Network


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

Hotspot


Dive into the research topics where Andreas Maderna is active.

Publication


Featured researches published by Andreas Maderna.


Cancer Research | 2009

RDEA119/BAY 869766: A Potent, Selective, Allosteric Inhibitor of MEK1/2 for the Treatment of Cancer

Cory Iverson; Gary Larson; Chon Lai; Li-Tain Yeh; Claudia Dadson; Paul Weingarten; Todd Appleby; Todd Vo; Andreas Maderna; Jean-Michel Vernier; Robert Hamatake; Jeffrey N. Miner; Barry Quart

The RAS-RAF-mitogen-activated protein kinase/extracellular signal-regulated kinase (ERK) kinase (MEK)-ERK pathway provides numerous opportunities for targeted oncology therapeutics. In particular, the MEK enzyme is attractive due to high selectivity for its target ERK and the central role that activated ERK plays in driving cell proliferation. The structural, pharmacologic, and pharmacokinetic properties of RDEA119/BAY 869766, an allosteric MEK inhibitor, are presented. RDEA119/BAY 869766 is selectively bound directly to an allosteric pocket in the MEK1/2 enzymes. This compound is highly efficacious at inhibiting cell proliferation in several tumor cell lines in vitro. In vivo, RDEA119/BAY 869766 exhibits potent activity in xenograft models of melanoma, colon, and epidermal carcinoma. RDEA119/BAY 869766 exhibits complete suppression of ERK phosphorylation at fully efficacious doses in mice. RDEA119/BAY 869766 shows a tissue selectivity that reduces its potential for central nervous system-related side effects. Using pharmacokinetic and pharmacodynamic data, we show that maintaining adequate MEK inhibition throughout the dosing interval is likely more important than achieving high peak levels because greater efficacy was achieved with more frequent but lower dosing. Based on its longer half-life in humans than in mice, RDEA119/BAY 869766 has the potential for use as a once- or twice-daily oral treatment for cancer. RDEA119/BAY 869766, an exquisitely selective, orally available MEK inhibitor, has been selected for clinical development because of its potency and favorable pharmacokinetic profile.


Chemistry: A European Journal | 2013

Total Syntheses and Biological Reassessment of Lactimidomycin, Isomigrastatin and Congener Glutarimide Antibiotics

Kévin Micoine; Peter Persich; Josep Llaveria; My-Hanh Lam; Andreas Maderna; Frank Loganzo; Alois Fürstner

Lactimidomycin (1) was described in the literature as an exquisitely potent cell migration inhibitor. Encouraged by this claim, we developed a concise and scalable synthesis of this bipartite glutarimide-macrolide antibiotic, which relies on the power of ring-closing alkyne metathesis (RCAM) for the formation of the unusually strained 12-membered head group. Subsequent deliberate digression from the successful path to 1 also brought the sister compound isomigrastatin (2) as well as a series of non-natural analogues of these macrolides into reach. A careful biological re-evaluation of this compound collection showed 1 and progeny to be potently cytotoxic against a panel of cancer cell lines, even after one day of compound exposure; therefore any potentially specific effects on tumor cell migration were indistinguishable from the acute effect of cell death. No significant cell migration inhibition was observed at sub-toxic doses. Although these findings cannot be reconciled with some reports in the literature, they are in accord with the notion that lactimidomycin is primarily a ribosome-binder able to effectively halt protein biosynthesis at the translation stage.


Bioorganic & Medicinal Chemistry Letters | 2011

Imidazo(1,5-a)quinoxalines as irreversible BTK inhibitors for the treatment of rheumatoid arthritis

Kyung-Hee Kim; Andreas Maderna; Mark E. Schnute; Martin Hegen; Shashi Mohan; Joy S. Miyashiro; Laura Lin; Evelyn Li; Sean Keegan; Jennifer Lussier; Christopher Wrocklage; Cheryl Nickerson-Nutter; Arthur J. Wittwer; Holly Soutter; Nicole Caspers; Seungil Han; Ravi G. Kurumbail; Kyri Dunussi-Joannopoulos; John Douhan; Allan Wissner

Imidazo[1,5-a]quinoxalines were synthesized that function as irreversible Brutons tyrosine kinase (BTK) inhibitors. The syntheses and SAR of this series of compounds are presented as well as the X-ray crystal structure of the lead compound 36 in complex with a gate-keeper variant of ITK enzyme. The lead compound showed good in vivo efficacy in preclinical RA models.


Molecular Cancer Therapeutics | 2015

Tumor Cells Chronically Treated with a Trastuzumab–Maytansinoid Antibody–Drug Conjugate Develop Varied Resistance Mechanisms but Respond to Alternate Treatments

Frank Loganzo; Xingzhi Tan; Matthew Sung; Guixian Jin; Jeremy Myers; Eugene Melamud; Fang Wang; Veronica Diesl; Sylvia Musto; My-Hanh Lam; William Hu; Manoj Charati; Kiran Khandke; Kenny Sung Kyoo Kim; Mike Cinque; Judy Lucas; Edmund I. Graziani; Andreas Maderna; Christopher J. O'Donnell; Kim Arndt; Hans-Peter Gerber

Antibody–drug conjugates (ADC) are emerging as clinically effective therapy. We hypothesized that cancers treated with ADCs would acquire resistance mechanisms unique to immunoconjugate therapy and that changing ADC components may overcome resistance. Breast cancer cell lines were exposed to multiple cycles of anti-Her2 trastuzumab–maytansinoid ADC (TM-ADC) at IC80 concentrations followed by recovery. The resistant cells, 361-TM and JIMT1-TM, were characterized by cytotoxicity, proteomic, transcriptional, and other profiling. Approximately 250-fold resistance to TM-ADC developed in 361-TM cells, and cross-resistance was observed to other non–cleavable-linked ADCs. Strikingly, these 361-TM cells retained sensitivity to ADCs containing cleavable mcValCitPABC-linked auristatins. In JIMT1-TM cells, 16-fold resistance to TM-ADC developed, with cross-resistance to other trastuzumab-ADCs. Both 361-TM and JIMT1-TM cells showed minimal resistance to unconjugated mertansine (DM1) and other chemotherapeutics. Proteomics and immunoblots detected increased ABCC1 (MRP1) drug efflux protein in 361-TM cells, and decreased Her2 (ErbB2) in JIMT1-TM cells. Proteomics also showed alterations in various pathways upon chronic exposure to the drug in both cell models. Tumors derived from 361-TM cells grew in mice and were refractory to TM-ADC compared with parental cells. Hence, acquired resistance to trastuzumab–maytansinoid ADC was generated in cultured cancer cells by chronic drug treatment, and either increased ABCC1 protein or reduced Her2 antigen were primary mediators of resistance. These ADC-resistant cell models retain sensitivity to other ADCs or standard-of-care chemotherapeutics, suggesting that alternate therapies may overcome acquired ADC resistance. Mol Cancer Ther; 14(4); 952–63. ©2015 AACR.


Journal of the American Chemical Society | 2014

Synthesis, molecular editing, and biological assessment of the potent cytotoxin leiodermatolide.

Damien Mailhol; Jens Willwacher; Nina Kausch-Busies; Elizabeith E. Rubitski; Zhanna Sobol; Maik Schuler; My-Hanh Lam; Sylvia Musto; Frank Loganzo; Andreas Maderna; Alois Fürstner

It was by way of total synthesis that the issues concerning the stereostructure of leiodermatolide (1) have recently been solved; with the target now being unambiguously defined, the mission of synthesis changes as to secure a meaningful supply of this exceedingly scarce natural product derived from a deep-sea sponge. To this end, a scalable route of 19 steps (longest linear sequence) has been developed, which features a catalytic asymmetric propargylation of a highly enolizable β-keto-lactone, a ring closing alkyne metathesis and a modified Stille coupling as the key transformations. Deliberate digression from this robust blueprint brought a first set of analogues into reach, which allowed the lead qualities of 1 to be assessed. The acquired biodata show that 1 is a potent cytotoxin in human tumor cell proliferation assays, distinguished by GI50 values in the ≤3 nM range even for cell lines expressing the Pgp efflux transporter. Studies with human U2OS cells revealed that 1 causes mitotic arrest, micronucleus induction, centrosome amplification and tubulin disruption, even though no evidence for direct tubulin binding has been found in cell-free assays; moreover, the compound does not seem to act through kinase inhibition. Indirect evidence points at centrosome declustering as a possible mechanism of action, which provides a potentially rewarding outlook in that centrosome declustering agents hold promise of being inherently selective for malignant over healthy human tissue.


Molecular Cancer Therapeutics | 2015

Novel Anti-TM4SF1 Antibody–Drug Conjugates with Activity against Tumor Cells and Tumor Vasculature

Alberto Visintin; Kelly M. Knowlton; Edyta Tyminski; Chi-Iou Lin; Xiang Zheng; Kimberly Marquette; Sadhana Jain; Lioudmila Tchistiakova; Dan Li; Christopher J. O'Donnell; Andreas Maderna; Xianjun Cao; Robert Dunn; William B. Snyder; Anson K. Abraham; Mauricio Leal; Shoba Shetty; Anthony Barry; Leigh Zawel; Anthony J. Coyle; Harold F. Dvorak; Shou-Ching Jaminet

Antibody–drug conjugates (ADC) represent a promising therapeutic modality for managing cancer. Here, we report a novel humanized ADC that targets the tetraspanin-like protein TM4SF1. TM4SF1 is highly expressed on the plasma membranes of many human cancer cells and also on the endothelial cells lining tumor blood vessels. TM4SF1 is internalized upon interaction with antibodies. We hypothesized that an ADC against TM4SF1 would inhibit cancer growth directly by killing cancer cells and indirectly by attacking the tumor vasculature. We generated a humanized anti-human TM4SF1 monoclonal antibody, v1.10, and armed it with an auristatin cytotoxic agent LP2 (chemical name mc-3377). v1.10-LP2 selectively killed cultured human tumor cell lines and human endothelial cells that express TM4SF1. Acting as a single agent, v1.10-LP2 induced complete regression of several TM4SF1-expressing tumor xenografts in nude mice, including non–small cell lung cancer and pancreas, prostate, and colon cancers. As v1.10 did not react with mouse TM4SF1, it could not target the mouse tumor vasculature. Therefore, we generated a surrogate anti-mouse TM4SF1 antibody, 2A7A, and conjugated it to LP2. At 3 mpk, 2A7A-LP2 regressed several tumor xenografts without noticeable toxicity. Combination therapy with v1.10-LP2 and 2A7A-LP2 together was more effective than either ADC alone. These data provide proof-of-concept that TM4SF1-targeting ADCs have potential as anticancer agents with dual action against tumor cells and the tumor vasculature. Such agents could offer exceptional therapeutic value and warrant further investigation. Mol Cancer Ther; 14(8); 1868–76. ©2015 AACR.


Bioorganic & Medicinal Chemistry Letters | 2013

New cytotoxic benzosuberene analogs. Synthesis, molecular modeling and biological evaluation

Zecheng Chen; Andreas Maderna; Sai Chetan K. Sukuru; Melissa Wagenaar; Christopher J. O’Donnell; My-Hanh Lam; Sylvia Musto; Frank Loganzo

In this Letter we describe the synthesis and biological evaluation of new benzosuberene analogs with structural modifications on the B-ring. The focus was initially to probe the chemical space around the B-ring C-8 position. This position was readily available for derivatization chemistry using our recently developed new synthesis for this compound class. Furthermore, we describe two new B-ring analogs, one containing a diene and the other a cyclic ether group. Both new analogs show excellent potencies in tumor cell proliferation assays. In addition, we describe molecular modeling studies that provide a binding rationale for reference compound 8 in the colchicine binding site using the known colchicine crystal structure. We also examine whether the cell based potency data obtained with selected new analogs are supported by modeling results.


Cancer Research | 2014

Abstract 4837: Extracellular proteolytic cleavage of peptide-linked antibody-drug conjugates promotes bystander killing of cancer cells

My-Hanh Lam; Judy Lucas; Andreas Maderna; Hallie Wald; Megan Wojciechowicz; Russell Dushin; Bryan Peano; Fang Wang; Jeremy Myers; Xingzhi Tan; Sylvia Musto; Manoj Charati; Hans-Peter Gerber; Frank Loganzo

Antibody drug conjugates (ADCs) are designed to deliver cytotoxics to tumor cells via binding to surface antigen followed by internalization and intracellular drug release. ADC linkers are typically categorized as non-cleavable or cleavable; a cleavable linker example is Y_mcValCitPABC_X, with antibody Y, a dipeptide sequence with self-immolative PABC spacer, and payload X. This linker is known to be cleaved by endosomal/lysosomal proteases such as cathepsins, releasing attached payload. In addition to intracellular processing of this linker, we report that conditioned media of cultured tumor cell lines is sufficient to promote extracellular cleavage of ADCs with peptide-linked payloads. Cultured cell lines N87 (gastric) and U87 (glioblastoma), and patient-derived xenograft PA0165 (pancreatic) adapted to in vitro culture, were plated either in standard 2D culture or in 3D Cultrex embedded culture. After 3 - 7 days, conditioned media from cells was transferred onto MDA-MB-468 or HT29 cells, and then ADCs (Y_mcValCitPABC_Aur) were added to cultures. ADCs were non-targeting IgG conjugated via cleavable dipeptide-PABC linker to auristatin tubulin inhibitor. Minimal cytotoxicity was observed with ADC alone on 468 or HT29 cells. However, in the presence of conditioned media from N87, U87, or PA0165 cells plus the ADC, cytotoxicity was observed in the recipient cells (up to 31, 22, 56% growth inhibition respectively at 100 nM ADC). Moreover, in all cases, the magnitude of the response was greatest when cells providing conditioned media were grown in 3D culture (up to 56, 48, 70%, respectively). In contrast, minimal response was observed using conditioned media from other cancer cell lines (ie HCC2429, 1 - 17%). Additional analyses were conducted by incubating conditioned media from these cells with a dipeptide-based cleavable substrate with fluorescent probe and measuring released product in a plate-based assay. Conditioned media promoted fluorescence, suggesting proteolytic enzymes secreted by cells. An ELISA confirmed the presence of cathepsins in conditioned media. Complementing these studies, proteolytic activity was detected in the interstitial fluid derived from tumors grown in athymic mice. Fluid extracted from xenograft tumors (cultured cancer lines and patient-derived tumors) was analyzed for proteolytic activity using cleavable-fluorescent linker-probe in a plate assay. The majority of samples demonstrated proteolytic activity. These data are consistent with reported secretion of cathepsins by cancer cells and we now show that these proteases may mediate extracellular release of cytotoxic payloads from ADCs containing peptide-based cleavable linkers. This activity is magnified when cells are grown in 3D culture and is observed in tumor xenografts grown in vivo. This response may provide a beneficial bystander effect of ADCs on antigen negative cells in a heterogenous tumor population. Citation Format: My-Hanh Lam, Judy Lucas, Andreas Maderna, Hallie Wald, Megan Wojciechowicz, Russell Dushin, Bryan Peano, Fang Wang, Jeremy Myers, Xingzhi Tan, Sylvia Musto, Manoj Charati, Hans-Peter Gerber, Frank Loganzo. Extracellular proteolytic cleavage of peptide-linked antibody-drug conjugates promotes bystander killing of cancer cells. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4837. doi:10.1158/1538-7445.AM2014-4837


Cancer Research | 2017

Abstract 3095: The development of CPI as a novel, next-generation DNA-targeting payload for ADCs

Jennifer Kahler; Maureen Dougher; Jane Xu; Matthew David Doroski; Andreas Maderna; Russell Dushin; Stephane Thibault; Mauricio Leal; Madan Katragadda; Christopher J. O'Donnell; Matthew Sung; Puja Sapra

DNA targeting drugs represent one of cornerstones of anti-cancer therapy for both hematologic and solid tumor indications. Low potency anti-DNA compounds (e.g. platins, anthracyclines) are included in many standard-of-care (SOC) regimens, however their modest activity and overall toxicity profiles limit their therapeutic potential. To increase the therapeutic window for DNA-damaging agents, high potency anti-DNA compounds with enhanced anti-tumor activity have been delivered to tumors as payloads of targeting modalities such as antibody-drug conjugates (ADCs). Herein, we describe the development of a novel DNA-damaging compound comprised of a dimeric structure of cyclopropylpyrrolo[e]indolones (CPIs) that was designed to alkylate DNA and generate toxic interstrand crosslinks (ICLs). In response to the CPI-induced formation of ICLs, CPI treatment of cells primarily activates the Fancomia anemia DNA damage response pathway, whereas other successful DNA-damaging ADC payloads such as calicheamicin activate double-strand break response pathways. CPI shows ~860-fold greater potency than calicheamicin in a panel of cell lines derived from a broad spectrum of tumor indications. Importantly, this new CPI payload retains potent activity in calicheamicin- and SOC-resistant tumor models (including overcoming overexpression of drug efflux pumps). When evaluated as payloads on anti-CD33 targeting ADCs, the CPI conjugate showed dramatically improved efficacy over the corresponding calicheamicin conjugates in MDR+ tumor models. As a site-specific conjugate, the CPI ADC shows enhanced in vivo stability and possesses a wider therapeutic window than the corresponding conventional calicheamicin conjugate and other leading DNA-damaging conjugates on the CD33 platform. Citation Format: Jennifer Kahler, Maureen Dougher, Jane Xu, Matthew Doroski, Andreas Maderna, Russell Dushin, Stephane Thibault, Mauricio Leal, Madan Katragadda, Christopher J. O9Donnell, Matthew Sung, Puja Sapra. The development of CPI as a novel, next-generation DNA-targeting payload for 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 3095. doi:10.1158/1538-7445.AM2017-3095


Cancer Research | 2015

Abstract 1697: Therapeutic targeting the NOTCH3 receptor with antibody drug conjugates

Kenneth G. Geles; Yijie Gao; Latha Sridharan; Andreas Giannakou; Ting-Ting Yamin; Jonathan Golas; Judy Lucas; Manoj Charati; Xiantang Li; Magali Guffroy; Tim Nichols; Kai Wang; Max Follettie; Andreas Maderna; Lioudmila Tchistiakova; Hans-Peter Gerber; Puja Sapra

Activation and mutation of the NOTCH signaling pathway is oncogenic in many tissue types and the target of multiple anti-cancer therapies currently in clinical development. Initial therapeutic strategies designed to target the NOTCH pathway have focused on inhibition of aberrant signaling, but can have undesirable side-effects or insufficient anti-tumor activity. Antibody drug conjugates (ADCs) are an emerging therapeutic modality that equips antibodies with potent cytotoxic payloads that can be directly delivered to tumor cells. We have developed and compared anti-NOTCH3 ADCs using two different classes of therapeutic anti-NOTCH3 antibodies that target the juxtamembrane Negative Regulatory Region (NRR). The first class is unable to stabilize the NOTCH3-NRR in an auto-inhibitory conformation in the presence of ligand and does not block ADAM protease cleavage of the receptor. The second class contains an antibody that exhibits potent signaling inhibition by stabilizing the NRR in an inactive state. Despite antagonizing NOTCH3 signaling, the inhibitory anti-NOTCH3 antibody was unable to regress preclinical tumor xenografts with active NOTCH3 signaling. To enhance their potency, both classes of anti-NOTCH3 antibodies were conjugated to an auristatin-based microtubule inhibitor through a cleavable linker. Unexpectedly, the inhibitory anti-NOTCH3 antibody demonstrated more rapid trafficking to the lysosome than the non-inhibitory antibody suggesting that the two antibodies have distinct internalization routes with important implications for NOTCH3-ADC pharmacology. Pharmacodynamic biomarker analysis demonstrated anti-NOTCH3 ADCs disrupted the mitotic spindle apparatus, induced cell cycle arrest and triggered apoptosis. Anti-NOTCH3 ADCs exhibited robust anti-tumor activity and induce prolonged tumor regressions in preclinical models of breast, lung and ovarian cancer regardless of their ability to block signaling. Furthermore, anti-NOTCH3 ADC treatment was able to regress OVCAR3 ovarian tumor xenografts that were refractory to a platinum-based agent or relapsed anti-VEGF therapy. Our studies demonstrate that anti-NOTCH3 ADCs had enhanced efficacy compared to other NOTCH signaling inhibitors and also allowed targeting of tumors that over-expressed NOTCH3 but were not necessarily addicted to its signaling. The safety and efficacy of the non-inhibitory anti-NOTCH3 ADC, PF-06650808, is currently being examined in a Ph1 clinical trial (Protocol B7501001). Citation Format: Kenneth G. Geles, Yijie Gao, Latha Sridharan, Andreas Giannakou, Ting-Ting Yamin, Jonathan Golas, Judy Lucas, Manoj Charati, Xiantang Li, Magali Guffroy, Tim Nichols, Kai Wang, Max Follettie, Andreas Maderna, Lioudmila Tchistiakova, Hans-Peter Gerber, Puja Sapra. Therapeutic targeting the NOTCH3 receptor with antibody drug conjugates. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1697. doi:10.1158/1538-7445.AM2015-1697

Collaboration


Dive into the Andreas Maderna's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge