽  
Yoichi Tanaka ◽  
...  

Abstract HM1.24 is a type II transmembrane glycoprotein that was originally identified as a myeloma cell-specific antigen and expected to be an attractive target molecule for immunotherapy of multiple myeloma (MM). Our previous studies have demonstrated that HM1.24-specific cytotoxic T lymphocytes (CTL) can be induced by the stimulation with dendritic cells (DC) pulsed with HM1.24-derived peptides such as HM1.24-126 (KLQDASAEV) and HM1.24-165 (APQLLIVLL) in normal HLA-A2 and/or A24 individuals. Recent clinical studies have shown that autologous peripheral blood stem cell transplantation (PBSCT) significantly improves the remission rate and overall survival of patients with MM. However, additional therapeutic strategies are necessary to eradicate residual disease. To explore the possibility of HM1.24-targeting cellular immunotherapy after PBSCT, we evaluated the ability of PBSC harvests as a source of DC and investigated the efficacy of these DC to induce HM1.24 peptide-specific CTL. Eight MM patients with HLA-A2+ and/or A24+ type undergoing autologous PBSCT were studied. PBSC were collected after high-dose cyclophosphamide (100 mg/kg) and granulocyte colony-stimulating factor (400 μg/m2/day) mobilization using a COBE Spectra. The median percentage of CD34+ cells in the harvest product was 1.5% (range 0.61–13.0). PBSC were cryopreserved in 5% DMSO and 6% hydroxyethyl starch until use. DC were derived by culture of plastic-adherent mononuclear cells from thawed PBSC in medium containing granulocyte-macrophage colony-stimulating factor and interleukin (IL) -4 for 6 days, and then in the presence of tumor necrosis factor-alpha for 1 day. To generate HM1.24-specific CTL, 1x106 cells of non-adherent fraction from thawed PBSC were stimulated with 1x105 DC pulsed with 10 μg/mL of HM1.24-126 or HM1.24-165 peptides. Cells were stimulated weekly with the peptide-pulsed DC in the presence of IL-2. After two rounds of stimulation, CD8+ cells were enriched using MACS column. EBV-transformed B cells (HLA-A2+, A24+) were used as target cells. Specific recognition of peptide-pulsed target cells by CTL was determined by ELISPOT assay for interferon-gamma. These CTL were also tested for cytotoxic activity using Granzyme B ELISPOT assay. Phenotypically mature DC (CD1a+, CD40+, CD80+, CD83+, CD86+) were generated from all PBSC harvests of MM patients. CD8+ cell expansion was observed in 6 of 8 products. HM1.24-126 and HM1.24-165 peptides successfully induced peptide-specific CTL in 4 of 4 HLA-A2 patients and 2 of 4 HLA-A24 patients, respectively. More importantly, these HM1.24-specific CTL showed cytotoxic activity against myeloma cell lines such as ARH-77 (HLA-A2+) and TSPC-1 (HLA-A24+) in an HLA-restricted and antigen-specific manner. No response was observed against K562 cells, indicating that this cytotoxicity was not mediated by non-specific NK activity. These results suggest the existence of functional DC and HM1.24-specific CTL precursors in PBSC harvests of patients with MM and provide the basis for cellular immunotherapy with these HM1.24-derived peptides using PBSC harvests in combination with PBSCT.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1739-1739
Author(s):  
Martha Q. Lacy ◽  
Sumithra Mandrekar ◽  
Brian Kabat ◽  
Angela Dispenzieri ◽  
Suzanne Hayman ◽  
...  

Abstract Introduction: Immunotherapy is most likely to work in a setting of low tumor burden, making vaccine strategies attractive as maintenance therapy post autologous peripheral blood stem cell transplantation (PBSCT) for multiple myeloma (MM). However, it is difficult to ascertain the effects of the vaccines from delayed responses to the transplant. A novel immunotherapeutic, APC8020 (Mylovenge), was studied as consolidation therapy after for MM post PBSCT (Mayo vaccine trial). Mylovenge is prepared from autologous antigen presenting cells, including dendritic cells, partially purified from an unmobilized leukapheresis product by gradient density isolation and then incubated for two days with autologous serum containing M protein obtained pretransplant. We report long term results of the Mayo vaccine trial compared retrospectively to a consecutive cohort of MM patients who underwent autologous stem cell transplant at Mayo Clinic during the same time period. Methods: The Mayo vaccine trial patients had transplants between July of 1998 and May of 2001. Using these cutoff dates we analyzed 151 total MM patients, 27 from the vaccine trial (9 newly diagnosed and 18 relapsed) and 124 from database (DB) (50 newly diagnosed patients and 74 relapsed). The median (range) of follow-up for alive patients in vaccine trial is 6.5 years (2.9 – 8), and in the DB is 7.1 years (6 – 8). Two-sided Fisher’s exact tests and stratified log rank tests (with newly diagnosed or relapsed as the stratum) were used to compare baseline patient characteristics and time to event distributions (overall survival – OS, progression free survival – PFS, and time to progression – TTP) between the two groups. Results: The median (range) of age in vaccine trial and DB was 56 (30–69) and 57 (36–71) years respectively. There were no significant differences in the known prognostic factors including PCLI, B2M, and CRP. The median (95% confidence interval - CI) TTP for the vaccine trial and DB patients was 1.5 years (1.3 – 2.4) and 1.6 years (1.3 – 1.8); stratified log rank p value = 0.46. The median (95% CI) PFS for the vaccine trial and DB patients was 1.5 years (1.3 – 2.4) and 1.5 years (1.1 – 1.8), stratified logrank p value = 0.30. The median (95% CI) OS for the vaccine trial and DB patients was 5.3 years (4.0 -N/A), and 3.4 years (2.7 – 4.6); stratified logrank p value = 0.02. Conclusions: Despite the fact that no difference was seen in TTP or PFS, post-transplant vaccine therapy was associated with prolonged survival. This is similar to what has been reported in other tumor systems including prostate cancer and glioblastoma multiforme. This approach warrants further investigation, including preclinical studies aimed at optimizing the immune response and randomized trials to define the role of vaccine therapy in myeloma. Figure Figure


Sign in / Sign up

Export Citation Format

Share Document