scholarly journals Relationship between Bone Marrow PD-1 and PD-L1 Expression and the Presence of Osteolytic Bone Disease in Multiple Myeloma Patients

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3183-3183
Author(s):  
Federica Costa ◽  
Marina Bolzoni ◽  
Rosanna Vescovini ◽  
Fabrizio Accardi ◽  
Anna Benedetta Dalla Palma ◽  
...  

Abstract Alterations of the bone marrow (BM) immune-microenvironment characterize the progression of monoclonal gammopathies and the development of osteolytic bone disease in multiple myeloma (MM). MM patients exhibit immune dysfunctions as impaired dendritic, NK and T cells, whereas the onset of MM osteolytic lesions is associated to an increased prevalence of Th17 cells. Recently, the pathophysiological role of the programmed cell death protein 1 (PD-1)/PD-1 ligand (PD-L1) pathway together with an increase of myeloid derived suppressor cells (MDSCs) in the induction of tumor tolerance and immune evasion has been underlined with a therapeutic relevance. However, unclear data on the expression profile of PD-1/PD-L1 axis in MM patients have been reported and it is not known if this axis could be related with the presence of osteolytic bone disease. In order to elucidate these aspects, we analyzed a total cohort of 80 consecutives patients with monoclonal gammopathies, including 15 monoclonal gammopathy of undetermined significance (MGUS), 23 smoldering MM (SMM), 21 newly diagnosed MM (MMD) and 21 relapsed/refractory MM (MMR) patients. The presence of bone disease was checked by low-dose computerized tomography (CT) with or without positron emission tomography (PET) scan and by X-rays skeletal survey in 11 MM patients. 87% of MM patients displayed osteolytic lesions. High bone disease (HBD) was defined by the presence of 3 or more osteolytic lesions (62% of our cohort). Patients without bone lesions or with minus of 3 lesions were defined as low bone disease (LBD). BM mononuclear cells were analyzed by flow cytometry, evaluating plasma cells (PCs) (CD138+), monocytes (CD14+) and T cells (total CD3+, CD3+CD4+ and CD3+CD8+). PD-L1 (CD274) expression was evaluated on CD138+ and CD14+ cells, and PD-1 (CD279) on CD3+, CD4+ and CD8+ cells. Lastly, in a subgroup of patients we analysed MDSC populations, including both granulocytic (gMDSCs) (CD11b+HLA-DR-CD14-CD15+) and monocytic MDSCs (mMDSCs), (CD11b+HLA-DR-/lowCD14+CD15-). The percentage of BM CD3+PD-1+ cells increased from MGUS to MMR patients with a significant trend (p=0.004). Indeed, patients with active MM (MMD and MMR) showed both higher % of CD3+PD-1+ cells (median value: 48.5% vs 40.6%, p=0.001) and PD-1 median fluorescence intensity (MFI) on CD3+ (median value: 18.9 vs 16.7 MFI, p=0.024) as compared to patients with SMM and MGUS. CD4+PD-1+, but not CD8+PD-1+ cells are increased in active MM compared to SMM and MGUS patients (p=0.023). On the other hand, any significant difference was not observed in the PD-L1 expression on both BM CD138+ and CD14+ cells across patient groups. The percentage of BM MDSC populations did not significantly change across the different monoclonal gammopathies (p=0.14); moreover, comparing MM with SMM and MGUS patients, the % of gMDSCs was significantly reduced (median %: 12.5% vs 17%, p=0.04) and the % of mMDSCs was increased (median %: 0.36% vs. 0.24%) without reaching a statistical significance. Focusing on MM bone disease, we found that osteolytic MM patients displayed higher CD4+/CD8+ ratio compared to non-osteolytic ones (p=0.043). Regarding the PD-1 expression, the % of CD3+PD-1+ cells did not change (p=0.192), whereas the % of CD8+PD-1+ cells was significantly reduced in osteolytic patients compared to non-osteolytic ones (p=0.016). Consistently, among the BM CD8+ cells, a significant decrease of %PD-1+ cells was found in HBD vs LBD MM patients (median value: 46.9% vs 57.4%, p=0.045). Interestingly, when compared to LBD MM patients, HBD MM patients displayed a significant reduction of PD-L1 expression on both BM CD138+ PCs (median MFI value: 13.3 vs 21.6 MFI, p=0.007) and CD14+ cells (median MFI value: 15.4 vs 23.8 MFI, p=0.007). In a multivariate analysis, PD-1+ expression on CD8+ cells and PD-L1 MFI on CD138+ were significant independent factors related to the presence of HBD. In conclusion, our study indicates that the frequency of PD-1+ T cells increases across the progression of the monoclonal gammopathies. On the other hand, for the first time, we show in MM patients a significant relationship between the presence of extensive osteolytic bone disease and a reduced expression profile of BM PD-1/PD-L1 axis on CD8+ and CD138+ cells. We hypothesize that a less immune-suppressive profile could be related to the development of osteolysis consistent with the negative cross talk existing between PD-1/PD-L1 axis and Th17 cells. Disclosures Aversa: Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees; Astellas: Honoraria; Basilea: Honoraria, Membership on an entity's Board of Directors or advisory committees; Merck: Honoraria; Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees. Giuliani:Takeda Pharmaceutical Co: Research Funding; Janssen Pharmaceutica: Other: Avisory Board, Research Funding; Celgene Italy: Other: Avisory Board, Research Funding.

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4481-4481
Author(s):  
Denise Toscani ◽  
Martina Chiu ◽  
Giuseppe Taurino ◽  
Emanuela Vicario ◽  
Valentina Marchica ◽  
...  

Abstract Multiple myeloma (MM) cells are characterized by tight dependence on the bone marrow (BM) microenvironment that exerts a permissive role on cell growth and survival. In turn, MM cells markedly modify their microenvironment leading, in particular, to the development of osteolytic bone lesions. Recently, we demonstrated that metabolic alterations is a major feature of MM cells showing that BM plasma of MM patients is characterized by lower levels of Glutamine (Gln) and higher levels of Glutamate (Glu) and ammonium when compared with patients with smoldering MM (SMM) and Monoclonal Gammopathy of Uncertain Significance (MGUS). In the majority of MM patients MM cells are Gln-addicted since they strictly depend on extracellular Gln, do not express Glutamine Synthetase (GS), the enzyme that synthetizes Gln from Glu and ammonium, and are endowed with high levels of the Gln transporter ASCT2. Based on this evidence, we have hypothesized that the peculiar Gln metabolism of MM cells may have a significant impact on the relationship with the bone microenvironment and contribute to the development of osteolytic lesions. We firstly characterized a panel of human MM cell lines (HMCLs) for their GS expression and response to decreasing levels of Gln. The majority of HMCLs, which did not express GS, consumed large amounts of extracellular Gln but secreted nearly half of the amino acid as Glu. Two HMCLs, MM1.S and U266, with a sizable GS expression, were less sensitive to Gln deprivation and secreted less Glu in the extracellular space compared with GS-negative HMCLs. Consistently, the activity of the Glu exchanger x-CT (the product of SLC7A11 gene) was lower in GS-positive than in GS-negative cells. The response to Gln starvation was then studied in mesenchymal stromal cell line (MSC), as well as in osteoblastic (HOBIT) and pre-osteocytic cells (HOB-01). HOBIT and HOB-01 were more sensitive to Gln depletion than MSC. Indeed, while MSC showed a low EC50 for Gln (0.064mM), which is 10-times lower than the physiological blood Gln concentration (around 0.6 mM), the EC50 values of HOBIT and HOB-01 cells were 0.250 mM and 0.297mM, respectively. Furthermore, L-methionine sulfoximine (MSO), an irreversible inhibitor of GS, emphasized the effects of Gln deprivation on all the cell lines tested. Indeed, Gln deprivation enhanced the expression of GS, suggesting that both stromal and osteoblastic cells exploit the enzyme to counteract Gln deprivation. On the basis of these data, we assessed the effects of Gln and Glu on osteogenic differentiation by incubating MSC, either immortalized or primary, with an osteogenic medium containing different concentrations of Gln and Glu. After 2 weeks, compared with cells differentiated in high Gln/high Glu conditions, MSC incubated in the presence of decreased Gln and increased Glu showed lower osteogenic ability, as assessed by real time PCR and ALP staining. Lastly, MSC co-cultured for 72 hours with GS-negative, but not with GS-positive HMCLs, showed reduced viability and increased GS expression. Lastly, to put in a translational perspective these in vitro observations, we analyzed the BM plasma levels of Gln and Glu in a cohort of 41 patients with newly diagnosed MM, including 9 smoldering MM (SMM) and 32 active MM patients (20 of them with osteolytic bone disease, 12 of them without bone disease). All 20 osteolytic MM patients had more than three osteolytic lesions. We found that MM patients had lower Gln levels and higher Glu levels than SMM patients. Moreover, when compared with MM patients without bone disease, MM patients with bone disease showed lower levels of Gln and higher levels of Glu. The results of these analyses are being continuously updated increasing the number of samples tested. Overall, these results indicate that MM cells are able to create a low-Gln/high-Glu bone marrow microenvironment that sustains GS expression in bone cells and impairs their differentiation and viability. Thus, the peculiar metabolic milieu in the MM bone microenvironment affects the relationship between neoplastic and bone cells and may contribute to the development of osteolytic bone disease in MM patients. Disclosures Aversa: Astellas: Honoraria; Merck: Honoraria; Pfizer: Honoraria, Membership on an entity's Board of Directors or advisory committees; Basilea: Honoraria, Membership on an entity's Board of Directors or advisory committees; Gilead: Honoraria, Membership on an entity's Board of Directors or advisory committees. Giuliani:Takeda Pharmaceutical Co: Research Funding; Celgene Italy: Other: Avisory Board, Research Funding; Janssen Pharmaceutica: Other: Avisory Board, Research Funding.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 8-9
Author(s):  
Christie P.M. Verkleij ◽  
Marloes Broekmans ◽  
Amy Wong ◽  
Sonja Zweegman ◽  
Raluca Verona ◽  
...  

Introduction: New immunotherapies directed against CD38, SLAMF7 or BCMA have significantly improved the outcome of multiple myeloma (MM) patients. However, most patients eventually relapse, underscoring the need for additional immunotherapeutic targets. We have previously shown that expression levels of GPRC5D, an orphan G protein-coupled receptor, are significantly higher on MM cells, compared to normal plasma cells or other immune cells. We also showed that the novel GPRC5DxCD3 bispecific antibody (BsAb) JNJ-7564, has promising anti-MM activity in patient-derived BM samples (Verkleij et al., EHA 2019). To elucidate which factors contribute to the observed heterogeneity in ex vivo response, we analyzed the impact of tumor and patient characteristics on efficacy of JNJ-7564. We further investigated whether tumor-intrinsic factors may be determinants of response by also testing in these assays JNJ-7957, a BCMA-targeting BsAb that differs from JNJ-7564 only in the tumor-antigen-binding domain. Methods: Bone marrow (BM) samples obtained from 13 newly diagnosed (ND), 17 daratumumab-naive relapsed/refractory (DARA-naive RR; median of 3 prior therapies) and 15 daratumumab-refractory (DARA-R, median of 6 prior therapies) MM patients were analyzed for tumor- and immune cell composition, and subsequently incubated with JNJ-7564 (0.00128-4.0 µg/mL) or JNJ-7957 (0.8 µg/mL). After 48 hours, MM cell lysis was assessed by flow cytometry. Luciferase-transduced MM cell lines were incubated with JNJ-7564 (0.032-4.0 µg/mL) in the presence of healthy peripheral blood mononuclear cells (PBMCs), purified CD4+CD25- T-cells or regulatory T-cells (Tregs). After 48 hours, MM cell lysis was assessed by bioluminescence assay. Results: We found no difference in JNJ-7564 efficacy with respect to disease stage (NDMM vs DARA-naive RRMM vs DARA-R MM, P=0.48). Importantly, the presence of high-risk cytogenetic abnormalities [del(17p), t(4;14) and t(14;16)] did not impair JNJ-7564 efficacy. The level of target expression was an important determinant of response, as evidenced by superior MM cell lysis in samples with higher than median GPRC5D expression, when compared to lower GPRC5D expression (Fig. 1A). Inferior MM cell lysis was observed in older patients (>67 years), in samples with low T-cell counts or low effector:target (E:T) ratios, and in those with a high frequency of PD-1+ T-cells, HLA-DR+ activated T-cells, or Tregs. These determinants of response also affected JNJ-7564-mediated T-cell activation and degranulation. To further analyze the impact of Tregs, we performed additional cell line experiments. Purified Tregs impaired T-cell proliferation, and were significantly less potent to kill MM cells when redirected by JNJ-7564, compared to CD4+CD25- T-cells (Fig. 1B). This was accompanied by reduced secretion of IFN-γ, TNF-α, IL-2 and granzyme B. To evaluate the impact of BM stromal cells (BMSCs) on JNJ-7564 activity, MM cell lines were co-incubated with PBMCs and patient-derived BMSCs. Direct cell-cell contact hampered MM cell lysis, while indirect contact (transwell) did not affect JNJ-7564 activity. Direct contact also decreased secretion of TNF-α and IL-2, and reduced GPRC5D expression on MM cells, contributing to BMSC-mediated resistance to JNJ-7564. Finally, we simultaneously evaluated the single agent activity of both JNJ-7564 and JNJ-7957 (0.8 µg/mL, dose whereby a plateau in MM cell lysis was observed with both BsAbs) in 40 BM samples. MM cell lysis induced by both agents was strongly correlated (Fig. 1C). In 6 samples, both agents exhibited poor activity (<45% lysis), whereas in 9 samples very good activity was observed (>80% lysis). Comparison of characteristics between these groups showed that a low E:T ratio (Fig. 1D) and high frequency of Tregs (Fig. 1E) significantly impaired efficacy of both BsAbs, suggesting patient-specific factors can determine response to T-cell redirectors targeting different antigens. Conclusion: We show that tumor-related factors, such as GPRC5D expression, as well as differences in the composition of the BM microenvironment, including E:T ratio, frequency of PD-1+ or HLA-DR+ T-cells or immune-suppressing Tregs or BMSCs, contribute to the variability in response to JNJ-7564. Our data indicate that strategies aiming at optimizing E:T ratio (e.g. induction therapy) or Treg depletion, may improve response to T-cell redirecting antibodies in MM. Disclosures Wong: Jhonson & Jhonson: Current Employment. Zweegman:Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees, Research Funding; Takeda: Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Membership on an entity's Board of Directors or advisory committees; Sanofi: Membership on an entity's Board of Directors or advisory committees. Verona:Johnson & Johnson: Current Employment, Current equity holder in publicly-traded company. Adams:Johnson & Johnson: Ended employment in the past 24 months. Mutis:Janssen Pharmaceuticals: Research Funding; Genmab: Research Funding; Takeda: Research Funding; Onkimmune: Research Funding; Gadeta: Research Funding. van de Donk:Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees, Research Funding; Amgen: Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Membership on an entity's Board of Directors or advisory committees, Research Funding; Novartis: Membership on an entity's Board of Directors or advisory committees; Bayer: Membership on an entity's Board of Directors or advisory committees; Takeda: Membership on an entity's Board of Directors or advisory committees; Ferrer: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 20-21
Author(s):  
Yaya Chu ◽  
Julie-An Talano ◽  
Lee Ann Baxter-Lowe ◽  
Carolyn A. Keever-Taylor ◽  
Erin Morris ◽  
...  

Background: CD3/CD19 cell depletion (Barfiled RC, et al, Cytotherapy, 2004), αβ T-cell/CD19 cell depletion (Locatelli F, et al, Blood, 2017), CD34+ positive selection (Aversa F, et al, NEJM, 1998) are designed to deplete T cells and reduce AGVHD following allogeneic stem cell transplantation (AlloSCT). These approaches achieved low rates of AGVHD, but the grafts had few T and B cells. To improve immune reconstitution we undertook an alternative approach to addback small numbers and percentages of immune cells in the final HSCT product. We previously reported a very low incidence of AGVHD in pediatric recipients receiving CD34 enriched HPC products with peripheral blood mononuclear cells (PBMNC) addback containing a fixed dose of 2 x 105 CD3/kg from MUD donors (Geyer/Cairo et al, BJH, 2012). Recently we demonstrated that despite a 5 log depletion of T cells, PBMNC addback (fixed at 2 x 105 CD3/kg) facilitated rapid hematopoietic engraftment, high levels of donor chimerism and immune reconstitution with a low probability of Grade II-IV AGVHD. Patients had a 1 yr OS of 90% following familial haploidentical (FHI) CD34 Enriched Stem Cell Transplantation in patients with SCD (Cairo, JAMA Pediatr, 2020). Objective: To determine the final immune cell concentration following CD34 enrichment and PBMNC (2 x 105 CD3/kg) addback and determine the effect on engraftment and T and NK cell immune reconstitution. Methods: Patients and/or their guardians signed written informed consents and/or assents (NCT NCT02675959). CD34+ enrichment was performed using a CD34+ reagent system (CliniMACS; Miltenyi Biotec). Mononuclear cells (2 × 105 CD3 cells/kg of recipient body weight) were removed from the leukapheresis collection prior to CD34+ enrichment and were cryopreserved as a source of MNC addback (T cells). The addback products were analyzed for CD3+CD56- T cells, CD3-CD56+ NK cells, CD3+CD56+ NKT cells, Lin-CD123+ HLA-DR+ DC cells and Lin-CD11c+ HLA-DR+ DC cells by multicolor flow cytometry analysis. Th1/Th2 cytokines were measured by multiplex assays. T cell activity was measured by viral T cells IFN-g and plasma cytokines. NK function was measured by NK receptor expression by flow cytometry analysis and in vitro cytotoxicity. Results: We identified in the PBMNC addback, mean+SEM white blood cell (WBC) percentage of: CD3+ CD56- T cells = 56.4±5%; CD3- CD56+ NK cells = 4.6±1%; CD3+ CD56+ NKT cells = 5.1±0.6%; CD19+ B cells = 29.9±3.5%. Lin- WBC consisted of: CD123+ HLA-DR+ DC cells = 18.4±8.2%; CD11c+ HLA-DR+ DC cells = 6.0±3.0%. There were 20.0+9.1e6 T cells, 1.1+0.3e6 NK cells, 1.6+0.7 e6 NKT cells, 8.6+2.5e6 B cells, 1.2+0.6e6 CD123+DC and 0.8+0.5e6 CD11c DC in the final infused products (Fig.1). We found that percentages of IFN-g+ in CD4 cells in response to CMV (pp65), ADV (hexon) and EBV (BZLF1), ranged from 0.2%+0.1% to 0.5%+0.1%, while percentages of IFN-g+ in CD8 cells in response to the antigens ranged from 0.7%+0.3% to 3.7%+1.8% when examined at days 180, 270 and 365. NK (CD3- CD56+) reconstitution was extremely rapid and occurred as early as day 30 (35.5±8.6%, 2710+1624.4 cells/ul total cells; p<0.01 vs pre-t). There were no significant differences pre-HSCT vs day 365 in plasma cytokines (Th1 and Th2) and growth factors released including IFN-g, TNF-a, IL-18, IL-4, IL-5, IL-6, IL-10, G-CSF, MCP-1 and MIP1a. There was also robust expression of NK receptor expression including NK cytotoxicity receptors, NK KIR receptors, and C-type lectin-like receptors at day 30 as compared to pre-HSCT. NK cytotoxicity, as measured using PBMC cells from recipients at different time points against K562 (E:T=10:1), was also significantly increased at day 30 (26.2±2.8%) and day 180 (28.3±3%) vs pre-HSCT (16.1±2.1%) (p<0.01). As a NK cell activation marker, CD107a expression and granzyme B levels in gated NK cells peaked at day 30. Conclusion: PBMNC addback to CD34 enriched HPC products, with a final dose of 2 × 105 CD3 cells/kg, led to stem cell products with a diverse mixture of T, NK, NKT, DC1, and DC2 cells. Immune reconstitution following PBMNC addback to CD34 enriched cells resulted in excellent CD4 and CD8 responses to CMV, ADV and EBV, and rapid functional NK cell reconstitution (Supported by FDA R01FD004090 (MSC)). Disclosures Baxter-Lowe: CHLA: Current Employment, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties: Patents related to HLA typing, Research Funding. Johnson:Miltenyi Biotec: Research Funding; Cell Vault: Research Funding. Cairo:Miltenyi: Research Funding; Technology Inc/Miltenyi Biotec: Research Funding; Jazz Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees, Research Funding, Speakers Bureau; Nektar Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees, Research Funding.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 783-783
Author(s):  
Jacalyn Rosenblatt ◽  
Irit Avivi ◽  
Baldev Vasir ◽  
Tami Katz ◽  
Lynne Uhl ◽  
...  

