scholarly journals Tumor associated antigen specific T cells with nivolumab are safe and persist in vivo in rel/ref Hodgkin Lymphoma

Author(s):  
Hema Dave ◽  
Madeline Terpilowski ◽  
Mimi Mai ◽  
Keri Toner ◽  
Melanie Grant ◽  
...  

Hodgkin Lymphoma (HL) Reed Sternberg cells express tumor associated antigens (TAA) that are potential targets for cellular therapies. We recently demonstrated that TAA specific T cells (TAA-T) targeting WT1, PRAME and Survivin were safe and associated with prolonged time to progression in solid tumors. Hence, we evaluated whether TAA-T when given alone or with nivolumab were safe and could elicit anti-tumor effects in vivo in patients with relapsed/refractory (r/r) HL. Ten patients were infused TAA-T (8 autologous; 2 allogeneic) for active HL(n=8) or as adjuvant therapy after hematopoietic stem cell transplant (n=2) at cumulative doses ranging from 0.5 X107 to 4 X107cells/m2. Six patients received nivolumab priming before TAA-T and continued until disease progression or unacceptable toxicity. All 10 products recognized 1 or more TAAs, were polyfunctional and did not demonstrated autoreactivity. Patients were monitored for safety for six weeks following the TAA-T and for response until disease progression. The infusions were safe with no clear dose limiting toxicities. Patients receiving TAA-T as adjuvant therapy remain in continued remission at 2+ years. Of the 8 patients with active disease,1 patient had a complete response and 7 had stable disease at 3 months, 3 of whom remain with stable disease at 1 year. Antigen spreading and long-term persistence of TAA-T in vivo were observed in responding patients. Nivolumab priming impacted the TAA-T recognition and persistence. In conclusion, treatment of r/r HL patients with TAA-T alone or in combination with nivolumab was safe and produced promising results (clinicaltrials.gov NCT022039303 and NCT03843294).

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 18-18 ◽  
Author(s):  
Hema Dave ◽  
Mimi Mai ◽  
Madeline Terpilowski ◽  
Keri Toner ◽  
Maja Stanojevic ◽  
...  

