scholarly journals Pharmacodynamics and molecular correlates of response to glofitamab in relapsed/refractory non-Hodgkin lymphoma

Author(s):  
Ann-Marie E. Bröske ◽  
Koorosh Korfi ◽  
Anton Belousov ◽  
Sabine Wilson ◽  
Chia-Huey Ooi ◽  
...  

Glofitamab, a novel CD20xCD3, T-cell-engaging bispecific antibody, demonstrated single-agent activity in study NP30179, a first-in-human, phase 1 trial in relapsed/refractory B-cell non-Hodgkin lymphoma. Preclinical studies showed that glofitamab leads to T-cell activation, proliferation and tumor cell killing upon binding to CD20 on malignant cells. Here, we provide evidence of glofitamab's clinical activity, including pharmacodynamics, mode of action and factors associated with clinical response, by evaluating biomarkers in patient samples from the dose-escalation part of this trial. Patients enrolled in NP30179 received single-dose obinutuzumab pretreatment (1000 mg; Gpt) 7 days prior to intravenous glofitamab (5 µg-25 mg). Glofitamab treatment lasted ≤12 cycles once every 2 or 3 weeks. Blood samples were collected at predefined time points per clinical protocol; T-cell populations were evaluated centrally by flow cytometry, and cytokine profiles were analyzed. Immunohistochemical and genomic biomarker analyses were performed on tumor biopsies. Pharmacodynamic modulation was observed with glofitamab treatment including dose-dependent induction of cytokines, and T-cell margination, proliferation and activation in peripheral blood. Gene expression analysis of pre-treatment tumor biopsies indicated that tumor cell intrinsic factors like TP53 signaling are associated with resistance to glofitamab, but may also be interlinked with a diminished effector T-cell profile in resistant tumors and thus represent a poor prognostic factor per se. This integrative biomarker data analysis provides clinical evidence of glofitamab's mode of action, supports optimal biological dose selection, and will further guide clinical development. This trial was registered at www.clinicaltrials.gov as #NCT03075696.

1995 ◽  
Vol 40 (6) ◽  
pp. 390-396 ◽  
Author(s):  
Gijsbert C. de Gast ◽  
Inez-Anne Haagen ◽  
Anja A. van Houten ◽  
Sigrid C. Klein ◽  
Ashley J. Duits ◽  
...  

2016 ◽  
Vol 12 (2) ◽  
pp. 101-106 ◽  
Author(s):  
Stephen M. Ansell

Tumor-specific cytotoxic T cells have the capacity to target and eradicate malignant B cells in patients with Hodgkin and non-Hodgkin lymphoma; however, multiple mechanisms, including regulatory T cells, immunosuppressive ligands, and immune exhaustion, suppress an effective antitumor immune response. One mechanism that is used by malignant cells to inhibit the immune response is overexpression of programmed death ligand 1 or 2 (PD-L1 or PD-L2) on the cancer cell surface. These ligands interact with the programmed death-1 (PD-1) receptor expressed on intratumoral T cells and provide an inhibitory signal, thereby suppressing the antitumor immune response. Monoclonal antibodies that block PD-1 signaling prevent T-cell inhibition and promote a T-cell–mediated antilymphoma response. Blocking antibodies that are directed against PD-1 or PD-L1 are currently being tested in patients with lymphoma and have shown remarkable efficacy, particularly in patients with relapsed Hodgkin lymphoma. On the basis of the promising activity of this approach, PD-1 inhibitors are being used as single-agent therapy in patients with relapsed Hodgkin lymphoma, and these inhibitors are also being tested in combination with standard chemotherapy or targeted agents in ongoing clinical trials.


1995 ◽  
Vol 40 (6) ◽  
pp. 390-396 ◽  
Author(s):  
Gijsbert C. de Gast ◽  
Inez-Anne Haagen ◽  
Anja A. van Houten ◽  
Sigrid C. Klein ◽  
Ashley J. Duits ◽  
...  

2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 8014-8014 ◽  
Author(s):  
T. F. Gajewski ◽  
D. Niedzwiecki ◽  
J. Johnson ◽  
G. Linette ◽  
C. Bucher ◽  
...  

8014 Background: Ras-based signaling is thought to be critical in the genesis of melanoma. Farneslytransferase (FT) inhibitors (FTIs) have been developed as a pharmacologic strategy to inhibit Ras function. Additional farnesylated proteins are also important for the malignant process, and FTIs inhibit melanoma cell line proliferation in vitro. These considerations motivated the development of a phase II trial of the FTI R115777 in patients with melanoma. Farnesylated proteins are also important for T cell activation. The interest in future combinations of targeted agents and immunotherapeutics in this disease prompted analysis of T cell function ex vivo. Methods: A 3-stage design was pursued with a maximum of 40 patients planned and early stopping if there were no responders in the first 14, or fewer than 2 responders in the first 28 patients. Eligibility included intact organ function, PS≤1, no prior chemotherapy, at most 1 prior immunotherapy, no brain metastases, and presence of at least 2 cutaneous lesions amenable to excisional biopsy. R115777 (300 mg orally) was administered twice per day for 21 days of a 28-day cycle. Patients were evaluated every 2 cycles by RECIST criteria. Blood was obtained pre-treatment and during week 7 for analysis of HDJ-2 farnesylation and for T cell IFN-γ production in response to SEA. In addition, tumor biopsies were performed pre- and post-treatment when feasible to directly measure FT activity. Results: 14 patients were enrolled. 2 patients had grade 3 toxicities, which included myelosuppression, nausea/vomiting, elevated BUN, and anorexia. There were no clinical responses, and only 4 patients went on to a second course of treatment. All analyzed patients showed HDJ-2 gel shift in peripheral blood cells, as well as marked inhibition of FT activity (by 85–98%) in tumor tissue. T cell production of IFN-γ was also suppressed. Conclusions: Despite potent target inhibition, the FTI R115777 showed no evidence for clinical activity as a single agent in this cohort of 14 metastatic melanoma patients. Inhibition of T cell function has implications for future combination therapies and suggests the possibility for FTIs as candidate immunosuppressive agents. New therapeutic approaches for melanoma, or logically selected combination therapies, are needed. [Table: see text]


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4793-4793 ◽  
Author(s):  
Gerhard Zugmaier ◽  
Dirk Nagorsen ◽  
Matthias Klinger ◽  
Ralf C Bargou ◽  
Mariele Goebeler ◽  
...  

Abstract Abstract 4793 Blinatumomab is a CD19/CD3-bispecific antibody construct of the bispecific T cell engager (BiTE®) class showing as single agent a high rate and duration of responses in patients with relapsed non-Hodgkin lymphoma (NHL) and B-precursor acute lymphocytic leukemia (ALL). Blinatumomab has a favorable safety profile with exception of a subset of patients developing neurological adverse events (AEs) during the first days of treatment, such as confusion, speech impairment or cerebellar symptoms. Thus far, all relevant neurological AEs (11 out of 48 patients) were transient, fully reversible and resolved without sequelae within 3 to 72 hours after stop of infusion. In no case, pathological findings were seen upon cranial magnetic resonance imaging. Despite treatment discontinuation, 4 patients with neurological AEs have achieved an objective lymphoma remission. Analysis of cerebrospinal fluid (CSF) taken within hours after stop of infusion showed detectable levels of blinatumomab in the majority of affected patients, while in one patient without neurological symptoms no blinatumomab was detectable in CSF during infusion. Moreover, increased levels of albumin and T lymphocytes in CSF support a disturbance of the blood brain barrier (BBB) as a possible underlying event. Analyses of patient serum samples for angiopoetin-2 and S100b are ongoing to investigate whether levels of the endothelial stress and BBB integrity marker, respectively, correlate with neurological AEs. In a retrospective analysis of 39 NHL patients, a baseline B cell to T cell (B:T) ratio in peripheral blood at or below 1:10 was identified as the only predictive factor for the subsequent occurrence of neurological AEs. The predictive value was then prospectively confirmed in 8 additional patients. Of note, ALL patients –despite very low B:T ratios– rarely showed neurological AEs, which may relate to previous intrathecal chemotherapy depleting target cells in the brain. Potential mechanisms for the neuroprotective effect of peripheral B cells are being investigated. In conclusion, we identified a simple measure to prospectively identify patients at risk of developing neurological AEs after onset of blinatumomab treatment. Mitigating measures are currently tested in these high-risk patients in order to avoid discontinuation of treatment. Disclosures: Zugmaier: Micromet: Employment, Equity Ownership. Nagorsen:Micromet: Employment, Equity Ownership. Klinger:Micromet: Employment, Equity Ownership. Bargou:Micromet: Consultancy, Patents & Royalties. Baeuerle:Micromet: Employment, Equity Ownership. Kufer:Micromet: Employment, Equity Ownership, Patents & Royalties.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2833-2833 ◽  
Author(s):  
Ruth Pettengell ◽  
Pier Luigi Zinzani ◽  
Geetha Narayanan ◽  
Fernando Hurtado de Mendoza ◽  
Raghunadharao Digumarti ◽  
...  

Abstract Abstract 2833 Introduction: No therapy with reliable, durable efficacy exists for patients with aggressive non-Hodgkin lymphoma (aNHL) who relapse following at least two lines of therapy. Pixantrone dimaleate (PIX) is a novel aza-anthracenedione, structurally similar to anthracyclines and mitoxantrone, and forms stable DNA adducts. Unlike anthracyclines and mitoxantrone, PIX does not bind iron, minimally promotes reactive oxygen species formation, and is substantially less cardiotoxic in preclinical models. On the basis of promising early clinical activity and acceptable safety, we conducted a phase 3 trial with PIX in patients with aNHL and ≥2 relapses. Patients and Methods: This randomized, multicenter, controlled, open-label study enrolled patients with aNHL (de novo or transformed) with ≥1 prior anthracycline-containing regimen and ≥2 relapses. Patients were randomized to PIX 85 mg/m2 on days 1, 8, and 15 of a 28-day cycle, for up to 6 cycles, or to investigator's choice of single-agent comparator (COMP): vinorelbine, oxaliplatin, ifosfamide, etoposide, mitoxantrone, or, in the US only, gemcitabine or rituximab. Both groups were followed for 18 months after last treatment. The primary endpoint, CR/CRu rate in ITT population, was assessed by an independent assessment panel. Other efficacy endpoints were overall response rate (ORR), duration of response, progression-free survival (PFS), and overall survival (OS). Final end-of-study results are reported here. Results: Planned enrollment was set at 320 patients; however, due to slow accrual, a total of 140 patients were randomized (n=70 per group) to this study. Median number of treatment cycles for PIX group was 4 vs 3 for COMP. Median duration of treatment for patients in the PIX group was about one month longer than in the COMP group (3.8 vs 2.6 months). The end-of-study CR/CRu rate was 24% (16% CR, 8%CRu) for PIX vs 7% (no CRs, 7% CRu only) for COMP (P = 0.009); ORR (CR/CRu/PR) was 40% vs 14% (P = 0.001). After treatment ended, 3 patients in the PIX group achieved CR with no subsequent therapy. Two of the 3 patients converted from SD to CR and 1 from PR to CRu. Median CR/CRu duration was 9.6 months for PIX vs 4.0 months for COMP (P = 0.081). For survival endpoints, there was a 40% improvement in PFS with a median of 5.3 months vs 2.6 months (HR = 0.60, log-rank P = 0.005) and a 21% improvement in OS with a median of 10.2 months for PIX vs 7.6 months for COMP (HR = 0.79, log-rank P = 0.251). Exploratory subgroup analyses of CR/CRu and ORR were consistently higher for PIX patients and suggest stronger efficacy with the subgroups sex (female), <3 prior regimens, no prior rituximab, and ≥1 yr from 1st to 2nd line therapy. During treatment, neutropenia and leukopenia were the most common (≥10%) grade 3/4 adverse events and the incidence of febrile neutropenia was 7.4% for PIX vs 3% for COMP. Percentage of patients with cardiac disorder SAEs was 8.8% for PIX vs 4.5% for COMP. There were 11 grade 1/2 and 2 grade 3 LVEF events for PIX vs 7 grade 1/2 for COMP. Conclusions: In this phase 3 study, patients treated with PIX achieved superior efficacy when compared with other agents, as assessed by CR/CRu rate, ORR, and PFS, and had a positive trend in OS. Pixantrone had a tolerable safety profile in heavily pretreated patients with relapsed aggressive NHL. Disclosures: Pettengell: Cell Therapeutics, Inc: Honoraria. Coiffier:Cell Therapeutics, Inc: Advisory Board. Schiller:Sunesis: Consultancy; Celgene, Genzyme, Millenium, CTI, Antisoma, Novartis: Research Funding. Rizzieri:Cell Therapeutics, Inc: Membership on an entity's Board of Directors or advisory committees. Cernohous:Cell Therapeutics, Inc: Employment. Wang:Cell Therapeutics, Inc: Employment. Singer:Cell Therapeutics, Inc: Employment, Membership on an entity's Board of Directors or advisory committees; DiaKine Therapeutics, Inc: Membership on an entity's Board of Directors or advisory committees.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1560-1560 ◽  
Author(s):  
Daruka Mahadevan ◽  
Eric Vick ◽  
Bryan Huber ◽  
Carla Morales ◽  
Laurence Cooke ◽  
...  

Abstract Introduction: Peripheral T-cell non-Hodgkin Lymphoma (PTCL) is heterogeneous and rare, accounting for 15% of non-Hodgkin lymphoma (NHL). PTCL is very aggressive, has a poor prognosis compared to B-NHL and novel therapeutic strategies are needed. Aurora A and B (AAK and ABK) are members of a highly conserved family of serine/threonine protein kinases that play key regulatory roles during normal and malignant mitosis. A recent study (SWOG1108, S1108) showed alisertib, a small molecular inhibitor of AAK/ABK to have a 30% response rate as a single-agent in relapsed/refractory PTCL (Barr et al. 2015, JCO). The novel mechanism of alisertib action provides a platform for synthetic lethal drug combinations. PI3Kinases are central signaling mediators which activate cell proliferation, immune regulation and lead to the activation of AAK/ABK through the mTOR pathway. MLN1117, a PI3KCa inhibitor is anti-proliferative and has anti-tumor activity in mouse models and in early phase clinical trials. The addition of MLN1117 to alisertib has the benefit of blocking the PI3K/mTOR and the MEK/ERK pathways. PD-L1 (CD274) is an immune checkpoint ligand that binds PD-1 and down-regulates the activity of active T-cells. PD-L1 expression is a mechanism of immune escape, allowing cancers to evade the immune system and represents a negative prognostic factor. Multiple receptor-mediated signaling pathways which regulate NF-κB, MAPK, PI3K/mTOR, and JAK/STAT are involved in PD-L1 induction. Using this mechanistic rationale, we investigated cellular drivers of PTCL in cell culture, a mouse xenograft model and in patient samples. We hypothesized a novel therapeutic strategy where aurora inhibition provides a synthetic lethal platform to target PI3K and PD-L1 signaling in PTCL. Methods : Immunohistochemistry (IHC) on 15 PTCL patient samples from S1108 was evaluated for Ki67, PD-1, and PD-L1 with reactive lymph node controls. Fluorescent microscopy provided quantification of PD-1, PD-L1 and Ki67. Cytokine arrays (RayBiotech) were used to evaluate cytokines that upregulate PD-L1 in S1108 samples. MTS assays were conducted with single agent alisertib, MLN1117, idelalisib and vincristine (VCR) in CRL-2396, TIB-48 and Jurkat T-NHL cell lines. Combination studies were conducted to ascertain IC50's and synergism (Chou-Talalay). A mouse CRL-2396 PTCL xenograft model was analyzed for anti-tumor activity of alisertib, alisertib + MLN1117, and alisertib + MLN1117 + VCR. The endpoints were response-to-therapy versus PD-1/PDL-1 expression by IHC and mechanisms of synergy-synthetic lethality. Results: IHC of PTCL pre-alisertib (S1108) revealed PD-1 and PD-L1 expression were low while Ki-67 was high. PD-L1:PD-1 staining ratio was increased at 8.9 fold (p=0.0037). Cytokine profiling (pre-alisertib and post-alisertib day 8, n=12) indicated IFNg (n=2 inc, n=4 dec, n=6 NC); TNFa (n=4 inc, n=4 dec, n=4 NC); IL-2 (n=4 inc, n=3 dec, n=5 NC); GMCSF (n=3 inc, n=3 dec, n=6 NC). IC50's for alisertib 42.0 nM; MLN1117 22.16 mM, idelalisib (inactive) and VCR 1.09 nM. Combination indices (CI) indicated strong synergism for alisertib + MLN1117, alisertib + VCR and alisertib + MLN1117 + VCR. The mouse PTCL revealed significant decreases in mean tumor volume between alisertib alone versus control (mean decrease 497 mm3, p=0.042), alisertib + MLN1117 (mean decrease 1288 mm3 p<0.001) and alisertib + MLN1117 + VCR (mean decrease 2817.5 mm3, p<0.001). Mouse PTCL tumors are being evaluated PD-1/PD-L1 expression and PI3K signaling in PTCL. Conclusion: PD-L1 is induced by chemokines and inactivates PI3K via SHP-2. However, PI3K activity induces PD-L1. Furthermore, alisertib activates PI3K/AKT hence targeting PI3K plus aurora (+VCR) abrogates PI3K activity and PD-L1 induction. Finally, PD-1/ PD-L1 IHC ratio may be useful in stratifying PTCL patients in prospective trials. The mechanistic interactions of aurora inhibition in the context of PI3K inhibition with PD-L1 blockade in under investigation (Figure 1). Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1351-1351 ◽  
Author(s):  
Brad Kahl ◽  
Nancy L. Bartlett ◽  
John P. Leonard ◽  
Kristen Ganjoo ◽  
Michael E. Williams ◽  
...  

Abstract Background: Bendamustine (Treanda®) is a purine analog/alkylator hybrid with single-agent activity in patients with relapsed/refractory non-Hodgkin lymphoma (NHL), chronic lymphocytic leukemia (CLL), multiple myeloma, and breast cancer. The objective of this phase 3 multicenter study was to evaluate the efficacy and safety of single-agent bendamustine HCl in patients with relapsed, rituximab-refractory NHL. Methods: Eligible patients had previously treated, rituximab-refractory, indolent-histology NHL. Rituximab-refractory was defined as no response or progression within 6 months of first dose of rituximab induction, completion of rituximab maintenance therapy or progression before the next scheduled rituximab dose, or completion of a combination of rituximab and chemotherapy. Patients received bendamustine 120 mg/m2 intravenously over 60 minutes on days 1 and 2 every 21 days for 6 cycles (2 additional cycles at investigator discretion; max 8). Tumor response was determined by modified International Working Group Response Criteria for NHL. The primary endpoint was overall response rate (ORR) with secondary endpoints of response duration (RD) and progression-free survival (PFS). Results: Results for the first 38 of 100 enrolled patients are presented. The median age was 60 years, 63% were male, and 63% had stage III/IV disease. Histologies included follicular (53%), CLL/small lymphocytic lymphoma (26%), and marginal zone (21%) lymphoma. Patients had received a median of 3 prior courses of treatment (range 1–10) and 2 prior rituximab-containing courses (range 1–6). Prior alkylator or purine analog-based treatment was administered to 79% and 37% of patients, respectively. Eighteen (47%) patients were considered to be chemoresistant. Four (11%) patients received prior radioimmunotherapy. The ORR in the primary analysis population was 84%, including 29% complete responses (CR), 3% CR unconfirmed, and 53% partial responses. The median RD was 9.3 months and the median PFS was 9.7 months. The primary hematologic toxicity was reversible myelosuppression; grade 3/4 hematologic side effects included leukopenia (60%), neutropenia (60%), thrombocytopenia (24%), lymphocytopenia (95%), and anemia (5%). Febrile neutropenia was not observed. Common nonhematologic adverse events (grades 1/2, 3, 4) were nausea (68%, 5%, 0%), vomiting (42%, 0%, 0%), and fatigue (42%, 13%, 3%). Conclusions: In this multicenter study, single-agent bendamustine was well tolerated and produced a high rate (84%) of durable responses. It is the first chemotherapeutic agent to demonstrate significant clinical activity in rituximab-refractory indolent lymphoma. These results compare favorably with prior reports of radioimmunotherapy in this patient population. An updated analysis of all 100 patients will be available for the annual meeting.


Sign in / Sign up

Export Citation Format

Share Document