Abstract Abstract 783 Autologous stem cell transplantation (ASCT) for multiple myeloma (MM) offers a unique setting to explore the role of immunotherapeutic strategies in eradicating malignancy. Patients achieve tumor cytoreduction following ASCT, however ultimately experience disease relapse from a persistent reservoir of chemotherapy resistant disease. Cancer vaccines that educate host immunity to target myeloma cells can be used to eradicate residual disease following ASCT. Our group has developed a cancer vaccine whereby dendritic cells (DCs) are fused with autologous tumor cells. DC/MM fusion cells present a broad array of tumor antigens in the context of DC derived costimulatory molecules. We are conducting a clinical trial in which patients with MM undergo ASCT followed by post-transplant vaccination with 3 doses of DC/MM fusions (cohort 1). A second cohort of patients receive an additional vaccination prior to stem cell collection in order to induce the expansion of tumor specific lymphocytes that are collected in the stem cell product (cohort 2). The infusion of educated lymphocytes provides a platform for subsequent post-transplant vaccination. To date, 26 patients have been enrolled in cohort 1 and 9 patient have been enrolled in cohort 2. Adherent mononuclear cells were isolated from leukapheresis collections and cultured with GM-CSF and IL-4 for 5-7 days, then exposed to TNFα for 48-72 hours to generate mature DCs. DCs expressed co-stimulatory (mean CD86 70%) and maturation markers (mean CD83 55%). MM cells were isolated from bone marrow and were identified by their expression of CD38 or CD138. DC and MM cells were co-cultured with PEG and fusion cells were quantified by determining the percentage of cells that co-express unique DC and myeloma antigens. Mean yield of the DC and myeloma preparations was 1.72 × 108 and 6.6 × 107 cells, respectively. Mean fusion efficiency was 38% and the mean cell dose generated was 3.6 × 106 fusion cells. Mean viability of the DC, myeloma, and fusion preparations was 87%, 87%, and 78%, respectively. As a measure of their potency as antigen presenting cells, fusion cells potently stimulated allogeneic T cell proliferation in vitro. Mean stimulation indexes were 13, 60, and 32 for T cells stimulated by myeloma cells, DCs, and fusion cells at an APC: T cell ratio of 1:10. Adverse events judged to be potentially vaccine related were mild, and included injection site reactions, pruritis, myalgias, fever, chills, and tachycardia. ASCT was associated with suppression of measures of cellular immunity. Circulating CD4 cells were depressed in the post-transplant period and CD4:CD8 ratios remained inverted for greater than 10 months. Similarly, 65% of patients had a positive DTH response to candida antigen prior to transplant while only 21% demonstrated a positive response in the early post-transplant period. T cell response to PHA mitogen was transiently depressed post-transplant with mean stimulation indexes of 79, 10, 26, 36, and 63 prior to transplant, 1, 2, 3, and 6 months post-transplant, respectively. Consistent with these findings, in vitro T cell responses to tetanus toxoid were blunted in the post-transplant period. In contrast, a significant increase in circulating tumor reactive lymphocytes was noted, as determined by T cell expression of IFN by CD4 and CD8 cells following ex vivo coculture with autologous myeloma cell lysate (Mean percentage of tumor reactive CD8 cells was 1 and 7.7 pre and post-transplant, respectively; mean percentage of CD4 cells was 0.9 and 3.2). A further amplification of tumor reactive lymphocytes was seen with vaccination in a subset of patients (mean percentage of CD4 and CD8 tumor reactive T cells was 6.4 and 13.4, respectively). In the post-transplant period, regulatory T cells fell to minimal levels. To date, 23 patients have completed follow up and were evaluable for clinical response. 3 patients achieved CR at 1 month following ASCT. Of note, an additional 7 patients obtained a CR following completion of vaccinations, suggesting a role for post-transplant immunotherapy in mediating elimination of disease. In summary, fusion cell vaccination in conjunction with ASCT was well tolerated, stimulated anti-tumor immunity and was associated with the induction of post-transplant complete response. Disclosures: Richardson: Millenium (Research Funding and Advisory Board), Celgene, Keryx, BMS, Merck, Johnson and Johnson (All Advisory Board): Membership on an entity's Board of Directors or advisory committees, Research Funding. Anderson:Millenium (Research Funding and Advisory Board: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees; Keryx: Membership on an entity's Board of Directors or advisory committees; BMS: Membership on an entity's Board of Directors or advisory committees; Merck: Membership on an entity's Board of Directors or advisory committees; Johnson and Johnson: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3425-3425
Author(s):  
Eduardo Espada ◽  
Ann E. Woolley ◽  
Jason Avigan ◽  
Edouard Forcade ◽  
Maria V.D. Soares ◽  
...  

Abstract Introduction: BK virus (BKV) seropositivity is highly prevalent (80-90%) in healthy adults, but BKV rarely causes significant clinical disease. However, immunologic control of BKV is often compromised after allogeneic hematopoietic stem cell transplantation (HSCT) and BKV reactivation can cause clinical disease, including hemorrhagic cystitis, in 5-68% of patients in the first year after HSCT. Immune reconstitution of BKV-specific T cells after HSCT has not been well defined, and the role of BKV-specific T cells in response to clinical infection has not been studied. Methods: From the DFCI 2010-2011 HSCT cohort (Rorije et al, BBMT 2014), 33 adult patients were selected who had urinary symptoms, were tested for urine BKV by PCR, and were diagnosed with BKV disease (cases; n=16) or did not have BKV reactivation (controls; n=17). Cases and controls were matched for cyclophosphamide use, acute GvHD and stem cell source. BKV-specific T cells were analyzed in 180 prospectively cryopreserved peripheral blood mononuclear cells (PBMC) samples (average 5.5/patient) across several timepoints (pre-HSCT and 1, 3, 6, 9, 12, 18 and 24 months post-HSCT) using cytokine flow cytometry (CFC). PBMC were stimulated for 6 hours with overlapping 15-mer peptides derived from BKV LT and VP1 proteins in the presence of brefeldin A, monensin and anti-CD28/49d. Results were compared with no-pepmix negative controls and Staphylococcal enterotoxin B positive controls. After stimulation, cells were stained with fluorochrome-conjugated antibodies specific for CD3, CD4, CD8, CD45RA, CCR7, CD57, CD107a, IFNγ, TNFα, IL-2, perforin (PERF) and granzyme B (GZMB). Laboratory assessments were blinded to clinical disease status. Results: Median age of the cohort was 51 years (IQR; 39-61); 79% were male and acute GvHD occurred in 64%. Conditioning included cyclophosphamide in 46% and ATG in 24%; cell source was peripheral blood stem cells in 81%, cord blood in 15% and bone marrow in 3%. Median time from HSCT to BKV testing was 60 days (23-181); median time to first positive test was 79 days (26-196) in BKV+ cases. Median duration of symptoms was 53 days (11-92) in cases, compared to 5 days in controls. The percentage of patients with BKV-specific T cells across timepoints is summarized in Figure 1. The frequency of BKV-specific degranulating (CD107a) T cells peaked at 6mo, where they could be detected in 37.5% and 58.3% of patients by reactivity of CD8+ and CD4+ T cells, respectively. Detection of BKV-specific T cells producing any cytokines (cytokine+) peaked at 9 mo (CD8+ in 19% and CD4+ in 38.1% of patients). BKV-specific IFNγ+, IL-2+, and TNFα+ T cells all peaked at 12mo after HSCT. Subsequently, detection of BKV-specific CD8+ T cells declined sharply while BKV-specific CD4+ T cells were maintained at higher levels 24 months after HSCT. Comparing BKV-specific immune responses in cases and controls, IFNγ+ CD8+ cells reconstituted faster in patients with clinical disease (first detected at 3 months vs. 9 mo), and were present in more patients at 9 months: 41.7% vs. 11.1%. A similar trend was noted for IL-2+ CD8+, IFNγ+ CD4+, and IL-2+ CD4+ T cells (Figure 2). BKV-specific CD8+ T cells were predominately central memory (CM) and terminally differentiated effector memory and their cytokine profile increased gradually after transplant; 19% produced at least two cytokines at 1 mo, compared to 71% at 12 mo. BKV-specific CD4+ T cells were predominately CM and effector memory and their cytokine profile also increased after transplant; 3.5% produced at least two cytokines at 1 mo, compared to 35% at 12 mo and 69.5% at 24 mo. PERF and/or GZMB were detected in 45-74% of BKV-specific CD107a CD8+ cells. CD107a CD8+ cells also produced IFNγ. Conclusion: BKV-specific T cells gradually reconstitute in the first 9 months after allogeneic HSCT. 30-40% of patients with clinical symptoms and 50-60% of patients with confirmed BKV disease had detectable BKV-specific T cells but BKV-specific immunity recovers more rapidly in patients with BKV disease. As CD8 and CD4 BKV-specific immunity recovers, T cells gradually become more functional with a memory phenotype. Our cohort is currently being expanded to assess the correlation between the development and expansion of BKV-specific T cells and clinical outcomes. Disclosures Koreth: kadmon corp: Membership on an entity's Board of Directors or advisory committees; takeda pharmaceuticals: Membership on an entity's Board of Directors or advisory committees; amgen inc: Consultancy; LLS: Research Funding; prometheus labs inc: Research Funding; millennium pharmaceuticals: Research Funding. Armand:Otsuka: Research Funding; BMS: Consultancy, Research Funding; Infinity: Consultancy; Sequenta: Research Funding; Roche: Research Funding; Merck & Co., Inc.: Consultancy, Research Funding; Sigma Tau: Research Funding; Tensha: Research Funding. Soiffer:GentiumSpA/Jazz Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees. Ritz:Kiadis: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 5230-5230 ◽  
Author(s):  
Matias Autio ◽  
Suvi-Katri Leivonen ◽  
Marja-Liisa Karjalainen-Lindsberg ◽  
Teijo Pellinen ◽  
Sirpa Leppa

Introduction Based on the cell of origin, diffuse large B-cell lymphoma (DLBCL) is divided into germinal center B-cell (GCB) and activated B-cell (ABC) like subtypes, which differ in their gene expression profiles and clinical presentation with the ABC DLBCLs showing a worse outcome in response to R-CHOP immunochemotherapy. However, composition of the tumor microenvironment (TME) of these molecular subtypes has not been characterized. Methods We used Hans algorithm to determine the molecular subtypes (GCB vs non-GCB) and multiplexed immunohistochemistry (mIHC) to characterize tumor infiltrating T-cell phenotypes, including cytotoxic T-cells (CTLs; CD8, Granzyme B, OX40, Ki67), T regulatory cells (Tregs; CD3, CD4, FoxP3), Th1 effector cells (CD3, CD4, TBET) and T-cell immune checkpoint (CD3, CD4, CD8, PD1, TIM3, LAG3) in 165 primary DLBCLs. The findings were correlated with the expression of human leukocyte antigens (HLA) I and II (beta-2 microglobulin (B2M), HLA-ABC and HLA-DR), and outcome of the patients treated with R-CHOP-like immunochemotherapy. Results In the whole cohort, 82 (50%) cases were classified as GCB and 83 (50%) as non-GCB DLBCLs. In the GCB subtype, cytotoxic T-cells were more often PD1+, and T-cells FoxP3+than in the non-GCB subtype (Figure 1A-B). Furthermore, GCB DLBCLs tended to be more commonly HLA-DR+(p=0.102). In the non-GCB DLBCLs in turn, HLA I positivity was more frequent (B2M, P=0.007; HLA-ABC, p=0.108), cytotoxic T-cells more often granzyme B+(Figure 1C), T-cells TBET+(p=0.018) and LAG3+TIM3+(p=0.033). A high proportion of granzyme B+cells (p=0.002), PD1+cells (p=0.02) and TIM3+CD4+T-cells (p=0.006) from all cells translated to adverse overall survival (OS) in the patients with non-GCB DLBCL, all independent of the IPI. In contrast, a high proportion of TIM3+cells (p=0.015), and FOXP3+TBET+T-cells (p=0.005) from all cells were associated with poor OS in the patients with GCB DLBCL, also independent of the IPI. Conclusions TME differs significantly between GCB and non-GCB DLBCLs and has subtype-specific prognostic impact on survival. Figure 1 Disclosures Leppa: Roche: Honoraria, Research Funding; Takeda: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Bayer: Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen-Cilag: Research Funding; Novartis: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1973-1973 ◽  
Author(s):  
Luca Bernabei ◽  
Alfred L. Garfall ◽  
J. Joseph Melenhorst ◽  
Simon F Lacey ◽  
Edward A. Stadtmauer ◽  
...  

Abstract Background: Autologous T cells expressing a chimeric antigen receptor (CAR) specific for B-cell maturation antigen (CART-BCMA cells) show activity in refractory MM, but relapses remain common. Anti-PD-1 antibodies (Abs) augment CAR T cell activity pre-clinically, and induced CAR T cell re-expansion and responses in DLBCL patients progressing after CD19-specific CAR T cells (Chong et al, Blood 2017). The IMiDs lenalidomide (len) and pomalidomide (pom) may enhance efficacy, but also toxicity, of both CAR T cells and PD-1 inhibitors in MM. Elotuzumab (elo) has clinical anti-MM activity in combination with IMiDs and dexamethasone (dex), and synergizes with anti-PD-1 Ab in pre-clinical models. Methods: We previously described outcomes of 25 subjects enrolled on our phase 1 study of CART-BCMA cells in relapsed/refractory MM (Cohen et al, ASH 2017, #505). We identified and retrospectively reviewed 5 subjects who progressed after CART-BCMA and received a PD-1 inhibitor (pembrolizumab (pembro)) combination as their next therapy. Responses were assessed by IMWG criteria. CART-BCMA levels were assessed by flow cytometry and qPCR pre-treatment, 2-4 weeks after first pembro dose, then q4 weeks until progression. Pembro dosing was 200mg every 3 weeks; dex dosing was 20-40mg/week. Results: Characteristics of the 5 subjects are in the Table. Median prior lines was 9; all had high-risk cytogenetics. All were refractory to pom, 2 to pembro/pom/dex, and 1 to elo. Best response to CART-BCMA was PR in 2, MR in 2, and PD in 1. Median time from CART-BCMA to pembro-based therapy was 117 days. All patients still had CART-BCMA cells detectable by qPCR, with 2 (pts. 07 and 21) still detectable by flow, at initiation of salvage therapy. The first pt. (02) received pembro/pom/dex and had MR but progressed at 2 months, with no detectable CART-BCMA re-expansion. The second pt. (07) had rapidly-progressing kappa light chain MM 2 months post-CART-BCMA and had previously progressed on pembro/pom/dex. He started elo/pembro/pom/dex and had MR at day 12 (free kappa 1446 to 937 mg/L), associated with robust expansion of CART-BCMA cells (875.64 to 20505.07 copies/µg DNA by qPCR; 0.7% to 6.4% of peripheral CD3+ cells by flow). Re-expanded CART-BCMA cells were predominantly CD8+ and highly activated (89% HLA-DR+, up from 18% pre-therapy). This response was short-lived, however, with progression 1 week later, and return of CART-BCMA levels to baseline at week 5. Three subsequent subjects then received elo/pembro/dex with either len or pom; with 2 MR and 1 SD, and PFS of 3 to 4 months. None had re-expansion of CART-BCMA cells. Non-specific immune modulation was observed and included altered CD4:CD8 T cell ratio (n=5), increased NK cell/decreased T cell frequency (n=4), and HLA-DR upregulation on CAR-negative T cells (n=2). More detailed phenotyping of CART and other immune cells, including PD-1 expression, is ongoing. With regard to toxicity, pt. 02 had self-limiting low-grade fevers and myalgias 4 weeks after pembro/pom/dex, associated with mild elevation in ferritin/CRP, suggestive of mild CRS. No other CRS was noted, including pt. 07 despite CART-BCMA re-expansion. One patient (17) developed recurrent expressive aphasia starting 2 months after elo/pembro/pom/dex, without signs of CRS and no observed expansion of CART-BCMA cells in blood or CSF. This resolved with stopping therapy and brief steroid taper. Conclusions: This study demonstrates that a PD1-inhibitor combination can induce CAR T cell re-expansion and anti-MM response in a MM patient progressing after CART-BCMA therapy. Since this patient previously progressed on pembro/pom/dex, the observed clinical activity was likely related to the CAR T cells, with elotuzumab also possibly contributing. However, this effect was very transient; re-expansion occurred infrequently (1/5 patients); and neurotoxicity was observed (though its relationship to the CAR T cells is unclear). This makes it difficult to endorse this specific salvage regimen. Nonetheless, this proof-of-principle observation suggests that a subset of patients may respond to checkpoint blockade or other immune-modulating approaches following BCMA CAR T cell therapy, meriting further study. Table. Table. Disclosures Garfall: Kite Pharma: Consultancy; Novartis: Research Funding; Amgen: Research Funding; Bioinvent: Research Funding. Melenhorst:novartis: Patents & Royalties, Research Funding; Incyte: Research Funding; Shanghai UNICAR Therapy, Inc: Consultancy; Casi Pharmaceuticals: Consultancy; Parker Institute for Cancer Immunotherapy: Research Funding. Lacey:Parker Foundation: Research Funding; Tmunity: Research Funding; Novartis Pharmaceuticals Corporation: Patents & Royalties; Novartis Pharmaceuticals Corporation: Research Funding. Stadtmauer:Janssen: Consultancy; AbbVie, Inc: Research Funding; Amgen: Consultancy; Takeda: Consultancy; Celgene: Consultancy. Vogl:Karyopharm Therapeutics: Consultancy. Plesa:Novartis: Research Funding. Young:Novartis: Patents & Royalties, Research Funding. Levine:Novartis: Consultancy, Patents & Royalties, Research Funding; Tmunity Therapeutics: Equity Ownership, Research Funding; Incysus: Consultancy; Cure Genetics: Consultancy; CRC Oncology: Consultancy; Brammer Bio: Consultancy. June:Immune Design: Membership on an entity's Board of Directors or advisory committees; Immune Design: Membership on an entity's Board of Directors or advisory committees; Tmunity Therapeutics: Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties, Research Funding; Celldex: Consultancy, Membership on an entity's Board of Directors or advisory committees; Tmunity Therapeutics: Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Patents & Royalties, Research Funding; Novartis Pharmaceutical Corporation: Patents & Royalties, Research Funding; Novartis Pharmaceutical Corporation: Patents & Royalties, Research Funding. Milone:Novartis: Patents & Royalties. Cohen:Bristol Meyers Squibb: Consultancy, Research Funding; Celgene: Consultancy; Novartis: Research Funding; Poseida Therapeutics, Inc.: Research Funding; Kite Pharma: Consultancy; GlaxoSmithKline: Consultancy, Research Funding; Seattle Genetics: Consultancy; Janssen: Consultancy; Oncopeptides: Consultancy.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1819-1819
Author(s):  
Homare Eda ◽  
Loredana Santo ◽  
Diana D. Cirstea ◽  
Samantha Pozzi ◽  
Miriam Canavese ◽  
...  

Abstract Abstract 1819 Objectives: Osteoblasts (OB) and osteoclasts (OC), are an integral part of the bone microenvironment, and play a crucial role in myeloma growth and survival. Their imbalance results in osteolytic disease and elucidating the mechanisms underlying osteolytic lesions is important not only for the improvement of osteolytic bone disease but also for the treatment of multiple myeloma (MM). The osteocyte-secreted protein sclerostin, encoded by the SOST gene, is a potent inhibitor of osteoblastogenesis. It is regarded as a good target for osteoporosis treatment, but its role in MM remains to be determined. Our objective was to study the role of sclerostin in MM bone disease and determine if sclerostin directed strategies were a reasonable approach in MM. Methods and Results: Sclerostin concentration in patients' blood plasma and MM cell line supernatant stimulated by IL-6, FGF-2, TNFalpha, BMP7 and TGFbeta was detected by ELISA (ALPCO immunoassays). Increased level of sclerostin was detected in MM patient plasma (n=20, median: 4.73 ng/mL, range: 1.5–19.5 ng/mL). Plasma concentrations were significantly higher (p<0.01) when compared to sclerostin concentration in the plasma of leukemia patients (n=3), gastric cancer patients (n=40) and healthy volunteers (n=4). High sclerostin levels were not associated with extent of bone disease but rather correlated with tumor burden (High B2M, creatinine and LDH, and low Hb) suggesting an autocrine loop for sclerostin production. Because sclerostin is derived from mature OB or orteocytes, we measured levels during OB differentiation but we were unable to detect increased levels. We then measured sclerostin levels in RPMI-8226 MM cell line supernatant either alone or stimulated by cytokines D Systems). Conclusions: These data demonstrate that increased sclerostin levels in MM patients inhibit osteoblastogenesis and stimulate osteoclastogenesis. Taken together, sclerostin may be good target to inhibit myeloma bone disease and help restore normal bone homeostasis. Disclosures: Raje: Celgene: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; Millenium: Membership on an entity's Board of Directors or advisory committees; Astra Zeneca: Research Funding; Amgen: Membership on an entity's Board of Directors or advisory committees; Acetylon: Research Funding.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 776-776
Author(s):  
Claire Roddie ◽  
Maeve A O'Reilly ◽  
Maria A V Marzolini ◽  
Leigh Wood ◽  
Juliana Dias Alves Pinto ◽  
...  

Introduction: 2nd generation CD19 CAR T cells show unprecedented efficacy in B-ALL, but several challenges remain: (1) scaling manufacture to meet patient need and (2) feasibility of generating products from lymphopenic patients post allogeneic stem cell transplant (allo-SCT). To overcome these issues we propose: (1) use of the CliniMACS Prodigy (Miltenyi Biotec), a semi-automated cGMP platform that simplifies CAR T cell manufacture and (2) the use of matched donor T cells to overcome the challenge posed by patient lymphopenia, albeit this may come with a heightened risk of graft versus host disease (GvHD). CARD (NCT02893189) is a Phase I study of matched donor derived CD19 CAR T cells generated on the CliniMACS Prodigy in 14 adult patients with relapsed/refractory (r/r) B ALL following allo-SCT. We additionally explore the requirement for lymphodepletion (LD) in the allogeneic CAR T cell setting and report on the incidence of GvHD with this therapy. Methods: Manufacturing: CARD utilises non-mobilised matched donor leucapheresate to manufacture 2nd generation CD19CAR T cells using a closed CliniMACS® Prodigy/ TransACTTM process. Study design: Eligible subjects are aged 16-70y with r/r B ALL following allo SCT. Study endpoints include feasibility of CD19CAR T cell manufacture from allo-SCT donors on the CliniMACS Prodigy and assessments of engraftment and safety including GvHD. To assess the requirement for LD prior to CD19CAR T cells in lymphopenic post-allo-SCT patients, the study is split into Cohort 1 (no LD) and Cohort 2 (fludarabine (30 mg/m2 x3) and cyclophosphamide (300mg/m2 x3)). To mitigate for the potential GvHD risk, cell dosing on study mirrors conventional donor lymphocyte infusion (DLI) schedules and is based on total CD3+ (not CAR T) cell numbers: Dose 1=1x106/kg CD3+ T cells; Dose 2= 3x106/kg CD3+ T cells; Dose 3= 1x107/kg CD3+ T cells. Results: As of 26 July 2019, 17 matched allo SCT donors were leukapheresed and 16 products were successfully manufactured and QP released. Patient demographics are as follows: (1) median patient age was 43y (range 19-64y); (2) 4/17 had prior blinatumomab and 5/17 prior inotuzumab ozogamicin; (3) 7/17 had myeloablative allo SCT and 10/17 reduced intensity allo SCT of which 6/17 were sibling donors and 12/17 were matched unrelated donors. No patients with haploidentical transplant were enrolled. To date, 12/16 patients have received at least 1 dose of CD19CAR T cells: 7/16 on Cohort 1 and 5/16 on Cohort 2 (2/16 are pending infusion on Cohort 2 and 2/16 died of fungal infection prior to infusion). Median follow-up for all 12 patients is 22.9 months (IQR 2.9-25.9; range 0.7 - 25.9). At the time of CAR T cell infusion, 7/12 patients were in morphological relapse with &gt;5% leukemic blasts. Despite this, CD19CAR T cells were administered safely: only 2/12 patients experienced Grade 3 CRS (UPenn criteria), both in Cohort 1, which fully resolved with Tocilizumab and corticosteroids. No patients experienced ≥Grade 3 neurotoxicity and importantly, no patients experienced clinically significant GvHD. In Cohort 1 (7 patients), median peak CAR expansion by flow was 87 CD19CAR/uL blood whereas in Cohort 2 (5 patients to date), median peak CAR expansion was 1309 CD19CAR/uL blood. This difference is likely to reflect the use of LD in Cohort 2. CAR T cell persistence by qPCR in Cohort 1 is short, with demonstrable CAR in only 2/7 treated patients at Month 2. Data for Cohort 2 is immature, but this will also be reported at the meeting in addition to potential mechanisms underlying the short persistence observed in Cohort 1. Of the 10 response evaluable patients (2/12 pending marrow assessment), 9/10 (90%) achieved flow/molecular MRD negative CR at 6 weeks. 2/9 responders experienced CD19 negative relapse (one at M3, one at M5) and 3/9 responders experienced CD19+ relapse (one at M3, one at M9, one at M12). 4/10 (40%) response evaluable patients remain on study and continue in flow/molecular MRD negative remission at a median follow up of 11.9 months (range 2.9-25.9). Conclusions: Donor-derived matched allogeneic CD19 CAR T cells are straightforward to manufacture using the CliniMACS Prodigy and deliver excellent early remission rates, with 90% MRD negative CR observed at Week 6 in the absence of severe CAR associated toxicity or GvHD. Peak CAR expansion appears to be compromised by the absence of LD and this may lead to a higher relapse rate. Updated results from Cohorts 1 and 2 will be presented. Disclosures Roddie: Novartis: Consultancy; Gilead: Consultancy, Speakers Bureau; Celgene: Consultancy, Speakers Bureau. O'Reilly:Kite Gilead: Honoraria. Farzaneh:Autolus Ltd: Equity Ownership, Research Funding. Qasim:Autolus: Equity Ownership; Orchard Therapeutics: Equity Ownership; UCLB: Other: revenue share eligibility; Servier: Research Funding; Bellicum: Research Funding; CellMedica: Research Funding. Linch:Autolus: Membership on an entity's Board of Directors or advisory committees. Pule:Autolus: Membership on an entity's Board of Directors or advisory committees. Peggs:Gilead: Consultancy, Speakers Bureau; Autolus: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 779-779 ◽  
Author(s):  
Zinaida Good ◽  
Jay Y. Spiegel ◽  
Bita Sahaf ◽  
Meena B. Malipatlolla ◽  
Matthew J. Frank ◽  
...  

Axicabtagene ciloleucel (Axi-cel) is an autologous anti-CD19 chimeric antigen receptor (CAR) T-cell therapy approved for the treatment of relapsed or refractory diffuse large B-cell lymphoma (r/r DLBCL). Long-term analysis of the ZUMA-1 phase 1-2 clinical trial showed that ~40% of Axi-cel patients remained progression-free at 2 years (Locke et al., Lancet Oncology 2019). Those patients who achieved a complete response (CR) at 6 months generally remained progression-free long-term. The biological basis for achieving a durable CR in patients receiving Axi-cel remains poorly understood. Here, we sought to identify CAR T-cell intrinsic features associated with CR at 6 months in DLBCL patients receiving commercial Axi-cel at our institution. Using mass cytometry, we assessed expression of 33 surface or intracellular proteins relevant to T-cell function on blood collected before CAR T cell infusion, on day 7 (peak expansion), and on day 21 (late expansion) post-infusion. To identify cell features that distinguish patients with durable CR (n = 11) from those who developed progressive disease (PD, n = 14) by 6 months following Axi-cel infusion, we performed differential abundance analysis of multiparametric protein expression on CAR T cells. This unsupervised analysis identified populations on day 7 associated with persistent CR or PD at 6 months. Using 10-fold cross-validation, we next fitted a least absolute shrinkage and selection operator (lasso) model that identified two clusters of CD4+ CAR T cells on day 7 as potentially predictive of clinical outcome. The first cluster identified by our model was associated with CR at 6 months and had high expression of CD45RO, CD57, PD1, and T-bet transcription factor. Analysis of protein co-expression in this cluster enabled us to define a simple gating scheme based on high expression of CD57 and T-bet, which captured a population of CD4+ CAR T cells on day 7 with greater expansion in patients experiencing a durable CR (mean±s.e.m. CR: 26.13%±2.59%, PD: 10.99%±2.53%, P = 0.0014). In contrast, the second cluster was associated with PD at 6 months and had high expression of CD25, TIGIT, and Helios transcription factor with no CD57. A CD57-negative Helios-positive gate captured a population of CD4+ CAR T cells was enriched on day 7 in patients who experienced progression (CR: 9.75%±2.70%, PD: 20.93%±3.70%, P = 0.016). Co-expression of CD4, CD25, and Helios on these CAR T cells highlights their similarity to regulatory T cells, which could provide a basis for their detrimental effects. In this exploratory analysis of 25 patients treated with Axi-cel, we identified two populations of CD4+ CAR T cells on day 7 that were highly associated with clinical outcome at 6 months. Ongoing analyses are underway to fully characterize this dataset, to explore the biological activity of the populations identified, and to assess the presence of other populations that may be associated with CAR-T expansion or neurotoxicity. This work demonstrates how multidimensional correlative studies can enhance our understanding of CAR T-cell biology and uncover populations associated with clinical outcome in CAR T cell therapies. This work was supported by the Parker Institute for Cancer Immunotherapy. Figure Disclosures Muffly: Pfizer: Consultancy; Adaptive: Research Funding; KITE: Consultancy. Miklos:Celgene: Membership on an entity's Board of Directors or advisory committees; BMS: Membership on an entity's Board of Directors or advisory committees; Kite-Gilead: Membership on an entity's Board of Directors or advisory committees, Research Funding; AlloGene: Membership on an entity's Board of Directors or advisory committees; Precision Bioscience: Membership on an entity's Board of Directors or advisory committees; Miltenyi Biotech: Membership on an entity's Board of Directors or advisory committees; Becton Dickinson: Research Funding; Adaptive Biotechnologies: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees, Research Funding; Juno: Membership on an entity's Board of Directors or advisory committees. Mackall:Vor: Other: Scientific Advisory Board; Roche: Other: Scientific Advisory Board; Adaptimmune LLC: Other: Scientific Advisory Board; Glaxo-Smith-Kline: Other: Scientific Advisory Board; Allogene: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Apricity Health: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Unum Therapeutics: Equity Ownership, Membership on an entity's Board of Directors or advisory committees; Obsidian: Research Funding; Lyell: Consultancy, Equity Ownership, Other: Founder, Research Funding; Nektar: Other: Scientific Advisory Board; PACT: Other: Scientific Advisory Board; Bryologyx: Other: Scientific Advisory Board.


Sign in / Sign up

Export Citation Format

Share Document