scholarly journals A Novel Carcinoembryonic Antigen T-Cell Bispecific Antibody (CEA TCB) for the Treatment of Solid Tumors

2016 ◽  
Vol 22 (13) ◽  
pp. 3286-3297 ◽  
Author(s):  
Marina Bacac ◽  
Tanja Fauti ◽  
Johannes Sam ◽  
Sara Colombetti ◽  
Tina Weinzierl ◽  
...  
2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A553-A553
Author(s):  
Elaine Shum ◽  
Matthew Reilley ◽  
Yana Najjar ◽  
Adil Daud ◽  
John Thompson ◽  
...  

BackgroundXmAb20717 is a humanized bispecific monoclonal antibody that simultaneously targets PD-1 and CTLA-4. We report updated data on patients treated at the recommended expansion dose from an ongoing, multicenter, Phase 1, dose-escalation and -expansion study of intravenous XmAb20717 in patients with selected advanced solid tumors that progressed after treatment with all standard therapies or with no standard therapeutic options.MethodsA maximum tolerated dose was not reached in dose escalation. XmAb20717 10 mg/kg every 2 weeks (Q2W) was selected as the expansion dose, based on consistent T-cell proliferation in peripheral blood indicative of dual PD-1/CTLA-4 checkpoint blockade, and response to treatment (RECIST[1.1]).1 Parallel expansion cohorts included ~20 patients each with melanoma, renal cell carcinoma (RCC), non-small cell lung cancer (NSCLC), castration-resistant prostate cancer (CRPC), and a basket of tumor types without an FDA-approved checkpoint inhibitor (CI). Patients treated with 10 mg/kg in dose escalation were pooled with expansion cohorts for analysis of clinical activity and safety.ResultsAs of 9 June 2021, 110 patients, ranging in age from 39 to 89 years and 66.4% male, were treated, and 5 were continuing treatment. Patients had received a median of 4 prior systemic treatment regimens, including CI therapy for 64.5%. The objective response rate was 13.0% (10/77 patients evaluable for efficacy), including 1 complete response (melanoma [confirmed]) and 9 partial responses (confirmed: 1 melanoma, 2 RCC, 2 CRPC, 1 ovarian cancer; unconfirmed: 1 melanoma, 2 NSCLC). The CRPC responders (2/7 with RECIST-measurable disease) had confirmed PSA decreases ≥ 50% from baseline (to 0.02 and 0.3 ng/mL); neither had progression on bone scans. All responders had prior CI exposure, except those with CRPC. Robust CD4 and CD8 T-cell activation was seen. Low baseline tumoral expression of myeloid recruitment genes, including IL-8, was associated with clinical benefit. Grade ≥ 3 immunotherapy-related adverse events in ≥ 3 patients included rash (16.4%), transaminase elevations (9.1%), hyperglycemia (4.5%), acute kidney injury (3.6%), amylase and lipase increased (2.7%), and lipase increased (2.7%).ConclusionsPreliminary data indicate 10 mg/kg XmAb20717 Q2W was associated with complete and partial responses in multiple tumor types and was generally well-tolerated in these heavily pretreated patients with advanced cancer. Changes in T-cell populations in the periphery and tumor are consistent with robust dual checkpoint blockade. These findings support further development of XmAb20717 in advanced solid tumors, including metastatic prostate cancer.Trial RegistrationNCT03517488ReferencesShum E, Daud A, Reilley M, et al. Preliminary safety, pharmacokinetics/pharmacodynamics, and antitumor activity of XmAb20717, a PD-1 x CTLA-4 bispecific antibody, in patients with advanced solid tumors. JITC 2020;8(3):A247-8.Ethics ApprovalThe study was approved by each institution’s IRB.


2018 ◽  
Vol 36 (5_suppl) ◽  
pp. 69-69 ◽  
Author(s):  
Martin Wermke ◽  
Juergen Alt ◽  
John S. Kauh ◽  
Jonathan Back ◽  
Yacine Salhi ◽  
...  

69 Background: HER2 is overexpressed in many solid tumors and is a validated therapeutic target. GBR 1302 is a HER2xCD3 bispecific antibody engineered (using Glenmark’s BEAT® platform) to direct T-cells to HER2-expressing tumor cells. GBR1302-101 (NCT02829372) is an ongoing, multicenter, open-label, first-in-human study of GBR 1302 in subjects with HER2-positive cancers to evaluate the safety, tolerability, and preliminary efficacy of GBR 1302, and to elucidate the mechanism(s) by which it redirects T-cells to tumor and enhances cytolytic activity of cytotoxic T-cells. Methods: Adults with progressive HER2-positive solid tumors with no available standard or curative treatment receive intravenous GBR 1302 on Day 1 and Day 15 in 28-day treatment cycles at escalating dose levels, starting at 1 ng/kg. The first 4 cohorts consist of a single subject; subsequent cohorts enroll using a 3+3 design. The primary and secondary efficacy and safety endpoints of this trial will be reported at the end of the study. Preliminary pharmacodynamic (PD) data are reported for cellular biomarkers and cytokines as assessed by FACS and ELISA in peripheral blood. Results: Beginning at 30 ng/kg dosing of GBR 1302 (Cohort 4), numbers of peripheral blood CD3, CD4, and CD8 positive T-cell populations decreased within 6 hours of initiating administration, but recovered to levels at or above baseline by 48 hours. A parallel, transient increase was observed in peripheral blood cytokines (IL-2, IL-6, IL-10, IFN-γ, TNF-α). At doses greater than 30 ng/kg, more pronounced cytokine increases were observed, which normalized at 12 hours. At the highest dose level for which data are available (n = 8 subjects; Cohort 5), changes from baseline in cytokine expression at ~340 hours were greater by ~60-fold for IL-6, ~30-fold for IL-2, ~3-fold for IFN-γ, ~5-fold for TNF-α, and ~18-fold for IL-10. Two subjects treated at 100 ng/kg experienced Grade 1 cytokine release syndrome, evidenced by short-lived fever spikes. Dose escalation is ongoing. Conclusions: Preliminary PD data indicate changes in peripheral T-cell populations and inflammatory cytokines following GBR 1302 treatment. Clinical trial information: NCT02829372.


2016 ◽  
Vol 5 (8) ◽  
pp. e1203498 ◽  
Author(s):  
Marina Bacac ◽  
Christian Klein ◽  
Pablo Umana

2016 ◽  
Vol 34 (15_suppl) ◽  
pp. TPS2592-TPS2592 ◽  
Author(s):  
Kenji Hashimoto ◽  
Ayesh Perera ◽  
Yoshitaka Ogita ◽  
Mikiko Nakamura ◽  
Takahiro Ishiguro ◽  
...  

2020 ◽  
Vol 3 (2) ◽  
pp. 126-145 ◽  
Author(s):  
Jing Zhang ◽  
Jizu Yi ◽  
Pengfei Zhou

ABSTRACT A bispecific antibody (bsAb) can simultaneously bind two different epitopes or antigens, allowing for multiple mechanistic functions with synergistic effects. BsAbs have attracted significant scientific attentions and efforts towards their development as drugs for cancers. There are 21 bsAbs currently undergoing clinical trials in China. Here, we review their platform technologies, expression and production, and biological activities and bioassay of these bsAbs, and summarize their structural formats and mechanisms of actions. T-cell redirection and checkpoint inhibition are two main mechanisms of the bsAbs that we discuss in detail. Furthermore, we provide our perspective on the future of bsAb development in China, including CD3-bsAbs for solid tumors and related cytokine release syndromes, expression and chemistry, manufacturing and controls, clinical development, and immunogenicity.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A625-A625
Author(s):  
Christianne Groeneveldt ◽  
Priscilla Kinderman ◽  
Diana JM van den Wollenberg ◽  
Ruben L van den Oever ◽  
Jim Middelburg ◽  
...  

BackgroundThe use of T cell-engaging CD3-bispecific antibodies (CD3-bsAbs) is a promising immunotherapeutic strategy for cancer. Although this therapy has reached clinical practice for hematological malignancies, the absence of sufficient infiltrating T cells is a major barrier for efficacy in solid tumors.1 Oncolytic viruses are emerging as anti-cancer therapeutics, and accumulating evidence demonstrates their applicability to sensitize tumors for immune checkpoint immunotherapy.2 In this study, we exploited oncolytic reovirus as a strategy to enhance the efficacy of CD3-bsAbs in immune-silent, solid tumors.MethodsThe mutant p53 and K-ras induced murine pancreatic cancer model KPC3 resembles human pancreatic ductal adenocarcinomas with a desmoplastic tumor microenvironment, low T cell density, and resistance to immunotherapy. Immune-competent mice with established, subcutaneous KPC3 tumors were intratumorally injected with an optimized regimen of oncolytic reovirus (type 3 Dearing strain) and the reovirus-induced changes in the tumor microenvironment and lymphoid organs were analyzed over time by NanoString analysis, RT-qPCR and multicolor flow cytometry. The efficacy of combination with systemically injected CD3-bsAbs was evaluated in KPC3 and B16.F10 murine tumor models and the close-to-patient HER2+ BT474 breast cancer model with cell surface-expressed TRP1 and HER2 as target antigens, respectively. Primary outcome was tumor size, measured with caliper three times a week in a blinded-manner.ResultsReplication-competent reovirus induced an early IFN-signature, followed by a strong influx of CD8+ T cells (2.6-fold increase, p=0.0092). Viral replication declined after seven days and was associated with systemic activation of lymphocytes. Tumor-infiltrating T cells were mostly reovirus-specific and served as effector cells for the subsequently systemically administered CD3-bsAbs. The combination of reovirus and CD3-bsAbs induced regressions up to 70% in all mice with large, established KPC3, B16.F10, and BT474 tumors and significantly prolonged survival. Importantly, the employment of reovirus as a pre-conditioning regimen performed significantly better than the simultaneous or preceding administration of bsAbs. This combination treatment also induced regressions of non-injected distant lesions, suggesting that this therapy might be effective for metastatic disease.Abstract 590 Figure 1Reovirus sensitizes tumors for CD3-bsAb therapyReovirus-induced interferon signaling leads to increased T cell influx and subsequent effective CD3-bispecific antibody therapy in solid tumorsConclusionsOncolytic reovirus administration represents an effective strategy to induce a local IFN response and strong T cell influx, thereby sensitizing the tumor microenvironment for subsequent CD3-bsAb therapy (figure 1). Our data advocate for the inclusion of oncolytic viruses as a pre-conditioning strategy in T cell engaging antibody trials for solid tumors. Since both CD3-bispecific antibodies and oncolytic viruses are in advanced clinical development as monotherapies, efficient translation of this combination seems feasible.AcknowledgementsThis work was financially supported by the Dutch Cancer Society Bas Mulder Award 11056 (to NvM), a PhD fellowship from Leiden University Medical Center (to CG) and the Support Casper campaign by the Dutch foundation ‘Stichting Overleven met Alvleesklierkanker’ (supportcasper.nl) project numbers SOAK 17.04 and 19.03.Ethics ApprovalAll mouse studies were approved by the institutional Animal Welfare Body of Leiden University Medical Center and carried out under project licenses AVD1160020187004 or AVD116002015271, issued by the competent authority on animal experiments in the Netherland (named CCD).ReferencesBenonisson H, Altıntaş I, Sluijter M, Verploegen S, Labrijn AF, Schuurhuis DH, Houtkamp MA, Verbeek JS, Schuurman J and van Hall T. CD3-Bispecific antibody therapy turns solid tumors into inflammatory sites but does not install protective memory. Mol Cancer Ther 2019; 18(2):312–322.Groeneveldt, C, van Hall, T, van der Burg, SH, ten Dijke, P and van Montfoort, N. Immunotherapeutic potential of TGF-β inhibition and oncolytic viruses. Trends Immunol 2020; 41(5):406–420.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. TPS3112-TPS3112
Author(s):  
Kenji Hashimoto ◽  
Ayesh Perera ◽  
Naofumi Sugaya ◽  
Yoshitaka Ogita ◽  
Mikiko Nakamura ◽  
...  

TPS3112 Background: Bispecific antibodies to facilitate T-cell directed cytotoxicity (TDCC) is a proven therapy strategy in cancer. ERY974 is a humanized IgG4 bispecific antibody designed to simultaneously bind to cytotoxic T-cell CD3 receptors and GPC3 (a glycoprotein expressed on cell surface of several tumors) to elicit T-cell activation and TDCC. The objectives of this multi-country, phase 1 study of ERY974 is to determine the maximum tolerated dose (MTD) and to perform a preliminary assessment of anti-tumor activity in patients with solid tumors expressing GPC3. Methods: ERY974 is dosed IV weekly. All patients receive premedication with dexamethasone (DEX) prior to 1st and 2nd ERY974 dose. DE uses an accelerated titration design (ATD), then a modified continual reassessment method (mCRM) described by one-parameter logistic model, to determine MTD, where DLT occurrence rate is 0.25. Combining ATD and mCRM is to permit rapid dose escalation whilst minimizing patient numbers exposed to sub-therapeutic doses, and to accurately determine MTD. Once grade 2 (G2) cytokine release syndrome (CRS) is observed, DEX is increased. If ≥G2 CRS is again observed, then at all subsequent doses the 1st dose of ERY974 is fixed at the last dose level when < G2 CRS was not seen, DE proceeds with the 2nd dose. ATD commences with n = 1, increasing to n = 3 once drug-related ≥G2 toxicity is seen. mCRM starts after 1st dose limiting toxicity (DLT), with the modifications of at least 3 patients required to dose escalate and up to 1.5x increment to minimize risk of toxicity. CE has 3 arms: GPC3+ gastric/gastroesophageal junction adenocarcinoma; GPC3+ squamous esophageal cancer; and other GPC3+ tumors. A 2-stage design is used to allow CE to stop early for futility. Subjects are adults with histologically confirmed, measurable malignant solid tumors and/or metastatic disease not amenable to standard therapy, and life expectancy ≥3 months. Patients with > 1cm or > 1 brain metastasis, current/previous interstitial lung disease, and acute/chronic infection are excluded. 3 cohorts have been completed without DLT. Cohort 4 began in January 2017. Clinical trial information: NCT02748837.


Sign in / Sign up

Export Citation Format

Share Document