scholarly journals Human immunocompetent Organ-on-Chip platforms allow safety profiling of tumor-targeted T-cell bispecific antibodies

eLife ◽  
2021 ◽  
Vol 10 ◽  
Author(s):  
S Jordan Kerns ◽  
Chaitra Belgur ◽  
Debora Petropolis ◽  
Marianne Kanellias ◽  
Riccardo Barrile ◽  
...  

Traditional drug safety assessment often fails to predict complications in humans, especially when the drug targets the immune system. Here, we show the unprecedented capability of two human Organs-on-Chips to evaluate the safety profile of T-cell bispecific antibodies (TCBs) targeting tumor antigens. Although promising for cancer immunotherapy, TCBs are associated with an on-target, off-tumor risk due to low levels of expression of tumor antigens in healthy tissues. We leveraged in vivo target expression and toxicity data of TCBs targeting folate receptor 1 (FOLR1) or carcinoembryonic antigen (CEA) to design and validate human immunocompetent Organs-on-Chips safety platforms. We discovered that the Lung-Chip and Intestine-Chip could reproduce and predict target-dependent TCB safety liabilities, based on sensitivity to key determinants thereof, such as target expression and antibody affinity. These novel tools broaden the research options available for mechanistic understandings of engineered therapeutic antibodies and assessing safety in tissues susceptible to adverse events.

2021 ◽  
Author(s):  
S. Jordan Kerns ◽  
Chaitra Belgur ◽  
Debora B. Petropolis ◽  
Riccardo Barrile ◽  
Marianne Kanellias ◽  
...  

AbstractTraditional drug safety assessment often fails to predict complications in humans, especially when the drug targets the immune system. Here, we show the unprecedented capability of two Organs-on-Chips to evaluate the safety profile of T-cell bispecific antibodies (TCBs) targeting tumor antigens. Although promising for cancer immunotherapy, TCBs are associated with an on-target, off-tumor risk due to low levels of expression of tumor antigens in healthy tissues. We leveraged in vivo target expression and toxicity data of TCBs targeting folate receptor 1 (FOLR1) or carcinoembryonic antigen (CEA) to design and validate human immunocompetent Organs-on-Chips safety platforms. We discovered that the Lung-Chip and Intestine-Chip could reproduce and predict target-dependent TCB safety liabilities, based on sensitivity to key determinants thereof, such as target expression and antibody affinity. These novel tools broaden the research options available for mechanistic understandings of engineered therapeutic antibodies and assessing safety in tissues susceptible to adverse events.


1995 ◽  
Vol 40 (3) ◽  
pp. 182-190 ◽  
Author(s):  
M. Belen Moreno ◽  
Julie A. Titus ◽  
Michael S. Cole ◽  
J. Yun Tso ◽  
Nhat Le ◽  
...  

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1709-1709
Author(s):  
Eva Alvarez ◽  
Esther Moga ◽  
Jorge Sierra ◽  
Javier Briones

Abstract Dendritic cells (DCs) are the main antigen presenting cells and play a pivotal role in the stimulation of T-cell immune responses. DCs cultured in the presence of a single tumor antigen can elicit an immune response against tumor cells expressing that antigen. However, simultaneous use of several tumor antigens may be advantageous since polyclonal activation of T cells against different tumor antigens may be a better approach to eradicate tumor cells. In this sense, fusions of dendritic and tumor cells (FCs) show a broad spectrum of tumor antigens, both known and unidentified, to be presented by class I and II MHC. Although prophylactic vaccines were successful in murine models, the results in the therapeutic setting have been unsatisfactory. We hypothesised that enhancing costimulation of FCs would help to break tumor tolerance once the tumor is established. To this purpose, we transduced FCs with a recombinant adenovirus encoding CD40L (AdvCD40L or AdvGFP as control) and we studied the therapeutic antitumoral effect of the administration of FC-CD40L in a murine model of myeloma. DCs obtained from day 7-bone marrow cultures of Balb/c mice were fused with tumor cells, a syngeneic murine myeloma cell line (4TOO). FCs hybrids were generated with PEG and selected after culturing in HAT medium plus GM-CSF for 7 days. FC were quantified by determining the percentage of cells that coexpress specific DC (CD11c) and tumor markers (CD138). Mean fusion efficiency was 30% (20–40%) and FCs expressed moderate levels of CD80, CD83, CD86, CD54, CD40 and MHC II and did not express CD40L. FC-CD40L showed a significant increase of expression of costimulatory molecules (CD80, CD86, CD54, and MHC II) compared to FC-GFP (p=0.011). Moreover, in a syngeneic mixed lymphocyte reaction, FC-CD40L induced a two-fold higher T-cell proliferation than FC-GFP or FC alone. In addition, FC-CD40L had improved migration to lymphoid tissues, preferentially to spleen, in comparison with FC-GFP (2.8% versus 1.6%). The antitumor effect of FC-CD40L was analyzed in vivo. Mice (n=10 per group) were injected i.v. with 2.5×105 tumor cells and treated with irradiated FC, FC-GFP or FC-CD40L (1×106 cells each) on days 2, 6 and 10 after tumor challenge. 40% of mice treated with FC-CD40L had long-term survival (>120 days). In contrast, all of mice treated with FC or FC-GFP died between days 25 and 35 (p=0.012). In parallel, treatment with mixed cells (not fused DC+ tumor cells), mix transduced with AdvGFP, or mix transduced with AdvCD40L did not provide any significant antitumor effect. We conclude that FCs transduced with AdvCD40L better stimulate in vitro and in vivo immune responses than FC alone and may provide a new strategy for treating patients with multiple myeloma or lymphoma.


2015 ◽  
Vol 112 (14) ◽  
pp. E1754-E1762 ◽  
Author(s):  
Diego Chowell ◽  
Sri Krishna ◽  
Pablo D. Becker ◽  
Clément Cocita ◽  
Jack Shu ◽  
...  

Despite the availability of major histocompatibility complex (MHC)-binding peptide prediction algorithms, the development of T-cell vaccines against pathogen and tumor antigens remains challenged by inefficient identification of immunogenic epitopes. CD8+ T cells must distinguish immunogenic epitopes from nonimmunogenic self peptides to respond effectively against an antigen without endangering the viability of the host. Because this discrimination is fundamental to our understanding of immune recognition and critical for rational vaccine design, we interrogated the biochemical properties of 9,888 MHC class I peptides. We identified a strong bias toward hydrophobic amino acids at T-cell receptor contact residues within immunogenic epitopes of MHC allomorphs, which permitted us to develop and train a hydrophobicity-based artificial neural network (ANN-Hydro) to predict immunogenic epitopes. The immunogenicity model was validated in a blinded in vivo overlapping epitope discovery study of 364 peptides from three HIV-1 Gag protein variants. Applying the ANN-Hydro model on existing peptide-MHC algorithms consistently reduced the number of candidate peptides across multiple antigens and may provide a correlate with immunodominance. Hydrophobicity of TCR contact residues is a hallmark of immunogenic epitopes and marks a step toward eliminating the need for empirical epitope testing for vaccine development.


2000 ◽  
Vol 192 (11) ◽  
pp. 1637-1644 ◽  
Author(s):  
Cassian Yee ◽  
John A. Thompson ◽  
Patrick Roche ◽  
David R. Byrd ◽  
Peter P. Lee ◽  
...  

Current strategies for the immunotherapy of melanoma include augmentation of the immune response to tumor antigens represented by melanosomal proteins such as tyrosinase, gp100, and MART-1. The possibility that intentional targeting of tumor antigens representing normal proteins can result in autoimmune toxicity has been postulated but never demonstrated previously in humans. In this study, we describe a patient with metastatic melanoma who developed inflammatory lesions circumscribing pigmented areas of skin after an infusion of MART-1–specific CD8+ T cell clones. Analysis of the infiltrating lymphocytes in skin and tumor biopsies using T cell–specific peptide–major histocompatibility complex tetramers demonstrated a localized predominance of MART-1–specific CD8+ T cells (>28% of all CD8 T cells) that was identical to the infused clones (as confirmed by sequencing of the complementarity-determining region 3). In contrast to skin biopsies obtained from the patient before T cell infusion, postinfusion biopsies demonstrated loss of MART-1 expression, evidence of melanocyte damage, and the complete absence of melanocytes in affected regions of the skin. This study provides, for the first time, direct evidence in humans that antigen-specific immunotherapy can target not only antigen-positive tumor cells in vivo but also normal tissues expressing the shared tumor antigen.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 9-10
Author(s):  
Jamie Oakley ◽  
Evelyn K. Williams ◽  
Christina Caruso ◽  
Yumiko Sakurai ◽  
Reginald Tran ◽  
...  

Hyperleukocytosis, most commonly defined as a white blood cell (WBC) count > 100,000/μL, is an oncologic emergency in acute leukemia that can lead to leukostasis, which occurs when leukemia cells obstruct the microvasculature resulting in significant morbidity and mortality from neurologic (CNS hemorrhage, thrombosis) or pulmonary (respiratory distress, hypoxia) symptoms. The underlying mechanisms are poorly understood but are thought to be related to increased blood viscosity, secondary to high WBC count, leukemia cell aggregation, and the abnormal mechanical properties, size, and cell-cell interactions of leukemia cells. Leukapheresis is a commonly used therapy for rapid cytoreduction in symptomatic patients, but the procedure is not without risks. No existing methods reliably predict leukostasis or guide treatment including the commonly used WBC count, which only loosely correlates with leukostasis and does not accurately describe the blood viscosity at the microvascular level. Importantly, while hematocrit/hemoglobin levels (Hgb) are known to be major contributors to blood viscosity, they have not been systematically assessed in leukostasis risk, and Hgb often decreases as leukemic cell counts rise, complicating the issue. Incorporating Hgb levels may better predict leukostasis and assist physicians balancing the risk of hyperleukocytosis compared to the interventions themselves. To that end, we investigated how the differing presentations of acute leukemia lead to microvessel occlusion, thereby affecting effective blood viscosity at the microvascular level using "microvasculature-on-a-chip" devices that mimic the microvascular geometry (Figure 1) developed by our laboratory. This physiologically relevant microvascular model allows for in vitro investigation as in vivo studies are nearly impossible due to difficulty in visualizing and manipulating the animal microvasculature and cell counts. The devices were microfabricated using polydimethylsiloxane (PDMS). Acute T-cell lymphoblastic (Jurkat) and acute monocytic (THP-1) cell lines were maintained via standard cell culture conditions. Red cells from healthy donors were isolated and mixed with leukemia cells to achieve target Hgb and WBC levels. Various physiologic leukemia "mixtures" were then perfused under physiologic microcirculatory flow conditions through the microvascular device and microchannels occlusion was tracked via videomicroscopy (Figure 2). With T-cell leukemia, Hgb levels affected the risk of "in vitro leukostasis." Specifically, with severe anemia and WBC count less than the hyperleukocytosis range, time to microchannel occlusion was longer, and was more dependent on Hgb rather than WBC count. However, in cases with severe anemia and WBC counts > 100k/μL, WBC count exhibited a stronger effect on occlusion with little dependence on Hgb (Figure 3). At Hgb > 8g/dL, microchannel occlusion was dependent on WBC count regardless of hyperleukocytosis or not. In contrast, our data to date shows that with myeloid leukemia, in vitro leukostasis is not associated with Hgb levels, and is consistent with how myeloid leukemias in vivo cause leukostasis symptoms at lower WBC counts than lymphoid leukemias, not only due to size but also adhesive interactions. These data suggest when determining risk for leukostasis, WBC count should not be the sole determinant. Here we show Hgb levels affect microvascular blood viscosity and propensity for microvascular occlusion, but it appears to have a greater impact with T-cell leukemias versus myeloid leukemias (Figure 4). These studies indicate Hgb is an important clinical parameter for leukostasis risk in acute leukemia and will help inform guidelines for leukapheresis and even phlebotomy, a much simpler and safer procedure, to mitigate hyperviscosity in acute leukemia. These results can also impact decisions regarding the need for red blood cell transfusions, which iatrogenically increase blood viscosity. Studies incorporating patient myeloid and lymphoid leukemia cells and microvasculature-on-chip devices integrating live endothelium to assess leukemia cell adhesion are ongoing. Figure Disclosures Lam: Sanguina, Inc: Current equity holder in private company.


2020 ◽  
Vol 12 (534) ◽  
pp. eaax1315 ◽  
Author(s):  
Brian H. Santich ◽  
Jeong A. Park ◽  
Hoa Tran ◽  
Hong-Fen Guo ◽  
Morgan Huse ◽  
...  

T cell–bispecific antibodies (BsAbs) couple cytotoxic T lymphocytes to tumor cells, inducing their destruction. Although there are more than 60 classes of BsAbs in development, the relative importance of parameters such as interdomain spacing or spatial configuration is largely unknown. Here, we dissected a symmetric dual bivalent BsAb platform (IgG-[L]-scFv: antitumor IgG with anti-CD3 scFv fused to the light chains) to explore the importance of valency and spatial configuration for BsAb-induced T cell cytotoxicity. Our results revealed that placing tumor and T cell binding domains on the same side of a BsAb (cis-configuration) elicited substantially stronger antitumor activity, in vitro and in vivo, compared to positioning them on opposite sides (trans-configuration). Moreover, using two cis-modules in the same BsAb further improved cytotoxicity (up to 2000-fold). In addition, separating antigen-binding components with a single Ig domain (CL) markedly enhanced cytokine release and in vivo tumor responses compared to smaller (G4S1) or larger (CH1-CH2-CH3) spacers. These findings provide guidelines for improving BsAb function and highlight the importance of spatial configuration and dual bivalency as development parameters.


Pharmaceutics ◽  
2020 ◽  
Vol 12 (3) ◽  
pp. 215
Author(s):  
Sara Nava ◽  
Daniela Lisini ◽  
Simona Frigerio ◽  
Simona Pogliani ◽  
Serena Pellegatta ◽  
...  

Dendritic cells (DC) are the most potent antigen-presenting cells, strongly inducers of T cell-mediated immune responses and, as such, broadly used as vaccine adjuvant in experimental clinical settings. DC are widely generated from human monocytes following in vitro protocols which require 5–7 days of differentiation with GM-CSF and IL-4 followed by 2–3 days of activation/maturation. In attempts to shorten the vaccine’s production, Fast-DC protocols have been developed. Here we reported a Fast-DC method in compliance with good manufacturing practices for the production of autologous mature dendritic cells loaded with antigens derived from whole tumor lysate, suitable for the immunotherapy in glioblastoma patients. The feasibility of generating Fast-DC pulsed with whole tumor lysate was assessed using a series of small-scale cultures performed in parallel with clinical grade large scale standard method preparations. Our results demonstrate that this Fast protocol is effective only in the presence of PGE2 in the maturation cocktail to guarantee that Fast-DC cells exhibit a mature phenotype and fulfill all requirements for in vivo use in immunotherapy approaches. Fast-DC generated following this protocol were equally potent to standard DC in inducing Ag-specific T cell proliferation in vitro. Generation of Fast-DC not only reduces labor, cost, and time required for in vitro clinical grade DC development, but can also minimizes inter-preparations variability and the risk of contamination.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 5432-5432
Author(s):  
Mauro Castellarin ◽  
Joseph A. Fraietta ◽  
Jihyun Lee ◽  
John Scholler ◽  
Yangbing Zhao ◽  
...  

Abstract Chimeric antigen receptor (CAR) engineered T cells have been used clinically to improve outcomes in patients with hematopoietic malignancies owing to the ability of CAR T cells to recognize tumor antigens and kill malignant cells. CAR T cells possess the antigen recognizing capability of an antibody through the single chain variable fragment (scFv) and their cytotoxicity is enhanced through signaling via the intracellular domains of T cell receptors and co-activating receptors such as CD3zeta and 4-1BB, respectively. Thus, CAR expressing T cells are able to detect cancer cells through tumor antigens and can become activated to unleash their cytotoxic potentials in a non-MHC restricted manner. Therapeutic side-effects can occur when T-cell receptor targeting is misdirected to the incorrect tissue causing potentially serious on-target off-tumor cytotoxicity. Factors that influence CAR targeting include expression levels of tumor-associated antigen in normal tissue and the binding affinities of scFvs. Our first step in developing an in vivo, on target, off-tumor, CAR T cell toxicity model was to generate mice with tunable expression of a human tumor antigen in normal tissue. NSG mice were IV injected with recombinant adeno-associated virus serotype 8 (rAAV8) to deliver a truncated human ErbB2 (Her2/neu or CD340) gene and a Katushka fluorescent reporter that were driven by the liver-specific promoter, thyroxine binding globulin (TBG). AAV8 genomic copies (GCs) were injected at varying dilutions of 1.5 x 1012 GC/mouse, 7.25 x 1011 GC/mouseand 1.5x1010 GC/mouse to induce a range of expression of ErbB2 in the liver. Katushka expression was visualized in vivo using the IVIS small animal imager. ErbB2 gene expression was detected using reverse transcription polymerase chain reaction (qRT-PCR) and the ErbB2 protein was detected using western blots and immunohistochemistry (IHC). Our data has shown that expression levels of ErbB2 and the Katushka reporter positively correlated with the number of AAV8 GCs that were injected. This enabled us to obtain ErbB2 expression levels in the liver comparable to the levels seen in either ErbB2High tumors (eg. SK-OV3) or ErbB2Low tumors (eg. PC3 and HEK293T). To determine if affinity tuning of scFvs will allow CAR T cells to discriminate between high and low ErbB2 expression in the liver, T cells were engineered to co-express the click beetle red (CBR) reporter and either a high-affinity scFv, anti-ErbB2 CAR (4D5) or a low-affinity scFv, anti-ErbB2 CAR (4D5-5). These T cells were then IV injected into NSG mice that had either high or low ErbB2-expressing livers. Although these experiments were ongoing at the time of abstract submission, we will show our results on T cell trafficking in the liver, which will be visualized by IHC and by in vivo imaging using the IVIS small animal imager. Liver toxicity will be assessed by histological examination and by measuring liver function via standard enzymatic testing of blood. Furthermore, we aim to show whether affinity tuning of scFvs will allow CAR T cells to selectively recognize and target ErbB2High tumors while sparing ErbB2Low normal tissue. This will be performed by inoculating ErbB2high SK-OV3 tumor cells into mice with ErbB2Low livers followed by IV injection with either 4D5 or 4D5-5 CAR T cells. We expect that the low-affinity anti-ErbB2 CAR (4D5-5) T cells will target the ErbB2High SK-OV3 tumor cells and cause tumor regression while preserving function in the ErbB2Low liver. If so, then we will have shown that our pre-clinical mouse model can be used to identify on-target off-tumor CAR T cell toxicity, which will aid in improving the safety profile and clinical outcomes of future CAR T cell therapies. Disclosures Scholler: Novartis: Patents & Royalties. Zhao:Novartis: Patents & Royalties, Research Funding. June:Novartis: Patents & Royalties, Research Funding.


Sign in / Sign up

Export Citation Format

Share Document