scholarly journals Different Doses of Allergen Exposure on Dendritic Cells Determine Their Genetic/Epigenetic Regulation and T Cell Differentiation

2019 ◽  
Author(s):  
Jianjun Chen ◽  
Ying Wang ◽  
Zizhong Yu ◽  
Yue Zhou ◽  
Yun Zhu ◽  
...  

Abstract Background: It has been reported that when DCs are exposed to high-dose antigens, they can induce Th0 cells into regulatory cells (Treg) and Th1 cells. When DCs are subjected to low-dose allergen, they can drive to Th2 cells. However, the mechanisms of such dose-effect relationship are poorly understood so far. Methods: Bone marrow immature DCs (imDCs) were generated from mice and stimulated with OVA of different concentrations (0, 10, 100, 1000, 10000 μg/ml, respectively). The mDCs were then seeded and cocultured with naïve T cells for 3 days, and then the markers of different T cell subgroups were flow cytometrically tested. RNA-seq detection and DNA methylation of DCs were performed. Results: When DCs were stimulated with low-dose (10ug/ml), 77 genes were up-regulated and 87 genes down-regulated. Most activated genes were related to ribosome synthesis and ion channel inhibition but not to the immune responses and Th2 activation. At the medium-dose (100μg/ml), 339 genes were up-regulated and 168 genes down-regulated. Most activated genes involved cytokine synthesis and regulation of immune responses. At high-dose (10000μg/ml), 2794 genes were up-regulated and 1156 genes down-regulated. Tumor necrosis factor signaling pathway, MAPK signaling pathway, antigen processing and presentation signaling pathway were mostly up-regulated. The related co-stimulators, co-inhibitory molecules, inhibitory cytokines, negative regulating enzymes were highly expressed. The monocarbate, coenzyme, fatty acid, glucolipid, starch, sucrose and other metabolism-related signaling pathways were down-regulated. Conclusion: The profiles of DNA methylation and RNA synthesis of DCs varied with different doses of OVA, which serves to induce T cells to differentiate in various directions.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 3267-3267
Author(s):  
Lauren T. Southerland ◽  
Jian-Ming Li ◽  
Sohrab Hossain ◽  
Cynthia Giver ◽  
Wayne Harris ◽  
...  

Abstract Background: The severe morbidity and mortality associated with bone marrow transplantation (BMT) is caused by uninhibited immune responses to alloantigen and suppressed immune responses to pathogens. Vasoactive Intestinal Peptide (VIP) is an immunomodulatory neuropeptide produced by T-cells and nerve fibers in peripheral lymphoid organs that suppresses immune responses by induction of tolerogenic dendritic cells. In order to determine the immunoregulatory effects of VIP, we examined T-cell immune responses to allo- and viral-antigens in VIP knockout (KO) mice and mouse BMT recipients of hematopoietic cells from VIP KO donors. Methods: VIP KO mice and VIP WT littermates were infected with lethal or sub-lethal doses (5 × 104− 5 × 105 PFU) of murine cytomegalovirus (mCMV) and the T-cell response to viral antigen was measured by flow cytometry for mCMV peptide-MHC class 1-tetramer+ CD8+ T-cells. We transplanted 5 × 106 BM plus 1 × 106 splenocytes (SP) either from VIP KO or VIP WT donors in an C57BL/6 to F1(BL/6 × Balb/c) allo-BMT model and assessed survival, GvHD, donor T-cell expansion, chimerism, and response to mCMV vaccination and mCMV infection. Results: B-cell, αβ and γδ T-cell, CD8+ T-cell, CD11b+ myeloid cell, and dendritic cell numbers were equivalent between VIP KO and WT mice, while VIP KO mice had higher number of CD4+ and CD4+CD62L+CD25+ T-cells. Non-transplanted VIP KO mice survived mCMV infection better compared to VIP WT, with a brisker anti-viral T-cell response in the blood. In the allogeneic BMT setting, recipients of VIP KO BM plus VIP KO SP had more weight loss and lower (40%) 100 day post-transplant survival compared to the recipients of VIP KO BM plus WT SP (80% survival), recipients of WT BM plus KO SP (100% survival), and recipients of WT BM plus WT SP (80% survival). Recipients of VIP KO grafts had a significantly greater anti-mCMV response that peaked four days earlier than the tetramer response of mice transplanted with WT cells. This increased anti-viral response to vaccination correlated with a greater and more rapid T-cell response to secondary viral challenge. Conclusions: These experiments suggest that the absence of all VIP in the body, or the absence of VIP in a transplanted immune system, enhances anti-viral immunity and allo-immune responses. Modulation of the VIP pathway is a novel method to regulate post-transplant immunity. Figure 1: VIP knockout(KO) mice have an increased CMV tetramer response. VIP KO and VIP WT mice were infected (day 0) with either a sub-lethal low dose (5 × 10^4 PFU) or a lethal high dose (5 × 10^5 PFU) of CMV. Peripheral blood was stained for T cell markers and tetramer and analyzed by flow cytometry. On day 3, high dosed VIP KO mice had a higher number of tetramer positive CD8 T cells and better survival than WT mice (all high dose VIP WT died prior to day 10). VIP KO mice had a significant increase in tetramer positive CD8 T cells between days 3 and 10. *** p<0.01, difference between VIP KO and VIP WT littermate at designated dose level and day. Figure 1:. VIP knockout(KO) mice have an increased CMV tetramer response. VIP KO and VIP WT mice were infected (day 0) with either a sub-lethal low dose (5 × 10^4 PFU) or a lethal high dose (5 × 10^5 PFU) of CMV. Peripheral blood was stained for T cell markers and tetramer and analyzed by flow cytometry. On day 3, high dosed VIP KO mice had a higher number of tetramer positive CD8 T cells and better survival than WT mice (all high dose VIP WT died prior to day 10). VIP KO mice had a significant increase in tetramer positive CD8 T cells between days 3 and 10. *** p<0.01, difference between VIP KO and VIP WT littermate at designated dose level and day.


2002 ◽  
Vol 70 (1) ◽  
pp. 153-162 ◽  
Author(s):  
Amy R. Tvinnereim ◽  
Sara E. Hamilton ◽  
John T. Harty

ABSTRACT Understanding how existing antivector immunity impacts live vaccine delivery systems is critical when the same vector system may be used to deliver different antigens. We addressed the impact of antivector immunity, elicited by immunization with attenuated actA-deficient Listeria monocytogenes, on the CD8+-T-cell response to a well-characterized lymphocytic choriomeningitis virus epitope, NP118-126, delivered by infection with recombinant L. monocytogenes. Challenges of immune mice with actA-deficient and with wild-type recombinant L. monocytogenes generated similar numbers of CD8+ T cells specific for the NP118-126 epitope. High-dose immunization with actA-deficient L. monocytogenes resulted in substantial numbers of CD8+ T cells specific for the L. monocytogenes LLO91-99 epitope in the effector and memory stages of the T-cell response. Challenge of these immune mice with recombinant L. monocytogenes resulted in rapid control of the infection and decreased CD8+-T-cell responses against both the secreted and nonsecreted form of the recombinant antigen compared to the response of naïve mice. In contrast, mice immunized with a low dose of actA-deficient L. monocytogenes had ∼10-fold fewer effector and memory T cells specific for LLO91-99 and a substantially higher CD8+-T-cell response against the recombinant antigen after challenge with recombinant L. monocytogenes. Although mice immunized with low-dose actA-deficient L. monocytogenes had a substantial recall response to LLO91-99, which reached the same levels by 5 to 7 days postchallenge as that in high-dose-immunized mice, they exhibited decreased ability to control L. monocytogenes replication. Thus, the level of antivector immunity impacts the control of infection and efficiency of priming responses against new antigens introduced with the same vector.


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.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 999-999 ◽  
Author(s):  
Jennifer N. Brudno ◽  
Victoria Shi ◽  
David Stroncek ◽  
Stefania Pittaluga ◽  
Jennifer A. Kanakry ◽  
...  

Abstract Background: Chimeric antigen receptors (CARs) are fusion proteins that combine antigen-recognition domains and T-cell signaling domains. T cells genetically modified to express CARs directed against the B-cell antigen CD19 can cause remissions of B-cell malignancies. Most CARs in clinical use contain components derived from murine antibodies. Immune responses have been reported to eliminate CAR T cells in clinical trials, especially after second infusions of CAR T cells (C. Turtle et al., Journal of Clinical Investigation, 2016). These immune responses could be directed at the murine components of CARs. Such immune responses might limit the persistence of the CAR T cells, and anti-CAR immune responses might be an especially important problem if multiple infusions of CAR T cells are administered. Development of fully-human CARs could reduce recipient immune responses against CAR T cells. Methods: We designed the first fully-human anti-CD19 CAR (HuCAR-19). The CAR is encoded by a lentiviral vector. This CAR has a fully-human single-chain variable fragment, hinge and transmembrane regions from CD8-alpha, a CD28 costimulatory domain, and a CD3-zeta T-cell activation domain. We conducted a phase I dose-escalation trial with a primary objective of investigating the safety of HuCAR-19 T cells and a secondary objective of assessing anti-lymphoma efficacy. Low-dose chemotherapy was administered before HuCAR-19 T-cell infusions to enhance CAR T-cell activity. The low-dose chemotherapy consisted of cyclophosphamide 300 mg/m2 daily for 3 days and fludarabine 30 mg/m2 daily for 3 days on the same days as cyclophosphamide. HuCAR-19 T cells were infused 2 days after the end of the chemotherapy regimen. Patients with residual lymphoma after a first treatment were potentially eligible for repeat treatments if dose-limiting toxicities did not occur with the first treatment. Repeat infusions were given at the same dose level as the first infusion or 1 dose level higher than the first infusion. Findings: A total of 11 HuCAR-19 T-cell infusions have been administered to 9 patients; 2 patients received 2 infusions each. So far, there is an 86% overall response rate (Table). Grade 3 adverse events (AEs) included expected cytokine-release syndrome toxicities such as fever, tachycardia, and hypotension. Corticosteroids were used to treat toxicity in Patient 3. The interleukin-(IL)-6 receptor antagonist tocilizumab was used to treat toxicity in Patient 4, and both tocilizumab and corticosteroids were used to treat toxicity in Patient 8. Only 1 of 8 evaluable patients, Patient 3, has experienced significant neurological toxicity to date. This patient experienced encephalopathy that was associated with a cerebrospinal fluid (CSF) white blood cell count of 165/mm3. Almost all of the CSF white cells were CAR T cells, and the CSF IL-6 level was elevated. All toxicities have resolved fully in all patients. In Patient 1, tumor biopsies revealed a complete loss of CD19 expression by lymphoma cells after 2 HuCAR-19 T-cell infusions, which to our knowledge is the first documented complete loss of CD19 expression by lymphoma after anti-CD19 CAR T-cell therapy. This loss of CD19 expression was associated with lymphoma progression. After first CAR-19 T-cell infusions, HuCAR-19 cells were detectable in the blood of every patient. The median peak number of blood CAR+ cells was 26/microliter (range 3 to 1005 cells/microliter). Blood HuCAR-19 cells were detected after second infusions in the blood of both patients who received second infusions. Patient 1 obtained a partial response after a second infusion after only obtaining stable disease after a first infusion. We detected elevations of inflammatory cytokines including IL-6, interferon gamma, and IL-8 in the serum of patients experiencing clinical toxicities consistent with cytokine-release syndrome. Interpretation: T cells expressing HuCAR-19 have substantial activity against advanced lymphoma, and infusions of HuCAR-19 T cells caused reversible toxicities attributable to cytokine-release syndrome. Disclosures Kochenderfer: Kite Pharma: Patents & Royalties, Research Funding; bluebird bio: Patents & Royalties, Research Funding.


2005 ◽  
Vol 79 (8) ◽  
pp. 4877-4885 ◽  
Author(s):  
Yvonne M. Mueller ◽  
Constantinos Petrovas ◽  
Paul M. Bojczuk ◽  
Ioannis D. Dimitriou ◽  
Brigitte Beer ◽  
...  

ABSTRACT Interleukin-15 (IL-15) in vitro treatment of peripheral blood mononuclear cells (PBMC) from human immunodeficiency virus (HIV)-infected individuals specifically enhances the function and survival of HIV-specific CD8+ T cells, while in vivo IL-15 treatment of mice preferentially expands memory CD8+ T cells. In this study, we investigated the in vivo effect of IL-15 treatment in 9 SIVmac251-infected cynomolgus macaques (low dose of IL-15, 10 μg/kg of body weight, n = 3; high dose of IL-15, 100 μg/kg, n = 3; control [saline], n = 3; dose administered twice weekly for 4 weeks). IL-15 treatment induced a nearly threefold increase in peripheral blood CD8+CD3− NK cells. Furthermore, CD8+ T-cell numbers increased more than twofold, mainly due to an increase in the CD45RA−CD62L− and CD45RA+CD62L− effector memory CD8+ T cells. Expression of Ki-67 in the CD8+ T cells indicated expansion of CD8+ T cells and not redistribution. IL-15 did not affect CD4+ T-cell, B-cell, and CD14+ macrophage numbers. No statistically significant differences in changes from baseline in the viral load were observed when control-, low-dose-, and high-dose-treated animals were compared. No clinical adverse effects were observed in any of the animals studied. The selective expansion of effector memory CD8+ T cells and NK cells by IL-15 further supports IL-15's possible therapeutic use in viral infections such as HIV infection.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 684-684 ◽  
Author(s):  
Daniel W. Lee ◽  
Maryalice Stetler-Stevenson ◽  
Constance M. Yuan ◽  
Terry J. Fry ◽  
Nirali N Shah ◽  
...  

CD19 chimeric antigen receptor (CAR) T cells have shown significant promise in multiple early phase trials including our own (Lancet 385:517-28). We manufacture CAR T cells containing CD28 and CD3z domains in 7 days using a retroviral platform. Several challenges remain to its widespread use: 1) reduction in the incidence of grade 4 cytokine release syndrome (CRS) and 2) incorporation with standard salvage regimens. Here, we update our experience with 39 patients. In the first 21 patients we defined the maximally tolerated dose as 1x106 CAR T cells/kg, grade 4 CRS occurred in 16%, and noted that severity of CRS correlated with disease burden. We stratified the current cohort (n=18) by disease burden. Subjects 1-21 and subsequent patients with low burden disease (Arm 1: isolated CNS disease or <25% marrow blasts) received a low dose preparative regimen of fludarabine (25 mg/m2/day Days-4 to -2) and cyclophosphamide (900 mg/m2 Day-2). Those with high burden disease (Arm 2: ³25% marrow blasts, circulating blasts or lymphomatous disease) received a high dose regimen to reduce tumor burden prior to cell infusion in an attempt to decrease severity of CRS. Arm 2 regimens were individualized based on prior therapies and risk from comorbidities. FLAG (n=6), ifosfamide/etoposide per AALL0031 (IE; n=2) and high dose fludarabine (30 mg/m2/day Days -6 to -3) with cyclophosphamide (1200 mg/m2/day Days -4 and -3) (HD flu/cy; n=3) were used. All products in the second cohort met cell dose though contaminating monocytes tended to inhibit maximal growth and transduction (see companion abstract by Stroncek). All patients received 1x106 CAR T cells/kg. Using grading criteria and an algorithm for early intervention to prevent grade 4 CRS (Blood 124:188-95) no grade 3 and only 1 grade 4 (5.6%) CRS occurred. Having significant comorbidities, Pt 34 was electively intubated for airway protection, did not require vasopressors, and rapidly recovered after tocilizumab and steroids. A brief seizure occurred, though he had a history of seizures. None others in the current cohort had neurotoxicity. Using intent to treat analysis, the complete response (CR) rate was 59% overall and 61% in ALL. 13/16 (81%) low burden and 10/22 (46%) high burden ALL patients had a CR across both cohorts. Low burden patients treated on either cohort had similar CR rate of 8/10 (80%) and 5/6 (83%). Although not statistically significant and underpowered, 7/11 (64%) high burden patients treated with low dose flu/cy had a CR while 3/11 (27%) had a CR with high dose regimens. Specifically, 3/6 (50%) receiving FLAG achieved MRD-CR while none receiving IE or HD flu/cy responded. 8/8 with primary refractory ALL had MRD-CR regardless of disease burden or preparative regimen raising the prospect that T cell fitness in these patients was superior to others. Of the 20 patients achieving an MRD-CR, the median leukemia free survival (LFS) is 17.7 months with 45.5% probability of LFS beginning at 18 months. Only 3 did not have a subsequent hematopoietic stem cell transplant as their referring oncologist determined the risk of such was unacceptable. Two relapsed with CD19-leukemia at 3 and 5 months, while 1 remains in CR with detectable CAR T cells at 5 months. Reliance on multiple infusions of cells is problematic as 0/5 CD19+ patients receiving a second dose responded. Preclinical models have demonstrated that T cell exhaustion has a role in limiting the efficacy of CAR T cells. We evaluated CAR products and the T cells used to generate them for phenotypic markers of exhaustion and will present data evaluating the relationship between these and response. Our results demonstrate that CD19 CAR T cell therapy is safe and effective with aggressive supportive care and use of an early intervention algorithm to prevent severe CRS and provides a potential for cure in primary refractory ALL. Table. Patient Characteristics, Response, and Toxicity Pt Age/ Sex/Risk # Relapses Arm/Prep Regimen(if Arm 2) Marrow Blasts Response CRS Grade Pre-Therapy Post CAR 22 17M 3 1 20 0 MRD- 2 23 13M 2 2 IE 99 98 SD 0 24 12M MLL 2 1 8.5 3 CR 1 25 25F 1 2 FLAG 95 0 MRD- 2 26 4M DS 2 2 IE (60%) 89 NA PD 0 27 8F 2 2 FLAG 77 69 SD 0 28 4M 2 2 FLAG (60%) 99 99 PD 0 29 12M PR 1 0.15 0 MRD- 1 30 15M Ph+ CNS2 3 1 0.08 0 MRD- 1 31 22M 3 2 FLAG 97 99 SD 0 32 15M CNS2 3 2 FLAG 0.04 + Lymphoma 0 MRD- 2 33 6M PR 1 0.15 0 MRD- 0 34 14M DS 3 2 Arm 1 Flu/Cy 90 0 MRD- 4 35 25M 2 2 HD Flu/Cy 30 87 PD 2 36 6M 2 1 1.5 91 PD 0 37 4F MLL 1 2 HD Flu/Cy 90 99 SD 0 38 7M 1 2 HD Flu/Cy 99 99 SD 1 Disclosures Off Label Use: Off-label use of tocilizumab will be discussed in managing cytokine release syndrome.. Rosenberg:Kite Pharma: Other: CRADA between Surgery Branch-NCI and Kite Pharma. Mackall:Juno: Patents & Royalties: CD22-CAR.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 48-48
Author(s):  
Tanaya Shree ◽  
Michael S. Khodadoust ◽  
Debra K. Czerwinski ◽  
Matthew J. Frank ◽  
Sara Beygi ◽  
...  

Introduction: Local treatment with intratumoral CpG (a toll-like receptor 9 agonist, SD-101) and low-dose radiation can elicit antitumor immune responses and global tumor reduction in patients with low-grade lymphoma (Frank, Cancer Discov, 2018). Ibrutinib compromises B-cell survival by inhibiting Bruton's tyrosine kinase, but also modulates T-cells by inhibiting interleukin-2-inducible T-cell kinase. In a mouse model of lymphoma, ibrutinib plus intratumoral CpG was curative of systemic disease, an effect that was T-cell dependent (Sagiv-Barfi, Blood, 2015). Thus, we initiated a phase I/II clinical trial combining oral ibrutinib, intratumoral CpG and local low-dose radiation in adults with recurrent low-grade lymphoma (NCT02927964). Methods: Enrolled patients received intratumoral injections of CpG (SD-101, 3mg) weekly for 5 doses, starting on the second day of a 2-day course of local radiation (4Gy total) to the same site. Daily oral ibrutinib (560mg) began on day 9. Treatment-emergent adverse events (AEs), ibrutinib dose modifications and adherence were recorded at every visit. Revised Lugano criteria (Cheson et al., JCO, 2014) were used to assess response to therapy, based on CT scans at 3, 6, 12, 18, and 24 months. Fine needle aspirates (FNAs) were obtained from CpG-injected and non-injected nodal tumor sites pre- and post-treatment and analyzed by flow cytometry and single-cell RNA sequencing (scRNAseq). When available, viably preserved tumor and peripheral blood cells were used for in vitro immune response assays. Results: As of July 16, 2020, 18 patients had been treated, with a median follow-up of 12 months. Ten were male and 8 were female. All but one had a diagnosis of follicular lymphoma; one patient had marginal zone lymphoma. All were previously treated with an average of 2 prior lines of therapy. AEs were consistent with known effects of ibrutinib (including diarrhea and rash) and of CpG (including fever and flu-like reaction) with no unexpected AEs to suggest synergistic toxicity. There were no grade 4 or 5 events. AEs led to ibrutinib dose reduction or discontinuation in 2 patients and dose interruption in 6 patients. At the time of analysis, 9 of 18 evaluable patients had achieved a partial response (50% ORR) and 12 of 18 patients experienced at least a 30% reduction in the distant uninjected lesions (Figure 1A). Most responses have been maintained for at least 6 months, many longer (Figure 1B). Flow cytometry revealed decreased T follicular helper cells and increased CD4 and/or CD8 effector T-cells, CD137+ activated T-cells, and NK cells in CpG-injected tumors. Abscopal immune effects in distant non-injected lesions included an increase in Granzyme B+ CD8 T-cells, most prominent after the addition of ibrutinib. scRNAseq data showed significant transcriptional shifts in tumor cells and in tumor-infiltrating T-cells, including signatures of interferon response and T cell activation and cytotoxicity. Finally, in vitro assays showed tumor-specific immune responses in peripheral blood T-cells of all 6 evaluable patients tested thus far. Conclusion: Early data suggest that the combination of oral ibrutinib, intratumoral CpG, and local low-dose radiation is safe and can generate systemic antitumor immune responses and systemic tumor shrinkage in low-grade lymphoma. Disclosures Khodadoust: Seattle Genetics: Consultancy; Kyowa Kirin: Consultancy. Levy:Quadriga: Membership on an entity's Board of Directors or advisory committees; Beigene: Membership on an entity's Board of Directors or advisory committees; GigaGen: Membership on an entity's Board of Directors or advisory committees; Teneobio: Membership on an entity's Board of Directors or advisory committees; Sutro: Membership on an entity's Board of Directors or advisory committees; Checkmate: Membership on an entity's Board of Directors or advisory committees; Nurix: Membership on an entity's Board of Directors or advisory committees; Dragonfly: Membership on an entity's Board of Directors or advisory committees; Abpro: Membership on an entity's Board of Directors or advisory committees; Apexigen: Membership on an entity's Board of Directors or advisory committees; Spotlight: Membership on an entity's Board of Directors or advisory committees; 47 Inc.: Membership on an entity's Board of Directors or advisory committees; XCella: Membership on an entity's Board of Directors or advisory committees; Immunocore: Membership on an entity's Board of Directors or advisory committees; Walking Fish: Membership on an entity's Board of Directors or advisory committees; Viracta: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3703-3703
Author(s):  
Debra K Czerwinski ◽  
Joshua Brody ◽  
Holbrook E Kohrt ◽  
Richard T. Hoppe ◽  
Ranjana H. Advani ◽  
...  

Abstract Abstract 3703 Introduction Local intratumoral injection of CpG ologonucleotides in conjunction with local low dose radiotherapy can induce regression of uninjected distant sites of disease in patients with follicular lymphoma (FL) (Brody et. al JCO). This maneuver activates the Toll-like receptor 9 (TLR9) within the tumor cells and in adjacent antigen presenting cells in the local tumor environment, such as dendritic cells and macrophages. A systemic anti-tumor T cell immune response is thereby induced. Our original phase I/II trial was conducted in patients who had been previously treated with standard therapies and had recurred with multiple sites of disease. We observed significant clinical responses as well as anti-tumor T cell immune responses as measured in vitro using peripheral blood T cells or T cells obtained from a pleural effusion adjacent to a responding tumor site. Methods We now extended the trial of in situ vaccination to newly diagnosed patients prior to any other therapy, a group that may have a more intact immune system. Patients were eligible if they had FL grades I-IIIa, stage III or IV and were not in need of immediate treatment. One involved lymph node was biopsied and viable suspensions of tumor cells were prepared and cryopreserved for use as stimulators in immune assays. A second site received low dose XRT (2Gy on each of two successive days) together with 10 weekly injections of 18mg of CpG ologonucleotide (PF-3512676, Pfizer) all into the same tumor site, beginning on the second day of XRT. Peripheral blood lymphocytes (PBL) were obtained prior to each injection and two weeks after the last injection. These were cryopreserved and used as responder cells for assays of T cell immune responses. We sought to determine the kinetics of anti-tumor T cell immune responses as well as which type of T cell response that was the most informative. Tumor cells were thawed and activated for 3 days with CpG and soluble CD40L. They were then incubated for 5 days with autologous PBL T cells. Fresh stimulator cells were then added for a final overnight culture. Responding T cells were stained for subtype (CD4, CD8, CD56, CD45RO), for their expression of activation markers (CD25, CD137 and CD278), for their expression of intracellular cytokines (IFN-g, TNF and IL-2), and for their expression of cytotoxic enzymes (perforin and granzyme B). The cells were then analyzed by multiparamer flow cytometry (BD LSRII). For measurement of perforin and granzymeB, the cells were gated on CD8+/CD3+/CD56- to exclude NK cells. Results In response to in situ vaccination, all patients made anti-tumor immune responses. Some generated only CD4 responses, some only CD8 responses and others made both CD4 and CD8 T cell responses. Representative data are shown in the figures below. We found that for CD4 responses the activation marker CD278 (ICOS) was particularly informative, and usually restricted to the CD45RO+ memory subset. For CD8 responses, the most robust readout was intracellular expression of the combination of Perforin and Granzyme B. CD8 Immune responses became positive as early as two weeks after the start of vaccination. CD4 responses became positive by four weeks of vaccination. As in our previous trial, we observed clinical responses, with regression of tumors at uninjected/untreated sites of disease. An evaluation of the magnitude and duration of these clinical responses and their relation to immune parameters awaits a more extended time of followup. Conclusions In situ vaccination efficiently induced immune responses in previously untreated patients with FL. These responses occurred within two weeks after initiation of vaccination. The immune responses was heterogeneous and included both CD4 and CD8 T cells. The most robust measures of T cell response will be correlated with clinical outcome. The identification of a limited panel of correlative immune response markers and an understanding of the response kinetics will allow a focused approach to immuno-monitoring in future clinical trials. Disclosures: Advani: Pharmacyclics: Research Funding.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Yao Wang ◽  
Chuan Tong ◽  
Hanren Dai ◽  
Zhiqiang Wu ◽  
Xiao Han ◽  
...  

AbstractInsufficient eradication capacity and dysfunction are common occurrences in T cells that characterize cancer immunotherapy failure. De novo DNA methylation promotes T cell exhaustion, whereas methylation inhibition enhances T cell rejuvenation in vivo. Decitabine, a DNA methyltransferase inhibitor approved for clinical use, may provide a means of modifying exhaustion-associated DNA methylation programmes. Herein, anti-tumour activities, cytokine production, and proliferation are enhanced in decitabine-treated chimeric antigen receptor T (dCAR T) cells both in vitro and in vivo. Additionally, dCAR T cells can eradicate bulky tumours at a low-dose and establish effective recall responses upon tumour rechallenge. Antigen-expressing tumour cells trigger higher expression levels of memory-, proliferation- and cytokine production-associated genes in dCAR T cells. Tumour-infiltrating dCAR T cells retain a relatively high expression of memory-related genes and low expression of exhaustion-related genes in vivo. In vitro administration of decitabine may represent an option for the generation of CAR T cells with improved anti-tumour properties.


Sign in / Sign up

Export Citation Format

Share Document