Influenza (flu) Vaccine Administration to Patients with Multiple Myeloma (MM) Prior to Autologous T-Cell Harvest Leads to Better Reconstitution of Flu Immunity After High Dose Melphalan and Autologous Stem Cell Transplant (ASCT) and Reinfusion of Primed Ex-Vivo Co-Stimulated Autologous T-Cells and Post-ASCT Second Flu Vaccination Than Post-ASCT Flu Vaccination Alone.

Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 797-797 ◽  
Author(s):  
Edward A. Stadtmauer ◽  
Dan T. Vogl ◽  
Nicole A. Aqui ◽  
Aaron P. Rapoport ◽  
Kenyetta Macdonald ◽  
...  

Abstract Abstract 797 Introduction: Epidemics of influenza A virus strains have been associated with a mortality rate of 0.1% and hospitalization of approximately 200,000 people per year in the United States. The recent pandemic of swine-origin H1N1 has further emphasized the need to protect the population. Patients with multiple myeloma (MM) are particularly vulnerable to flu infection from altered humoral and cellular immunity from the disease as well as from the immunosuppressive chemotherapies, corticosteroids and radiation frequently used for treatment. In particular, high dose melphalan and ASCT, a common and efficacious treatment as part of first-line or salvage therapy for MM, is associated with post-ASCT immune suppression by virtue of low T-cell numbers, poor T-cell function and immune paresis. Despite anti-infection vaccination after ASCT, pneumococcal pneumonia and influenza are common. In an attempt to improve this outcome we previously showed that adoptive transfer of in-vivo PCV vaccine-primed and ex-vivo (anti-CD3/anti-CD28) co-stimulated autologous T cells (aT-cells) early (within 14 days) post-ASCT increased CD4 and CD8 T cell counts and induced pneumococcal conjugate vaccine-directed T and B-cell responses [Rapoport et al, Nature Medicine, 2005]. Patients who did not receive pre-collection vaccination did not show this response despite infusion of co-stimulated T-cells and post-ASCT vaccination. In the current clinical trial we investigated whether pre-harvest flu vaccination would result in a similar improvement in immunity. Methods: We performed a randomized open-label single center study of two influenza vaccine schedules for patients with high-risk MM responding after conventional induction therapy. Of 21 patients enrolled, 11 were randomly assigned to Group X and received pre- and d+14 post-transplant flu vaccine with the commercially available influenza vaccine (Fluzone) produced by Aventis Pasteur. The 10 patients in Group Y received only post-transplant vaccination. All patients received T-cell harvest, high dose melphalan and ASCT and aT-cell infusion. Immunological assessment (T-cell and B-cell subsets, T-cell repertoire, T-ELISPOT, CFSE-CBC, antibody HAI, B-ELISPOT) was conducted d+60, 100 and 180. The primary study endpoint is the immune response to influenza as measured by the serotype-specific influenza antibody response on day 100 using a standard hemagglutination inhibition assay (HAI) optimized for the vaccine administered each year. Results: The median age was 55 (range 37-68), 13 M, 8 F, 18 Caucasian, 2 African American, 1 Hispanic, IgG 57%, IgA 33%, light chain 10%. With a median follow-up of 1 year, 86% (18/21) patients are alive with 48% (10/21) alive in remission. No difference in these parameters was seen between both Groups. HAI titers, however were higher at all three time points (d+60, 100, 180) for Group X when compared to Group Y both for H3N2 (p= 0.04) and for H1N1 (p=0.07). HAI titers for Group Y remained near baseline throughout all time points while Group X peaked day 60 and remained elevated day 180. Analysis of other immune assessment is ongoing and will be presented. Conclusion: Preliminary analysis of this randomized trial of flu vaccine schedule for MM patients undergoing ASCT suggests superior flu immune reconstitution when vaccine is administered to prime autologous T-cells prior ASCT and aT-cell infusion. This result adds to our observation that an approach of vaccine priming and co-stimulated autologous T-cell infusion after high dose melphalan and ASCT for MM augments an otherwise poor response in this patient population to pneumococcal and influenza vaccination. Investigation of this approach as a platform for anti-myeloma immune therapy is ongoing. Disclosures: June: University of Pennsylvania: Patents & Royalties.

2020 ◽  
Vol 4 (17) ◽  
pp. 4195-4207
Author(s):  
Shih-Feng Cho ◽  
Liang Lin ◽  
Lijie Xing ◽  
Yuyin Li ◽  
Kenneth Wen ◽  
...  

Abstract We investigated here the novel immunomodulation and anti–multiple myeloma (MM) function of T cells engaged by the bispecific T-cell engager molecule AMG 701, and further examined the impact of AMG 701 in combination with immunomodulatory drugs (IMiDs; lenalidomide and pomalidomide). AMG 701 potently induced T-cell–dependent cellular cytotoxicity (TDCC) against MM cells expressing B-cell maturation antigen, including autologous cells from patients with relapsed and refractory MM (RRMM) (half maximal effective concentration, <46.6 pM). Besides inducing T-cell proliferation and cytolytic activity, AMG 701 also promoted differentiation of patient T cells to central memory, effector memory, and stem cell–like memory (scm) phenotypes, more so in CD8 vs CD4 T subsets, resulting in increased CD8/CD4 ratios in 7-day ex vivo cocultures. IMiDs and AMG 701 synergistically induced TDCC against MM cell lines and autologous RRMM patient cells, even in the presence of immunosuppressive bone marrow stromal cells or osteoclasts. IMiDs further upregulated AMG 701–induced patient T-cell differentiation toward memory phenotypes, associated with increased CD8/CD4 ratios, increased Tscm, and decreased interleukin 10–positive T and T regulatory cells (CD25highFOXP3high), which may downregulate T effector cells. Importantly, the combination of AMG 701 with lenalidomide induced sustained inhibition of MM cell growth in SCID mice reconstituted with human T cells; tumor regrowth was eventually observed in cohorts treated with either agent alone (P < .001). These results strongly support AMG 701 clinical studies as monotherapy in patients with RRMM (NCT03287908) and the combination with IMiDs to improve patient outcomes in MM.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2410-2410
Author(s):  
James R. Berenson ◽  
Ivan M. Borrello ◽  
Ravi Vij ◽  
Asad Bashey ◽  
Thomas Martin ◽  
...  

