CD3-Bispecific Antibody Therapy Turns Solid Tumors into Inflammatory Sites but Does Not Install Protective Memory

2018 ◽  
Vol 18 (2) ◽  
pp. 312-322 ◽  
Author(s):  
Hreinn Benonisson ◽  
Işıl Altıntaş ◽  
Marjolein Sluijter ◽  
Sandra Verploegen ◽  
Aran F. Labrijn ◽  
...  
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.


Cancers ◽  
2021 ◽  
Vol 13 (2) ◽  
pp. 287
Author(s):  
Jim Middelburg ◽  
Kristel Kemper ◽  
Patrick Engelberts ◽  
Aran F. Labrijn ◽  
Janine Schuurman ◽  
...  

Immunotherapy of cancer with CD3-bispecific antibodies is an approved therapeutic option for some hematological malignancies and is under clinical investigation for solid cancers. However, the treatment of solid tumors faces more pronounced hurdles, such as increased on-target off-tumor toxicities, sparse T-cell infiltration and impaired T-cell quality due to the presence of an immunosuppressive tumor microenvironment, which affect the safety and limit efficacy of CD3-bispecific antibody therapy. In this review, we provide a brief status update of the CD3-bispecific antibody therapy field and identify intrinsic hurdles in solid cancers. Furthermore, we describe potential combinatorial approaches to overcome these challenges in order to generate selective and more effective responses.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A861-A861
Author(s):  
Jifang Gong ◽  
Lin Shen ◽  
Zhi Dong ◽  
Dan Liu ◽  
June Xu ◽  
...  

BackgroundHER2 potently inhibits innate immunity through cGAS–STING signaling [Ref],meanwhile HER2 antibody induced ADCP will also lead to macrophage mediated immune suppression. Both preclinical and clinical studies have suggested a coordination of engagement of innate and adaptive immunity with the combination of an anti-HER2 antibody and an immune checkpoint blockade. KN026 is a novel bispecific antibody that simultaneously binds to two distinct HER2 epitopes. KN046 is a novel bispecific antibody that blocks both PD-L1 interaction with PD-1/CD80 and CTLA-4 interaction with CD80/CD86. Here we reported the interim results from an ongoing phase Ib dose escalation and expansion study assessing the safety, tolerability and preliminary efficacy for KN026 in combination with KN046 in Patients with HER2 aberrated solid tumors.MethodsThis study enrolled pts with solid tumors who failed available standard of care, HER2 aberration status confirmed locally (HER2 mutation, HER2 amplification and/or HER2 overexpression). Eligible pts received combination of KN026 and KN046 at three dose levels until disease progression, unacceptable toxicity or withdrawal of informed consent (DL1: KN026 20 mg/kg Q2W + KN046 3 mg/kg Q2W; DL2: KN026 20 mg/kg Q2W with loading on Days 1, 8 of Cycle 1 + KN046 5 mg/kg Q3W; DL3: KN026 30 mg/kg Q3W with loading on Days 1, 8 of Cycle 1 + KN046 5 mg/kg Q3W). Tumor response was evaluated Q8W per RECIST 1.1. Primary endpoint was DLT and key secondary endpoints were efficacy parameters (ORR, DOR, PFS).ResultsAs of the Sep. 08, 2020, 25 pts were enrolled into DL1 (n = 20, 3 for dose escalation), DL2 (n = 3) and DL3 (n = 2) (mGC/GEJ 15 pts; mCRC 8 pts; other solid tumors 2 pts). 15 pts remained on the study treatment and 10 pts discontinued treatment due to disease progression (n=5), death (n=2) and other reasons (n=3). 18 pts had HER2-positive status (12 of 18 failed previous trastuzumab therapy), 2 pts had HER2 mutation and 5 pts had HER2 low expression (without FISH amplification). No DLTs were observed. No pts experienced LVEF decreased or other clinically meaningful cardiac AEs. Treatment-related TEAEs occurred in 23 (92%) pts, of which 6 (24%) pts experienced grade 3 or above treatment-related TEAEs. 11 (44%) pts experienced irAEs, majority were of grade 1 or 2 except that 1 patient experienced grade 3 immune-mediated endocrinopathy. The most common (frequency ≥ 15%) KN026 or KN046 related TEAEs were infusion related reaction (n=11, 44.0%), anaemia (n=9, 36.0%), white blood cell count decreased (n=6, 24.0%), diarrhea (n=5, 20.0%), AST increased (n=5, 20.0%), platelet count decreased (n=5, 20.0%), rash (n=5, 20.0%) and ALT increased (n=4, 16.0%). The objective response rate in pts with HER2-positive tumors (n = 14 efficacy evaluable pts) was 9/14 (64.3%, 95% CI 35.1~87.2%) and disease control rate 13/14 (92.9%, 95% CI 66.1~99.8%). 4 out of 5 pts with HER2 mutation or low expression achieved SD including one patient with SD for more than 24 weeks. 2 death cases due to disease progression were reported, both only received one cycle of KN026 plus KN046 due to COVID-19 restriction.ConclusionsKN026 combined with KN046 is well tolerated and has demonstrated preliminary albeit profound anti-tumor activity in HER2-positive solid tumors.Trial RegistrationClinical trial information: NCT04040699ReferenceShiying Wu, Qian Zhang, Fei Zhang, et al. HER2 recruits AKT1 to disrupt STING signalling and suppress antiviral defence and antitumour immunity. Nature Cell Biology 2019;21:1027–1040.


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.


2019 ◽  
Author(s):  
Danielle Dettling ◽  
Eilene Kwok ◽  
Lucy Quach ◽  
Aakash Datt ◽  
Jeremiah D. Degenhardt ◽  
...  

Antibodies ◽  
2019 ◽  
Vol 8 (3) ◽  
pp. 43 ◽  
Author(s):  
Qiong Wang ◽  
Yiqun Chen ◽  
Jaeyoung Park ◽  
Xiao Liu ◽  
Yifeng Hu ◽  
...  

With the current biotherapeutic market dominated by antibody molecules, bispecific antibodies represent a key component of the next-generation of antibody therapy. Bispecific antibodies can target two different antigens at the same time, such as simultaneously binding tumor cell receptors and recruiting cytotoxic immune cells. Structural diversity has been fast-growing in the bispecific antibody field, creating a plethora of novel bispecific antibody scaffolds, which provide great functional variety. Two common formats of bispecific antibodies on the market are the single-chain variable fragment (scFv)-based (no Fc fragment) antibody and the full-length IgG-like asymmetric antibody. Unlike the conventional monoclonal antibodies, great production challenges with respect to the quantity, quality, and stability of bispecific antibodies have hampered their wider clinical application and acceptance. In this review, we focus on these two major bispecific types and describe recent advances in the design, production, and quality of these molecules, which will enable this important class of biologics to reach their therapeutic potential.


Sign in / Sign up

Export Citation Format

Share Document