Rituximab-drug Conjugate Incorporating Auristatin E via A Quaternary Ammonium Linker Inducing Potent Antitumor Activity Against Non-Hodgkin’s B-cells

Author(s):  
Xin-Yue Hu ◽  
Lin-Lin Wang ◽  
Yue Sun ◽  
A-long Cui

Background: Rituximab represents a drug used for standard Non-Hodgkin’s B-cell lymphoma therapy; however, it displays limited clinical efficacy. Antibody-drug conjugate (ADC) is one of the potential strategies to increase the antitumor activity of an antibody, with improved cytotoxicity directly resulting from the delivery of a molecular warhead. Currently, the warhead monomethyl auristatin E (MMAE) has been widely applied in the study of ADCs, conjugated to a carbamate-based linker (MC-VC-PABC). However, the hydrophobic drug-linker (MC-VC-PABC-MMAE) may lead to ADC aggregation, ultimately resulting in decreased activity. Objective: In this study, we developed a hydrophilic drug-linker MC-VC-PABQ-AE linked to rituximab.. If the replacement of the tertiary amine in AE for a secondary amine in MMAE represents a characteristic modification, the change of antitumor activity of two corresponding anti-CD20 ADC is unknown, requiring further verification. Method: The structural elucidation of MC-VC-PAB-AE was displayed by high-resolution mass spectra. The average drug antibody ratio (DAR) of rituximab-VC-AE/MMAE ADCs was performed by HIC-HPLC. The cell cycle arrest analysis of two ADCs was detected by flow cytometry, and the antitumor activity of two ADCs was evaluated in vitro against Ramos and Daudi cells. Results: The average drug antibody ratio (DAR) of two ADCs was approximately 4.0. The activities of rituximab-VC-AE could be increased in CD20 positive B-lymphoma cell lines, most notably due to the higher cell viability inhibitory rates and apoptosis rates compared to rituximab-VC-MMAE. Conclusions: A hydrophilicity linker of ADC was developed and studied. Rituximab-VC-AE may potentially be used against CD20-positive cells, and the therapeutic efficacy and safety bring about further investigations.

2021 ◽  
Vol 4 (3) ◽  
pp. 175-184
Author(s):  
Hu Li ◽  
Xiao Zhang ◽  
Zhenyi Xu ◽  
Lingrui Li ◽  
Wenchao Liu ◽  
...  

Abstract Background ERBB2 is a proto-oncogene of multiple cancers including breast and gastric cancers with HER2 protein overexpression or gene amplification and has been proven clinically as a valid target for these cancers. HER2-targeting agents such as Herceptin®, Kadcyla® and ENHERTU® have been approved by the FDA for the treatment of breast cancer, but these drugs still face the challenge of acquired resistance and/or severe adverse reactions in clinical use. Therefore, there is significant unmet medical need for developing new agents that are more effective and safer for patients with advanced HER2-positive solid tumors including breast and gastric cancers. Methods We report here the making of MRG002, a novel HER2-targeted antibody drug conjugate (ADC), and preclinical characterization including pharmacology, pharmacodynamics and toxicology and discuss its potential as a novel agent for treating patients with HER2-positive solid tumors. Results MRG002 exhibited similar antigen binding affinity but much reduced antibody-dependent cellular cytotoxicity (ADCC) activity compared to trastuzumab. In addition to potent in vitro cytotoxicity, MRG002 showed tumor regression in both high- and medium-to-low HER2 expressing in vivo xenograft models. Furthermore, MRG002 showed enhanced antitumor activity when used in combination with an anti-PD-1 antibody. Main findings from toxicology studies are related to the payload and are consistent with literature report of other ADCs with monomethyl auristatinE. Conclusion MRG002 has demonstrated a favorable toxicity profile and potent antitumor activities in the breast and gastric PDX models with varying levels of HER2 expression, and/or resistance to trastuzumab or T-DM1. A phase I clinical study of MRG002 in patients with HER2-positive solid tumors is ongoing (CTR20181778).


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 610-610 ◽  
Author(s):  
Kevin J. Hamblett ◽  
Jeremy Barton ◽  
Charles G. Cerveny ◽  
Jamie B. Andreyka ◽  
Kim M. Kissler ◽  
...  

Abstract Hodgkin/Reed-Sternberg (H-RS) and anaplastic large cell lymphoma (ALCL) cells express high levels of the cell surface receptor CD30, a member of the TNF receptor superfamily. To augment the antitumor activity of anti-CD30 based therapies, a novel synthetic anti-mitotic agent, monomethyl auristatin E (MMAE), has been conjugated to the anti-CD30 antibody cAC10. MMAE was synthetically modified to include a maleimide for conjugation to an antibody and a protease cleavable Val-Cit peptide linker to release the drug from the antibody. The in vitro and in vivo activity of SGN-35, the resulting antibody-drug conjugate, is outlined. SGN-35 has an average of four MMAE molecules per antibody. Cell surface binding, internalization and intracellular release of MMAE were monitored by immunofluoresence and isotope labeling methods. The H-RS cell line L540cy translocates SGN-35 to lysosomes within 16 hours. Analysis of the cellular contents demonstrated that MMAE is rapidly and efficiently liberated from the antibody, and MMAE accumulates inside the cells. After treating L540cy cells with 200 ng/mL of SGN-35 (6 nM MMAE), the intracellular concentration of MMAE reaches 800 nM after 24 hours, rising to nearly 1000 nM after 72 hours. From 24 to 72 hours, about 70% of the intracellular drug is liberated MMAE, while 30% is still bound to the antibody. In vitro potency of SGN-35 was measured by incubating various concentrations of SGN-35 with CD30+ tumor cells and measuring cell viability in culture. SGN-35 had potent, selective in vitro activity with IC50s ranging from 4 to 35 ng/mL for CD30+ cell lines, and an IC50 > 1000 ng/mL for the CD30− line WSU-NHL. This demonstrates that the conjugation of the novel cytotoxic agent MMAE to an antibody can be accomplished without compromising the antibody’s specific binding and internalization. SCID mice were implanted with L540cy and treated with intravenous doses of SGN-35. In mice bearing L540cy xenografts, complete tumor regressions were achieved at doses as low as 2 mg/kg q4dx4. SGN-35 is also highly effective in treating mice with disseminated L540cy implants, at well tolerated doses. Based on the potent cytotoxicity, efficacy in xenograft models, and tolerability, SGN-35 is being advanced toward clinical development in CD30-positive hematologic malignancies.


2021 ◽  
Vol 10 (6) ◽  
pp. 1323
Author(s):  
Victor Yip ◽  
M. Violet Lee ◽  
Ola M. Saad ◽  
Shuguang Ma ◽  
S. Cyrus Khojasteh ◽  
...  

Polatuzumab vedotin (or POLIVY®), an antibody–drug conjugate (ADC) composed of a polatuzumab monoclonal antibody conjugated to monomethyl auristatin E (MMAE) via a cleavable dipeptide linker, has been approved by the United States Food and Drug Administration (FDA) for the treatment of diffuse large B-cell lymphoma (DLBCL). To support the clinical development of polatuzumab vedotin, we characterized the distribution, catabolism/metabolism, and elimination properties of polatuzumab vedotin and its unconjugated MMAE payload in Sprague Dawley rats. Several radiolabeled probes were developed to track the fate of different components of the ADC, with 125I and 111In used to label the antibody component and 3H to label the MMAE payload of the ADC. Following a single intravenous administration of the radiolabeled probes into normal or bile-duct cannulated rats, blood, various tissues, and excreta samples were collected over 7–14 days post-dose and analyzed for radioactivity and to characterize the metabolites/catabolites. The plasma radioactivity of polatuzumab vedotin showed a biphasic elimination profile similar to that of unconjugated polatuzumab but different from unconjugated radiolabeled MMAE, which had a fast clearance. The vast majority of the radiolabeled MMAE in plasma remained associated with antibodies, with a minor fraction as free MMAE and MMAE-containing catabolites. Similar to unconjugated mAb, polatuzumab vedotin showed a nonspecific distribution to multiple highly perfused organs, including the lungs, heart, liver, spleen, and kidneys, where the ADC underwent catabolism to release MMAE and other MMAE-containing catabolites. Both polatuzumab vedotin and unconjugated MMAE were mainly eliminated through the biliary fecal route (>90%) and a small fraction (<10%) was eliminated through renal excretion in the form of catabolites/metabolites, among which, MMAE was identified as the major species, along with several other minor species. These studies provided significant insight into ADC’s absorption, distribution, metabolism, and elimination (ADME) properties, which supports the clinical development of POLIVY.


2018 ◽  
Vol 19 (10) ◽  
pp. 3179 ◽  
Author(s):  
Hongling Gu ◽  
Na Li ◽  
Jiangkun Dai ◽  
Yaxi Xi ◽  
Shijun Wang ◽  
...  

A series of novel bivalent β-carboline derivatives were designed and synthesized, and in vitro cytotoxicity, cell apoptosis, and DNA-binding affinity were evaluated. The cytotoxic results demonstrated that most bivalent β-carboline derivatives exhibited stronger cytotoxicity than the corresponding monomer against the five selected tumor cell lines (A549, SGC-7901, Hela, SMMC-7721, and MCF-7), indicating that the dimerization at the C3 position could enhance the antitumor activity of β-carbolines. Among the derivatives tested, 4B, 6i, 4D, and 6u displayed considerable cytotoxicity against A549 cell line. Furthermore, 4B, 6i, 4D, and 6u induced cell apoptosis in a dose-dependent manner, and caused cell cycle arrest at the S and G2/M phases. Moreover, the levels of cytochrome C in mitochondria, and the expressions of bcl-2 protein, decreased after treatment with β-carbolines, which indicated that 6i and 6u could induce mitochondria-mediated apoptosis. In addition, the results of UV-visible spectral, thermal denaturation, and molecular docking studies revealed that 4B, 6i, 4D, and 6u could bind to DNA mainly by intercalation.


RSC Advances ◽  
2018 ◽  
Vol 8 (43) ◽  
pp. 24376-24385 ◽  
Author(s):  
Wen-Bin Kuang ◽  
Ri-Zhen Huang ◽  
Yi-Lin Fang ◽  
Gui-Bin Liang ◽  
Chen-Hui Yang ◽  
...  

A series of novel 2-chloro-3-(1H-benzo[d]imidazol-2-yl)quinoline derivatives were designed and synthesized as antitumor agents under the combination principle. The antitumor activity and mechanisms were then evaluated.


2012 ◽  
Vol 2012 ◽  
pp. 1-13 ◽  
Author(s):  
Jai-Sing Yang ◽  
Chia-Chun Wu ◽  
Chao-Lin Kuo ◽  
Yu-Hsuan Lan ◽  
Chin-Chung Yeh ◽  
...  

We investigated the molecular mechanisms of cell cycle arrest and apoptotic death induced bySolanum lyratumextracts (SLE) or diosgenin in WEHI-3 murine leukemia cellsin vitroand antitumor activityin vivo. Diosgenin is one of the components of SLE. Our study showed that SLE and diosgenin decreased the viable WEHI-3 cells and inducedG0/G1phase arrest and apoptosis in concentration- or time-dependent manners. Both reagents increased the levels of ROS production and decreased the mitochondrial membrane potential (ΔΨm). SLE- and diosgenin-triggered apoptosis is mediated through modulating the extrinsic and intrinsic signaling pathways. Intriguingly, the p53 inhibitor (pifithrin-α), anti-Fas ligand (FasL) mAb, and specific inhibitors of caspase-8 (z-IETD-fmk), caspase-9 (z-LEHD-fmk), and caspase-3 (z-DEVD-fmk) blocked SLE- and diosgenin-reduced cell viability of WEHI-3 cells. Thein vivostudy demonstrated that SLE has marked antitumor efficacy against tumors in the WEHI-3 cell allograft model. In conclusion, SLE- and diosgenin-inducedG0/G1phase arrest and triggered extrinsic and intrinsic apoptotic pathways via p53 activation in WEHI-3 cells. SLE also exhibited antitumor activityin vivo. Our findings showed that SLE may be potentially efficacious in the treatment of leukemia in the future.


2015 ◽  
Vol 51 (53) ◽  
pp. 10624-10627 ◽  
Author(s):  
João P. M. Nunes ◽  
Maurício Morais ◽  
Vessela Vassileva ◽  
Eifion Robinson ◽  
Vineeth S. Rajkumar ◽  
...  

A next generation maleimide–ADC is shown to have excellent stability in blood serum, as well as high potency and selectivity in vitro.


2018 ◽  
Author(s):  
Marc Hillairet de Boisferon ◽  
Caroline Mignard ◽  
Coralie Durix ◽  
Toshimitsu Uenaka ◽  
Katherine Rybinski ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document