Abstract Background: T cells from myeloma subjects can be activated and expanded ex vivo using the Xcellerate™ Process, in which peripheral blood mononuclear cells are incubated with anti-CD3 and anti-CD28 antibody-coated magnetic beads (Xcyte™-Dynabeads®). In a previous study (Borrello et al., ASCO 2004), Xcellerated T Cells administered to myeloma subjects following high dose chemotherapy and autologous stem cell transplantation led to accelerated lymphocyte recovery and restoration of the T cell receptor repertoire. In the current study, subjects with relapsed or refractory myeloma were randomized to Xcellerated T Cells with or without one cycle of fludarabine prior to Xcellerated T Cells. Fludarabine is being used to assess the influence of lymphoablation on the anti-tumor and immune reconstitution effects of T cell therapy; it has previously been reported to have no significant activty in myeloma (Kraut et al., Invest. New Drugs, 1990). Methods: Approximately 30 subjects are planned to receive treatment. Each receives a single dose of 60–100 x 109 Xcellerated T Cells. Subjects on the fludarabine arm receive a single cycle (5 days at 25 mg/m2), completed 4 days prior to the Xcellerated T Cell infusion. Results: 17 subjects have been enrolled and 13 treated to date, with median last f/u visit of 28 days (range 0–140). Xcellerated T Cells were successfully manufactured in all subjects, with T cell expansion 136 ± 61 fold (mean ± SD), with 79.2 ± 13.8 x 109 cells infused, and final product 98.0 ± 2.0% T cells (n=13). There have been no reported serious adverse events related to Xcellerated T Cells. In the fludarabine arm, lymphocytes decreased from 1,228 ± 290/mm3 (mean ± SEM) to 402 ± 164 following fludarabine, and then increased to 1,772 ± 278 on Day 14 following T cell infusion (n=7). In the non-fludarabine arm, lymphocyte counts increased from 1,186 ± 252 to 3,204 ± 545 on Day 14 (n=4). Lymphocytes were comprised of both CD4+ and CD8+ T cells. Increases were observed in NK cells from 77 ± 26 to 121 ± 25, monocytes from 166 ± 44 to 220 ± 30 and platelets from 218 ± 16 to 235 ± 24 by Day 14 (n=11). In the non-fludarabine arm, neutrophils increased from 3.6 ± 0.9 to 4.8 ± 0.6 on Day 1. On the fludarabine arm, 3 of 6 subjects developed Grade 4 neutropenia and one developed Grade 3 thrombocytopenia. Seven subjects were evaluable for serum M-protein measurements to Day 28. One of three fludarabine treated subjects had an M-protein decrease of 38%. Conclusions: Xcellerated T Cells were well-tolerated and led to increased lymphocytes, including T cells and NK cells. Increases in other hematologic parameters, including neutrophils and platelets were also observed. In this patient population, fludarabine is lymphoablative and also can cause neutropenia and thrombocytopenia. The fludarabine schedule has been decreased from 5 to 3 days. A decrease in M-protein has been observed in one of three fludarabine-treated subjects; data on additional subjects will be presented.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3671-3671
Author(s):  
Jochen Greiner ◽  
Susanne Hofmann ◽  
Krzysztof Giannopoulos ◽  
Markus Rojewski ◽  
Anna Babiak ◽  
...  

Abstract Abstract 3671 Poster Board III-607 For effective elimination of malignant cells by specific T cells a co-activation of CD4- and CD8-positive T cells might be important. We performed two RHAMM-R3 peptide vaccination trials using 300μg and 1000μg for patients with AML, MDS and multiple myeloma overexpressing RHAMM. Similar mild toxicity of both cohorts was found, only mild drug-related adverse events were observed such as erythema and induration of the skin. In the 300μg cohort we detected in 7/10 (70 %) patients specific immune responses and also positive clinical effects in 5/10 (50 %) patients. In the high dose peptide vaccination trial (1000μg peptide) 4/9 (44 %) patients showed positive immune responses. These patients showed an increase of CD8+RHAMM-R3 tetramer+/CD45RA+/CCR7-/CD27-/CD28- effector T cells and an increase of R3-specific CD8+ T cells. In the higher peptide dose cohort three patients showed positive clinical effects. However, higher doses of peptide do not improve the frequency and intensity of immune responses in this clinical trial and might induce immune tolerance. In this work, we investigated the co-existence of serological immune responses against RHAMM detected by a RHAMM-specific ELISA of patients with AML, MDS and multiple myeloma treated in these two peptide vaccination trials. We correlated these results to specific T cell responses of CD8-positive T cells measured by ELISpot assays for interferon gamma and Granzyme B, tetramer staining and chromium release assays. Moreover, these results were compared to the frequency of regulatory T cells. 4/19 patients have a positive serological immune response in ELISA assay, all of these patients developed also strong specific CD8-positive T cell responses during peptide vaccination detected by ELISpot assays and tetramer staining. As expected, peptide vaccination did not result in the induction of humoral immune responses. In further ELISA assays we measured IL-2 and IL-10 levels in the sera of the patients before and three weeks after four vaccinations. While IL-10 levels remained at a rather low level over the time of vaccination, we detected an increase of IL-2 up to the five-fold of the initial levels in four of ten patients. Moreover, we performed a proteome array to detect cytokine and chemokine regulation in sera of patients vaccinated in these two trials during and after RHAMM-R3 peptide vaccination. 36 cytokines, chemokines and acute phase proteins were measured and both cohorts vaccinated with different peptide doses were compared. Taken together, RHAMM-R3 peptide vaccination induced both immunological and clinical responses. Co-existence of immune responses of CD4-positive T cells against the target RHAMM seems to be important for an induction of strong immune responses of CD8-positive T cells. Disclosures: No relevant conflicts of interest to declare.


JCI Insight ◽  
2019 ◽  
Vol 4 (4) ◽  
Author(s):  
Alfred L. Garfall ◽  
Edward A. Stadtmauer ◽  
Wei-Ting Hwang ◽  
Simon F. Lacey ◽  
Jan Joseph Melenhorst ◽  
...  

Cancers ◽  
2020 ◽  
Vol 12 (12) ◽  
pp. 3713
Author(s):  
Christie P. M. Verkleij ◽  
Amy Jhatakia ◽  
Marloes E. C. Broekmans ◽  
Kristine A. Frerichs ◽  
Sonja Zweegman ◽  
...  

The CD38-targeting antibody daratumumab mediates its anti-myeloma activities not only through Fc-receptor-dependent effector mechanisms, but also by its effects on T-cell immunity through depletion of CD38+ regulatory T-cells, regulatory B-cells, and myeloid-derived suppressor cells. Therefore, combining daratumumab with modulators of other potent immune inhibitory pathways, such as the PD-1/PD-L1 axis, may further improve its efficacy. We show that multiple myeloma (MM) cells from relapsed/refractory patients have increased expression of PD-L1, compared to newly diagnosed patients. Furthermore, PD-1 is upregulated on T-cells from both newly diagnosed and relapsed/refractory MM patients, compared to healthy controls. In short-term experiments with bone marrow samples from MM patients, daratumumab-mediated lysis was mainly associated with the MM cells’ CD38 expression levels and the effector (NK-cells/monocytes/T-cells)-to-target ratio, but not with the PD-L1 expression levels or PD-1+ T-cell frequencies. Although PD-1 blockade with nivolumab did not affect MM cell viability or enhanced daratumumab-mediated lysis in short-term ex vivo experiments, nivolumab resulted in a mild but clear increase in T-cell numbers. Moreover, with a longer treatment duration, PD-1 blockade markedly improved anti-CD38 antibody-mediated cytotoxicity in vivo in murine CD38+ tumor models. In conclusion, dual targeting of CD38 and PD-1 may represent a promising strategy for treating MM and other CD38-positive malignancies.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4555-4555
Author(s):  
Thomas Stuebig ◽  
Michael Lioznov ◽  
Ulrike Fritsche-Friedland ◽  
Haefaa Alchalby ◽  
Christine Wolschke ◽  
...  

Abstract Abstract 4555 Introduction: Activating and inhibitory killer immunoglobulin like receptors (KIR) are predominantly expressed on natural killer (NK) cells. KIR mismatch allogeneic stem cell transplantation (alloSCT) has been reported to provide beneficial effects for Multiple Myeloma (MM). However, their recovery in MM patients remains poorly understood. We, therefore, analysed KIR recovery in 90 MM patients after alloSCT. Methods: KIR expression (CD158a/h, CD158b/b2, CD158e1/e2) on NK cells and T cell subsets was measured by flow cytometry at different time points after alloSCT. Results: During the first 90 days after alloSCT NK cells represent the largest lymphocyte subset. Activating receptors like NKp30 and NKp44 showed a fluctuating expression while members of the KIR family were expressed at a constant rate (20% of NK cells). There was no significant difference in the early post transplantation period (day 0–90) compared to later time points (day 360). In contrast, T cells showed increased KIR expression during the first 30 days after alloSCT, which was highly significant for CD158e (p=0,0001). After 30 days the expression declined to baseline. Furthermore, T cell activation marker HLA-DR reached its highest expression between days 60 and 90 when KIR receptors were expressed at their lowest level (27% vs. 8%, p < 0,0001). Conclusions: We conclude that KIR receptors were differentially expressed on NK and T cells. Because KIR receptors are constantly expressed by NK cells and NK cells are the most frequent lymphocyte populations early after alloSCT, NK cells may be useful for KIR mismatch cellular therapy. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 766-766 ◽  
Author(s):  
Aaron P. Rapoport ◽  
Edward A. Stadtmauer ◽  
Dan T. Vogl ◽  
Brendan M Weiss ◽  
Gwendolyn K. Binder-Scholl ◽  
...  

Abstract Background Despite recent therapeutic advances, multiple myeloma (MM) remains primarily an incurable cancer. Patients experiencing rapid recovery of T cells post autologous stem cell transplant (auto-SCT) may have improved outcomes, and spontaneous cellular responses to tumor can occur, suggesting immune mediated control of tumor is possible. We and others have investigated therapeutic cancer vaccines that have shown promise in pilot studies, in particular following post-transplant infusion of activated autologous T cells. However, efficacy of these approaches may be limited by thymic selection which restricts the repertoire of T cell receptors (TCRs) to low affinity TCRs that cannot recognize the low level of antigen present on most tumor cells. We hypothesized that incorporation of affinity-enhanced tumor antigen-specific TCRs into autologous T cells infused post-transplant would overcome this limitation and improve response rates in the post auto-SCT setting. Methods We report interim results of a Phase II clinical trial (NCT01352286) to evaluate the safety and activity of autologous T cells genetically engineered to express an affinity-enhanced TCR that recognizes the NY-ESO-1/LAGE-1 peptide complex HLA‐A*0201‐SLLMWITQC; these cells are infused in the setting of profound lymphodepletion that accompanies high dose chemotherapy administered during auto-SCT. Patients with high risk or relapsed MM, who are HLA‐A*0201 positive, and whose tumor is positive for NY-ESO-1 and/or LAGE-1 by RT-PCR are eligible. CD25 depleted CD4 and CD8 T cells are activated and expanded using anti-CD3/CD28 antibody conjugated microbeads, and genetically modified with a lentiviral vector containing the TCR construct at a multiplicity of infection of 1. Engineered T cells are administered four days after high dose melphalan and two days following auto-SCT, at a dose range of 1-10 billion total cells with a minimum gene modification requirement of 10%. Patients are evaluated for MM responses in accordance with the IMWG criteria at 6 weeks, and 3 and 6 months post infusion. At 3 months, patients start lenalidomide maintenance. The initial 6 patient phase is complete and a 20 patient extension phase is ongoing. Results Prior to enrollment on study, patients had received a median of 3 prior therapies including 6 with prior transplant. 50% of tumors contained high risk chromosomal abnormalities, and NY-ESO-1 expression is correlated with adverse prognosis. 20 patients (average age of 57) have been infused with an average of 2.3 X 109 engineered T cells (range 4.5 X 108-3.9 X 109); this reflects an average clinical scale transduction efficiency of 34% (range 18% – 49%). Infusions have been well tolerated, and the majority of adverse events were related to the high dose melphalan. Possibly related SAEs were neutropenia and thrombocytopenia, and GI and metabolism disorders including diarrhea, colitis, hyponatremia and hypomagnesemia. 10, 4, 2, and 2 patients have reached the 1 year, 9 month, 6 month and 3 month assessment timepoints, respectively, and 17/20 patients are alive. Best response by day 100 is sCR/CR in 2/15 (13%), nCR in 10/15 (67%), and PR in 3/15% (20%), which compares favorably to historic responses in patients undergoing first or second transplant. Engineered T cells expanded and persisted in blood and marrow at 180 days by Q-PCR and flow-cytometry in all but one case (Figure). 7 patients progressed after day 100, which was accompanied either by loss of engineered T cells or loss of tumor antigen. Detailed phenotyping and functional analysis of engineered T cells, and correlates with clinical responses, is underway. Summary This is the first clinical evaluation of engineered T cells in the MM setting. Infusions are safe, well tolerated, and are associated with encouraging responses in a high risk myeloma population. A study evaluating the engineered T cells in a non-transplant study is underway. Disclosures: Stadtmauer: Celgene: Consultancy. Binder-Scholl:Adaptimmune: Employment. Smethurst:Adaptimmune: Employment. Brewer:Adaptimmune: Employment. Bennett:Adaptimmune: Employment. Gerry:Adaptimmune: Employment. Pumphrey:Adaptimmune: Employment. Tayton-Martin:Adaptimmune: Employment. Ribeiro:Adaptimmune: Employment. Levine:Novartis: cell and gene therapy IP, cell and gene therapy IP Patents & Royalties. Jakobsen:Adaptimmune: Employment. Kalos:Novartis corporation: CART19 technology, CART19 technology Patents & Royalties; Adaptive biotechnologies: Member scientific advisory board , Member scientific advisory board Other. June:Novartis: Patents & Royalties, Research Funding.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 812-812 ◽  
Author(s):  
Pawel Muranski ◽  
Sarah I Davies ◽  
Sawa Ito ◽  
Eleftheria Koklanaris ◽  
Jeanine Superata ◽  
...  

Abstract Background: Reactivation of latent viruses post-allogeneic stem cell transplant (SCT) negatively affects outcomes and increases non-relapse mortality. We have reported over 80% early CMV reactivation rate and significant additional costs in recipients of T cell depleted (TCD) SCT. Ex vivo generated multi-virus specific T cells (MVSTs) are effective as a therapy of active infection, but have not been evaluated as prophylaxis early after transplant. In a Phase I study (NIH 14-H-0182) we transferred MVSTs targeting immunodominant viral proteins of CMV, Epstein-Barr virus (EBV), BK and adenovirus (Ad) immediately post SCT as a novel approach to prevent viral reactivation post-SCT. Methods: All subjects were enrolled in HLA-matched T cell depleted (TCD) transplant protocol (NIH 13-H-0144). MVSTs cells were manufactured from SCT sibling donors by stimulation of elutriated lymphocytes for 14 days with seven overlapping peptide libraries (pepmixes) pulsed onto autologous dendritic cells (DCs) in presence of IL-7, IL-15 and IL-2. MVSTs were infused as early as possible (day 0 to +60) post SCT. A Phase I 3+3 dose escalation design was used as follows: Cohort 1 - 1x10e5 total nucleated cells (TNC)/kg, Cohort 2 - 5x10e5 TNC/kg, Cohort 3 - 1x10e6 TNC/kg. Three additional subjects received MVST cells manufactured using pepmix-pulsed mononuclear cells as stimulators (Cohort 3A; 1x10e6TNC/kg) under an amended protocol. The primary safety endpoint at day 42 post infusion was the occurrence of dose limiting toxicity (DLT) defined as Grade IV GVHD or any other severe adverse even (SAE) deemed to be at least "probably" or "definitely" related to the MVST infusion. Patients were followed to day +100 post SCT for secondary outcomes of efficacy, immune reconstitution and GVHD biomarkers (ST2, REG3). CDR3 sequencing (ImmunoSEQ) was performed on selected MVST products and peripheral blood samples post MVST and compared to control SCT recipients. GVHD biomarkers were analyzed pre- and post-treatment. Results: Twelve subjects were treated: nine received MVSTs generated using DCs and three subjects using mononuclear cells (cohort 3A). Median time from SCT to MVST administration was 13 days (range D +2 to +52 post-SCT). Median time to MVST for subjects in Cohort 3A was 3 days (range 2-7). There were no immediate infusion-related adverse events or DLT at the highest dose level. De novo grade II-III aGVHD post-MVST infusion was seen in three subjects (one in Cohort 1 and two in Cohort 3A), but GVHD biomarker elevation predated MVST infusion. CMV reactivation post-MVST occurred in 6 out of 12 subjects (50%) vs. 45 out of 52 patients (50% vs 87%) in a historical group of recipients of T cell depleted SCT. In all cases CMV reactivation occurred in the context of high dose steroids and in two subjects MVSTs were derived from CMV seronegative donors with minimal anti-CMV activity. One subject experienced rapidly rising AdV viremia (asymptomatic) and received an additional infusion of MVSTs. We saw self-limiting low level EBV replication in 8 cases and one BK viremia, but no disease. CDR3 sequencing of MVST products and serial peripheral blood from subjects revealed a robust contribution of ex vivo expanded cells to the overall repertoire, in contrast to untreated controls where the repertoire of (sham) MVST cell products generated from the transplant donors did not significantly overlap with the immune repertoire in peripheral blood of TCD-SCT recipients in the early post SCT period. Only at day +180 some convergence of repertoires became visible indicating spontaneous immune reconstitution (Figure). Detailed CDR3 analysis of cytokine-captured CMV pp65 and IE-1 specific CD4+ and CD8+ T cells was performed in a representative subject clearly demonstrating the detrimental effect of high-dose steroids on frequency of anti-viral T cells, precipitating CMV reactivation. Conclusions: This is the first report demonstrating safety and feasibility of using MVST immediately post-SCT to rapidly reconstitute anti-viral immunity and ameliorate the detrimental impact of the early viral reactivation in SCT recipient. No DLTs were seen and a minimal risk of aGVHD was observed, as there was no correlation with GVHD biomarkers. As revealed by serial CDR3 sequencing, MVSTs robustly contributed to the T cell repertoire. Our results suggest efficacy of this strategy in reducing viral reactivation. A Phase II study is warranted. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1725-1725
Author(s):  
Min Wang ◽  
Eleftherios Papoutsakis

Abstract Introduction. Ex vivo expanded T cells are increasingly employed in a variety of immunotherapy protocols. The success of such protocols requires large numbers of biologically active T cells. Several activation and culture protocols have been examined, but none in significant or molecular detail to allow more targeted protocol interventions. In this study, our aim was to better understand early T-cell activation using a comprehensive, microarray-based transcriptional analysis, in conjunction with targeted protein-level and functional analyses. Methods. Three biological experiments were transcriptionally analyzed. T cells were activated with anti-CD3/anti-CD28 Mab, cultured for 96 hours, and samples collected at 0, 4, 10, 48 and 96 hours. mRNA levels at each time point were compared to that of 0 hour. Genes were selected as follows: a minimum 1.8-fold difference in at least 6 time points of the 12 (3x4) time points. Data were subject to Gene Ontology (GO) analysis, EASE, to discover enriched biological themes. Interferon-γ (IFNG) and CCL20 ELISA assays and NF-κB p65 (active complex) assay by flow cytometry were carried out to confirm select findings of the transcriptional analysis. Results. 2340 genes passed the gene-selection criteria. GO analysis showed that the GO groups Immune Response, Regulation of Apoptosis, and Mitochondrion are among the most over-represented groups among differentially expressed genes during T-cell activation. The upregulated Immune Response group contained many genes coding cytokines typically produced by helper T cells. Highly upregulated genes in this category include IL2, CCL20, CXCL11, CXCL9, CXCL10, CD83, IFNG, CCL3, CSF2, IL23A, XCL2, TNF, CCL4, TNFSF6 and ICOS. Although several of these findings would have been expected, our data include a more extensive gene list and suggest potential use of these cytokines as T-cell ex vivo culture supplements. We singled out CCL20, sharing a very similar transcription pattern with IL2, and IFNG, for further analysis. ELISA assays of IFNG and CCL20 showed that the kinetics of secreted protein levels were in agreement with transcriptional data. In the GO group Regulation of Apoptosis, the downregulated gene cluster was enriched by pro-apoptotic genes, while the upregulated cluster was enriched by anti-apoptotic genes. Our data indicated that NF-κB complex activity changed upon T-cell activation. This was verified by NF-κB p65 Phosflow assay, showing that phosphorylated NF-κB p65 was suppressed up to 10 hours, and then increased until hour 96. Although it is known that mitochondrial hyperpolarization is important in T-cell activation, our transcriptional data suggest more profound changes in mitochondrial biology. Genes in this category include several having oxidoreductase activity, such as SOD2, SCO1, NDUFAB1, GLUD2, consistent with reported increases in intracellular ROS levels upon T-cell activation, and suggest the potential use of anti-oxidants in ex vivo T-cell expansion. Conclusion. Our data and analysis provide a better understanding of T-cell activation and identify several potential targets that can be explored to benefit T-cell expansion protocols for immunotherapy applications. Although there is much known about T-cell activation, modern genomic tools provide an extraordinary opportunity to verify, extend and enrich prior knowledge, and, discover new players and processes not previously associated with T-cell activation.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1530-1530
Author(s):  
Helene Sicard ◽  
Jean-Francois Rossi ◽  
Philippe Rousselot ◽  
Alexis Collette ◽  
Christine Paiva ◽  
...  

Abstract Background: IPH1101, a chemically-synthesized, structural analogue of γδ T lymphocyte natural phosphoantigens, combined with low doses of IL-2 induces a highly selective proliferation of γδ T cells, non conventional lymphocytes bearing potent effector and regulatory immune functions. Today, IPH1101 is tested in Phase II clinical trials in different indications and settings (alone or in combination), as the first specific γδ T cell-mediated immunotherapy. Whereas γδT cells from healthy donors always proliferate in response to IPH1101 + IL-2, cells from cancer patients often present moderate to very strong impaired proliferative capacity. The reasons for this defect are still unclear, but might result from a suppressive effect induced by the tumour itself. Thus, defective γδ T cell responses might be partly disease-dependent. In some cases, the decreased ability to respond to IPH1101 might also correlate with the stage of the disease or the nature of previous or ongoing treatments. In order to explore in which cancer indications or clinical settings γδ T cell pharmacology is impaired or fully maintained, we have set up a quantitative standardized in vitro “IPH1101 sensitivity test” that requires only a small sample of patient’s PBMC. The objective of this ongoing ex-vivo observational study is to identify types of cancers and settings for which γδ T cell immunotherapy using IPH1101 treatment may be beneficial. Here are reported results from 3 hematological indications: multiple myeloma (MM), follicular lymphoma (FL) and chronic myeloid leukaemia (CML) receiving long term imatinib treatment. Method: Patients with MM, FL and CML have been enrolled at 2 French sites. A small sample of blood (20 mL) is sufficient to prepare PBMCs and culture them in the presence of IPH1101 and IL-2. Results on the extent of in vitro amplification of cells by IPH1101 are available within 8 days and are expressed as % of γδ T cells in the culture and total amplification rate of γδ T cells. Results: Nineteen patients with MM, 31 with FL and 19 with CML receiving imatinib were evaluable in this study. One patient by indication was found strictly not sensitive to IPH1101 stimulation ex vivo, meaning that their γδT cells showed no signs of proliferation in culture. Samples from patients with MM and, to a lesser extent, FL showed a clear impairment of their proliferative response as compared to samples from healthy individuals, with 32% and 10% of MM and FL samples, respectively, demonstrating an impaired response. Finally, in imatinib-treated CML patients, 94% PBMC samples had γδ T cells with full proliferative capacity in response to IPH1101. Conclusion: Ex vivo, γδ T cells from multiple myeloma and follicular lymphoma patients show a low proportion of moderate to strong impairment of their proliferative capacity in response to their specific stimulus IPH1101. For future γδ T cell based immunotherapy trials in these indications, it might be beneficial to consider selecting patients based on such a criteria. In CML patients undergoing long-term treatment with imatinib, γδ T cells have maintained full ability to respond to their specific stimulus (despite the well-known immunosuppressive effects of imatinib). Thus, these results confirm the biological feasibility of combining imatinib and IPH1101 in a clinical trial in CML.


Sign in / Sign up

Export Citation Format

Share Document