Abstract 2578: Efficacy of anti-CD22 antibody drug-conjugates in Ramos and Raji tumor xenograft models

Author(s):  
Chin Pan ◽  
Colin Chong ◽  
Orville Cortez ◽  
Alison Witte ◽  
Ayesha Nazeer ◽  
...  
2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Hung-Ju Hsu ◽  
Chao-Ping Tung ◽  
Chung-Ming Yu ◽  
Chi-Yung Chen ◽  
Hong-Sen Chen ◽  
...  

AbstractMesothelin (MSLN) is an attractive candidate of targeted therapy for several cancers, and hence there are increasing needs to develop MSLN-targeting strategies for cancer therapeutics. Antibody–drug conjugates (ADCs) targeting MSLN have been demonstrated to be a viable strategy in treating MSLN-positive cancers. However, developing antibodies as targeting modules in ADCs for toxic payload delivery to the tumor site but not to normal tissues is not a straightforward task with many potential hurdles. In this work, we established a high throughput engineering platform to develop and optimize anti-MSLN ADCs by characterizing more than 300 scFv CDR-variants and more than 50 IgG CDR-variants of a parent anti-MSLN antibody as candidates for ADCs. The results indicate that only a small portion of the complementarity determining region (CDR) residues are indispensable in the MSLN-specific targeting. Also, the enhancement of the hydrophilicity of the rest of the CDR residues could drastically increase the overall solubility of the optimized anti-MSLN antibodies, and thus substantially improve the efficacies of the ADCs in treating human gastric and pancreatic tumor xenograft models in mice. We demonstrated that the in vivo treatments with the optimized ADCs resulted in almost complete eradication of the xenograft tumors at the treatment endpoints, without detectable off-target toxicity because of the ADCs’ high specificity targeting the cell surface tumor-associated MSLN. The technological platform can be applied to optimize the antibody sequences for more effective targeting modules of ADCs, even when the candidate antibodies are not necessarily feasible for the ADC development due to the antibodies’ inferior solubility or affinity/specificity to the target antigen.


2019 ◽  
Author(s):  
Deng Pan ◽  
Yubo Tang ◽  
Jiao Tong ◽  
Chengmei Xie ◽  
Jiaxi Chen ◽  
...  

AbstractBackgroundAntibodies targeting abnormally glycosylated proteins have been ineffective in treating cancer. Antibody-drug conjugates are emerging as an efficient option, which allow specific delivery of drugs into tumors. We and others have dissected the abnormally glycosylated tandem repeat region of MUC1 glycoprotein as three site-specific glycosylated neoantigen peptide motifs (PDTR, GSTA, GVTS) for monoclonal antibody binding.MethodsInternalization of monoclonal antibodies was studied by immunofluorescence staining and colocalization with lysosomal markers in live cells. Antibody positivity in tumor and peritumoral tissue samples were studied by immunohistochemistry. The efficacy of anti-MUC1 ADCs were evaluated with various cancer cell lines and mouse tumor xenograft model.ResultsWe describe an anti-MUC1 ADC by conjugating GSTA neoantigen-specific 16A with monomethyl auristatin E (MMAE). 16A-MMAE showed potent antitumoral efficacy with IC50 ranging from 0.2 to 49.4 nM toward multiple types of cancer cells. In vivo, 16A-MMAE showed dose-dependent inhibition of tumor growth in mouse xenograft of NCI-H838 NSCLC cell line, with minimum effective dose at 1 mg/kg. At the dose of 3 mg/kg, 16A-MMAE did not cause significant toxicity in a transgenic mouse expressing human MUC1.ConclusionsThe high antitumoral efficacy of 16A-MMAE suggest that aberrant glycosylated MUC1 neoantigen is a target with high positivity in multiple cancer types for ADC development. Personalized therapy may be achieved by development of glycosite-specific antibody-drug conjugates.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 526-526
Author(s):  
Andrew G. Polson ◽  
Suzanna Clark ◽  
Changchun Du ◽  
Dan Eaton ◽  
Kristi Elkins ◽  
...  

Abstract Antibody-drug conjugates (ADCs), potent cytotoxic drugs linked to antibodies via specialized chemical linkers, provide a means to increase the effectiveness of chemotherapy by targeting the drug to neoplastic cells while reducing side effects. Our research has focused on ADCs that contain the maytansinoid DM1, monomethylauristatin E (MMAE), or monomethylauristatin F (MMAF), all of which are potent inhibitors of microtubule polymerization. These drugs are attached to the antibody via several different linker formats (Table 1). Table 1 Linker-drug Linker Mechanism Linked via Drug Cell Permeable PAB-vc-MC-MMAE Cleavage of peptide bond Cysteine Yes MC-MMAF Uncleavable Cysteine No SPP-DM1 Reduction of disulfide bond Lysine Yes MCC-DM1 Uncleavable Lysine Yes We identified seven cell surface proteins expressed in non-Hodgkin lymphoma (NHL) with limited expression patterns in normal tissue (CD19, CD20, CD21, CD22, CD72, CD79, and CD180), as potential therapeutic targets for ADCs. ADCs directed to any of these seven targets are effective in xenograft models when containing linkers that can be cleaved in the tumor microenvironment or when internalized by tumor cells. However, ADCs with uncleavable linkers, i.e., linkers that are active when antibody is internalized and degraded within cells, are effective only when targeted to CD22 or CD79b. This suggests that these antigens provide the desired intracellular targeting of the cytotoxic drug. Further, we demonstrated that the uncleavable-linker anti-CD22 and anti-CD79b ADCs result in complete tumor regression with no recurrence in some xenograft models of NHL. In vitro experiments revealed that sensitivity of a given cell line to the ADCs anti-CD22-MCC-DM1 and anti-CD79b-MCC-DM1, correlated more with sensitivity of the cell line to the corresponding free drug, than to the amount of surface expression of the target or the amount of internalized ADC. We conducted pilot safety studies in rats (non-binding species) and cynomolgus monkeys (binding species). In rats, ADCs with uncleavable linkers caused less target-independent hepatic and hematologic toxicity than their counter parts with cleavable linkers, likely due to their decreased systemic release of free drug. Because of these results, we evaluated three ADCs with uncleavable linkers (anti-CD22-MCC-DM1, anti-cynomolgus monkey CD79b-MCC-DM1, and anti-CD22-MC-MMAF) at ∼30 mg/kg q3 weeks for 2 doses in cynomolgus monkeys. Normal B-cells were depleted in circulation and in tissue, particularly in germinal centers of lymphoid tissues; an expected pharmacological effect. Other findings included clinically tolerated and reversible elevations in liver enzymes (MCC-DM1 and MC-MMAF conjugates) and decreases in platelet counts (MCC-DM1 conjugates); and minimal to mild sciatic nerve degeneration (MCC-DM1 conjugates). The data demonstrate that anti-CD22 and anti-CD79b ADCs with uncleavable linkers have potent efficacy in xenograft models of NHL, favorable safety profiles in pilot animal studies, and suggest that for specific targets, uncleavable linkers provide a promising mechanism to improve margins of safety in humans.


2019 ◽  
Vol 30 (5) ◽  
pp. 1356-1370 ◽  
Author(s):  
Dian Su ◽  
Jinhua Chen ◽  
Ely Cosino ◽  
Josefa dela Cruz-Chuh ◽  
Helen Davis ◽  
...  

2015 ◽  
Author(s):  
Esther C.W. Breij ◽  
Sandra Verploegen ◽  
Andreas Lingnau ◽  
Edward N. van den Brink ◽  
Maarten Janmaat ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document