Background: Hodgkin Lymphoma (HL) Reed Sternberg cells express tumor associated antigens (TAA) that are potential targets for cellular therapies. We recently demonstrated that TAA specific T cells (TAA-T) targeting WT1, PRAME and Survivin were safe and associated with prolonged time to progression in solid tumors (Hont JCO 2019). Hence, we evaluated whether TAA-T cells are safe and elicit anti-tumor effects in patients with relapsed/refractory (rel/ref) HL. We further evaluated the safety of Nivolumab following the TAA-T infusion and its effect on the persistence of the TAA-T cells in vivo. Methods: TAA-T products were generated from patients or healthy donors on 2 trials (NCT02203903; NCT03843294). Thirteen patients underwent procurement for product generation and 10 patients (2 allogeneic; 8 autologous) were infused TAA-T for rel/ref HL or as consolidation after autologous hematopoietic stem cell transplant (HSCT) at cumulative doses ranging from 0.5 X107 to 4 X107cells/m2. Patients were monitored for six weeks for safety and for response until disease progression. Seven patients received Nivolumab starting at 8 weeks after the first TAA-T infusion until disease progression or unacceptable toxicity. Results: TAA-T products (n=10) were polyclonal CD3+ T cells (Median 97%; 80.9-99.5%), comprised predominantly of CD4+ helper T cells (Median 10.5%; 1.74-20%) and CD8+ cytotoxic T cells (Median 70%; 29.3-87.5%). Specificity of TAA-T products was tested using Interferon-ϒ(INFϒ)-enzyme-linked immunospot (ELIspot) assay and defined as ≥ 2x spot-forming cells (SFC)/2.5X105cells against the tumor antigen as compared to irrelevant control antigen Actin(Figure 1). The median TAA specificity of the products was 2 antigens (range 0-3). All products were polyfunctional secreting INF-ϒ and TNF-α upon restimulation with tumor antigens (Fig 1). Median age of patients was 36yrs (range16-53). Patients had received a median 6 lines of therapy including HSCT prior to receiving TAA-T. Median follow-up post TAA-T#1 was 6 months (range 32 days-2.5yrs). There were no dose limiting toxicities observed within the 6 week safety monitoring period. In patients receiving Nivolumab post TAA-T, there were no increased immune related events over expected. One patient had Grade 3 seizures, possibly related to Nivolumab, 2 patients developed hypothyroidism requiring thyroid supplements and one patient developed myositis and discontinued Nivolumab after 5 months. The 2 patients who received TAA-T (1 donor derived and one autologous) as consolidation post HSCT achieved a continued complete remission (CCR) for 2+ years. Of the 8 patients with rel/ref HL at the time of infusion, 1 had disease progression at 6 weeks. He then received Nivolumab off protocol and achieved complete remission (CR) but developed Grade 4 GVHD. The remaining 7 patients had stable disease (SD) at 6 weeks. At a median follow-up of 6 months (32 days-2.5 years), 1 patient had progressive disease(PD) at 3 months, 1 patient had a complete metabolic response at 6 months and proceeded to allogeneic HSCT for definitive cure. 2 patients had PD at 6 months and the other 2 patients continue with SD at 6 months and remain on Nivolumab (Fig 1). All patients with objective responses (stable disease or better) recovered functional TAA-T cells in the peripheral blood at 3 months as detected by anti-Interferon-ϒ ELISPOT and reported as mean SFC/1 X105 cells for WT1(14±SD18.1); PRAME (17.4±15.3) and Survivin (4.5±7) compared to those with progressive disease with mean SFC/1 X105 cells for WT1 1.4(±2.3); PRAME (6.7±15.5) and Survivin (0.8±1.2). To evaluate TAA-T persistence, unique T cell receptor clonotypes defined in the TAA-T product and not present at baseline were detected in the peripheral blood 6 weeks post TAA-T, long-term persistence data and evaluating the effect on the TCR repertoire when adding nivolumab are pending. Conclusion: TAA-T cells given in combination with Nivolumab were safe when administered to patients with rel/ref HL with prolonged clinical responses (ranging from SD to CCR) observed in multiply relapsed patients. Disclosures Glenn: Genentech: Research Funding. Hanley:Mana Therapeutics: Honoraria, Other: Board Member; Cellevolve: Honoraria, Other: Board(Scientific Advisory Board). Bollard:Mana Therapeutics: Other: IP.


2020 ◽  
Vol 4 (14) ◽  
pp. 3357-3367 ◽  
Author(s):  
Rafet Basar ◽  
May Daher ◽  
Nadima Uprety ◽  
Elif Gokdemir ◽  
Abdullah Alsuliman ◽  
...  

Abstract Virus-specific T cells have proven highly effective for the treatment of severe and drug-refractory infections after hematopoietic stem cell transplant (HSCT). However, the efficacy of these cells is hindered by the use of glucocorticoids, often given to patients for the management of complications such as graft-versus-host disease. To address this limitation, we have developed a novel strategy for the rapid generation of good manufacturing practice (GMP)–grade glucocorticoid-resistant multivirus-specific T cells (VSTs) using clustered regularly interspaced short palindromic repeats (CRISPR)–CRISPR-associated protein 9 (Cas9) gene-editing technology. We have shown that deleting the nuclear receptor subfamily 3 group C member 1 (NR3C1; the gene encoding for the glucocorticoid receptor) renders VSTs resistant to the lymphocytotoxic effect of glucocorticoids. NR3C1-knockout (KO) VSTs kill their targets and proliferate successfully in the presence of high doses of dexamethasone both in vitro and in vivo. Moreover, we developed a protocol for the rapid generation of GMP-grade NR3C1 KO VSTs with high on-target activity and minimal off-target editing. These genetically engineered VSTs promise to be a novel approach for the treatment of patients with life-threatening viral infections post-HSCT on glucocorticoid therapy.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4828-4828
Author(s):  
Lauren S. Maeda ◽  
Richard T. Hoppe ◽  
Saul A. Rosenberg ◽  
Sandra J. Horning ◽  
Ranjana H. Advani

Abstract Abstract 4828 Purpose: Stanford V is an abbreviated combined modality approach for the treatment of advanced stage Hodgkin lymphoma (HL). This regimen was developed with the aim of shortening the duration of chemotherapy, limiting the radiotherapy (RT) to a modified involved field and thereby potentially reducing short and long term toxicity while maintaining or improving cure rates. Specifically the chemotherapy regimen has significantly lower cumulative doses of adriamycin, bleomycin and alkylating agents compared to other standard regimens such as ABVD or escalated BEACOPP. We have previously reported excellent outcomes with this regimen with a freedom from progression (FFP) of 89% and overall survival (OS) of 96% (Horning, S.J., et al., J Clin Oncol 2002, 20:630-7). The purpose of this study was to determine the outcome of patients (pts) treated with secondary therapy after failing Stanford V. Methods: Pts with advanced stage HL who had either refractory disease or had relapsed after primary therapy with Stanford V, were retrospectively identified from the HL database. We analyzed this group of patients for risk factors, salvage therapy, and treatment outcome. Results: Between May 1989 and March 2003, 167 pts were treated on protocol. At a median follow-up of 12.8 years the outcomes are excellent with a 10-year FFP and OS of 87% and 93%, respectively. Therapy failed in 19 pts (11%) of which 16 relapsed and 3 did not complete the intended treatment (disease progression n=2, and muscle pain and hyponatremia n=1). The median age of pts who failed therapy was 31 years (range 21 – 58) with a median time to progression of 5.1 months (range 0.2 – 41.4). 11 pts relapsed at 0 to 12 months from completion of therapy and 8 pts relapsed at > 12 months. At initial diagnosis 5 had stage I/II disease with bulky mediastinum, 5 stage III and 9 stage IV disease. The International Prognostic Score (IPS) at initial diagnosis was 0–1 (n=4), 2–3 (n=10) and 4–7 (n=5). 13/19 (68%) pts relapsed outside the RT field, 2 infield, 3 both infield and outside and 1 unknown. 7/19 pts in whom therapy failed had bulky disease and of these 5 failed outside the RT field. Relapse was detected clinically in 12 pts, and on surveillance positron emission tomography scan performed every 3 to 6 months in 5 pts who were asymptomatic (2 pts unknown). 14/19 (74%) pts received secondary therapy with a platinum-containing regimen (ICE or DHAP) with an overall response rate (ORR) of 91% (complete response [CR] n=1, partial response [PR] n=9, progressive disease [PD] n=1, unknown response n=3), followed by an autologous hematopoietic stem cell transplant. 5 pts were treated with non-transplant regimens consisting of chemotherapy with MOPP/ABV + RT (n=2), ChlVPP (n=1), oral cyclophosphamide (n=1) and procarbazine/alkeran/adriamycin/etoposide (n=1), with an ORR of 80% (CR n=4). Reasons for non-transplant therapy were neuropathy (n=1), pt preference (n=1), liver disease (n=1), and unknown (n=2). 11 of the 19 pts in whom Stanford V failed died (disease progression n=3, second malignancy n=2, graft failure n=1, infection n=1, liver failure n=1, cardiac arrest n=1, suicide n=1 and unknown n=1). At a median follow-up of 8 years, the disease-specific survival (DSS), FFP and OS for pts with refractory or relapsed disease after Stanford V was 84%, 63% and 42%, respectively. Outcome of pts who relapsed within a year was worse than pts who relapsed > 1 year with an OS of 36% versus 50%, respectively. There was no difference in FFP for these groups, 64% versus 63%, respectively. Conclusions: The outcome of pts with advanced HL is excellent with the Stanford V regimen. For the 11% of pts in whom front line therapy fails, secondary therapy is effective with a DSS of > 80%. The majority of pts (84%) failed at distant sites suggesting that more aggressive upfront chemotherapy may have been beneficial in these pts. Future efforts will aim at identifying this subset upfront. Pts who relapse within a year have a worse outcome despite salvage and for this subgroup, newer therapies are warranted. Disclosures: Horning: Genentech: Employment.


2020 ◽  
Vol 4 (14) ◽  
pp. 3443-3456
Author(s):  
Gloria Castellano-González ◽  
Helen M. McGuire ◽  
Fabio Luciani ◽  
Leighton E. Clancy ◽  
Ziduo Li ◽  
...  

Abstract Invasive fungal infections are a major cause of disease and death in immunocompromised hosts, including patients undergoing allogeneic hematopoietic stem cell transplant (HSCT). Recovery of adaptive immunity after HSCT correlates strongly with recovery from fungal infection. Using initial selection of lymphocytes expressing the activation marker CD137 after fungal stimulation, we rapidly expanded a population of mainly CD4+ T cells with potent antifungal characteristics, including production of tumor necrosis factor α, interferon γ, interleukin-17, and granulocyte-macrophage colony stimulating factor. Cells were manufactured using a fully good manufacturing practice–compliant process. In vitro, the T cells responded to fungal antigens presented on fully and partially HLA-DRB1 antigen–matched presenting cells, including when the single common DRB1 antigen was allelically mismatched. Administration of antifungal T cells lead to reduction in the severity of pulmonary and cerebral infection in an experimental mouse model of Aspergillus. These data support the establishment of a bank of cryopreserved fungus-specific T cells using normal donors with common HLA DRB1 molecules and testing of partially HLA-matched third-party donor fungus-specific T cells as a potential therapeutic in patients with invasive fungal infection after HSCT.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 223-223
Author(s):  
Ulrike Gerdemann ◽  
Usha L Katari ◽  
Jacqueline Keirnan ◽  
John A. Craddock ◽  
Janet Salinas ◽  
...  

Abstract Abstract 223 We have previously demonstrated that small numbers of ex vivo-expanded, trivirus-specific T cells targeting Epstein Barr virus (EBV), cytomegalovirus (CMV), and Adenovirus (Adv) are safe, proliferate in vivo and protect human subjects against all 3 viruses following HSCT. However, broader implementation is limited by the need for infectious virus (EBV) to establish an EBV-transformed B lymphoblastoid cell line (EBV-LCL), for clinical grade adenoviral vector, and by prolonged (6wks for EBV-LCL and 6wks for T cells) and complex manufacture. Moreover, competition between viral antigens limits extension to additional viruses. We now evaluate whether it is possible to make clinically effective T cell lines using methods that exclude all viral components and utilize simplified manufacturing technology. With NHLBI-Production Assistance for Cellular Therapies (PACT) support, 29 clinical-grade rCTL lines have been generated. From an initial 15×106 PBMCs, we prepared a median of 214±88 × 106 T cells (range 100–420×106) over 9–11 days by using dendritic cells (DCs) nucleofected with DNA plasmids encoding immunogenic EBV (LMP2, EBNA1 and BZLF1), Adv (Hexon and Penton), and CMV (pp65 and IE1) antigens, and expanding them with IL4+7 in gas permeable (G-Rex) devices. The rCTL lines were polyclonal, comprising both CD4+ (33±3%) and CD8+ (60.5±3%) cells, that expressed the activation and memory markers CD45RO+ CD62L+ (64.3±26.6%) and CD45RO+ CD62L- (17.4±14%). Twenty lines generated from donors that were seropositive for all three viruses demonstrated activity against all 3 targets - CMV (IE1: 359±100; pp65: 637±177 SFC/2×105), EBV (LMP2: 217±60, EBNA1: 67±19 and BZLF1: 111±31) and Adv (Hexon: 265±74, Penton: 191±53) - while 9 lines generated from donors who were CMV seronegative demonstrated activity exclusively against EBV (LMP2: 197±70, EBNA1: 145±51 and BZLF1: 239±84) and Adv (Hexon: 271±96, Penton: 254±90). None of the lines reacted against recipient PHA blasts (median spontaneous Cr51 release of 0% at a 20:1 effector to target cell ratio). To date we have administered these lines to 10 recipients of allogeneic HSCT. Five patients received dose level (DL) 1 (5×106/m2), 2 received DL2 (1×107/m2) and 3 had DL3 (2×107/m2) of this phase I/II study. Three patients were infused as treatment for CMV, 2 for Adv, 2 for EBV, 1 for EBV+Adv, and 2 for CMV+Adv. Our major anticipated concern was that these once stimulated, unselected rCTLs might cause GvHD in vivo, but that was not the case. One patient developed a skin rash 2 weeks after rCTLs but no other toxicity related to the infused cells was observed. Eight of the 10 treated patients including one patient with a biopsy-proven EBV lymphoma and the 3 patients with double reactivations had complete responses to rCTL therapy with a return of viral load to normal and resolution of all other symptoms. Response was associated with an increase in the frequency of virus-specific T cells detected in the peripheral blood against the infecting virus. For CMV there was an increase from a median of 0.5 to 96 and 1 to 277 SFC/4×105 IE1 and pp65-specific T cells respectively 3–6wks post-infusion; for Adv an increase from a mean of 0.5 to 137 and 0.5 to 99 SFC/4×105 Hexon and Penton-specific cells 2wks post-infusion, respectively, and for EBV an increase from 2.75 to 227, 1.5 to 39, and 1 to 188.5 SFC/4×105 EBNA1, LMP2, and BZLF1-specific T cells 2–4wks post-infusion, respectively. Two patients failed to respond. The first had a 3 year history of persistent CMV colitis despite high circulating CMV-specific precursors (297 IE1-specific and 193 pp65-specific T cells/4×105 PBMCs). Post rCTL we saw no increase in T cell precursor levels and no clinical improvement. The second was treated for an elevated EBV viral load but also had high pre-existing EBV-specific T cell precursors (60, 23, and 240 SFC/4×105 EBNA1, LMP2, and BZLF1-specific T cells). Again, post-rCTL we did not detect an increase in EBV-specific precursors and no response. Thus, infusion of rCTLs has been safe and in 8/10 patients was associated with the appearance of virus-reactive T cells directed against the infecting virus in peripheral blood and subsequent virus clearance. rCTLs have the potential to increase the availability of cell products for HSCT recipients and we are currently extending this platform to additional viruses, thereby broadening the spectrum of pathogens that can be targeted by adoptive transfer of a single T cell line. Disclosures: Off Label Use: IND cell therapy product.


Leukemia ◽  
2021 ◽  
Author(s):  
Julia Dahlhoff ◽  
Hannah Manz ◽  
Tim Steinfatt ◽  
Julia Delgado-Tascon ◽  
Elena Seebacher ◽  
...  

AbstractMultiple myeloma remains a largely incurable disease of clonally expanding malignant plasma cells. The bone marrow microenvironment harbors treatment-resistant myeloma cells, which eventually lead to disease relapse in patients. In the bone marrow, CD4+FoxP3+ regulatory T cells (Tregs) are highly abundant amongst CD4+ T cells providing an immune protective niche for different long-living cell populations, e.g., hematopoietic stem cells. Here, we addressed the functional role of Tregs in multiple myeloma dissemination to bone marrow compartments and disease progression. To investigate the immune regulation of multiple myeloma, we utilized syngeneic immunocompetent murine multiple myeloma models in two different genetic backgrounds. Analyzing the spatial immune architecture of multiple myeloma revealed that the bone marrow Tregs accumulated in the vicinity of malignant plasma cells and displayed an activated phenotype. In vivo Treg depletion prevented multiple myeloma dissemination in both models. Importantly, short-term in vivo depletion of Tregs in mice with established multiple myeloma evoked a potent CD8 T cell- and NK cell-mediated immune response resulting in complete and stable remission. Conclusively, this preclinical in-vivo study suggests that Tregs are an attractive target for the treatment of multiple myeloma.


2019 ◽  
Vol 3 (13) ◽  
pp. 2022-2034 ◽  
Author(s):  
Edward A. Stadtmauer ◽  
Thomas H. Faitg ◽  
Daniel E. Lowther ◽  
Ashraf Z. Badros ◽  
Karen Chagin ◽  
...  

Abstract This study in patients with relapsed, refractory, or high-risk multiple myeloma (MM) evaluated the safety and activity of autologous T cells engineered to express an affinity-enhanced T-cell receptor (TCR) that recognizes a peptide shared by cancer antigens New York esophageal squamous cell carcinoma-1 (NY-ESO-1) and L-antigen family member 1 (LAGE-1) and presented by HLA-A*02:01. T cells collected from 25 HLA-A*02:01-positive patients with MM expressing NY-ESO-1 and/or LAGE-1 were activated, transduced with self-inactivating lentiviral vector encoding the NY-ESO-1c259TCR, and expanded in culture. After myeloablation and autologous stem cell transplant (ASCT), all 25 patients received an infusion of up to 1 × 1010 NY-ESO-1 specific peptide enhanced affinity receptor (SPEAR) T cells. Objective response rate (International Myeloma Working Group consensus criteria) was 80% at day 42 (95% confidence interval [CI], 0.59-0.93), 76% at day 100 (95% CI, 0.55-0.91), and 44% at 1 year (95% CI, 0.24-0.65). At year 1, 13/25 patients were disease progression-free (52%); 11 were responders (1 stringent complete response, 1 complete response, 8 very good partial response, 1 partial response). Three patients remained disease progression-free at 38.6, 59.2, and 60.6 months post-NY-ESO-1 SPEAR T-cell infusion. Median progression-free survival was 13.5 months (range, 3.2-60.6 months); median overall survival was 35.1 months (range, 6.4-66.7 months). Infusions were well tolerated; cytokine release syndrome was not reported. No fatal serious adverse events occurred during study conduct. NY-ESO-1 SPEAR T cells expanded in vivo, trafficked to bone marrow, demonstrated persistence, and exhibited tumor antigen-directed functionality. In this MM patient population, NY-ESO-1 SPEAR T-cell therapy in the context of ASCT was associated with antitumor activity. This trial was registered at www.clinicaltrials.gov as #NCT01352286.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 1836-1836
Author(s):  
Shih-Shih Chen ◽  
Xiao J. Yan ◽  
Thomas M. Herndon ◽  
Clare C. Sun ◽  
Priyadarshini Ravichandran ◽  
...  

Abstract Chronic lymphocytic leukemia (CLL) cells actively participate in the formation of the tumor microenvironment (TME). The interplay of CLL cells and leukemia-supporting cells such as Th2 cells and regulatory T cells (Tregs) promotes a leukemia-supportive, immune-tolerant TME. In these supportive/tolerogenic niches of lymph nodes (LN) and bone marrow (BM), CLL cells slowly proliferate, and the rate of proliferation correlates with disease progression. However, how these dividing/recently-divided cells and other intraclonal CLL fractions interact with non-neoplastic cells to shape the TME to support growth and accelerate disease is unclear. To address these questions, matched LN and PB samples collected at day 13 after 2H2O ingestion from treatment-naïve patients (7 with stable disease and 7 with active disease) were sorted to determine in vivo growth rates of CLL cells within the proliferative fraction (PF, CXCR4DimCD5Bright), resting fraction (RF, CXCR4BrightCD5Dim) and intermediate fraction (IF, CXCR4IntCD5Int). In LN, PF cells had the highest 2H-DNA levels, and only the growth rate of PF but not IF or RF cells correlated with disease aggressiveness. In the PB, PF cells also had the highest growth rate; however, all 3 fractions (PF, IF, RF) had 2H-DNA levels that correlated with disease aggressiveness. Thus, PF cells from patients with aggressive clinical course undergo quicker transitions to the IF and then to the RF which might promote faster tissue homing and disease progression. Indeed, more PF cells from active than stable disease patients were found in the spleen (SP) and BM and less remained in PB, 18 hours post-injection into NSG mice. Gene expression profiling (GEP) was then performed on RF, IF, and PF from 7 paired PB and LN samples. GEP signatures of the PF from PB and LN were similar, consistent with 2H2O data that the PF are recent emigrants from TME. These GEP also inferred enhanced cell proliferation (CCND2, CDK2AP1), adhesion and motility (FERMT3, CD49d, CD11a, CD21), antigen presentation (CD1C), and promotion of T-cell trafficking (CCL3, CCL4) in LN CLL cells within the PF but not the IF or RF. Thus, CLL cells within the three intraclonal fractions might promote distinct biologic functions. To test this, we studied T cell responses stimulated by the CXCR4/CD5 fractions in vitro and in vivo. In an antigen-driven allo-MLR, the whole clone of CLL cells triggered the division of normal T cells, but PF induced the highest level of T-cell division that is ≥ 3 times more than any other fraction. Similarly, in an autologous polyclonal CD4 T cell response stimulated by anti-CD3/28 Dynabeads and IL-2, T-cell division was suppressed by unseparated CLL cells and each fraction. However, the least suppression was seen in T cells co-cultured with PF cells compared to those cultured alone or with IF or RF. In both settings, PF cells induced significantly more IL-4+ T cells, and RF cells triggered more Tregs. Similar numbers of Th1 cells were seen in all cultures. The RF and IF, but not the PF, produced the immunosuppressive cytokine IL-10. Finally, when dividing cases based on disease aggressiveness, significantly more T-cell division was triggered by PF and RF from active patients than stable patients; the percentages of Th1 and Th2 cells however were similar. These results were confirmed in vivo; in NSG mice injected with autologous T cells together with the PF, IF or RF sorted from 2 sets of active versus stable disease patients, PF from all 4 cases induced the highest levels of CLL B and T cell expansion in SP and BM, and RF from active but not stable disease patients triggered the growth of CLL T and B cells. In summary, CLL disease progression correlates with the rate of CLL cell division, and the rapidity that CLL cells home to the TME. The intraclonal fractions of CLL clones exhibit distinct biologic properties that can be further differentiated based on disease aggressiveness. This appears especially relevant for the development of a leukemia-supportive, immune-tolerant TME contributed by all 3 CXCR4/CD5 fractions, albeit by different mechanisms. The PF creates this by superior antigen presentation capacity and skewing T cell function to an immunosuppressive Th2 phenotype. For the IF and RF, this is done by inducing IL-10 secretion and amplifying Tregs. Together, these findings suggest the possibility of targeting specific subpopulations in CLL clones with distinct immunoregulatory modalities as a novel form of therapy. Disclosures Chen: Beigene: Research Funding; Pharmacyclics: Research Funding; Verastem: Research Funding. Wiestner:Pharmacyclics LLC, an AbbVie Company: Research Funding. Rai:Cellectis: Membership on an entity's Board of Directors or advisory committees; Pharmacyclics: Membership on an entity's Board of Directors or advisory committees; Roche/Genentech: Membership on an entity's Board of Directors or advisory committees. Chiorazzi:Janssen, Inc: Consultancy; AR Pharma: Equity Ownership.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2717-2717 ◽  
Author(s):  
Patrick B. Johnston ◽  
Lauren Pinter-Brown ◽  
Jaqueline Rogerio ◽  
Ghulam Warsi ◽  
Anne Graham ◽  
...  

Abstract Abstract 2717 Background: Dysregulation of the phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signal transduction pathway, which is central to cellular growth, proliferation, metabolism, survival, and angiogenesis, is implicated in Hodgkin lymphoma (HL) pathogenesis. In a previous phase II study of patients with relapsed/refractory lymphomas, the oral mTOR inhibitor everolimus showed promising clinical activity and acceptable toxicity in a subgroup of 19 heavily pretreated patients with HL (Johnston et al. Am J Hematol 2010;85:320–4). The purpose of this study was to confirm the safety and efficacy of everolimus 10 mg/day in patients with relapsed/refractory classical HL. Methods: In this multicenter, US, phase II study, patients aged ≥18 years with classical HL whose disease progressed after high-dose chemotherapy with autologous hematopoietic stem cell transplant (AHSCT) and/or a gemcitabine-, vinorelbine-, or vinblastine-containing regimen were treated with oral everolimus 10 mg/day until disease progression or unacceptable toxicity. Radiological response was assessed every 12 weeks using integrated positron emission tomography/computed tomography with contrast or computed tomography with contrast. The primary endpoint was the overall response rate (ORR) evaluated using modified response criteria for malignant lymphoma (Cheson et al. J Clin Oncol 2007;25:579–86). Secondary endpoints included median progression-free survival (PFS). Adverse events (AEs) were assessed throughout the study and for ≥4 weeks after the last everolimus dose. Results: The results from 37 patients in the safety population with evaluable data are reported in this abstract. Among these 37 patients, 35% were male, and the median age was 32 years. The median times from initial diagnosis to the first and most recent recurrence/relapse were 9 and 38 months, respectively. Of the 37 patients, 54% were pretreated with AHSCT and 95% were pretreated with a gemcitabine-, vinorelbine-, or vinblastine-containing regimen. The percentage of patients who experienced disease progression during prior therapies or discontinued their previous treatment due to disease progression was 73%. The median number of prior medication regimens was 4.5. Twenty-two patients discontinued treatment, including 11 due to disease progression and 8 due to AEs. The ORR was 35%, and an additional 27% of patients had stable disease (Table). The median PFS was 7.2 months (Figure). The most common hematologic AEs were thrombocytopenia (38%) and anemia (22%), and the most common nonhematologic AEs were fatigue (43%), cough (27%), headache (22%), dyspnea (22%), and rash (22%). Grade 3 or 4 drug-related AEs were noted in 12 (32%) patients and most commonly included thrombocytopenia (16%), neutropenia (8%), and anemia (8%). Serious AEs were experienced by 19% of patients; no serious AE occurred in more than 1 patient. Conclusion: Everolimus was associated with a favorable ORR and median PFS duration in highly pretreated patients with relapsed/refractory classical HL, confirming results of a previous study. The AE profile was consistent with that previously observed for everolimus; most AEs were grade 1 or 2 and manageable. These results suggest that further study evaluating everolimus in HL is warranted. Disclosures: Pinter-Brown: Genentech: Speakers Bureau; Spectrum: Honoraria; Celgene: Consultancy; Merck: Consultancy; Allos: Consultancy. Rogerio:Novartis Pharmaceuticals Corporation: Employment. Warsi:Novartis Pharmaceuticals Corporation: Employment. Graham:Novartis Pharmaceuticals Corporation: Employment. Ramchandren:Seattle Genetics: Research Funding; Novartis: Research Funding; Millennium: Research Funding; Celgene: Research Funding; Pfizer: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document