Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Nanomedicine ; 17: 71-81, 2019 04.
Article in English | MEDLINE | ID: mdl-30654182

ABSTRACT

MM-302 is an anti-HER2 antibody-targeted pegylated liposomal doxorubicin designed to deliver doxorubicin specifically to HER2-expressing solid tumors. The delivery and activity of MM-302 were evaluated in orthotopic, transgenic, and intravenous breast cancer models expressing varying levels of HER2 that metastasize to some of the most common sites of dissemination for breast cancer, namely, lung, liver, and brain. Metastatic burden was quantified by gross evaluation, immunohistochemistry (IHC), and bioluminescent imaging. Liposome delivery was quantified by IHC and ex vivo fluorescent imaging. Unlike its non-targeted counterpart, pegylated liposomal doxorubicin (PLD), MM-302 showed activity at controlling both primary and metastatic tumor burden in all models tested. The effect of HER2-targeting was greatest in the lung where lymphatic vessel density and MM-302 delivery were highest. Our data indicate that the therapeutic advantage of actively targeting a nanoliposome with an antibody is influenced by both target expression and the tumor microenvironment.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Breast Neoplasms/drug therapy , Doxorubicin/analogs & derivatives , Immunoconjugates/chemistry , Liposomes/chemistry , Single-Chain Antibodies/chemistry , Animals , Antibiotics, Antineoplastic/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Doxorubicin/therapeutic use , Drug Delivery Systems , Female , Mice , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/therapeutic use , Receptor, ErbB-2/metabolism , Tumor Microenvironment/drug effects
2.
Mol Cancer Ther ; 21(7): 1047-1059, 2022 07 05.
Article in English | MEDLINE | ID: mdl-35511740

ABSTRACT

ADAM metallopeptidase domain 9 (ADAM9) is a member of the ADAM family of multifunctional, multidomain type 1 transmembrane proteins. ADAM9 is overexpressed in many cancers, including non-small cell lung, pancreatic, gastric, breast, ovarian, and colorectal cancer, but exhibits limited expression in normal tissues. A target-unbiased discovery platform based on intact tumor and progenitor cell immunizations, followed by an IHC screen, led to the identification of anti-ADAM9 antibodies with selective tumor-versus-normal tissue binding. Subsequent analysis revealed anti-ADAM9 antibodies were efficiently internalized and processed by tumor cells making ADAM9 an attractive target for antibody-drug conjugate (ADC) development. Here, we describe the preclinical evaluation of IMGC936, a novel ADC targeted against ADAM9. IMGC936 is comprised of a high-affinity humanized antibody site-specifically conjugated to DM21-C, a next-generation linker-payload that combines a maytansinoid microtubule-disrupting payload with a stable tripeptide linker, at a drug antibody ratio of approximately 2.0. In addition, the YTE mutation (M252Y/S254T/T256E) was introduced into the CH2 domain of the antibody Fc to maximize in vivo plasma half-life and exposure. IMGC936 exhibited cytotoxicity toward ADAM9-positive human tumor cell lines, as well as bystander killing, potent antitumor activity in human cell line-derived xenograft and patient-derived xenograft tumor models, and an acceptable safety profile in cynomolgus monkeys with favorable pharmacokinetic properties. Our preclinical data provide a strong scientific rationale for the further development of IMGC936 as a therapeutic candidate for the treatment of ADAM9-positive cancers. A first-in-human study of IMGC936 in patients with advanced solid tumors has been initiated (NCT04622774).


Subject(s)
Immunoconjugates , ADAM Proteins , Cell Line, Tumor , Heterografts , Humans , Immunoconjugates/chemistry , Membrane Proteins/genetics , Xenograft Model Antitumor Assays
3.
Bioinformatics ; 26(3): 432-3, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19965880

ABSTRACT

MOTIVATION: DataPflex is a MATLAB-based application that facilitates the manipulation and visualization of multidimensional datasets. The strength of DataPflex lies in the intuitive graphical user interface for the efficient incorporation, manipulation and visualization of high-dimensional data that can be generated by multiplexed protein measurement platforms including, but not limited to Luminex or Meso-Scale Discovery. Such data can generally be represented in the form of multidimensional datasets [for example (time x stimulation x inhibitor x inhibitor concentration x cell type x measurement)]. For cases where measurements are made in a combinational fashion across multiple dimensions, there is a need for a tool to efficiently manipulate and reorganize such data for visualization. DataPflex accepts data consisting of up to five arbitrary dimensions in addition to a measurement dimension. Data are imported from a simple .xls format and can be exported to MATLAB or .xls. Data dimensions can be reordered, subdivided, merged, normalized and visualized in the form of collections of line graphs, bar graphs, surface plots, heatmaps, IC50's and other custom plots. Open source implementation in MATLAB enables easy extension for custom plotting routines and integration with more sophisticated analysis tools. AVAILABILITY: DataPflex is distributed under the GPL license (http://www.gnu.org/licenses/) together with documentation, source code and sample data files at: http://code.google.com/p/datapflex. SUPPLEMENTARY INFORMATION: Supplementary data available at Bioinformatics online.


Subject(s)
Computational Biology/methods , Information Storage and Retrieval/methods , Software , Computer Graphics , Database Management Systems , User-Computer Interface
4.
Mol Cancer Ther ; 20(1): 203-212, 2021 01.
Article in English | MEDLINE | ID: mdl-33177153

ABSTRACT

Several antibody-drug conjugates (ADC) showing strong clinical responses in solid tumors target high expression antigens (HER2, TROP2, Nectin-4, and folate receptor alpha/FRα). Highly expressed tumor antigens often have significant low-level expression in normal tissues, resulting in the potential for target-mediated drug disposition (TMDD) and increased clearance. However, ADCs often do not cross-react with normal tissue in animal models used to test efficacy (typically mice), and the impact of ADC binding to normal tissue antigens on tumor response remains unclear. An antibody that cross-reacts with human and murine FRα was generated and tested in an animal model where the antibody/ADC bind both human tumor FRα and mouse FRα in normal tissue. Previous work has demonstrated that a "carrier" dose of unconjugated antibody can improve the tumor penetration of ADCs with high expression target-antigens. A carrier dose was employed to study the impact on cross-reactive ADC clearance, distribution, and efficacy. Co-administration of unconjugated anti-FRα antibody with the ADC-improved efficacy, even in low expression models where co-administration normally lowers efficacy. By reducing target-antigen-mediated clearance in normal tissue, the co-administered antibody increased systemic exposure, improved tumor tissue penetration, reduced target-antigen-mediated uptake in normal tissue, and increased ADC efficacy. However, payload potency and tumor antigen saturation are also critical to efficacy, as shown with reduced efficacy using too high of a carrier dose. The judicious use of higher antibody doses, either through lower DAR or carrier doses, can improve the therapeutic window by increasing efficacy while lowering target-mediated toxicity in normal tissue.


Subject(s)
Antibodies/administration & dosage , Antibodies/pharmacology , Immunoconjugates/metabolism , Animals , Antibodies/toxicity , Cell Line, Tumor , Cross Reactions/immunology , Drug Carriers/chemistry , Female , Immunoconjugates/blood , Mice , Mice, SCID , Neoplasms/pathology , Treatment Outcome
5.
J Cell Biol ; 164(4): 535-46, 2004 Feb 16.
Article in English | MEDLINE | ID: mdl-14769859

ABSTRACT

Accurate chromosome segregation depends on precise regulation of mitosis by the spindle checkpoint. This checkpoint monitors the status of kinetochore-microtubule attachment and delays the metaphase to anaphase transition until all kinetochores have formed stable bipolar connections to the mitotic spindle. Components of the spindle checkpoint include the mitotic arrest defective (MAD) genes MAD1-3, and the budding uninhibited by benzimidazole (BUB) genes BUB1 and BUB3. In animal cells, all known spindle checkpoint proteins are recruited to kinetochores during normal mitoses. In contrast, we show that whereas Saccharomyces cerevisiae Bub1p and Bub3p are bound to kinetochores early in mitosis as part of the normal cell cycle, Mad1p and Mad2p are kinetochore bound only in the presence of spindle damage or kinetochore lesions that interfere with chromosome-microtubule attachment. Moreover, although Mad1p and Mad2p perform essential mitotic functions during every division cycle in mammalian cells, they are required in budding yeast only when mitosis goes awry. We propose that differences in the behavior of spindle checkpoint proteins in animal cells and budding yeast result primarily from evolutionary divergence in spindle assembly pathways.


Subject(s)
Chromosomes, Fungal/metabolism , Microtubules/metabolism , Saccharomyces cerevisiae/metabolism , Spindle Apparatus/metabolism , Animals , Aurora Kinases , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Chromosome Segregation , Genes, cdc , Intracellular Signaling Peptides and Proteins , Kinetochores/metabolism , Macromolecular Substances , Mad2 Proteins , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nuclear Proteins , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction/physiology
6.
Mol Biol Cell ; 14(11): 4557-68, 2003 Nov.
Article in English | MEDLINE | ID: mdl-13679521

ABSTRACT

Chromosome segregation at mitosis depends critically on the accurate assembly of kinetochores and their stable attachment to microtubules. Analysis of Saccharomyces cerevisiae kinetochores has shown that they are complex structures containing >/=50 protein components. Many of these yeast proteins have orthologs in animal cells, suggesting that key aspects of kinetochore structure have been conserved through evolution, despite the remarkable differences between the 125-base pair centromeres of budding yeast and the Mb centromeres of animal cells. We describe here an analysis of S. cerevisiae Ndc10p, one of the four protein components of the CBF3 complex. CBF3 binds to the CDEIII element of centromeric DNA and initiates kinetochore assembly. Whereas CDEIII binding by Ndc10p requires the other components of CBF3, Ndc10p can bind on its own to CDEII, a region of centromeric DNA with no known binding partners. Ndc10p-CDEII binding involves a dispersed set of sequence-selective and -nonselective contacts over approximately 80 base pairs of DNA, suggesting formation of a multimeric structure. CDEII-like sites, active in Ndc10p binding, are also present along chromosome arms. We propose that a polymeric Ndc10p complex formed on CDEII and CDEIII DNA is the foundation for recruiting microtubule attachment proteins to kinetochores. A similar type of polymeric structure on chromosome arms may mediate other chromosome-spindle interactions.


Subject(s)
Chromosomes, Fungal/metabolism , DNA-Binding Proteins/metabolism , Kinetochores/metabolism , Microtubules/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Base Sequence , Molecular Sequence Data , Mutation , Sequence Analysis, DNA
7.
Cancer Res ; 76(6): 1517-27, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26759238

ABSTRACT

Trastuzumab is the standard of care for HER2-positive breast cancer patients, markedly improving disease-free and overall survival. Combined with chemotherapy, it enhances patient outcomes, but cardiotoxicity due to the trastuzumab treatment poses a serious adverse effect. MM-302 is a HER2-targeted PEGylated liposome that encapsulates doxorubicin to facilitate its delivery to HER2-overexpressing tumor cells while limiting exposure to nontarget tissues, including the heart. In this study, we evaluated the feasibility and preclinical activity of combining MM-302 with trastuzumab. MM-302 and trastuzumab target different domains of the HER2 receptor and thus could simultaneously bind HER2-overexpressing tumor cells in vitro and in vivo. Furthermore, trastuzumab did not disrupt the mechanism of action of MM-302 in delivering doxorubicin to the n0ucleus and inducing DNA damage. Reciprocally, MM-302 did not interfere with the ability of trastuzumab to block prosurvival p-Akt signaling. Interestingly, coadministration of the two agents acutely increased the deposition of MM-302 in human xenograft tumors and subsequently increased the expression of the DNA damage marker p-p53. Finally, the combination of MM-302 and trastuzumab induced synergistic antitumor activity in HER2-overexpressing xenograft models of breast and gastric cancer. Collectively, our findings highlight a novel combination therapy that efficiently targets HER2-overexpressing cells through multiple mechanisms and support the ongoing investigation of combined MM-302/trastuzumab therapy for HER2-positive metastatic breast cancer in a randomized phase II clinical trial.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Receptor, ErbB-2/genetics , Stomach Neoplasms/drug therapy , Antibiotics, Antineoplastic/administration & dosage , Breast Neoplasms/metabolism , Cell Line, Tumor , DNA Damage/drug effects , Doxorubicin/administration & dosage , Doxorubicin/analogs & derivatives , Drug Synergism , Female , Humans , Polyethylene Glycols/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , Stomach Neoplasms/metabolism , Trastuzumab/administration & dosage , Tumor Suppressor Protein p53/metabolism
8.
Mol Cancer Ther ; 14(9): 2060-71, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26162690

ABSTRACT

Given the bulky nature of nanotherapeutics relative to small molecules, it is hypothesized that effective tumor delivery and penetration are critical barriers to their clinical activity. HER2-targeted PEGylated liposomal doxorubicin (MM-302, HER2-tPLD) is an antibody-liposomal drug conjugate designed to deliver doxorubicin to HER2-overexpressing cancer cells while limiting uptake into nontarget cells. In this work, we demonstrate that the administration and appropriate dose sequencing of cyclophosphamide can improve subsequent MM-302 delivery and enhance antitumor activity in preclinical models without negatively affecting nontarget tissues, such as the heart and skin. We demonstrate that this effect is critically dependent on the timing of cyclophosphamide administration. Furthermore, the effect was found to be unique to cyclophosphamide and related analogues, and not shared by other agents, such as taxanes or eribulin, under the conditions examined. Analysis of the cyclophosphamide-treated tumors suggests that the mechanism for improved MM-302 delivery involves the induction of tumor cell apoptosis, reduction of overall tumor cell density, substantial lowering of interstitial fluid pressure, and increasing vascular perfusion. The novel dosing strategy for cyclophosphamide described herein is readily translatable to standard clinical regimens, represents a potentially significant advance in addressing the drug delivery challenge, and may have broad applicability for nanomedicines. This work formed the basis for clinical evaluation of cyclophosphamide for improving liposome deposition as part of an ongoing phase I clinical trial of MM-302 in HER2-positive metastatic breast cancer.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/metabolism , Cyclophosphamide/pharmacology , Doxorubicin/analogs & derivatives , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antibiotics, Antineoplastic/administration & dosage , Apoptosis/drug effects , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cyclophosphamide/administration & dosage , Disease Models, Animal , Doxorubicin/administration & dosage , Drug Synergism , Female , Humans , Ifosfamide/administration & dosage , Ifosfamide/pharmacology , Mice , Polyethylene Glycols/administration & dosage , Positron-Emission Tomography , Tomography, X-Ray Computed , Tumor Microenvironment , Xenograft Model Antitumor Assays
9.
Mol Cancer Ther ; 12(9): 1816-28, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23723124

ABSTRACT

Numerous targeted nanotherapeutics have been described for potential treatment of solid tumors. Although attention has focused on antigen selection and molecular design of these systems, there has been comparatively little study of how cellular heterogeneity influences interaction of targeted nanoparticles with tumor cells. Antigens, such as HER2/ERBB2, are heterogeneously expressed across different indications, across patients, and within individual tumors. Furthermore, antigen expression in nontarget tissues necessitates optimization of the therapeutic window. Understanding the performance of a given nanoparticle under different regimens of antigen expression has the ability to inform patient selection and clinical development decisions. In this work, HER2-targeted liposomal doxorubicin was used as a model-targeted nanoparticle to quantitatively investigate the effect of HER2 expression levels on delivery of doxorubicin to the nucleus. We find quantitatively greater nuclear doxorubicin delivery with increasing HER2 expression, exhibiting a threshold effect at approximately 2 × 10(5) HER2 receptors/cell. Kinetic modeling indicated that the threshold effect arises from multiple low-affinity interactions between the targeted liposome and HER2. These results support previous data showing little or no uptake into human cardiomyocytes, which express levels of HER2 below the threshold. Finally, these results suggest that HER2-targeted liposomal doxorubicin may effectively target tumors that fall below traditional definitions of HER2-positive tumors, thereby expanding the potential population of patients that might benefit from this agent.


Subject(s)
Antibiotics, Antineoplastic/pharmacokinetics , Doxorubicin/pharmacokinetics , Neoplasms/drug therapy , Receptor, ErbB-2/genetics , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/therapeutic use , Cell Line, Tumor , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Drug Delivery Systems/methods , Humans , Liposomes , Mice , Molecular Targeted Therapy , Neoplasms/metabolism , Receptor, ErbB-2/metabolism
10.
Mol Biosyst ; 6(10): 1956-72, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20617251

ABSTRACT

Rheumatoid arthritis (RA) is a complex, multicellular disease involving a delicate balance between both pro- and anti-inflammatory cytokines which ultimately determines the disease phenotype. The simultaneous presence of multiple signaling molecules, and more specifically their relative levels, potentially influences the efficacy of directed therapies. Using the human U937 monocytic cell line, we generated a self-consistent dataset measuring 50 cytokines and 23 phosphoproteins in the presence of 6 small molecule inhibitors under 15 stimulatory conditions throughout a 24 hour time course. From this dataset, we are able to explore phosphoprotein and cytokine relationships, as well as evaluate the significance of cellular context on the ability of small molecule inhibitors to block inflammatory processes. We show that the ability of a p38 inhibitor to attenuate TNFalpha production is influenced by local levels of GM-CSF and IL-1beta, two cytokines known to be elevated in the joints of RA patients. Within the cell, compensatory mechanisms between signaling pathways are apparent, as selective p38 MAPK inhibition results in the increased phosphorylation of other MAPKs (ERK and JNK) and their downstream substrates (CREB, c-Jun, and ATF-2). Further, we demonstrate that TNFalpha-neutralizing antibodies have secondary effects on cytokine production, impacting more than just TNFalpha alone. p38 MAPK inhibition using a small molecule inhibitor also blocks production of anti-inflammatory cytokines including IL-10, IL-1ra and IL-2ra. Collectively, the impact of cell context on TNFalpha production and unintended blockade of anti-inflammatory cytokines may compromise the efficacy of p38 inhibitors in a clinical setting. The effort described in this work evaluates the effect of inhibitors on multiple endpoints (both intra- and extracellular), under a range of biologically relevant conditions, thus providing a unique means for differentiation of compounds and potential opportunity for improved pharmacological manipulation of disease endpoints in RA.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/blood , Interleukin-1beta/blood , Macrophages/metabolism , Monocytes/metabolism , Tumor Necrosis Factors/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Humans , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL