Network


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

Hotspot


Dive into the research topics where Charles G. Cerveny is active.

Publication


Featured researches published by Charles G. Cerveny.


Nature Biotechnology | 2003

Development of potent monoclonal antibody auristatin conjugates for cancer therapy

Svetlana O. Doronina; Brian E. Toki; Michael Torgov; Brian A. Mendelsohn; Charles G. Cerveny; Dana F. Chace; Ron L. DeBlanc; R Patrick Gearing; Tim D. Bovee; Clay B. Siegall; Joseph A. Francisco; Alan F. Wahl; Damon L. Meyer; Peter D. Senter

We describe the in vitro and in vivo properties of monoclonal antibody (mAb)-drug conjugates consisting of the potent synthetic dolastatin 10 analogs auristatin E (AE) and monomethylauristatin E (MMAE), linked to the chimeric mAbs cBR96 (specific to Lewis Y on carcinomas) and cAC10 (specific to CD30 on hematological malignancies). The linkers used for conjugate formation included an acid-labile hydrazone and protease-sensitive dipeptides, leading to uniformly substituted conjugates that efficiently released active drug in the lysosomes of antigen-positive (Ag+) tumor cells. The peptide-linked mAb-valine-citrulline-MMAE and mAb-phenylalanine-lysine-MMAE conjugates were much more stable in buffers and plasma than the conjugates of mAb and the hydrazone of 5-benzoylvaleric acid-AE ester (AEVB). As a result, the mAb-Val-Cit-MMAE conjugates exhibited greater in vitro specificity and lower in vivo toxicity than corresponding hydrazone conjugates. In vivo studies demonstrated that the peptide-linked conjugates induced regressions and cures of established tumor xenografts with therapeutic indices as high as 60-fold. These conjugates illustrate the importance of linker technology, drug potency and conjugation methodology in developing safe and efficacious mAb-drug conjugates for cancer therapy.We describe the in vitro and in vivo properties of monoclonal antibody (mAb)-drug conjugates consisting of the potent synthetic dolastatin 10 analogs auristatin E (AE) and monomethylauristatin E (MMAE), linked to the chimeric mAbs cBR96 (specific to Lewis Y on carcinomas) and cAC10 (specific to CD30 on hematological malignancies). The linkers used for conjugate formation included an acid-labile hydrazone and protease-sensitive dipeptides, leading to uniformly substituted conjugates that efficiently released active drug in the lysosomes of antigen-positive (Ag+) tumor cells. The peptide-linked mAb-valine-citrulline-MMAE and mAb-phenylalanine-lysine-MMAE conjugates were much more stable in buffers and plasma than the conjugates of mAb and the hydrazone of 5-benzoylvaleric acid-AE ester (AEVB). As a result, the mAb-Val-Cit-MMAE conjugates exhibited greater in vitro specificity and lower in vivo toxicity than corresponding hydrazone conjugates. In vivo studies demonstrated that the peptide-linked conjugates induced regressions and cures of established tumor xenografts with therapeutic indices as high as 60-fold. These conjugates illustrate the importance of linker technology, drug potency and conjugation methodology in developing safe and efficacious mAb-drug conjugates for cancer therapy.


Clinical Cancer Research | 2004

Effects of drug loading on the antitumor activity of a monoclonal antibody drug conjugate.

Kevin J. Hamblett; Peter D. Senter; Dana Chace; Michael M. C. Sun; Joel S. Lenox; Charles G. Cerveny; Kim M. Kissler; Starr X. Bernhardt; Anastasia K. Kopcha; Roger F. Zabinski; Damon L. Meyer; Joseph A. Francisco

Purpose: An antibody-drug conjugate consisting of monomethyl auristatin E (MMAE) conjugated to the anti-CD30 monoclonal antibody (mAb) cAC10, with eight drug moieties per mAb, was previously shown to have potent cytotoxic activity against CD30+ malignant cells. To determine the effect of drug loading on antibody-drug conjugate therapeutic potential, we assessed cAC10 antibody-drug conjugates containing different drug-mAb ratios in vitro and in vivo. Experimental Design: Coupling MMAE to the cysteines that comprise the interchain disulfides of cAC10 created an antibody-drug conjugate population, which was purified using hydrophobic interaction chromatography to yield antibody-drug conjugates with two, four, and eight drugs per antibody (E2, E4, and E8, respectively). Antibody-drug conjugate potency was tested in vitro against CD30+ lines followed by in vivo xenograft models. The maximum-tolerated dose and pharmacokinetic profiles of the antibody-drug conjugates were investigated in mice. Results: Although antibody-drug conjugate potency in vitro was directly dependent on drug loading (IC50 values E8<E4<E2), the in vivo antitumor activity of E4 was comparable with E8 at equal mAb doses, although the E4 contained half the amount of MMAE per mAb. E2 was also an active antitumor agent but required higher doses. The maximum-tolerated dose of E2 in mice was at least double that of E4, which in turn was twice that of E8. MMAE loading affected plasma clearance, as E8 cleared 3-fold faster than E4 and 5-fold faster than E2. Conclusions: By decreasing drug loading per antibody, the therapeutic index was increased demonstrating that drug loading is a key design parameter for antibody-drug conjugates.


Journal of Biological Chemistry | 2006

Lysosomal Trafficking and Cysteine Protease Metabolism Confer Target-specific Cytotoxicity by Peptide-linked Anti-CD30-Auristatin Conjugates *

May Kung Sutherland; Russell J. Sanderson; Kristine A. Gordon; Jamie B. Andreyka; Charles G. Cerveny; Changpu Yu; Timothy S. Lewis; Damon L. Meyer; Roger F. Zabinski; Svetlana O. Doronina; Peter D. Senter; Che-Leung Law; Alan F. Wahl

The chimeric anti-CD30 monoclonal antibody cAC10, linked to the antimitotic agents monomethyl auristatin E (MMAE) or F (MMAF), produces potent and highly CD30-selective anti-tumor activity in vitro and in vivo. These drugs are appended via a valine-citrulline (vc) dipeptide linkage designed for high stability in serum and conditional cleavage and putative release of fully active drugs by lysosomal cathepsins. To characterize the biochemical processes leading to effective drug delivery, we examined the intracellular trafficking, internalization, and metabolism of the parent antibody and two antibody-drug conjugates, cAC10vc-MMAE and cAC10vc-MMAF, following CD30 surface antigen interaction with target cells. Both cAC10 and its conjugates bound to target cells and internalized in a similar manner. Subcellular fractionation and immunofluorescence studies demonstrated that the antibody and antibody-drug conjugates entering target cells migrated to the lysosomes. Trafficking of both species was blocked by inhibitors of clathrin-mediated endocytosis, suggesting that drug conjugation does not alter the fate of antibody-antigen complexes. Incubation of cAC10vc-MMAE or cAC10vc-MMAF with purified cathepsin B or with enriched lysosomal fractions prepared by subcellular fractionation resulted in the release of active, free drug. Cysteine protease inhibitors, but not aspartic or serine protease inhibitors, blocked antibody-drug conjugate metabolism and the ensuing cytotoxicity of target cells and yielded enhanced intracellular levels of the intact conjugates. These findings suggest that in addition to trafficking to the lysosomes, cathepsin B and perhaps other lysosomal cysteine proteases are requisite for drug release and provide a mechanistic basis for developing antibody-drug conjugates cleavable by intracellular proteases for the targeted delivery of anti-cancer therapeutics.


Cancer Research | 2005

Preclinical antilymphoma activity of a humanized anti-CD40 monoclonal antibody, SGN-40

Che-Leung Law; Kristine A. Gordon; John P. Collier; Kerry Klussman; Julie A. McEarchern; Charles G. Cerveny; Bruce Mixan; Wyne P. Lee; Zhonghau Lin; Patricia Valdez; Alan F. Wahl; Iqbal S. Grewal

SGN-40 is a humanized IgG1 antihuman CD40 that is currently in a phase I clinical trial for the treatment of multiple myeloma. As surface CD40 expression on B-lineage cells is maintained from pro-B cells to plasma cells, SGN-40 may be applicable to treatment of other B-cell neoplasias, including non-Hodgkins lymphoma. In this study, we examined potential in vitro and in vivo anti-B-lineage lymphoma activity of SGN-40. Recombinant SGN-40 was expressed and purified from Chinese hamster ovary cells and characterized based on binding affinity, specificity, and normal B-cell stimulation. The ability of SGN-40 to target neoplastic B cells was examined in vitro by proliferation inhibition, cytotoxicity, and antibody-dependent cell cytotoxicity assays and in vivo by human lymphoma xenograft models. Recombinant SGN-40 showed high affinity, Kd of approximately 1 nmol/L, and specific binding to CD40. Whereas SGN-40 was a weak agonist in stimulating normal B-cell proliferation in the absence of IL-4 and CD40L, it delivered potent proliferation inhibitory and apoptotic signals to, and mediated antibody-dependent cytotoxicity against, a panel of high-grade B-lymphoma lines. These in vitro antilymphoma effects were extended to disseminated and s.c. xenograft CD40 tumor models. In these xenograft models, the antitumor activity of SGN-40 was comparable with that of rituximab. The preclinical in vitro and in vivo antilymphoma activity of SGN-40 observed in this study provides a rationale for the clinical testing of SGN-40 in the treatment of CD40+ B-lineage lymphomas.


Cancer Research | 2006

Lymphocyte Activation Antigen CD70 Expressed by Renal Cell Carcinoma Is a Potential Therapeutic Target for Anti-CD70 Antibody-Drug Conjugates

Che Leung Law; Kristine A. Gordon; Brian E. Toki; Andrew K. Yamane; Michelle A. Hering; Charles G. Cerveny; Joseph M. Petroziello; Maureen Ryan; Leia M. Smith; Ronald Simon; Guido Sauter; Ezogelin Oflazoglu; Svetlana O. Doronina; Damon L. Meyer; Joseph A. Francisco; Paul Carter; Peter D. Senter; John A. Copland; Christopher G. Wood; Alan F. Wahl

Metastatic renal cell carcinoma (RCC) is an aggressive disease refractory to most existing therapeutic modalities. Identifying new markers for disease progression and drug targets for RCC will benefit this unmet medical need. We report a subset of clear cell and papillary cell RCC aberrantly expressing the lymphocyte activation marker CD70, a member of the tumor necrosis factor superfamily. Importantly, CD70 expression was found to be maintained at the metastatic sites of RCC. Anti-CD70 antibody-drug conjugates (ADC) consisting of auristatin phenylalanine phenylenediamine (AFP) or monomethyl auristatin phenylalanine (MMAF), two novel derivatives of the anti-tubulin agent auristatin, mediated potent antigen-dependent cytotoxicity in CD70-expressing RCC cells. Cytotoxic activity of these anti-CD70 ADCs was associated with their internalization and subcellular trafficking through the endosomal-lysosomal pathway, disruption of cellular microtubule network, and G2-M phase cell cycle arrest. The efficiency of drug delivery using anti-CD70 as vehicle was illustrated by the much enhanced cytotoxicity of antibody-conjugated MMAF compared with free MMAF. Hence, ADCs targeted to CD70 can selectively recognize RCC, internalize, and reach the appropriate subcellular compartment(s) for drug release and tumor cell killing. In vitro cytotoxicity of these ADCs was confirmed in xenograft models using RCC cell lines. Our findings provide evidence that CD70 is an attractive target for antibody-based therapeutics against metastatic RCC and suggest that anti-CD70 ADCs can provide a new treatment approach for advanced RCC patients who currently have no chemotherapeutic options.


Clinical Cancer Research | 2004

Efficient elimination of B-lineage lymphomas by anti-CD20-auristatin conjugates.

Che-Leung Law; Charles G. Cerveny; Kristine A. Gordon; Kerry Klussman; Bruce Mixan; Dana Chace; Damon L. Meyer; Svetlana O. Doronina; Clay B. Siegall; Joseph A. Francisco; Peter D. Senter; Alan F. Wahl

The anti-CD20 antibody rituximab is useful in the treatment of certain B-cell malignancies, most notably non-Hodgkin’s lymphoma. Its efficacy has been increased when used in combination with chemotherapy, yet anti-CD20 monoclonal antibodies (mAbs) directly conjugated with drugs such as doxorubicin (Dox) have failed to deliver drug or to demonstrate antitumor activity. We have produced anti-CD20 antibody-drug conjugates that possess potent antitumor activity by using the anti-mitotic agent, monomethyl auristatin E (MMAE), linked via the lysosomally cleavable dipeptide, valine-citrulline (vc). Two anti-CD20 conjugates, rituximab-vcMMAE and 1F5-vcMMAE, were selectively cytotoxic against CD20+ B-lymphoma cell lines, with IC50 values ranging from 50 ng/mL to 1 μg/mL. Unlike rituximab, which showed diffuse surface localization, rituximab-vcMMAE capped and was internalized within 4 hours after binding to CD20+ B cells. Internalization of rituximab-vcMMAE was followed by rapid G2-M phase arrest and onset of apoptosis. Anti-CD20 antibody-drug conjugates prepared with Dox were internalized and localized as with rituximab-vcMMAE, yet these were not effective for drug delivery (IC50 > 50 μg/mL). Consistent with in vitro activity, rituximab-vcMMAE showed antitumor efficacy in xenograft models of CD20-positive lymphoma at doses where rituximab or rituximab-Dox conjugates were ineffective. These data indicate that anti-CD20–based antibody-drug conjugates are effective antitumor agents when prepared with a stable, enzyme-cleavable peptide linkage to highly potent cytotoxic agents such as MMAE.


Oncogene | 2004

Suppression subtractive hybridization and expression profiling identifies a unique set of genes overexpressed in non-small-cell lung cancer

Joseph M. Petroziello; Andrew K. Yamane; Lori Westendorf; Melissa C. Thompson; Charlotte Mcdonagh; Charles G. Cerveny; Che-Leung Law; Alan F. Wahl; Paul Carter

Expression array data for >3000 individual clones from two suppression subtractive hybridization libraries revealed 147 genes overexpressed in non-small-cell lung cancer (NSCLC) cell lines. Of these 147 genes, 30 genes have previously unknown cancer association and 65 genes have been associated with cancers other than NSCLC. The identification of 52 genes previously associated with NSCLC by different methodologies supports the validity of the strategy used here. Of the 147 genes, 19 have no prior named Unigene cluster designation, and are designated herein as L1 to L19. Quantitative real-time PCR and cancer profiling arrays were used as independent validation tools to confirm tumor overexpression for five of the ‘L’ genes in tumor cell lines and patient samples from NSCLC and other cancers. Follow-up studies for candidate NSCLC-associated genes can be useful in providing valuable insight into the etiology of lung cancer as well as providing potentially interesting diagnostic or therapeutic targets for further investigation.


Blood | 2003

cAC10-vcMMAE, an anti-CD30–monomethyl auristatin E conjugate with potent and selective antitumor activity

Joseph A. Francisco; Charles G. Cerveny; Damon L. Meyer; Bruce Mixan; Kerry Klussman; Dana Chace; Starr X. Rejniak; Kristine A. Gordon; Ronald DeBlanc; Brian E. Toki; Che-Leung Law; Svetlana O. Doronina; Clay B. Siegall; Peter D. Senter; Alan F. Wahl


Bioconjugate Chemistry | 2006

Enhanced activity of monomethylauristatin F through monoclonal antibody delivery: effects of linker technology on efficacy and toxicity.

Svetlana O. Doronina; Brian A. Mendelsohn; Tim D. Bovee; Charles G. Cerveny; Stephen C. Alley; Damon L. Meyer; Ezogelin Oflazoglu; Brian E. Toki; Russell J. Sanderson; Roger F. Zabinski; and Alan F. Wahl; Peter D. Senter


Bioconjugate Chemistry | 2005

Reduction−Alkylation Strategies for the Modification of Specific Monoclonal Antibody Disulfides

Michael M. C. Sun; Kevin S. Beam; Charles G. Cerveny; Kevin J. Hamblett; Richard S. Blackmore; Michael Torgov; Felicia G. M. Handley; Nathan Ihle; Peter D. Senter; Stephen C. Alley

Collaboration


Dive into the Charles G. Cerveny's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge