Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Immunol Methods ; 510: 113362, 2022 11.
Article in English | MEDLINE | ID: mdl-36174735

ABSTRACT

BACKGROUND: Antibodies and other recognition molecules direct cancer cell death by multiple types of immune cells. Therapy directed at only one target typically results in tumor regrowth because of tumor heterogeneity. Our goal is to direct therapy to multiple targets simultaneously. Our previous studies showed that multiple antibodies targeting mutated tumor proteins inhibited tumor growth when injected subcutaneously near the time of cancer cell implantation. METHODS: A cocktail of rabbit antibodies against B16-F10 cell surface related mutated proteins were generated. Implanted B16-F10 cells were allowed to grow to palpable size before treatment. Antibodies were administered using different routes of exposure. Free antibody was compared to antibody armed on mouse splenic white blood cells (WBCs). Binding of the antibody cocktail was determined for mouse and human WBCs. RESULTS: The antibody cocktail inhibited tumor growth and prolonged survival when administered as free antibody or armed on WBCs. The antibody cocktail armed on WBCs achieved similar tumor inhibition as free antibody but at a dose 1000-fold less. Armed WBCs achieved tumor inhibition by intravenous and subcutaneous administration. The antibody cocktail bound well to human WBCs and saturation dose was defined. Binding was stable under simulated in vivo condition in human plasma at 37 °C. CONCLUSIONS: Antibodies targeting multiple tumor mutated proteins inhibited tumor growth and prolonged survival. Effective antibody dose was reduced 1000-fold by arming WBCs. Rabbit antibodies saturated human WBCs using <1 mg per billion cells. A phase I trial in cancer patients using this strategy has been approved by the FDA.


Subject(s)
Melanoma, Experimental , Animals , Antibodies , Humans , Leukocytes/metabolism , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Rabbits
2.
Ann Clin Lab Sci ; 52(3): 349-358, 2022 May.
Article in English | MEDLINE | ID: mdl-35777796

ABSTRACT

OBJECTIVE: Tumor heterogeneity is a fundamental problem in treating cancer with monotargeting therapy, including chemical, antibody, and T cell therapies. Our goal is to target multiple mutated peptides found in a patient's cancer to increase antibody therapy effectiveness. METHODS: Tumor samples were derived from patients with neuroblastoma. Whole-exome sequencing was performed of tumor and normal cells. Mutated proteins with missense mutations were selected from the patient tumor. These mutated proteins were further selected for the presence of missense mutations in the outer cell surface. Peptides representing a mutated section of the proteins were used for vaccinating rabbits and generating anti-peptide antibodies. The binding of individual polyclonal antibodies (pAbs) and the mixtures of pAbs were determined against the patient's tumor as cultured neuroblastoma cells and in a murine xenograft model. Antibodies were prepared according to FDA requirements of a phase I clinical protocol. RESULTS: All of the generated rabbit pAbs bound with high affinity to the corresponding peptide used for vaccination. The pAbs also bound to low passage neuroblastoma cells. Mixed as cocktails, the pAbs had substantially increased binding to cells and bound well to the xenograft tissue. No binding was observed to the panel of normal human tissues. Preparation of pAbs by an academic lab to clinical-grade was approved by FDA for phase I clinical trial. CONCLUSION: We describe a new strategy to make customized antibodies for individual cancer patients and present the data required to meet FDA specifications to begin a phase I clinical trial.


Subject(s)
Antibodies , Neuroblastoma , Animals , Cell Line , Humans , Mice , Mutation/genetics , Neuroblastoma/genetics , Neuroblastoma/therapy , Peptides , Rabbits
3.
J Immunol Methods ; 499: 113157, 2021 12.
Article in English | MEDLINE | ID: mdl-34597620

ABSTRACT

OBJECTIVE: Our goal was to develop a simpler and less expensive method of obtaining human clinical-grade WBCs using an alternative method to continuous leukapheresis. Our purpose for the WBCs is to arm them with rabbit anticancer antibodies for a phase I clinical trial. METHODS: Using leukocyte reduction filters (LRFs) discarded from the blood bank, we evaluated multiple variables to maximize recovery of WBCs with the lowest contamination of RBCs. Using an optimized protocol, full-scale runs according to FDA current Good Manufacturing Practice (cGMP) standards were completed with immediate filtration of blood obtained from donors participating in our study. RESULTS: Forward flushing of the filter removed 85% to 95% of residual RBCs and platelets. When backward flushed with 800 mL, 95% of the WBCs recovered were contained in the first 400 mL. The number of recovered WBCs was in the range of 166-211 million/100 mL filtered blood. Subpopulations of WBCs recovered from the LRFs were in the same proportion as the donors' whole blood. Viability of recovered WBCs was 96-99%. Exogenous rabbit antibodies bound well to the recovered WBCs and were retained for at least 5 h without significant reduction. Three full scale runs of WBCs recovered from donor blood filtered through the LRF met all FDA specification of sterility, endotoxin levels, viability and stability. CONCLUSION: Using LRFs, high quality clinical grade WBCs are readily obtained in quantities of 0.2 to 1.2 billion cells from 100 mL to 450 mL (1 unit) of whole blood.


Subject(s)
Filtration , Leukocytes/cytology , Humans
4.
J Immunol Methods ; 478: 112720, 2020 03.
Article in English | MEDLINE | ID: mdl-31812660

ABSTRACT

BACKGROUND: Antibodies that target a single tumor antigen fail to cure stage IV cancer patients due to tumor heterogeneity and variable expression of antigen. Tumor cells with insufficient binding of antibody will not undergo antibody induced cytotoxicity. We describe targeting multiple tumor-specific antigens that resulted in homogeneous dense binding to mouse melanoma cells and significant tumor growth inhibition. METHODS: Surface-related tumor-specific mutations on B16-F10 cells were identified. Peptides containing the single amino acid mutation were synthesized for 9 different neoantigens. Rabbits were vaccinated with each of these peptides and high affinity polyclonal antibodies to each peptide were obtained. The 9 antibodies were combined as a cocktail and mice with implanted B16-F10 cells were treated with and without PD1 inhibitor. RESULTS: Even a single dose of the antibody cocktail inhibited tumor growth and prolonged survival. PD1 inhibitor alone had little effect on tumor growth. The antibody cocktail plus PD1 inhibition increased tumor response and 4 doses of the cocktail completely prevented tumor growth in 50% of the mice. Complete responses were durable. The complete responders were highly resistant to tumor re-challenge at 6 months. No adverse events were identified in the antibody treated mice. CONCLUSIONS: Multiple tumor-specific cell surface-related neoantigens were abundant in B16-F10 cells. Antibodies to 9 of these neoantigens had variable binding but when combined had dense homogeneous binding. Even one dose of this cocktail of 9 antibodies improved survival and when multiple doses were combined with PD1 inhibition 50% of the mice were rendered permanently tumor free.


Subject(s)
Antibodies/administration & dosage , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/administration & dosage , Melanoma, Experimental/therapy , Skin Neoplasms/therapy , Animals , Antibodies/immunology , Antibody Affinity/immunology , Antineoplastic Agents, Immunological/immunology , Cell Line, Tumor , Drug Combinations , Female , Humans , Melanoma, Experimental/immunology , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Rabbits , Skin Neoplasms/immunology
5.
J Immunol Methods ; 460: 51-62, 2018 09.
Article in English | MEDLINE | ID: mdl-29906453

ABSTRACT

The aim of this preclinical study was to evaluate T7 bacteriophage as a nanoparticle platform for expression of neoantigens that could allow rapid generation of vaccines for potential studies in human cancer patients. We have generated recombinant T7 phage vaccines carrying neoepitopes derived from mutated proteins of B16-F10 melanoma tumor cells. With the single mutated amino acid (AA) centered, peptides were expressed on the outer coat of T7 phage. All peptides with 11 and 34 AAs were successfully expressed. Trimers of the 11-AA peptides were successfully expressed in only 3 of 8 peptides. The 11-AA peptide was better in stimulating antibodies selective for the mutated region than the longer 34-AA peptide. We observed a dose response for vaccines which provides an initial framework of the minimum phage required for vaccination. A single injection with phage-peptide vaccines in both monomer and trimer formats produced significant immune responses in mice on day 21, as assessed by lymph node cell counts, next generation sequencing (NGS), and plasma titers against T7 phage and vaccine peptides. A trimer provided no additional serum response to the monomer format. Immunization of mice with a mixture of 8 different peptide vaccines resulted in antibodies to most of the peptides. It was encouraging that induced antibodies had higher binding to the mutated peptides compared to the corresponding normal peptides. The NGS of lymph node cells demonstrated a low B cell receptor diversity and clonal hyperpolarization in vaccine-draining lymph nodes in comparison to those in unvaccinated mice nodes. The NGS data also revealed phenomenal increase in IgG and other class-switched antibodies following vaccination. These results agree with the higher plasma titers of IgG antibodies against T7 phage and vaccine peptides. Antibodies bound whole B16-F10 cells, lysates and multiple bands on Western blot. This indicates that these vaccine peptides successfully induced antibodies that bind full proteins from which the vaccine peptides were derived. We demonstrate a preclinical platform for rapid production of vaccines that can deliver mutated peptides and stimulate an appropriate B cell response. We anticipate further research in utilizing the cells from a tumor or vaccine draining lymph node as a resource for therapeutic anticancer reagents.


Subject(s)
Antibodies, Neoplasm/immunology , B-Lymphocytes/immunology , Bacteriophage T7/immunology , Cancer Vaccines/immunology , Lymph Nodes/immunology , Melanoma, Experimental/immunology , Melanoma-Specific Antigens/immunology , Nanoparticles , Peptide Library , Animals , B-Lymphocytes/pathology , Cancer Vaccines/genetics , Cell Line, Tumor , Lymph Nodes/pathology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Melanoma-Specific Antigens/genetics , Mice , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
6.
Cancer Immunol Immunother ; 67(5): 729-738, 2018 May.
Article in English | MEDLINE | ID: mdl-29427082

ABSTRACT

A better understanding of antitumor immune responses is the key to advancing the field of cancer immunotherapy. Endogenous immunity in cancer patients, such as circulating anticancer antibodies or tumor-reactive B cells, has been historically yet incompletely described. Here, we demonstrate that tumor-draining (sentinel) lymph node (SN) is a rich source for tumor-reactive B cells that give rise to systemic IgG anticancer antibodies circulating in the bloodstream of breast cancer patients. Using a synergistic combination of high-throughput B-cell sequencing and quantitative immunoproteomics, we describe the prospective identification of tumor-reactive SN B cells (based on clonal frequency) and also demonstrate an unequivocal link between affinity-matured expanded B-cell clones in the SN and antitumor IgG in the blood. This technology could facilitate the discovery of antitumor antibody therapeutics and conceivably identify novel tumor antigens. Lastly, these findings highlight the unique and specialized niche the SN can fill in the advancement of cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , B-Lymphocytes/immunology , Breast Neoplasms/immunology , Clone Cells/immunology , Immunoglobulin G/immunology , Sentinel Lymph Node/immunology , Amino Acid Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cells, Cultured , Female , Humans , Sequence Homology
7.
J Immunol Methods ; 455: 14-23, 2018 04.
Article in English | MEDLINE | ID: mdl-29382466

ABSTRACT

Autoantibodies to breast and other cancers are commonly present in cancer patients. A method to rapidly produce these anti-cancer autoantibodies in the lab would be valuable for understanding immune events and to generate candidate reagents for therapy and diagnostics. The purpose of this report is to evaluate sentinel nodes (SNs) of breast cancer patients as a source of anti-cancer antibodies. Radiotracer lymphatic mapping in 29 patients with breast cancer confirmed the identity of the SNs which provided source cells for this study. Flow cytometry demonstrated ~28% of the MNCs were B cells and ~44% of the B cells were class switched memory B cells. EBV-induced proliferation of B cells yielded tumor binding antibodies from 3 wells per 1000 but cultures were too unstable for detailed evaluations. Hybridomas generated by electrofusion produced IgG (48%), IgM (34%) and IgA (18%) antibody isotypes which were screened for binding to a panel of breast cancer cells of the major molecular subtypes. Tumor lysate binding was observed in 28% of the hybridoma clones and 10% of these bound whole tumor cells with unique binding patterns. More detailed evaluation of selected clones showed binding to the patient's own tumor. SNs are removed from more than 100,000 breast cancer patients in the US per year. Samples from these lymph nodes represent a substantial opportunity to generate anticancer antibodies.


Subject(s)
Antibodies/isolation & purification , B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Breast Neoplasms/diagnosis , Epstein-Barr Virus Infections/diagnosis , Herpesvirus 4, Human/immunology , Sentinel Lymph Node/metabolism , Antigens, Neoplasm/immunology , Autoantibodies/blood , Autoantigens/immunology , Breast Neoplasms/immunology , Cell Extracts , Cell Transformation, Neoplastic , Epstein-Barr Virus Infections/immunology , Female , Flow Cytometry , Humans , Hybridomas , Immunologic Memory , Radioactive Tracers , Sentinel Lymph Node/immunology
8.
J Transl Med ; 15(1): 180, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28851380

ABSTRACT

BACKGROUND: Our research is focused on using the vaccine draining lymph node to better understand the immune response to cancer vaccines and as a possible source of anti-cancer reagents. We evaluated vaccine draining lymph nodes archived from a clinical study in melanoma patients and determined the reaction of B cells to the vaccine peptides. METHODS: Mononuclear cells (MNCs) were recovered from cryopreserved lymph nodes that were directly receiving drainage from multi-peptide melanoma vaccine. The patients were enrolled on a vaccine study (NCT00089219, FDA, BB-IND No. 10825). B cell responses in the vaccine-draining lymph nodes were studied under both stimulated and un-stimulated conditions. Cryopreserved cells were stimulated with CD40L, stained with multiple human cell-surface markers (CD19, CD27, IgM) to identify different categories of B cell sub populations with flow cytometry. Hybridomas were generated from the lymph node cells after CD40L-stimulation. Cells were fused to murine plasmacytoma P3X63.Ag8.653 using Helix electrofusion chamber. ELISA was used to evaluate hybridoma derived antibody binding to vaccine peptides. RESULTS: Viable MNCs were satisfactorily recovered from lymph nodes cryopreserved from six vaccine study patients 8-14 years previously. B cell ELISPOT demonstrated responses for each patient to multiple vaccine peptides. CD40L stimulation of lymph node cells increased the proportion of CD19+ CD27+ cells from 12 to 65% of the sample and increased the proportion of class-switched cells. Screening of IgG secreting clones demonstrated binding to melanoma vaccine peptides. CONCLUSIONS: B cells were successfully recovered and expanded from human cryopreserved vaccine-draining lymph nodes. Individual B cells were identified that secreted antibodies that bound to cancer vaccine peptides. The ability to reliably generate in vitro the same antibodies observed in the blood of vaccinated patients will facilitate research to understand mechanisms of human antibody activity and possibly lead to therapeutic antibodies.


Subject(s)
Antibodies, Neoplasm/immunology , Cancer Vaccines/immunology , Lymph Nodes/pathology , B-Lymphocytes/immunology , CD40 Ligand , Clone Cells , Enzyme-Linked Immunospot Assay , Flow Cytometry , Humans , Hybridomas/pathology , Immunoglobulin G/metabolism , Peptides/immunology , Protein Binding
9.
Vaccine ; 35(9): 1259-1265, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28161423

ABSTRACT

PURPOSE: Our research is focused on using vaccine draining lymph nodes as a source of immune cells to better understand the immune response and to attempt to generate new anti-cancer reagents. Following a vaccine, harvesting the lymph node can only be done once. We endeavored to determine the range of times that B cells secreting anti-KLH antibodies were present in the node of KLH-vaccinated mice. RESULTS: Following vaccination the total number of mononuclear cells (MNCs) increased in the vaccine-draining lymph node (VDN). The percentage of MNCs that were B cells nearly doubled. B cells recovered from the node that secreted anti-KLH antibodies were evident by day 7. The number continued to increase and then slowly decreased over the observed time range to 28days after vaccination. The VDN, compared to the spleen, the bone marrow and the nonVDN, contained a higher percentage of B cells that secreted anti-KLH antibodies. CONCLUSIONS: After a vaccine, there is a multi-week window of time when an increasing number of B cells are present in a VDN that secrete anti-KLH antibodies. These results support using the VDN as a source for B cells that secrete anti-vaccine antibodies.


Subject(s)
B-Lymphocytes/immunology , Hemocyanins/immunology , Lymph Nodes/cytology , Vaccines/immunology , Adjuvants, Immunologic , Animals , Antibody Formation , Antibody-Producing Cells , Hemocyanins/administration & dosage , Lymph Nodes/immunology , Lymphocyte Activation , Mice , Spleen/immunology , Time Factors , Vaccines/administration & dosage
10.
Sci Rep ; 6: 27060, 2016 06 03.
Article in English | MEDLINE | ID: mdl-27257138

ABSTRACT

The design of potent and specific peptide inhibitors to therapeutic targets is of enormous utility for both proof-of-concept studies and for the development of potential new therapeutics. Grb7 is a key signaling molecule in the progression of HER2 positive and triple negative breast cancers. Here we report the crystal structure of a stapled bicyclic peptide inhibitor G7-B1 in complex with the Grb7-SH2 domain. This revealed an unexpected binding mode of the peptide, in which the staple forms an alternative contact with the surface of the target protein. Based on this structural information, we designed a new series of bicyclic G7 peptides that progressively constrain the starting peptide, to arrive at the G7-B4 peptide that binds with an approximately 2-fold enhanced affinity to the Grb7-SH2 domain (KD = 0.83 µM) compared to G7-B1 and shows low affinity binding to Grb2-, Grb10- and Grb14-SH2 domains (KD > 100 µM). Furthermore, we determined the structure of the G7-B4 bicyclic peptide in complex with the Grb7-SH2 domain, both before and after ring closing metathesis to show that the closed staple is essential to the target interaction. The G7-B4 peptide represents an advance in the development of Grb7 inhibitors and is a classical example of structure aided inhibitor development.


Subject(s)
GRB7 Adaptor Protein/chemistry , Peptides, Cyclic/chemistry , Amino Acid Sequence , Crystallography, X-Ray , Drug Design , GRB7 Adaptor Protein/antagonists & inhibitors , Humans , Hydrogen Bonding , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Quaternary
11.
J Med Chem ; 58(19): 7707-18, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26359549

ABSTRACT

The Grb7 adaptor protein is a therapeutic target for both TNBC and HER2+ breast cancer. A nonphosphorylated cyclic peptide 1 (known as G7-18NATE) inhibits Grb7 via targeting the Grb7-SH2 domain, but requires the presence of phosphate ions for both affinity and specificity. Here we report the discovery of malonate bound in the phosphotyrosine binding pocket of the apo-Grb7-SH2 structure. Based on this, we carried out the rational design and synthesis of two analogues of peptide 1 that incorporate carboxymethylphenylalanine (cmF) and carboxyphenylalanine (cF) as mimics of phosphotyrosine (pY). Binding studies using SPR confirmed that affinity for Grb7-SH2 domain is improved up to 9-fold over peptide 1 under physiological phosphate conditions (KD = 2.1-5.7 µM) and that binding is specific for Grb7-SH2 over closely related domains (low or no detectable binding to Grb2-SH2 and Grb10-SH2). X-ray crystallographic structural analysis of the analogue bearing a cmF moiety in complex with Grb7-SH2 has identified the precise contacts conferred by the pY mimic that underpin this improved molecular interaction. Together this study identifies and characterizes the tightest specific inhibitor of Grb7 to date, representing a significant development toward a new Grb7-targeted therapeutic.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , GRB7 Adaptor Protein/antagonists & inhibitors , Peptides, Cyclic/chemistry , Phosphotyrosine/chemistry , Antineoplastic Agents/chemical synthesis , Binding Sites , Breast Neoplasms/drug therapy , Crystallography, X-Ray , Female , GRB7 Adaptor Protein/metabolism , Humans , Malonates/chemistry , Molecular Targeted Therapy , Peptides, Cyclic/chemical synthesis , Peptidomimetics , Phosphates/chemistry , Phosphates/metabolism , Protein Conformation , src Homology Domains
12.
J Immunol Methods ; 426: 35-41, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26210205

ABSTRACT

Antibody therapy of neuroblastoma is promising and our goal is to derive antibodies from patients with neuroblastoma for developing new therapeutic antibodies. The feasibility of using residual bone marrow obtained for clinical indications as a source of tumor cells and a source of antibodies was assessed. From marrow samples, neuroblastoma cells were recovered, grown in cell culture and also implanted into mice to create xenografts. Mononuclear cells from the marrow were used as a source to generate phage display antibody libraries and also hybridomas. Growth of neuroblastoma patient cells was possible both in vitro and as xenografts. Antibodies from the phage libraries and from the monoclonal hybridomas bound autologous neuroblastoma cells with some selectivity. It appears feasible to recover neuroblastoma cells from residual marrow specimens and to generate human antibodies that bind autologous neuroblastoma cells. Expansion of this approach is underway to collect more specimens, optimize methods to generate antibodies, and to evaluate the bioactivity of neuroblastoma-binding antibodies.


Subject(s)
Antibodies, Monoclonal/immunology , Neuroblastoma/immunology , Animals , Humans , Hybridomas , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Peptide Library , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
J Biol Chem ; 288(52): 36926-35, 2013 Dec 27.
Article in English | MEDLINE | ID: mdl-24217252

ABSTRACT

We investigated the role of protein-tyrosine phosphatase α (PTPα) in regulating signaling by the ErbB2 oncoprotein in mammary epithelial cells. Using this model, we demonstrated that activation of ErbB2 led to the transient inactivation of PTPα, suggesting that attenuation of PTPα activity may contribute to enhanced ErbB2 signaling. Furthermore, RNAi-induced suppression of PTPα led to increased cell migration in an ErbB2-dependent manner. The ability of ErbB2 to increase cell motility in the absence of PTPα was characterized by prolonged interaction of GRB7 with ErbB2 and increased association of ErbB2 with a ß1-integrin-rich complex, which depended on GRB7-SH2 domain interactions. Finally, suppression of PTPα resulted in increased phosphorylation of focal adhesion kinase on Tyr-407, which induced the recruitment of vinculin and the formation of a novel focal adhesion kinase complex in response to ErbB2 activation in mammary epithelial cells. Collectively, these results reveal a new role for PTPα in the regulation of motility of mammary epithelial cells in response to ErbB2 activation.


Subject(s)
Cell Movement/physiology , Epithelial Cells/enzymology , Focal Adhesion Kinase 1/metabolism , Mammary Glands, Human/enzymology , Receptor, ErbB-2/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Cell Line , Epithelial Cells/cytology , Focal Adhesion Kinase 1/genetics , Humans , Male , Mammary Glands, Human/cytology , Phosphorylation/physiology , Receptor, ErbB-2/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Signal Transduction/physiology , Vinculin/genetics , Vinculin/metabolism
14.
Cancer Immunol Immunother ; 62(8): 1397-410, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23736951

ABSTRACT

Phage display is a powerful method for target discovery and selection of ligands for cancer treatment and diagnosis. Our goal was to select tumor-binding antibodies in cancer patients. Eligibility criteria included absence of preexisting anti-phage-antibodies and a Stage IV cancer status. All patients were intravenously administered 1 × 10(11) TUs/kg of an scFv library 1 to 4 h before surgical resection of their tumors. No significant adverse events related to the phage library infusion were observed. Phage were successfully recovered from all tumors. Individual clones from each patient were assessed for binding to the tumor from which clones were recovered. Multiple tumor-binding phage-antibodies were identified. Soluble scFv antibodies were produced from the phage clones showing higher tumor binding. The tumor-homing phage-antibodies and derived soluble scFvs were found to bind varying numbers (0-5) of 8 tested normal human tissues (breast, cervix, colon, kidney, liver, spleen, skin, and uterus). The clones that showed high tumor-specificity were found to bind corresponding tumors from other patients also. Clone enrichment was observed based on tumor binding and DNA sequence data. Clone sequences of multiple variable regions showed significant matches to certain cancer-related antibodies. One of the clones (07-2,355) that was found to share a 12-amino-acid-long motif with a reported IL-17A antibody was further studied for competitive binding for possible antigen target identification. We conclude that these outcomes support the safety and utility of phage display library panning in cancer patients for ligand selection and target discovery for cancer treatment and diagnosis.


Subject(s)
Antibodies, Neoplasm/immunology , Neoplasms/immunology , Peptide Library , Single-Chain Antibodies/immunology , Adult , Amino Acid Sequence , Antibodies, Neoplasm/genetics , Antibodies, Neoplasm/metabolism , Antibody Affinity/immunology , Antibody Specificity/immunology , Enzyme-Linked Immunosorbent Assay , Female , Fluorescent Antibody Technique , Follow-Up Studies , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Infusions, Intravenous , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Molecular Sequence Data , Neoplasm Staging , Neoplasms/genetics , Neoplasms/metabolism , Protein Binding/immunology , Sequence Homology, Amino Acid , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism
15.
J Mol Recognit ; 25(1): 57-67, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22213451

ABSTRACT

Src-homology (SH2) domains are an attractive target for the inhibition of specific signalling pathways but pose the challenge of developing a truly specific inhibitor. The G7-18NATE cyclic peptide is reported to specifically inhibit the growth factor receptor bound protein 7 (Grb7) adapter protein, implicated in the progression of several cancer types, via interactions with its SH2 domain. G7-18NATE effectively inhibits the interaction of Grb7 with ErbB3 and focal adhesion kinase in cell lysates and, with the addition of a cell permeability sequence, inhibits the growth and migration of a number of breast cancer cell lines. It is thus a promising lead in the development of therapeutics targeted to Grb7. Here we investigate the degree to which G7-18NATE is specific for the Grb7-SH2 domain compared with closely related SH2 domains including those of Grb10, Grb14, and Grb2 using surface plasmon resonance. We demonstrate that G7-18NATE binds with micromolar binding affinity to Grb7-SH2 domain (K(D) = 4-6 µm) compared with 50-200 times lower affinity for Grb10, Grb14, and Grb2 but that this specificity depends critically on the presence of phosphate in millimolar concentrations. Other differences in buffer composition, including use of Tris or 2-(N-Morpholino)ethanesulfonic acid or varying the pH, do not impact on the interaction. This suggests that under cellular conditions, G7-18NATE binds with highest affinity to Grb7. In addition, our findings demonstrate that the basis of specificity of G7-18NATE binding to the Grb7-SH2 domain is via other than intrinsic structural features of the protein, representing an unexpected mode of molecular recognition.


Subject(s)
GRB7 Adaptor Protein/antagonists & inhibitors , GRB7 Adaptor Protein/chemistry , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Phosphates/chemistry , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/chemistry , Amino Acid Sequence , Breast Neoplasms/drug therapy , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Female , GRB10 Adaptor Protein/antagonists & inhibitors , GRB10 Adaptor Protein/chemistry , GRB2 Adaptor Protein/antagonists & inhibitors , GRB2 Adaptor Protein/chemistry , Humans , Molecular Sequence Data , Phosphates/metabolism , Protein Binding , Sensitivity and Specificity , Surface Plasmon Resonance/methods , src Homology Domains
16.
Breast Cancer Res Treat ; 133(2): 607-15, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22005836

ABSTRACT

Triple-negative breast cancer (TNBC) is a heterogeneous disease that is usually associated with poor prognosis, and frequently associated with the basal-like breast cancer gene expression profile. There are no targeted therapeutic modalities for this disease, and no useful biomarkers. High GRB7 RNA expression levels are associated with an elevated risk of recurrence in patients with operable TNBC treated with standard adjuvant anthracycline and taxane therapy. To determine whether GRB7 is involved in the pathobiology of TNBC, we evaluated the biological effects of GRB7 inhibition in a panel of triple-negative cell lines-MDA-MB-468, MDA-MB-231, HCC70, and T4-2. We found GRB7 inhibition reduced cell motility and invasion of these cell lines and promoted cell death by apoptosis in 3D culture. These data suggest that GRB7 itself, or GRB7-dependent pathways, may prove to be important therapeutic targets in this disease.


Subject(s)
Breast Neoplasms/pathology , GRB7 Adaptor Protein/physiology , Apoptosis/drug effects , Apoptosis/genetics , Breast Neoplasms/genetics , Cell Culture Techniques , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Survival/genetics , Female , GRB7 Adaptor Protein/antagonists & inhibitors , GRB7 Adaptor Protein/genetics , Humans , Neoplasm Invasiveness/genetics , Neoplasms, Basal Cell/genetics , Neoplasms, Basal Cell/pathology , Peptides, Cyclic/pharmacology , Receptor, ErbB-2/deficiency , Receptors, Estrogen/deficiency , Receptors, Progesterone/deficiency , Reproducibility of Results
17.
Biotechniques ; 52(4)2012 Apr 01.
Article in English | MEDLINE | ID: mdl-26307247

ABSTRACT

Antibodies are important drugs for treating cancer and there is strong rationale for using multiple antibodies to improve outcomes. We labeled two breast cancer binding antibodies, anti-ErbB2 and anti-EpCAM, with infrared fluorescence dyes of different wavelengths and determined their in vivo distribution in a breast cancer xenograft model using a near-infrared (NIR) fluorescence imaging system. Our data show that these two antibodies can be readily assessed simultaneously in mouse xenograft model. This will help guide design of dosing strategies for multiple antibodies and identify potential interaction that could affect pharmacokinetics and possible side effects.


Subject(s)
Antibodies, Neoplasm/chemistry , Fluorescent Dyes/chemistry , Neoplasms/immunology , Neoplasms/pathology , Optical Imaging/methods , Animals , Antibodies, Neoplasm/immunology , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/immunology , Cell Line, Tumor , Epithelial Cell Adhesion Molecule , Humans , Mice , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/immunology
18.
J Mol Biol ; 412(3): 397-411, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21802427

ABSTRACT

Growth-receptor-bound protein (Grb)7 is an adapter protein aberrantly overexpressed, along with the erbB-2 receptor in breast cancer and in other cancers. Normally recruited to focal adhesions with a role in cell migration, it is associated with erbB-2 in cancer cells and is found to exacerbate cancer progression via stimulation of cell migration and proliferation. The G7-18NATE peptide (sequence: WFEGYDNTFPC cyclized via a thioether bond) is a nonphosphorylated peptide that was developed for the specific inhibition of Grb7 by blocking its SH2 domain. Cell-permeable versions of G7-18NATE are effective in the reduction of migration and proliferation in Grb7-overexpressing cells. It thus represents a promising starting point for the development of a therapeutic against Grb7. Here, we report the crystal structure of the G7-18NATE peptide in complex with the Grb7-SH2 domain, revealing the structural basis for its interaction. We also report further rounds of phage display that have identified G7-18NATE analogues with micromolar affinity for Grb7-SH2. These peptides retained amino acids F2, G4, and F9, as well as the YDN motif that the structural biology study showed to be the main residues in contact with the Grb7-SH2 domain. Isothermal titration calorimetry measurements reveal similar and better binding affinity of these peptides compared with G7-18NATE. Together, this study facilitates the optimization of second-generation inhibitors of Grb7.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , GRB7 Adaptor Protein/chemistry , GRB7 Adaptor Protein/metabolism , Peptides, Cyclic/chemistry , Peptides, Cyclic/metabolism , Amino Acid Sequence , Crystallography, X-Ray , GRB7 Adaptor Protein/antagonists & inhibitors , Humans , Kinetics , Models, Molecular , Molecular Sequence Data , Peptide Library , Peptides, Cyclic/isolation & purification , Protein Binding , Protein Structure, Quaternary , Sequence Alignment
19.
Biopolymers ; 96(2): 181-8, 2011.
Article in English | MEDLINE | ID: mdl-20564009

ABSTRACT

Grb7 is an adapter protein found to be overexpressed in several breast and other cancer cell types along with ErbB2. Grb7 is normally an interaction partner with focal adhesion kinase and in cancer cells also aberrantly interacts with ErbB2. It is thus implicated in the migratory and proliferative potential of cancer cells. Previous studies have shown that the phage display-derived cyclic nonphosphorylated inhibitor peptide, G7-18NATE, when linked to Penetratin, is able to interfere with the interaction of Grb7 with its upstream binding partners and to impact on both cell migration and proliferation. Here we report the synthesis of a biotinylated G7-18NATE covalently attached to just the last seven residues of Penetratin (G7-18NATE-P-Biotin). We demonstrate that this construct is taken up efficiently into MDA-MB-468 breast cancer cells and colocalizes with Grb7 in the cytoplasm. We also used isothermal titration calorimetry to determine the binding affinity of G7-18NATE-P-Biotin to the Grb7-SH2 domain, and showed that it binds with micromolar affinity (K(d) = 14.4 microM), similar to the affinity of G7-18NATE (K(d) = 35.4 microM). Together this shows that this shorter G7-18NATE-P-Biotin construct is suitable for further studies of the antiproliferative and antimigratory potential of this inhibitor.


Subject(s)
Cytoplasm/metabolism , GRB7 Adaptor Protein/metabolism , Peptides, Cyclic/pharmacokinetics , Cell Line, Tumor , Humans , Peptides, Cyclic/pharmacology , Protein Binding , src Homology Domains
20.
Biotechnol Prog ; 25(6): 1780-7, 2009.
Article in English | MEDLINE | ID: mdl-19899107

ABSTRACT

Recent evidence suggests that most malignancies are driven by "cancer stem cells" sharing the signature characteristics of adult stem cells: the ability to self renew and to differentiate. Furthermore these cells are thought to be quiescent, infrequently dividing cells with a natural resistance to chemotherapeutic agents. These studies theorize that therapies, which effectively treat the majority of tumor cells but 'miss' the stem cell population, will fail, while therapies directed at stern cells can potentially eradicate tumors. In breast cancer, researchers have isolated 'breast cancer stem cells' capable of recreating the tumor in vivo and in vitro. Generated new tumors contained both additional numbers of cancer stem cells and diverse mixed populations of cells present in the initial tumor, supporting the intriguing self-renewal and differentiation characteristics. In the present study, an antibody phage library has been used to search for phage displayed-single chain antibodies (scFv) with selective affinity to specific targets on breast cancer stem cells. We demonstrate evidence of two clones binding specifically to a cancer stem cell population isolated from the SUMl59 breast cancer cell line. These clones had selective affinity for cancer stem cells and they were able to select cancer stem cells among a large population of non-stem cancer cells in paraffin-embedded sections. The applicability of these clones to paraffin sections and frozen tissue specimens made them good candidates to be used as diagnostic and prognostic markers in breast cancer patient samples taking into consideration the cancer stern cell concept in tumor biology.


Subject(s)
Breast Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Peptide Library , Single-Chain Antibodies/metabolism , Xenograft Model Antitumor Assays/methods , Animals , Cell Adhesion , Cell Line, Tumor , Female , Flow Cytometry , Humans , Immunoglobulin Variable Region , Mice , Mice, Inbred NOD , Mice, SCID , Microscopy, Fluorescence , Single-Chain Antibodies/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...