Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
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
2.
Mol Cancer Ther ; 19(11): 2235-2244, 2020 11.
Article in English | MEDLINE | ID: mdl-32967924

ABSTRACT

B7-H3, also referred to as CD276, is a member of the B7 family of immune regulatory proteins. B7-H3 is overexpressed on many solid cancers, including prostate cancer, renal cell carcinoma, melanoma, squamous cell carcinoma of the head and neck, non-small cell lung cancer, and breast cancer. Overexpression of B7-H3 is associated with disease severity, risk of recurrence and reduced survival. In this article, we report the preclinical development of MGC018, an antibody-drug conjugate targeted against B7-H3. MGC018 is comprised of the cleavable linker-duocarmycin payload, valine-citrulline-seco duocarmycin hydroxybenzamide azaindole (vc-seco-DUBA), conjugated to an anti-B7-H3 humanized IgG1/kappa mAb through reduced interchain disulfides, with an average drug-to-antibody ratio of approximately 2.7. MGC018 exhibited cytotoxicity toward B7-H3-positive human tumor cell lines, and exhibited bystander killing of target-negative tumor cells when cocultured with B7-H3-positive tumor cells. MGC018 displayed potent antitumor activity in preclinical tumor models of breast, ovarian, and lung cancer, as well as melanoma. In addition, antitumor activity was observed toward patient-derived xenograft models of breast, prostate, and head and neck cancer displaying heterogeneous expression of B7-H3. Importantly, MGC018 exhibited a favorable pharmacokinetic and safety profile in cynomolgus monkeys following repeat-dose administration. The antitumor activity observed preclinically with MGC018, together with the positive safety profile, provides evidence of a potentially favorable therapeutic index and supports the continued development of MGC018 for the treatment of solid cancers. GRAPHICAL ABSTRACT: http://mct.aacrjournals.org/content/molcanther/19/11/2235/F1.large.jpg.


Subject(s)
B7 Antigens/antagonists & inhibitors , Drug Evaluation, Preclinical , Immune Checkpoint Inhibitors/pharmacology , Immunoconjugates/pharmacology , Neoplasms/drug therapy , Animals , B7 Antigens/genetics , B7 Antigens/metabolism , Bystander Effect , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Monitoring , Gene Knockdown Techniques , Humans , Immune Checkpoint Inhibitors/chemistry , Immune Checkpoint Inhibitors/isolation & purification , Immunoconjugates/chemistry , Immunoconjugates/isolation & purification , Mice , Neoplasms/metabolism , Neoplasms/pathology , Treatment Outcome , Xenograft Model Antitumor Assays
3.
Mol Cancer Ther ; 17(8): 1761-1772, 2018 08.
Article in English | MEDLINE | ID: mdl-29866746

ABSTRACT

We have developed MGD007 (anti-glycoprotein A33 x anti-CD3), a DART protein designed to redirect T cells to target gpA33 expressing colon cancer. The gpA33 target was selected on the basis of an antibody-based screen to identify cancer antigens universally expressed in both primary and metastatic colorectal cancer specimens, including putative cancer stem cell populations. MGD007 displays the anticipated-bispecific binding properties and mediates potent lysis of gpA33-positive cancer cell lines, including models of colorectal cancer stem cells, through recruitment of T cells. Xenograft studies showed tumor growth inhibition at doses as low as 4 µg/kg. Both CD8 and CD4 T cells mediated lysis of gpA33-expressing tumor cells, with activity accompanied by increases in granzyme and perforin. Notably, suppressive T-cell populations could also be leveraged to mediate lysis of gpA33-expressing tumor cells. Concomitant with CTL activity, both T-cell activation and expansion are observed in a gpA33-dependent manner. No cytokine activation was observed with human PBMC alone, consistent with the absence of gpA33 expression on peripheral blood cell populations. Following prolonged exposure to MGD007 and gpA33 positive tumor cells, T cells express PD-1 and LAG-3 and acquire a memory phenotype but retain ability to support potent cell killing. In cynomolgus monkeys, 4 weekly doses of 100 µg/kg were well tolerated, with prolonged PK consistent with that of an Fc-containing molecule. Taken together, MGD007 displays potent activity against colorectal cancer cells consistent with a mechanism of action endowed in its design and support further investigation of MGD007 as a potential novel therapeutic treatment for colorectal cancer. Mol Cancer Ther; 17(8); 1761-72. ©2018 AACR.


Subject(s)
Colorectal Neoplasms/drug therapy , Immunotherapy/methods , Animals , Cell Line, Tumor , Colorectal Neoplasms/pathology , Female , Haplorhini , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis
4.
Clin Cancer Res ; 18(14): 3834-45, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22615450

ABSTRACT

PURPOSE: The goal of this research was to harness a monoclonal antibody (mAb) discovery platform to identify cell-surface antigens highly expressed on cancer and develop, through Fc optimization, potent mAb therapies toward these tumor-specific antigens. EXPERIMENTAL DESIGN: Fifty independent mAbs targeting the cell-surface immunoregulatory B7-H3 protein were obtained through independent intact cell-based immunizations using human tissue progenitor cells, cancer cell lines, or cell lines displaying cancer stem cell properties. Binding studies revealed this natively reactive B7-H3 mAb panel to bind a range of independent B7-H3 epitopes. Immunohistochemical analyses showed that a subset displayed strong reactivity to a broad range of human cancers while exhibiting limited binding to normal human tissues. A B7-H3 mAb displaying exquisite tumor/normal differential binding was selected for humanization and incorporation of an Fc domain modified to enhance effector-mediated antitumor function via increased affinity for the activating receptor CD16A and decreased binding to the inhibitory receptor CD32B. RESULTS: MGA271, the resulting engineered anti-B7-H3 mAb, mediates potent antibody-dependent cellular cytotoxicity against a broad range of tumor cell types. Furthermore, in human CD16A-bearing transgenic mice, MGA271 exhibited potent antitumor activity in B7-H3-expressing xenograft models of renal cell and bladder carcinoma. Toxicology studies carried out in cynomolgus monkeys revealed no significant test article-related safety findings. CONCLUSIONS: This data supports evaluation of MGA271 clinical utility in B7-H3-expressing cancer, while validating a combination of a nontarget biased approach of intact cell immunizations and immunohistochemistry to identify novel cancer antigens with Fc-based mAb engineering to enable potent antitumor activity.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Antigens, Surface/immunology , B7 Antigens/immunology , Neoplasms , Animals , Cell Line, Tumor , Epitopes/immunology , Humans , Immunoglobulin Fc Fragments/immunology , Immunotherapy , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasms/drug therapy , Neoplasms/immunology
5.
Methods Mol Biol ; 682: 3-13, 2011.
Article in English | MEDLINE | ID: mdl-21057916

ABSTRACT

Apoptosis, or programmed cell death, plays an important role in normal development and homeostasis of adult tissues. Apoptosis has also been linked to many disease states, including cancer. One of the biochemical hallmarks of apoptosis is the generation of free 3'-hydroxyl termini on DNA via cleavage of chromatin into single and multiple oligonuleosome-length fragments. The TdT-mediated dUTP-biotin nick end labeling (TUNEL) assay exploits this biochemical hallmark by labeling the exposed termini of DNA, thereby enabling visualization of nuclei containing fragmented DNA. This review outlines the general method for in situ TUNEL staining of cultured cells and tissue sections, and highlights recent improvements in the technique and limitations of the assay.


Subject(s)
Apoptosis , In Situ Nick-End Labeling/methods , Animals , Cell Adhesion , Cells, Cultured , Colorimetry
6.
Clin Cancer Res ; 16(5): 1673-81, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20179219

ABSTRACT

PURPOSE: RAV12 is a high affinity, internalizing, chimeric IgG1 monoclonal antibody that binds RAAG12, a novel primate-restricted N-linked carbohydrate epitope present on multiple cell surface proteins. RAAG12 is highly expressed on many adenocarcinomas, particularly those of gastrointestinal origin. A phase 1 dose-escalation safety and pharmacokinetics trial was conducted in patients with metastatic or recurrent adenocarcinomas. EXPERIMENTAL DESIGN: RAV12 was initially given i.v. weekly x4, then by fractionated dosing twice or thrice weekly. Thirty-three patients were treated in the dose escalation segment of the trial in the following cohorts: 0.3 mg/kg qw (6), 1.0 mg/kg qw (8), 1.5 mg/kg qw (7); and 0.5 mg/kg biw (3), 0.75 mg/kg biw (3), and 0.5 mg/kg tiw (6). Twenty patients were enrolled in a maximum tolerated dose cohort expansion at 0.75 mg/kg biw. RESULTS: Two clinical syndromes were associated with drug administration: abdominal cramping pain with diarrhea, and asymptomatic, self-limited increases of liver function tests. These effects were partially ameliorated with fractionated dosing. Pharmacokinetics was dose dependent. Maximum concentration was reduced, whereas area under the concentration versus time curve was maintained with fractionated dosing. One patient with colorectal cancer experienced a durable partial remission, with a time to progression (TTP) of >8 months. Three additional patients experienced a TTP of >4 months. CONCLUSIONS: RAV12 has activity in recurrent adenocarcinomas. However, the safety profile of the antibody seems to preclude the delivery of highly efficacious doses. Re-engineering the molecule to remove FcRn binding (while maintaining FcgammaR binding) and to humanize it may improve the toxicity profile and efficacy.


Subject(s)
Adenocarcinoma/drug therapy , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/pharmacokinetics , Antigens, Tumor-Associated, Carbohydrate/drug effects , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Immunohistochemistry , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Recurrence, Local/drug therapy
7.
Arch Pathol Lab Med ; 133(9): 1403-12, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19722746

ABSTRACT

CONTEXT: RAAG12 is a primate-restricted N-linked carbohydrate antigen present on multiple membrane-associated proteins. RAAG12 is recognized by the RAV12 monoclonal antibody. RAV12 binds to RAAG12-expressing gastrointestinal adenocarcinomas, modifies growth factor-mediated signaling, induces oncotic cell death in vitro, and has antitumor activity toward gastrointestinal tumor xenografts. OBJECTIVE: To determine the expression pattern of RAAG12 in normal and tumor tissue to identify indications for clinical study and potential safety issues. DESIGN: Immunohistochemistry of 36 normal human tissues and a broad range of tumor tissues to profile RAAG12 expression. RESULTS: More than 90% of colon, gastric, and pancreatic adenocarcinomas expressed RAAG12, and expression was uniform in most samples. Expression of RAAG12 at lower frequency and/or uniformity was observed in other cancers, including esophageal, ovarian, liver, breast, and prostate carcinomas and adenocarcinomas. Similar RAAG12 expression was observed between primary and metastatic colon adenocarcinomas. No staining was seen on cardiovascular, endocrine, neuromuscular, hematopoietic, or nervous system tissue from non-tumor-bearing individuals. RAAG12 was expressed on mucosal and glandular/ductal epithelium. The gastrointestinal tract mucosa and pancreatic/biliary ducts displayed the most uniform reactivity. RAAG12 exhibited differential subcellular localization in these normal, compared with tumor, tissues. Normal polarized epithelia primarily displayed apical membrane and cytoplasmic staining, whereas tumors exhibited whole membrane staining that increased with decreasing differentiation. CONCLUSIONS: High expression of RAAG12 on tumors of gastrointestinal origin suggests these cancers are appropriate targets for RAV12 therapy. Differential subcellular location of RAAG12 on normal epithelia may limit accessibility of RAV12 to the subset of normal tissues that exhibit antigen expression.


Subject(s)
Adenocarcinoma/immunology , Antibodies, Monoclonal/immunology , Antigens, Tumor-Associated, Carbohydrate/immunology , Neoplasms/immunology , Animals , Antigens, Tumor-Associated, Carbohydrate/metabolism , Cell Line, Tumor , Female , Humans , Male , Mice , Mice, SCID , Xenograft Model Antitumor Assays
8.
Curr Opin Pharmacol ; 8(5): 627-31, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18804182

ABSTRACT

The identification of novel cell surface antigens present on tumor cells is crucial for developing new cancer therapies. Intact, viable cancer cells, which display cancer-restricted antigens in their native conformation and cellular context, provide a rich source of novel antigens. Antibody-based technologies are being used to probe the surface of intact cancer cells for cancer-specific antigen targets. In addition to identifying new proteins, these approaches are generating monoclonal antibodies (MAbs) to cancer-specific epitopes and nonprotein targets not amenable to genomics-based approaches. The multiple cell-based approaches developing epitope-specific MAbs to cancer antigens is likely to usher in a new era of therapeutic MAb target discovery.


Subject(s)
Antibodies , Antigens, Surface/drug effects , Antigens, Surface/immunology , Antineoplastic Agents/pharmacology , Animals , Antibodies/chemistry , Antigens, Surface/chemistry , Genomics , Humans , Proteomics
9.
Methods Cell Biol ; 86: 241-55, 2008.
Article in English | MEDLINE | ID: mdl-18442650

ABSTRACT

Homogeneous, well-characterized cultures of kidney cells representative of defined cellular phenotypes comprising the developing and adult kidney provide important tools to investigate kidney biology. Further, the development of defined media for these culture systems provides opportunities to investigate the role of nutrients, hormones, and matrix components, as well as exogenous insults, in renal development, function, and toxicity. The current explosion in stem cell research has fueled an expanded effort to develop techniques to isolate and culture kidney progenitor and stem cells, which have the potential to treat various forms of renal disease. In this chapter, we outline methods to initiate and propagate long-term cultures of highly homogeneous fetal kidney epithelial progenitor cells. By utilizing a low calcium-containing serum-free culture medium together with a set of defined hormones and extracellular matrix, kidney epithelial progenitor cells can be cultured for more than 60 population doublings without loss of growth potential or phenotypic signs of differentiation. The cultures appear to represent early kidney epithelial progenitors based on cellular marker expression. The cells express the mRNA encoding the embryonic kidney mesenchyme/epithelial marker PAX-2, the stem cell protein CD133, the kidney embryonic progenitor protein CD24, as well as CD29 and CD44. The cells are negative for E-cadherin when grown under low calcium conditions (<0.05 mM); however, E-cadherin expression is induced when cells are cultured under normal calcium conditions (1.2 mM), suggesting that differentiation of the kidney epithelial progenitor culture can be modulated in part by altering the calcium concentration of the medium.


Subject(s)
Cell Culture Techniques , Epithelial Cells/cytology , Fetus/cytology , Kidney/cytology , Stem Cells/cytology , Adult , Animals , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Differentiation , Cells, Cultured , Culture Media/chemistry , Humans , Swine
10.
Mol Cancer Ther ; 6(3): 856-65, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17363480

ABSTRACT

RAV12 is a chimeric antibody that recognizes an N-linked carbohydrate antigen (RAAG12) strongly expressed on multiple solid organ cancers. More than 90% of tumors of colorectal, gastric, and pancreatic origin express RAAG12, and a majority of these tumors exhibit uniform RAAG12 expression. RAV12 exhibits potent cytotoxic activity in vitro against COLO 205 colon tumor cells via an oncotic cell death mechanism. RAV12-treated COLO 205 cells undergo morphologic changes consistent with oncosis, including cytoskeletal rearrangement, rapid plasma membrane swelling, and cell lysis. RAV12 inhibited the growth of RAAG12-expressing gastrointestinal tumor xenografts in athymic mice. In the case of SNU-16 tumor cells, twice weekly treatment of established s.c. tumors with 10 mg/kg RAV12 caused a approximately 70% suppression of tumor growth at the end of the study. This preclinical data has led to the initiation of a phase I/IIA clinical study of RAV12 in patients with metastatic or recurrent adenocarcinoma.


Subject(s)
Adenocarcinoma/drug therapy , Antibodies, Monoclonal/therapeutic use , Antigens, Tumor-Associated, Carbohydrate/immunology , Gastrointestinal Neoplasms/drug therapy , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , CHO Cells , Cricetinae , Cricetulus , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Female , Fetus , Gastrointestinal Neoplasms/immunology , Gastrointestinal Neoplasms/pathology , Glycosylation , Humans , Immunization , Kidney/metabolism , Kidney/pathology , Male , Mice , Mice, Inbred BALB C , Polymerase Chain Reaction , Repressor Proteins/immunology , Repressor Proteins/metabolism , Tissue Array Analysis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Zinc Fingers
SELECTION OF CITATIONS
SEARCH DETAIL
...