Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Nicholas C. Yoder is active.

Publication


Featured researches published by Nicholas C. Yoder.


Bioconjugate Chemistry | 2017

Effects of Drug–Antibody Ratio on Pharmacokinetics, Biodistribution, Efficacy, and Tolerability of Antibody–Maytansinoid Conjugates

Xiuxia Sun; Jose F. Ponte; Nicholas C. Yoder; Rassol Laleau; Jennifer Coccia; Leanne Lanieri; Qifeng Qiu; Rui Wu; Erica Hong; Megan Bogalhas; Lintao Wang; Ling Dong; Yulius Y. Setiady; Erin Maloney; Olga Ab; Xiaoyan Zhang; Jan Pinkas; Thomas A. Keating; Ravi S. Chari; Hans K. Erickson; John M. Lambert

Antibody-drug conjugates (ADCs) are being actively pursued as a treatment option for cancer following the regulatory approval of brentuximab vedotin (Adcetris) and ado-trastuzumab emtansine (Kadcyla). ADCs consist of a cytotoxic agent conjugated to a targeting antibody through a linker. The two approved ADCs (and most ADCs now in the clinic that use a microtubule disrupting agent as the payload) are heterogeneous conjugates with an average drug-to-antibody ratio (DAR) of 3-4 (potentially ranging from 0 to 8 for individual species). Ado-trastuzumab emtansine employs DM1, a semisynthetic cytotoxic payload of the maytansinoid class, which is conjugated via lysine residues of the antibody to an average DAR of 3.5. To understand the effect of DAR on the preclinical properties of ADCs using maytansinoid cytotoxic agents, we prepared a series of conjugates with a cleavable linker (M9346A-sulfo-SPDB-DM4 targeting folate receptor α (FRα)) or an uncleavable linker (J2898A-SMCC-DM1 targeting the epidermal growth factor receptor (EGFR)) with varying DAR and evaluated their biochemical characteristics, in vivo stability, efficacy, and tolerability. For both formats, a series of ADCs with DARs ranging from low (average of ∼2 and range of 0-4) to very high (average of 10 and range of 7-14) were prepared in good yield with high monomer content and low levels of free cytotoxic agent. The in vitro potency consistently increased with increasing DAR at a constant antibody concentration. We then characterized the in vivo disposition of these ADCs. Pharmacokinetic analysis showed that conjugates with an average DAR below ∼6 had comparable clearance rates, but for those with an average DAR of ∼9-10, rapid clearance was observed. Biodistribution studies in mice showed that these 9-10 DAR ADCs rapidly accumulate in the liver, with maximum localization for this organ at 24-28% percentage injected dose per gram (%ID/g) compared with 7-10% for lower-DAR conjugates (all at 2-6 h post-injection). Our preclinical findings on tolerability and efficacy suggest that maytansinoid conjugates with DAR ranging from 2 to 6 have a better therapeutic index than conjugates with very high DAR (∼9-10). These very high DAR ADCs suffer from decreased efficacy, likely due to faster clearance. These results support the use of DAR 3-4 for maytansinoid ADCs but suggest that the exploration of lower or higher DAR may be warranted depending on the biology of the target antigen.


Bioconjugate Chemistry | 2016

Understanding How the Stability of the Thiol-Maleimide Linkage Impacts the Pharmacokinetics of Lysine-Linked Antibody–Maytansinoid Conjugates

Jose F. Ponte; Xiuxia Sun; Nicholas C. Yoder; Nathan Fishkin; Rassol Laleau; Jennifer Coccia; Leanne Lanieri; Megan Bogalhas; Lintao Wang; Sharon D. Wilhelm; Wayne C. Widdison; Jan Pinkas; Thomas A. Keating; Ravi S. Chari; Hans K. Erickson; John M. Lambert

Antibody-drug conjugates (ADCs) have become a widely investigated modality for cancer therapy, in part due to the clinical findings with ado-trastuzumab emtansine (Kadcyla). Ado-trastuzumab emtansine utilizes the Ab-SMCC-DM1 format, in which the thiol-functionalized maytansinoid cytotoxic agent, DM1, is linked to the antibody (Ab) via the maleimide moiety of the heterobifunctional SMCC linker. The pharmacokinetic (PK) data for ado-trastuzumab emtansine point to a faster clearance for the ADC than for total antibody. Cytotoxic agent release in plasma has been reported with nonmaytansinoid, cysteine-linked ADCs via thiol-maleimide exchange, for example, brentuximab vedotin. For Ab-SMCC-DM1 ADCs, however, the main catabolite reported is lysine-SMCC-DM1, the expected product of intracellular antibody proteolysis. To understand these observations better, we conducted a series of studies to examine the stability of the thiol-maleimide linkage, utilizing the EGFR-targeting conjugate, J2898A-SMCC-DM1, and comparing it with a control ADC made with a noncleavable linker that lacked a thiol-maleimide adduct (J2898A-(CH2)3-DM). We employed radiolabeled ADCs to directly measure both the antibody and the ADC components in plasma. The PK properties of the conjugated antibody moiety of the two conjugates, J2898A-SMCC-DM1 and J2898A-(CH2)3-DM (each with an average of 3.0 to 3.4 maytansinoid molecules per antibody), appear to be similar to that of the unconjugated antibody. Clearance values of the intact conjugates were slightly faster than those of the Ab components. Furthermore, J2898A-SMCC-DM1 clears slightly faster than J2898A-(CH2)3-DM, suggesting that there is a fraction of maytansinoid loss from the SMCC-DM1 ADC, possibly through a thiol-maleimide dependent mechanism. Experiments on ex vivo stability confirm that some loss of maytansinoid from Ab-SMCC-DM1 conjugates can occur via thiol elimination, but at a slower rate than the corresponding rate of loss reported for thiol-maleimide links formed at thiols derived by reduction of endogenous cysteine residues in antibodies, consistent with expected differences in thiol-maleimide stability related to thiol pKa. These findings inform the design strategy for future ADCs.


mAbs | 2016

Microscale screening of antibody libraries as maytansinoid antibody-drug conjugates

Kalli C. Catcott; Molly A. McShea; Carl Uli Bialucha; Kathy Miller; Stuart W. Hicks; Parmita Saxena; Thomas G. Gesner; Mikias Woldegiorgis; Megan E. Lewis; Chen Bai; Michael S. Fleming; Seth Ettenberg; Hans K. Erickson; Nicholas C. Yoder

ABSTRACT Antibody-drug conjugates (ADCs) are of great interest as targeted cancer therapeutics. Preparation of ADCs for early stage screening is constrained by purification and biochemical analysis techniques that necessitate burdensome quantities of antibody. Here we describe a method, developed for the maytansinoid class of ADCs, enabling parallel conjugation of antibodies in 96-well format. The method utilizes ∼100 µg of antibody per well and requires <5 µg of ADC for characterization. We demonstrate the capabilities of this system using model antibodies. We also provide multiple examples applying this method to early-stage screening of maytansinoid ADCs. The method can greatly increase the throughput with which candidate ADCs can be screened in cell-based assays, and may be more generally applicable to high-throughput preparation and screening of different types of protein conjugates.


Cancer Discovery | 2017

Discovery and Optimization of HKT288, a Cadherin-6 Targeting ADC for the Treatment of Ovarian and Renal Cancer.

Carl Uli Bialucha; Scott D. Collins; Xiao Li; Parmita Saxena; Xiamei Zhang; Clemens Dürr; Bruno Lafont; Pierric Prieur; Yeonju Shim; Rebecca Mosher; David M. Lee; Lance Ostrom; Tiancen Hu; Sanela Bilic; Ivana Liric Rajlic; Vladimir Capka; Wei Jiang; Joel P. Wagner; GiNell Elliott; Artur Veloso; Jessica C. Piel; Meghan M. Flaherty; Keith Mansfield; Emily K. Meseck; Tina Rubic-Schneider; Anne Serdakowski London; William R. Tschantz; Markus Kurz; Duc Nguyen; Aaron Bourret

Despite an improving therapeutic landscape, significant challenges remain in treating the majority of patients with advanced ovarian or renal cancer. We identified the cell-cell adhesion molecule cadherin-6 (CDH6) as a lineage gene having significant differential expression in ovarian and kidney cancers. HKT288 is an optimized CDH6-targeting DM4-based antibody-drug conjugate (ADC) developed for the treatment of these diseases. Our study provides mechanistic evidence supporting the importance of linker choice for optimal antitumor activity and highlights CDH6 as an antigen for biotherapeutic development. To more robustly predict patient benefit of targeting CDH6, we incorporate a population-based patient-derived xenograft (PDX) clinical trial (PCT) to capture the heterogeneity of response across an unselected cohort of 30 models-a novel preclinical approach in ADC development. HKT288 induces durable tumor regressions of ovarian and renal cancer models in vivo, including 40% of models on the PCT, and features a preclinical safety profile supportive of progression toward clinical evaluation.Significance: We identify CDH6 as a target for biotherapeutics development and demonstrate how an integrated pharmacology strategy that incorporates mechanistic pharmacodynamics and toxicology studies provides a rich dataset for optimizing the therapeutic format. We highlight how a population-based PDX clinical trial and retrospective biomarker analysis can provide correlates of activity and response to guide initial patient selection for first-in-human trials of HKT288. Cancer Discov; 7(9); 1030-45. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 920.


Blood Advances | 2018

A CD123-targeting antibody-drug conjugate, IMGN632, designed to eradicate AML while sparing normal bone marrow cells

Yelena Kovtun; Gregory Jones; Sharlene Adams; Lauren Harvey; Charlene Audette; Alan Wilhelm; Chen Bai; Lingyun Rui; Rassol Laleau; Fenghua Liu; Olga Ab; Yulius Y. Setiady; Nicholas C. Yoder; Victor S. Goldmacher; Ravi V. J. Chari; Jan Pinkas; Thomas Chittenden

The outlook for patients with refractory/relapsed acute myeloid leukemia (AML) remains poor, with conventional chemotherapeutic treatments often associated with unacceptable toxicities, including severe infections due to profound myelosuppression. Thus there exists an urgent need for more effective agents to treat AML that confer high therapeutic indices and favorable tolerability profiles. Because of its high expression on leukemic blast and stem cells compared with normal hematopoietic stem cells and progenitors, CD123 has emerged as a rational candidate for molecularly targeted therapeutic approaches in this disease. Here we describe the development and preclinical characterization of a CD123-targeting antibody-drug conjugate (ADC), IMGN632, that comprises a novel humanized anti-CD123 antibody G4723A linked to a recently reported DNA mono-alkylating payload of the indolinobenzodiazepine pseudodimer (IGN) class of cytotoxic compounds. The activity of IMGN632 was compared with X-ADC, the ADC utilizing the G4723A antibody linked to a DNA crosslinking IGN payload. With low picomolar potency, both ADCs reduced viability in AML cell lines and patient-derived samples in culture, irrespective of their multidrug resistance or disease status. However, X-ADC exposure was >40-fold more cytotoxic to the normal myeloid progenitors than IMGN632. Of particular note, IMGN632 demonstrated potent activity in all AML samples at concentrations well below levels that impacted normal bone marrow progenitors, suggesting the potential for efficacy in AML patients in the absence of or with limited myelosuppression. Furthermore, IMGN632 demonstrated robust antitumor efficacy in multiple AML xenograft models. Overall, these findings identify IMGN632 as a promising candidate for evaluation as a novel therapy in AML.


Cancer Research | 2013

Abstract 5677: The anti-EGFR ADC, IMGN289 displays favorable pharmacokinetic properties.

Jose F. Ponte; Xiuxia Sun; Nicholas C. Yoder; Nathan Fishkin; Sharon Wilhelm; Susan J. Hawes; Wayne C. Widdison; Robert Mastico; Jan Pinkas; Ravi J. Chari; Robert J. Lutz; Hans K. Erickson

IMGN289 is an antibody-drug conjugate (ADC) in preclinical development for the treatment of EGFR-positive cancers. IMGN289 utilizes the same SMCC thioether linker and DM1 cytotoxic agent used in trastuzumab emtansine (T-DM1), and both conjugates contain approximately 3.5 DM1 molecules per antibody. The stability of the SMCC linker is exemplified by several reports that have shown maytansinoid release only after hydrolysis of the antibody backbone within cells and tissue to yield lysine-SMCC-DM1. Despite these findings, pharmacokinetic (PK) data for T-DM1 point to a faster clearance for conjugated antibody than for total antibody. Additionally, reports of payload release from non-maytansinoid, cysteine-linked ADCs via thiol-maleimide exchange in plasma have led to speculation of DM1 release from T-DM1, and of this occurring via a similar mechanism. Consequently, we conducted a series of studies in mice to examine the PK behavior of IMGN289, its unmodified antibody component (J2898A) and a version of IMGN289 (J2898A-(CH2)3-DM1) in which the SMCC linker is replaced with an all-carbon linker incapable of cleavage via thiol-maleimide exchange. PK was assessed in CD-1 mice administered a single bolus iv dose of the test article. Total antibody and ADC concentrations (species with at least one linked DM1) were measured at periodic intervals over 35 days using standard sandwich ELISA assays. We also employed radioactive assays to directly measure the antibody and ADC components in plasma. For total antibody detection, the antibody or ADC was labeled with the radioactive tracer, N-succinimidyl-2,3-[3H]propionate. For ADC detection, tritium was incorporated into the C20-methoxy group of its maytansinoid moiety. The PK profiles for the ADCs and their unmodified antibody were found to be similar using the ELISA and radioactive assays for total antibody, having only slight changes in the PK parameters for the ADC. Using the ELISA assays, we found the total antibody and ADC PK profiles were indistinguishable for IMGN289, in contrast to the reported PK for T-DM1 in mice. The profiles developed using the radioactive assays were in good agreement with these findings, supporting that the ELISA assays were reliable. To confirm our results were not unique to IMGN289, we repeated the experiments with two additional antibody-SMCC-DM1 conjugates and found similar results. Additionally, the PK profile for the all- carbon linked conjugate, J2898A-C3-[3H]DM1, was similar to that of J2898A-SMCC-[3H]DM1 conjugate, suggesting little if any thiol-maleimide exchange in the latter, consistent with the finding of similar total antibody and ADC PK profiles for Ab-SMCC-DM1 conjugates. In summary, IMGN289 appears to retain the PK properties of its antibody component and avoids the payload release via thiol-maleimide exchange described for cysteine-maleimide linked ADCs. Citation Format: Jose F. Ponte, Xiuxia Sun, Nicholas C. Yoder, Nathan Fishkin, Sharon Wilhelm, Susan J. Hawes, Wayne Widdison, Robert A. Mastico, Jan Pinkas, Ravi J. Chari, Robert J. Lutz, Hans K. Erickson. The anti-EGFR ADC, IMGN289 displays favorable pharmacokinetic properties. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 5677. doi:10.1158/1538-7445.AM2013-5677


Molecular Cancer Therapeutics | 2018

A DNA-Interacting Payload Designed to Eliminate Cross-Linking Improves the Therapeutic Index of Antibody–Drug Conjugates (ADCs)

Michael L. Miller; Manami Shizuka; Alan Wilhelm; Paulin Salomon; Emily E. Reid; Leanne Lanieri; Surina Sikka; Erin Maloney; Lauren Harvey; Qifeng Qiu; Katie Archer; Chen Bai; Dilrukshi Vitharana; Luke Harris; Rajeeva Singh; Jose F. Ponte; Nicholas C. Yoder; Yelena Kovtun; Katharine C. Lai; Olga Ab; Jan Pinkas; Thomas A. Keating; Ravi V. J. Chari

Tumor-selective delivery of cytotoxic agents in the form of antibody–drug conjugates (ADCs) is now a clinically validated approach for cancer treatment. In an attempt to improve the clinical success rate of ADCs, emphasis has been recently placed on the use of DNA–cross-linking pyrrolobenzodiazepine compounds as the payload. Despite promising early clinical results with this class of ADCs, doses achievable have been low due to systemic toxicity. Here, we describe the development of a new class of potent DNA-interacting agents wherein changing the mechanism of action from a cross-linker to a DNA alkylator improves the tolerability of the ADC. ADCs containing the DNA alkylator displayed similar in vitro potency, but improved bystander killing and in vivo efficacy, compared with those of the cross-linker. Thus, the improved in vivo tolerability and antitumor activity achieved in rodent models with ADCs of the novel DNA alkylator could provide an efficacious, yet safer option for cancer treatment. Mol Cancer Ther; 17(3); 650–60. ©2018 AACR.


Cancer Research | 2017

Abstract 38: Target validation, antibody discovery and preclinical data supporting ADAM9 as an antibody-drug conjugate therapeutic target for solid tumors

Juniper A. Scribner; Bhaswati Barat; Stuart W. Hicks; Nicholas C. Yoder; Thomas Son; Lusiana Widjaja; Gundo Diedrich; Sergey Gorlatov; Jeff Hooley; Ann Easton; Peter Lung; Anushka De Costa; Francine Chen; Michael Chiechi; Pam Li; Monica Licea; Timothy Hotaling; Michael Spliedt; Valentina Ciccarone; Nadia Gantt; James Tamura; Megan E. Fuller; Molly A. McShea; Scott Koenig; Syd Johnson; Paul A. Moore; Ezio Bonvini; Deryk Loo

Introduction: A target-unbiased approach based on intact cell immunizations with fetal progenitor cells and cancer stem cells, followed by an immunohistochemistry (IHC) screen for cancer-specific candidates, led to the identification of anti-ADAM9 (a disintegrin and metalloproteinase) mAbs with highly differential tumor-versus-normal tissue binding. ADAM9 is a cell surface protein over-expressed in multiple tumors, with a possible role in promotion and progression of cancer through multiple mechanisms, including modulation of adhesion and migration as well as processing of tumorigenic and pro-angiogenic factors. In this preclinical study, we performed target/mAb validation and evaluated the therapeutic potential of anti-ADAM9 antibody-drug conjugates (ADCs) toward ADAM9-expressing solid cancers. Methods: IHC was performed with anti-ADAM9 mAbs to confirm and extend available data of human normal and tumor tissue expression. Epitope mapping studies were conducted to define epitope-specificity. mAbs were also screened to identify those that efficiently internalized into tumor cells. In vitro cellular processing studies were performed to further evaluate the mAbs as ADC candidates. Selected mAbs were converted to ADCs via chemical conjugation to potent anti-microtubule (DM4) or DNA alkylating (DGN549) agents; in vitro cytotoxicity studies were conducted with tumor cell lines representing human cancer types that overexpress ADAM9. A lead mAb was then selected for humanization and affinity maturation to yield a development candidate. Results: Anti-ADAM9 mAbs exhibited strong reactivity toward the tumor epithelium of solid cancers, including pancreatic, kidney, prostate, bladder, breast, colon, lung, and ovarian cancer, but limited reactivity toward normal tissues. Anti-ADAM9 mAbs were efficiently internalized and processed by tumor cell lines, including lines with only modest ADAM9 expression. Anti-ADAM9 ADCs exhibited specific, dose-dependent cytotoxicity toward ADAM9-positive cancer cell lines in vitro, with IC 50 values in the sub-nanomolar range. Humanization and affinity maturation of the lead mAb yielded a development candidate that retains potent antitumor activity toward ADAM9-positive tumor cell lines and equivalent, high affinity binding to both human and cynomolgus monkey ADAM9. Conclusion: ADAM9 is a cell surface antigen that is over-expressed on a wide range of solid cancers. Anti-ADAM9 mAbs that were strongly reactive with representative tumors exhibited high affinity for the antigen and were efficiently internalized and processed by ADAM9-bearing tumor cells. Anti-ADAM9 ADCs demonstrated dose-dependent cytotoxicity in vitro toward a panel of ADAM9-positive tumor cell lines. Our findings demonstrate that an ADC targeting ADAM9 may serve as a potential therapeutic for ADAM9-expressing solid tumors. Citation Format: Juniper A. Scribner, Bhaswati Barat, Stuart W. Hicks, Nicholas C. Yoder, Thomas Son, Lusiana Widjaja, Gundo Diedrich, Sergey Gorlatov, Jeff Hooley, Ann Easton, Peter Lung, Anushka De Costa, Francine Chen, Michael Chiechi, Pam Li, Monica Licea, Timothy E. Hotaling, Michael Spliedt, Valentina Ciccarone, Nadia Gantt, James Tamura, Megan E. Fuller, Molly McShea, Scott Koenig, Syd Johnson, Paul A. Moore, Ezio Bonvini, Deryk Loo. Target validation, antibody discovery and preclinical data supporting ADAM9 as an antibody-drug conjugate therapeutic target for solid tumors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 38. doi:10.1158/1538-7445.AM2017-38


Cancer Research | 2017

Abstract 75: Comparison of site-specific and lysine-linked indolino-benzodiazepine antibody-drug conjugates (ADCs)

Chen Bai; Nicholas C. Yoder; Alan Wilhelm; Sharlene Adams; Kathleen R. Whiteman; Jenny Lee; Katie O'Callaghan; Erin Maloney; Manami Shizuka; Yelena Kovtun; Thomas A. Keating

ADCs are a promising modality for cancer therapy enabled by chemical conjugation of potent cytotoxic compounds to monoclonal antibodies. While many ADCs in clinical evaluation employ heterogeneous conjugation chemistries where the payload is linked through lysine or endogenous cysteine residues, there has recently been considerable interest in site-specific conjugation. ADCs prepared using site-specific methods are believed to have a wider therapeutic index compared to heterogeneous ADCs. We have previously shown that site-specific ADCs incorporating the maytansinoid-based tubulin inhibitor DM1 were less efficacious in vivo when compared to analogous lysine-linked conjugates (Yoder et al AACR 2015 Abstract #545). More recently, we presented results from the evaluation of 2.5-3.0 drug-to-antibody ratio (DAR) heterogeneous lysine-linked and 2 DAR site-specific ADCs using antibodies with engineered cysteines at position 442 in the heavy chains (known as CYSMABTM). These ADCs used the peptide-linked indolino-benzodiazepine DNA alkylator DGN549 (also known as IGN-P1) as the effector. Unlike the DM1 case, site-specific DGN549 ADCs were at least as active in vivo when compared to lysine-linked ADCs (Yoder et al AACR 2016 Abstract #2960). We have made further pharmacological comparisons between CYSMAB and lysine-linked DGN549 ADCs at matched payload doses using two different antibodies targeting distinct cell surface receptors. In the case of mAb1, CYSMAB and lysine-linked ADCs were comparably active. For mAb2, the CYSMAB ADC was more active than the lysine-linked ADC in some models and similarly active in others. The mAb1 CYSMAB ADC exhibited a significantly higher maximum tolerated dose (MTD) compared to the lysine-linked ADC. In contrast, the MTDs of the mAb2 ADCs were similar. However, the mAb2 CYSMAB conjugate was better tolerated in terms of median lethal dose. The mechanism for the improved tolerability of the mAb1 CYSMAB conjugate is not apparent. In an effort to understand whether it is a consequence of conjugation chemistry or DAR, we compared the tolerability of the mAb1 CYSMAB conjugate to that of a ~4 DAR site-specific analog and found that both factors contribute. To determine if our observations can be rationalized in terms of in vivo disposition, we compared the pharmacokinetics of mAb1 CYSMAB and lysine-linked ADCs at matched antibody doses. Intriguingly, the CYSMAB ADC showed slightly greater exposure. These results, along with our previous work on site-specific DM1 ADCs, indicate that in some cases site-specific conjugation can lead to improved efficacy and tolerability. However, generalizations cannot be made across different combinations of antibody, linker, and payload. We conclude that the advantages and disadvantages of site-specific conjugation should be carefully considered for every candidate ADC. Citation Format: Chen Bai, Nicholas C. Yoder, Alan Wilhelm, Sharlene Adams, Kathleen Whiteman, Jenny Lee, Katie O9Callaghan, Erin Maloney, Manami Shizuka, Yelena Kovtun, Thomas A. Keating. Comparison of site-specific and lysine-linked indolino-benzodiazepine antibody-drug conjugates (ADCs) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 75. doi:10.1158/1538-7445.AM2017-75


Cancer Research | 2017

Abstract 37: Novel antibody-drug conjugates targeting ADAM9-expressing solid tumors demonstrate potent preclinical activity

Stuart W. Hicks; Nicholas C. Yoder; Deryk Loo; Asli Muvaffak; Yinghui Zhou; Megan E. Fuller; Molly A. McShea; Marian Themeles; Katherine H. Mucciarone; Juniper A. Scribner; Bhaswati Barat; Thomas Sun; James Tamura; Francine Chen; Kerry Donahue; Tom Chittenden

ADAM9, also known as MDC9 or meltrin-γ, is a member of the ADAM (a disintegrin and metalloproteinase) family of proteases, which have been implicated in cytokine and growth factor shedding, and cell migration. Dysregulation of ADAM9 has been implicated in tumor progression and metastasis, as well as pathological neovascularization. ADAM9 overexpression has been shown to correlate with poor prognosis in prostate, renal, and pancreatic cancers. Using an immunization approach in which antibodies were raised to fetal progenitor and stem-like cancer cell lines followed by screening on tumor and normal tissues, we identified ADAM9 as a promising cell surface tumor target. FFPE-IHC expression analysis revealed that ADAM9 is overexpressed in multiple solid tumor indications relative to corresponding normal tissues. The overexpression of ADAM9 in tumors coupled with its restricted expression in normal tissues make ADAM9 an attractive target for antibody-drug conjugate (ADC) therapy. Here, we describe two ADCs both of which are based on a high affinity anti-ADAM9 antibody to selectively target ADAM9-expressing tumors. The first ADC utilizes the maytansine-derived microtubule disruptor, DM4, linked via a hindered disulfide hydrophilic linker (sulfo-SPDB). The second ADC exploits an ultra-potent DNA alkylating payload, DGN549, which is conjugated to two engineered cysteines via a peptide linker. Both conjugates bound with similar subnanomolar affinity to ADAM9-expressing cells. In vitro cytotoxicity studies showed that anti-ADAM9 ADCs can kill a broad panel of ADAM9-positve tumor cell lines, including lung, pancreatic, renal, prostate, and colon tumor cell lines. In particular, the anti-ADAM9-DGN549 conjugate was extremely potent with IC50 values ranging from 0.1 to 65 pM and was at least 2 logs more active than a non-targeting conjugate. Surprisingly, efficient in vitro cytotoxicity was observed at ADAM9 expression levels as low as a few thousand cell surface receptors per cell. Consistent with their in vitro activity, both anti-ADAM9 ADCs displayed compelling anti-tumor activity in xenograft models. In a CaLu3 non-small cell lung cancer xenograft model, anti-ADAM9-DM4 induced tumor growth delay at a single 1.25 mg Ab/kg dose. In the same model, a single intravenous dose of 0.25 mg Ab/kg of the anti-ADAM9-DGN549 produced durable complete remissions in 8/8 mice. A non-targeting DGN549 ADC was inactive even when dosed at 10 times that of the anti-ADAM9 ADC, demonstrating that targeted delivery of DGN549 through ADAM9 binding is required for activity. These data demonstrate that anti-ADAM9 ADCs exhibit antitumor activity against a broad panel of ADAM9-positive malignancies and cause durable remissions in preclinical models at doses expected to be clinically achievable. Anti-ADAM9 ADCs represent a promising therapeutic strategy to target a wide range of ADAM9-expressing tumors. Citation Format: Stuart W. Hicks, Nicholas C. Yoder, Deryk Loo, Asli Muvaffak, Yinghui Zhou, Megan E. Fuller, Molly A. McShea, Marian Themeles, Katherine H. Mucciarone, Juniper A. Scribner, Bhaswati Barat, Thomas Sun, James Tamura, Francine Z. Chen, Kerry A. Donahue, Tom Chittenden. Novel antibody-drug conjugates targeting ADAM9-expressing solid tumors demonstrate potent preclinical activity [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 37. doi:10.1158/1538-7445.AM2017-37

Collaboration


Dive into the Nicholas C. Yoder's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jan Pinkas

University of Massachusetts Amherst

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge