Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
J Immunother Cancer ; 12(3)2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38519055

ABSTRACT

BACKGROUND: Patients with relapsed/refractory B-cell non-Hodgkin lymphoma (R/R B-NHL) have a significant need for effective treatment options. Odronextamab is an Fc-silenced, human, CD20×CD3 bispecific antibody that targets CD20-expressing cells via T-cell-mediated cytotoxicity independent of T-cell/major histocompatibility complex interaction. Phase I results in patients with R/R B-NHL demonstrated that odronextamab monotherapy could achieve deep and durable responses with a generally manageable safety profile (ELM-1; NCT02290951). As part of a biomarker analysis of the same study, we investigated potential biomarkers and mechanisms of resistance to odronextamab. METHODS: Patients with R/R B-NHL enrolled in ELM-1 received one time per week doses of intravenous odronextamab for 4×21 day cycles, then doses every 2 weeks thereafter. Patient tumor biopsies were obtained at baseline, on-treatment, and at progression. Immune cell markers were analyzed by immunohistochemistry, flow cytometry, single-cell RNA sequencing, and whole genome sequencing. RESULTS: Baseline tumor biopsies showed that almost all patients had high proportions of B cells that expressed the CD20 target antigen, whereas expression of other B-cell surface antigens (CD19, CD22, CD79b) was more variable. Responses to odronextamab in patients with diffuse large B-cell lymphoma were not related to the relative level of baseline CD20 expression, cell of origin, or high-risk molecular subtype. A potential link was observed between greater tumor programmed cell death-ligand 1 expression and increased likelihood of response to odronextamab. Similarly, a trend was observed between clinical response and increased levels of CD8 T cells and regulatory T cells at baseline. We also identified an on-treatment pharmacodynamic shift in intratumoral immune cell subsets. Finally, loss of CD20 expression through inactivating gene mutations was identified as a potential mechanism of resistance in patients who were treated with odronextamab until progression, as highlighted in two detailed patient cases reported here. CONCLUSIONS: This biomarker analysis expands on clinical findings of odronextamab in patients with R/R B-NHL, providing verification of the suitability of CD20 as a therapeutic target, as well as evidence for potential mechanisms of action and resistance.


Subject(s)
Antibodies, Bispecific , Antineoplastic Agents , Lymphoma, Large B-Cell, Diffuse , Humans , Antineoplastic Agents/therapeutic use , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Treatment Outcome , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Antigens, CD20
2.
Sci Transl Med ; 14(670): eabn1082, 2022 11 09.
Article in English | MEDLINE | ID: mdl-36350988

ABSTRACT

Although many patients with diffuse large B cell lymphoma (DLBCL) may achieve a complete response to frontline chemoimmunotherapy, patients with relapsed/refractory disease typically have poor outcomes. Odronextamab, a CD20xCD3 bispecific antibody that provides "signal 1" through the activation of the T cell receptor/CD3 complex, has exhibited early, promising activity for patients with highly refractory DLBCL in phase 1 trials. However, not all patients achieve complete responses, and many relapse, thus representing a high unmet medical need. Here, we investigated whether adding a costimulatory "signal 2" by engaging CD28 receptors on T cells could augment odronextamab activity. We demonstrate that REGN5837, a bispecific antibody that cross-links CD22-expressing tumor cells with CD28-expressing T cells, enhances odronextamab by potentiating T cell activation and cytolytic function. In preclinical DLBCL studies using human immune system-reconstituted animals, REGN5837 promotes the antitumor activity of odronextamab and induces intratumoral expansion of reprogrammable T cells while skewing away from a dysfunctional state. Although REGN5837 monotherapy shows limited activity and no toxicity in primate studies, it augments T cell activation when dosed in combination with odronextamab. In addition, analysis of non-Hodgkin lymphoma clinical samples reveals an increase in CD28+CD8+ T cells after odronextamab treatment, demonstrating the presence of a population that could potentially be targeted by REGN5837. Collectively, our data demonstrate that REGN5837 can markedly enhance the antitumor activity of odronextamab in preclinical NHL models, and the combination of these two bispecific antibodies may provide a chemotherapy-free approach for the treatment of DLBCL.


Subject(s)
Antibodies, Bispecific , Antineoplastic Agents , Lymphoma, Large B-Cell, Diffuse , Lymphoma, Non-Hodgkin , Animals , Humans , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , CD28 Antigens , CD8-Positive T-Lymphocytes , Antigens, CD19 , Neoplasm Recurrence, Local/drug therapy , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Large B-Cell, Diffuse/drug therapy , Antineoplastic Agents/pharmacology , Sialic Acid Binding Ig-like Lectin 2/therapeutic use
3.
Br J Haematol ; 199(3): 366-370, 2022 11.
Article in English | MEDLINE | ID: mdl-35892294

ABSTRACT

Outcomes remain poor for patients with relapsed/refractory B-cell non-Hodgkin lymphoma (R/R B-NHL). While chimeric antigen receptor (CAR) T-cell therapy has revolutionised treatment, a significant proportion of patients relapse or fail to respond. Odronextamab is a CD20 × CD3 bispecific antibody that has demonstrated durable responses and a manageable safety profile in patients with R/R B-NHL in a first-in-human trial (NCT02290951). Here, we document two patients with diffuse large B-cell lymphoma refractory to CART-cell therapy. Both achieved complete responses that remain ongoing for ≥2 years following odronextamab. Neither patient experienced Grade ≥3 cytokine release syndrome or Grade ≥3 neurological adverse events during treatment.


Subject(s)
Antibodies, Bispecific , Antineoplastic Agents , Lymphoma, Follicular , Lymphoma, Large B-Cell, Diffuse , Receptors, Chimeric Antigen , Humans , Receptors, Chimeric Antigen/therapeutic use , Antigens, CD19 , Neoplasm Recurrence, Local/pathology , Immunotherapy, Adoptive/adverse effects , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Follicular/etiology , Cytokine Release Syndrome , Cell- and Tissue-Based Therapy , Receptors, Antigen, T-Cell/genetics
4.
Lancet Haematol ; 9(5): e327-e339, 2022 May.
Article in English | MEDLINE | ID: mdl-35366963

ABSTRACT

BACKGROUND: Odronextamab is a hinge-stabilised, fully human IgG4-based CD20 × CD3 bispecific antibody that binds CD3 on T cells and CD20 on B cells. We aimed to evaluate the safety and antitumour activity of odronextamab in patients with relapsed or refractory B-cell non-Hodgkin lymphoma. METHODS: This single-arm, multicentre, phase 1, dose-escalation and dose-expansion (ELM-1) trial was conducted at ten academic sites across the USA and Germany. Patients aged 18 years or older with CD20-positive relapsed or refractory B-cell malignancies who previously received CD20-directed antibody therapy and who had at least one measurable lesion, and an ECOG performance status of 0 or 1 were included. Patients received intravenous odronextamab, according to a step-up dosing schedule in cycle 1, followed by treatment once per week at target doses ranging from 0·1 mg to 320 mg during cycles 2-4 (each cycle was 21 days). After cycle 4, maintenance treatment occurred every 2 weeks until disease progression or unacceptable toxicity. The primary endpoint of safety was assessed by the incidence of adverse events and dose-limiting toxicities to determine the maximum tolerated dose or phase 2 dose of odronextamab, or both. Preliminary antitumour activity, as measured by objective response rate, was a secondary endpoint. This study is registered with ClinicalTrials.gov, NCT02290951. FINDINGS: From Feb 4, 2015, to Sept 25, 2021, 145 heavily pretreated patients (median of 3 (IQR 2-5] previous therapies) were enrolled (94 to the dose-escalation and 51 to the dose-expansion part of the study). The median age of patients was 67·0 years (IQR 57·0-73·0); 101 (70%) were male and 44 (30%) were female; most participants were White (119 [82%]) and not Hispanic or Latino (132 [91%]). 42 (29%) patients received previous CAR T therapy and 119 (82%) were refractory to the last line of therapy. Median duration of follow-up was 4·2 months (IQR 1·5-11·5). During dose escalation, odronextamab was administered up to the maximum dose of 320 mg once per week and no dose-limiting toxicities were observed. The recommended dose for expansion in patients with follicular lymphoma grade 1-3a was 80 mg and was 160 mg for patients with diffuse large B-cell lymphoma. Cytokine release syndrome and neurological treatment-emergent adverse events were predominantly low grade and did not result in treatment discontinuation. The most common grade 3 or worse treatment-emergent adverse events were anaemia (36 [25%]), lymphopenia (28 [19%]), hypophosphataemia (27 [19%]), neutropenia (27 [19%]), and thrombocytopenia (20 [14%]). Serious treatment-emergent adverse events occurred in 89 (61%) of 145 patients; the most frequent were cytokine release syndrome (41 [28%]), pyrexia (11 [8%]), pneumonia (nine [6%]), and infusion-related reaction (six [4%]). Four deaths were considered related to treatment (gastric perforation in a patient with gastric involvement by lymphoma, lung infection, pneumonia, and tumour-lysis syndrome). Objective response rate was 51% (95% CI 42-59; 72 of 142). In patients with follicular lymphoma who received odronextamab doses of 5 mg or higher, the objective response rate was 91% (95% CI 75-98; 29 of 32) and the complete response rate was 72% (95% CI 53-86; 23 of 32). In patients with diffuse large B-cell lymphoma without previous CAR T-cell therapy who received doses of 80 mg or higher, the objective response rate was 53% (eight of 15) and all responses were complete responses. In patients with diffuse large B-cell lymphoma who had previous CAR T-cell therapy and received doses of 80 mg or higher, the objective response rate was 33% (ten of 30) and complete response rate was 27% (eight of 30). INTERPRETATION: Odronextamab monotherapy showed a manageable safety profile and encouraging preliminary activity, including durable responses in heavily pretreated patients with B-cell non-Hodgkin lymphoma, supporting further clinical investigation in phase 2 and 3 trials. FUNDING: Regeneron Pharmaceuticals.


Subject(s)
Antibodies, Bispecific , Antineoplastic Agents , Lymphoma, Follicular , Lymphoma, Large B-Cell, Diffuse , Lymphoma, Non-Hodgkin , Aged , Antibodies, Bispecific/adverse effects , Antigens, CD20 , Antineoplastic Agents/therapeutic use , Cytokine Release Syndrome , Female , Humans , Lymphoma, Follicular/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Non-Hodgkin/drug therapy , Male , Middle Aged
5.
Oncologist ; 26(9): e1538-e1547, 2021 09.
Article in English | MEDLINE | ID: mdl-34037286

ABSTRACT

BACKGROUND: Weight loss is common in advanced gastric and gastroesophageal junction adenocarcinoma (G/GEA); however, the prognostic implications of weight loss during the first cycle (C1) of chemotherapy remain poorly characterized. In this study, we investigated the impact of early weight loss during systemic treatment as a potential prognostic factor for overall survival (OS) in patients with advanced G/GEA. MATERIALS AND METHODS: We performed a post hoc analysis of three phase III studies of ramucirumab. Patients were categorized into two groups: weight loss of ≥3% and <3% based on weight change during C1 (3-4 weeks) of treatment. OS by weight groups was assessed for each study and as a pooled meta-analysis. The effect of C1 weight change on patient survival was evaluated using univariate and multivariate Cox models. RESULTS: A total of 1,464 patients with weight data at the end of C1 were analyzed: REGARD (n = 311), RAINBOW (n = 591), and RAINFALL (n = 562). For all three studies, there were fewer patients in the weight loss ≥3% than <3% group. OS was numerically shorter for patients with weight loss of ≥3% than for patients with weight loss of <3% during C1 irrespective of treatment arm. Similar treatment independent effects of early weight loss on OS were observed in the meta-analysis. Overall, early weight loss ≥3% was associated with shorter survival in patients receiving active drug as well as placebo/best supportive care. CONCLUSION: This large post hoc analysis demonstrated that weight loss of ≥3% during C1 was a negative prognostic factor for OS in patients with advanced G/GEA. IMPLICATIONS FOR PRACTICE: This comprehensive analysis examining early weight loss during systemic treatment as a predictor of survival outcomes in patients with advanced gastric and gastroesophageal junction adenocarcinoma (G/GEA) includes a large sample size, reliable on-treatment data reported in well-conducted phase III clinical trials, and global representation of cancer patients with advanced G/GEA. Understanding the impact of on-treatment weight loss is clinically relevant and may represent an opportunity for targeted interventions.


Subject(s)
Stomach Neoplasms , Antineoplastic Combined Chemotherapy Protocols , Clinical Trials, Phase III as Topic , Esophagogastric Junction , Humans , Paclitaxel/therapeutic use , Prognosis , Stomach Neoplasms/drug therapy , Weight Loss
6.
BMC Cancer ; 16(1): 842, 2016 11 04.
Article in English | MEDLINE | ID: mdl-27809808

ABSTRACT

BACKGROUND: Gliomas are the most common primary brain tumors in adults and invariably carry a poor prognosis. Recent clinical studies have demonstrated the safety and compelling survival benefit when tumor treating fields (TTFields) are added to temozolomide for patients with newly diagnosed glioblastoma. TTFields are low-intensity, intermediate frequency (200 kHz) alternating electric fields, delivered directly to a patient's brain through the local application of non-invasive transducer arrays. Experimental simulations have demonstrated that TTFields distribute in a non-uniform manner within the brain. To ensure patients receive the maximal therapeutic level of TTFields at the site of their tumor, tumor burden is mapped and an optimal array layout is personalized using the NovoTAL software. The NovoTAL software utilizes magnetic resonance imaging (MRI) measurements for head size and tumor location obtained from axial and coronal T1 postcontrast sequences to determine the optimal paired transducer array configuration that will deliver the maximal field intensity at the site of the tumor. In clinical practice, physicians planning treatment with TTFields may determine that disease activity is more accurately represented in noncontrast-enhancing sequences. Here we present and discuss a series of 8 cases where a treating physician has utilized non-contrast enhancement and advanced imaging to inform TTFields treatment planning based on a clinical evaluation of where a patient is believed to have active tumor. This case series is, to our knowledge, the first report of this kind in the literature. CASE PRESENTATIONS: All patients presented with gliomas (grades 2-4) and ranged in age from 49 to 65 years; 5 were male and 3, female. Each patient had previously received standard therapy including surgery, radiation therapy and/or chemotherapy prior to initiation of TTFields. The majority had progressed on prior therapy. A standard pre- and postcontrast MRI scan was acquired and used for TTFields treatment planning. CONCLUSION: This paper details important approaches for integrating clinical considerations, nonmeasurable disease and advanced imaging into the treatment planning workflow for TTFields. As TTFields become integrated into standard care pathways for glioblastoma, this case series demonstrates that treatment planning beyond the extent of contrast enhancement is clinically feasible and should be prospectively compared to standard treatment planning in a clinical trial setting, in order to determine the impact on patient outcomes.


Subject(s)
Antineoplastic Agents/administration & dosage , Dacarbazine/analogs & derivatives , Glioma/diagnostic imaging , Glioma/drug therapy , Image Enhancement , Magnetic Resonance Imaging , Aged , Brain/pathology , Combined Modality Therapy , Dacarbazine/administration & dosage , Female , Glioma/pathology , Humans , Male , Middle Aged , Neoplasm Grading , Temozolomide , Treatment Outcome
7.
Annu Int Conf IEEE Eng Med Biol Soc ; 2016: 6461-6464, 2016 Aug.
Article in English | MEDLINE | ID: mdl-28269726

ABSTRACT

This paper reviews the state-of-the-art in simulation-based studies of Tumor Treating Fields (TTFields) and highlights major aspects of TTFields in which simulation-based studies could affect clinical outcomes. A major challenge is how to simulate multiple scenarios rapidly for TTFields delivery. Overcoming this challenge will enable a better understanding of how TTFields distribution is correlated with disease progression, leading to better transducer array designs and field optimization procedures, ultimately improving patient outcomes.


Subject(s)
Computer Simulation , Electric Stimulation Therapy/instrumentation , Electric Stimulation Therapy/methods , Phantoms, Imaging , Animals , Cell Line, Tumor , Head , Humans , Neoplasms/therapy , Transducers
8.
Annu Int Conf IEEE Eng Med Biol Soc ; 2016: 2357-2360, 2016 Aug.
Article in English | MEDLINE | ID: mdl-28268799

ABSTRACT

Tumor Treating Fields (TTFields) are low intensity alternating electric fields in the 100-500 KHz frequency range that are known to have an anti-mitotic effect on cancerous cells. In the USA, TTFields are approved by the Food and Drug Administration (FDA) for the treatment of glioblastoma (GBM) in both the newly diagnosed and recurrent settings. Optimizing treatment with TTFields requires a deep understanding of how TTFields distribute within the brain. To address this issue, simulations using realistic head models have been performed. However, the preparation of such models is time-consuming and requires a high level of expertise, limiting the usefulness of these models for systematic studies in which the testing of multiple cases is required. Here we present a platform for rapidly simulating TTFields distributions in multiple scenarios. This platform enables high throughput computational simulations to be performed, allowing comparison of field distributions within the head in multiple clinically relevant scenarios. The simulation setup is simple and intuitive, allowing non-expert users to run simulations and evaluate results, thereby providing a valuable tool for studying how to optimize TTFields delivery in the clinic.


Subject(s)
Brain Neoplasms/therapy , Electric Stimulation Therapy , Glioblastoma/therapy , Brain , Computer Simulation , Electricity , Head , Humans , Models, Theoretical
9.
Sci Rep ; 5: 18046, 2015 Dec 11.
Article in English | MEDLINE | ID: mdl-26658786

ABSTRACT

Tumor Treating Fields (TTFields) are low intensity, intermediate frequency, alternating electric fields. TTFields are a unique anti-mitotic treatment modality delivered in a continuous, noninvasive manner to the region of a tumor. It was previously postulated that by exerting directional forces on highly polar intracellular elements during mitosis, TTFields could disrupt the normal assembly of spindle microtubules. However there is limited evidence directly linking TTFields to an effect on microtubules. Here we report that TTFields decrease the ratio between polymerized and total tubulin, and prevent proper mitotic spindle assembly. The aberrant mitotic events induced by TTFields lead to abnormal chromosome segregation, cellular multinucleation, and caspase dependent apoptosis of daughter cells. The effect of TTFields on cell viability and clonogenic survival substantially depends upon the cell division rate. We show that by extending the duration of exposure to TTFields, slowly dividing cells can be affected to a similar extent as rapidly dividing cells.


Subject(s)
Chromosome Segregation/physiology , Mitosis/physiology , Neoplasms/pathology , Spindle Apparatus/pathology , Animals , Apoptosis/physiology , Cell Line, Tumor , Cell Survival/physiology , Electricity , Humans , MCF-7 Cells , Microtubules/metabolism , Microtubules/pathology , Neoplasms/metabolism , Rats , Rats, Inbred F344 , Tubulin/metabolism
10.
World J Surg Oncol ; 13: 316, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26558989

ABSTRACT

BACKGROUND: Optune™, previously known as the NovoTTF-100A System™, generates Tumor Treating Fields (TTFields), an effective anti-mitotic therapy for glioblastoma. The system delivers intermediate frequency, alternating electric fields to the supratentorial brain. Patient therapy is personalized by configuring transducer array layout placement on the scalp to the tumor site using MRI measurements and the NovoTAL System. Transducer array layout mapping optimizes therapy by maximizing electric field intensity to the tumor site. This study evaluated physician performance in conducting transducer array layout mapping using the NovoTAL System compared with mapping performed by the Novocure in-house clinical team. METHODS: Fourteen physicians (7 neuro-oncologists, 4 medical oncologists, and 3 neurosurgeons) evaluated five blinded cases of recurrent glioblastoma and performed head size and tumor location measurements using a standard Digital Imaging and Communications in Medicine reader. Concordance with Novocure measurement and intra- and inter-rater reliability were assessed using relevant correlation coefficients. The study criterion for success was a concordance correlation coefficient (CCC) >0.80. RESULTS: CCC for each physician versus Novocure on 20 MRI measurements was 0.96 (standard deviation, SD ± 0.03, range 0.90-1.00), indicating very high agreement between the two groups. Intra- and inter-rater reliability correlation coefficients were similarly high: 0.83 (SD ±0.15, range 0.54-1.00) and 0.80 (SD ±0.18, range 0.48-1.00), respectively. CONCLUSIONS: This user study demonstrated an excellent level of concordance between prescribing physicians and Novocure in-house clinical teams in performing transducer array layout planning. Intra-rater reliability was very high, indicating reproducible performance. Physicians prescribing TTFields, when trained on the NovoTAL System, can independently perform transducer array layout mapping required for the initiation and maintenance of patients on TTFields therapy.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/surgery , Electric Stimulation Therapy/instrumentation , Glioblastoma/pathology , Glioblastoma/surgery , Humans , Magnetic Resonance Imaging , Observer Variation , Reproducibility of Results , Scalp , Transducers
11.
J Oncol Pract ; 8(3): 173-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22942812

ABSTRACT

PURPOSE: Social media channels such as Twitter are gaining increasing acceptance as mechanisms for instantaneous scientific dialogue. Professional medical societies such as ASCO are using social media to expand the reach of scientific communications at and around their scientific meetings. This article examines the how Twitter use by oncologists expanded at the ASCO Annual Meetings from 2010 to 2011. METHODS: In both years, tweets that were specifically generated by physicians and that incorporated the official meeting hashtag were harvested from the public domain, and a discourse analysis was performed by three independent raters. Follow-up surveys were conducted to assess physician attitudes toward Twitter and its potential role in clinical practice. RESULTS: A combined total of 12,644 tweets were analyzed for 2010 and 2011. Although the number of physicians authoring tweets was small (14 in 2010, 34 in 2011), this group generated nearly 29% of the total meeting dialogue examined in this analysis in 2010 and 23% in 2011. Physicians used Twitter for reporting clinical news from scientific sessions, for discussions of treatment issues, for promotion, and to provide social commentary. The tangible impact of Twitter discussions on clinical practice remains unclear. CONCLUSION: Despite the 140-character limit, Twitter was successfully used by physicians at the 2010 and 2011 ASCO Annual Meetings to engage in clinical discussions, whether or not an author was on site as a live attendee. Twitter usage grew significantly from 2010 to 2011. Professional societies should monitor these phenomena to enhance annual meeting attendee user experience.

SELECTION OF CITATIONS
SEARCH DETAIL
...