1
|
Gupta MK, Vadde R. Delivery strategies of immunotherapies in the treatment of pancreatic cancer. IMMUNE LANDSCAPE OF PANCREATIC CANCER DEVELOPMENT AND DRUG RESISTANCE 2024:173-202. [DOI: 10.1016/b978-0-443-23523-8.00004-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
2
|
Röwer C, Olaleye OO, Bischoff R, Glocker MO. Mass Spectrometric ITEM-ONE and ITEM-TWO Analyses Confirm and Refine an Assembled Epitope of an Anti-Pertuzumab Affimer. Biomolecules 2023; 14:24. [PMID: 38254624 PMCID: PMC10813730 DOI: 10.3390/biom14010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Intact Transition Epitope Mapping-One-step Non-covalent force Exploitation (ITEM-ONE) analysis reveals an assembled epitope on the surface of Pertuzumab, which is recognized by the anti-Pertuzumab affimer 00557_709097. It encompasses amino acid residues NSGGSIYNQRFKGR, which are part of CDR2, as well as residues FTLSVDR, which are located on the variable region of Pertuzumab's heavy chain and together form a surface area of 1381.46 Å2. Despite not being part of Pertuzumab's CDR2, the partial sequence FTLSVDR marks a unique proteotypic Pertuzumab peptide. Binding between intact Pertuzumab and the anti-Pertuzumab affimer was further investigated using the Intact Transition Epitope Mapping-Thermodynamic Weak-force Order (ITEM-TWO) approach. Quantitative analysis of the complex dissociation reaction in the gas phase afforded a quasi-equilibrium constant (KD m0g#) of 3.07 × 10-12. The experimentally determined apparent enthalpy (ΔHm0g#) and apparent free energy (ΔGm0g#) of the complex dissociation reaction indicate that the opposite reaction-complex formation-is spontaneous at room temperature. Due to strong binding to Pertuzumab and because of recognizing Pertuzumab's unique partial amino acid sequences, the anti-Pertuzumab affimer 00557_709097 is considered excellently suitable for implementation in Pertuzumab quantitation assays as well as for the accurate therapeutic drug monitoring of Pertuzumab in biological fluids.
Collapse
Affiliation(s)
- Claudia Röwer
- Proteome Center Rostock, Medical Faculty and Natural Science Faculty, University of Rostock, 18057 Rostock, Germany
| | - Oladapo O. Olaleye
- Department of Analytical Biochemistry, Faculty of Science & Engineering, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Rainer Bischoff
- Department of Analytical Biochemistry, Faculty of Science & Engineering, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Michael O. Glocker
- Proteome Center Rostock, Medical Faculty and Natural Science Faculty, University of Rostock, 18057 Rostock, Germany
| |
Collapse
|
3
|
Ghafouri SR, Guvvala S, Jones C, Philipovskiy A. Recently approved treatment options for patients with metastatic triple-negative and HER2-neu-positive breast cancer. J Investig Med 2022; 70:1329-1341. [PMID: 35705261 DOI: 10.1136/jim-2021-002298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2022] [Indexed: 11/03/2022]
Abstract
Breast cancer (BC) is the most common cancer affecting women worldwide. In 2021, the estimated number of new breast cancer cases was 281 550 and about 43 500 women died from metastatic breast cancer (mBC). For women aged 20-59 years, mBC remains the leading cause of cancer death and is, therefore, an important public health concern. Only 5% of women initially present with metastatic disease. Approximately 20% of patients presenting with local or locoregional disease progress to mBC despite adjuvant therapy. Inspite of all the medicosurgical advancements, the overall prognosis for patients diagnosed with mBC remains poor, with median overall survival of approximately 31 months, although this varies based on tumor biology. In recent years, there has been significant progress in developing immunotargeted therapies such as antihuman epidermal growth factor receptor 2 (anti-HER2) or check point inhibitors that confirmed to have dramatically improve the prognosis of mBC, a historically unfavorable disease subset. Even with the major progress that has been made in understanding the biology of BC, challenges such as resistance frequency to therapies, unknown efficacy, concerns for safety of drug combination and toxicities still remain high. Therefore, a new targeted and more selective treatment approaches are the need of the hour. In this review, we aim to outline the most recently approved medications in treatment of Her2-positive and triple-negative breast cancers.
Collapse
Affiliation(s)
- Sayed Reshad Ghafouri
- Internal Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, USA
| | - Suvarna Guvvala
- Internal Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, Texas, USA
| | - Catherine Jones
- Texas Tech University Health Sciences Center School of Medicine, Lubbock, Texas, USA
| | | |
Collapse
|
4
|
Ziad A, Abdurahman A, Misako N. A comprehensive review on antibody-drug conjugates (ADCs) in the treatment landscape of non-small cell lung cancer (NSCLC). Cancer Treat Rev 2022; 106:102393. [DOI: 10.1016/j.ctrv.2022.102393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/02/2022] [Accepted: 04/10/2022] [Indexed: 11/28/2022]
|
5
|
Zhang S, Yan C, Millar DG, Yang Q, Heather JM, Langenbucher A, Morton LT, Sepulveda S, Alpert E, Whelton LR, Zarrella DT, Guo M, Minogue E, Lawrence MS, Rueda BR, Spriggs DR, Lu W, Langenau DM, Cobbold M. Antibody-Peptide Epitope Conjugates for Personalized Cancer Therapy. Cancer Res 2022; 82:773-784. [PMID: 34965933 DOI: 10.1158/0008-5472.can-21-2200] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 11/11/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022]
Abstract
Antibody-peptide epitope conjugates (APEC) are a new class of modified antibody-drug conjugates that redirect T-cell viral immunity against tumor cells. APECs contain a tumor-specific protease cleavage site linked to a patient-specific viral epitope, resulting in presentation of viral epitopes on cancer cells and subsequent recruitment and killing by CD8+ T cells. Here we developed an experimental pipeline to create patient-specific APECs and identified new preclinical therapies for ovarian carcinoma. Using functional assessment of viral peptide antigen responses to common viruses like cytomegalovirus (CMV) in patients with ovarian cancer, a library of 192 APECs with distinct protease cleavage sequences was created using the anti-epithelial cell adhesion molecule (EpCAM) antibody. Each APEC was tested for in vitro cancer cell killing, and top candidates were screened for killing xenograft tumors grown in zebrafish and mice. These preclinical modeling studies identified EpCAM-MMP7-CMV APEC (EpCAM-MC) as a potential new immunotherapy for ovarian carcinoma. Importantly, EpCAM-MC also demonstrated robust T-cell responses in primary ovarian carcinoma patient ascites samples. This work highlights a robust, customizable platform to rapidly develop patient-specific APECs. SIGNIFICANCE This study develops a high-throughput preclinical platform to identify patient-specific antibody-peptide epitope conjugates that target cancer cells and demonstrates the potential of this immunotherapy approach for treating ovarian carcinoma.
Collapse
Affiliation(s)
- Songfa Zhang
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases & Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Chuan Yan
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, Massachusetts
| | - David G Millar
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Qiqi Yang
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, Massachusetts
| | - James M Heather
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Adam Langenbucher
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | | | - Sean Sepulveda
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Eric Alpert
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, Massachusetts
| | - Lauren R Whelton
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, Massachusetts
| | - Dominique T Zarrella
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, Massachusetts
- Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, Massachusetts
| | - Mei Guo
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Eleanor Minogue
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Michael S Lawrence
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Bo R Rueda
- Vincent Center for Reproductive Biology, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, Massachusetts
- Obstetrics, Gynecology and Reproductive Biology, Harvard Medical School, Boston, Massachusetts
| | - David R Spriggs
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Weiguo Lu
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases & Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - David M Langenau
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
- Molecular Pathology Unit, Massachusetts General Hospital Research Institute, Charlestown, Massachusetts
| | - Mark Cobbold
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
- AstraZeneca, Gaithersburg, Maryland
| |
Collapse
|
6
|
Santos IDA, Grosche VR, Bergamini FRG, Sabino-Silva R, Jardim ACG. Antivirals Against Coronaviruses: Candidate Drugs for SARS-CoV-2 Treatment? Front Microbiol 2020; 11:1818. [PMID: 32903349 PMCID: PMC7438404 DOI: 10.3389/fmicb.2020.01818] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/10/2020] [Indexed: 01/08/2023] Open
Abstract
Coronaviruses (CoVs) are a group of viruses from the family Coronaviridae that can infect humans and animals, causing mild to severe diseases. The ongoing pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) represents a global threat, urging the development of new therapeutic strategies. Here we present a selection of relevant compounds that have been described from 2005 until now as having in vitro and/or in vivo antiviral activities against human and/or animal CoVs. We also present compounds that have reached clinical trials as well as further discussing the potentiality of other molecules for application in (re)emergent CoVs outbreaks. Finally, through rationalization of the data presented herein, we wish to encourage further research encompassing these compounds as potential SARS-CoV-2 drug candidates.
Collapse
Affiliation(s)
- Igor de Andrade Santos
- Laboratory of Virology, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
| | - Victória Riquena Grosche
- Laboratory of Virology, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
- Institute of Biosciences, Language and Exact Sciences, São Paulo State University, São José do Rio Preto, Brazil
| | | | - Robinson Sabino-Silva
- Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Ana Carolina Gomes Jardim
- Laboratory of Virology, Institute of Biomedical Science, Federal University of Uberlândia, Uberlândia, Brazil
- Institute of Biosciences, Language and Exact Sciences, São Paulo State University, São José do Rio Preto, Brazil
| |
Collapse
|
7
|
Goldenberg DM, Sharkey RM. Sacituzumab govitecan, a novel, third-generation, antibody-drug conjugate (ADC) for cancer therapy. Expert Opin Biol Ther 2020; 20:871-885. [PMID: 32301634 DOI: 10.1080/14712598.2020.1757067] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION We describe a new, third-generation of antibody-drug conjugates (ADCs) having a high drug payload against topoisomerase I, important for DNA function, and targeting selective tumor antigens, predominantly TROP-2. AREAS COVERED The historical development of ADCs is reviewed before presenting the current line of improved, third-generation ADCs targeting topoisomerase I, thus affecting DNA and causing double-stranded DNA breaks. Emphasis is given to explaining why sacituzumab govitecan represents a paradigm change in ADCs by achieving a high therapeutic index due to its novel target, TROP-2, an internalizing antigen/antibody, proprietary linker chemistry, and high drug payload, resulting in a high tumor concentration of the drug given in repeated doses with acceptable tolerability, particularly evidencing a lower percentage of 'late' diarrhea than its prodrug, irinotecan. PubMed was used for the primary search conducted. EXPERT OPINION The properties and clinical results of third-generation ADCs, based on sacituzumab govitecan, are discussed, including prospects for future applications, particularly combination therapies with PARP inhibitors and immune checkpoint inhibitors. Since one topoisomerase I ADC has just received regulatory approval for HER2+ breast cancer, and sacituzumab govitecan is under FDA review for accelerated approval in the therapy of triple-negative breast cancer, the prospects for these novel ADCs are discussed.
Collapse
Affiliation(s)
| | - Robert M Sharkey
- Center for Molecular Medicine and Immunology , Mendham, New Jersey, USA
| |
Collapse
|
8
|
Kommineni N, Pandi P, Chella N, Domb AJ, Khan W. Antibody drug conjugates: Development, characterization, and regulatory considerations. POLYM ADVAN TECHNOL 2019. [DOI: 10.1002/pat.4789] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Nagavendra Kommineni
- Department of PharmaceuticsNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad India
| | - Palpandi Pandi
- Department of PharmaceuticsNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad India
| | - Naveen Chella
- Department of PharmaceuticsNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad India
| | - Abraham J. Domb
- School of Pharmacy‐ Faculty of MedicineThe Hebrew University of Jerusalem Jerusalem Israel
| | - Wahid Khan
- Department of PharmaceuticsNational Institute of Pharmaceutical Education and Research (NIPER) Hyderabad India
| |
Collapse
|
9
|
Sheervalilou R, Shahraki O, Hasanifard L, Shirvaliloo M, Mehranfar S, Lotfi H, Pilehvar-Soltanahmadi Y, Bahmanpour Z, Zadeh SS, Nazarlou Z, Kangarlou H, Ghaznavi H, Zarghami N. Electrochemical Nano-biosensors as Novel Approach for the Detection of Lung Cancer-related MicroRNAs. Curr Mol Med 2019; 20:13-35. [DOI: 10.2174/1566524019666191001114941] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/22/2019] [Accepted: 09/03/2019] [Indexed: 12/12/2022]
Abstract
In both men and women around the world, lung cancer accounts as the
principal cause of cancer-related death after breast cancer. Therefore, early detection of
the disease is a cardinal step in improving prognosis and survival of patients. Today, the
newly-defined microRNAs regulate about 30 to 60 percent of the gene expression.
Changes in microRNA Profiles are linked to numerous health conditions, making them
sophisticated biomarkers for timely, if not early, detection of cancer. Though evaluation
of microRNAs in real samples has proved to be rather challenging, which is largely
attributable to the unique characteristics of these molecules. Short length, sequence
similarity, and low concentration stand among the factors that define microRNAs.
Recently, diagnostic technologies with a focus on wide-scale point of care have recently
garnered attention as great candidates for early diagnosis of cancer. Electrochemical
nano-biosensors have recently garnered much attention as a molecular method,
showing great potential in terms of sensitivity, specificity and reproducibility, and last but
not least, adaptability to point-of-care testing. Application of nanoscale materials in
electrochemical devices as promising as it is, brings multiplexing potential for conducting
simultaneous evaluations on multiple cancer biomarkers. Thanks to their enthralling
properties, these materials can be used to improve the efficiency of cancer diagnostics,
offer more accurate predictions of prognosis, and monitor response to therapy in a more
efficacious way. This article presents a concise overview of recent advances in the
expeditiously evolving area of electrochemical biosensors for microRNA detection in
lung cancer.
Collapse
Affiliation(s)
| | - Omolbanin Shahraki
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Leili Hasanifard
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Shirvaliloo
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Mehranfar
- Department of Genetics and Immunology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Hajie Lotfi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Younes Pilehvar-Soltanahmadi
- Cellular and Molecular Research Center, Research Institute for Cellular and Molecular Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Zahra Bahmanpour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sadaf Sarraf Zadeh
- Neurosciences Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Ziba Nazarlou
- Material Engineering Department, College of Science Koç University, Istanbul 34450, Turkey
| | - Haleh Kangarlou
- Department of Physics, Urmia Branch, Islamic Azad University, Urmia, Iran
| | - Habib Ghaznavi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Nosratollah Zarghami
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
10
|
Liu-Kreyche P, Shen H, Marino AM, Iyer RA, Humphreys WG, Lai Y. Lysosomal P-gp-MDR1 Confers Drug Resistance of Brentuximab Vedotin and Its Cytotoxic Payload Monomethyl Auristatin E in Tumor Cells. Front Pharmacol 2019; 10:749. [PMID: 31379564 PMCID: PMC6650582 DOI: 10.3389/fphar.2019.00749] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/11/2019] [Indexed: 12/31/2022] Open
Abstract
Antibody-drug conjugates (ADCs) are composed of an antibody linked to cytotoxic anticancer payloads. ADCs recognize tumor-specific cell surface antigens and are internalized into lysosomes where proteolytic enzymes release the cytotoxic payloads. Efflux transporters on plasma membrane that play a significant role on multi-drug resistance in chemotherapy can be internalized on lysosomal membrane and sequester the cytotoxic payloads. In the present study, ATP binding cassette (ABC) efflux transporters including breast cancer resistance protein (BCRP), P-glycoprotein (P-gp-MDR1), multidrug resistance protein (MRP) 2, MRP3 and MRP4 in lysosomal, and plasma membrane of tumor cells were quantified by targeted quantitative proteomics. The cytotoxicity of brentuximab vedotin and its cytotoxic payload monomethyl auristatin E (MMAE) to the tumor cell lines in the presence and absence of elacridar (P-gp-MDR1 inhibitor) or chloroquine (lysosomotropic agent) were evaluated. MMAE is a substrate for P-gp-MDR1, as the apparent efflux ratio in MDR1 transfected MDCK cell monolayers was 44.5, and elacridar abolished the MMAE efflux. Cell lines that highly express P-gp-MDR1 show higher EC50s toward the cell killing effects of MMAE. Co-incubation with chloroquine or elacridar resulted in left shift of MMAE EC50 by 2.9-16-fold and 4.2-22-fold, respectively. Similarly co-incubation with chloroquine or elacridar or in combination of chloroquine and elacridar increased cytotoxic effects of brentuximab vedotin by 2.8- to 21.4-fold on KM-H2 cells that express a specific tumor antigen CD30 and P-gp-MDR1. These findings demonstrate important roles of P-gp-MDR1 on cytotoxic effects of brentuximab vedotin and its payload MMAE. Collectively, ABC transporter-mediated drug extrusion and/or sequestration needs to be early assessed for selection of optimal payloads and linkers when developing ADCs.
Collapse
Affiliation(s)
- Peggy Liu-Kreyche
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Lawrenceville, NJ, United States
| | - Hong Shen
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Lawrenceville, NJ, United States
| | - Anthony M Marino
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Lawrenceville, NJ, United States
| | - Ramaswamy A Iyer
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Lawrenceville, NJ, United States
| | - W Griffith Humphreys
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Lawrenceville, NJ, United States
| | - Yurong Lai
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Lawrenceville, NJ, United States
| |
Collapse
|
11
|
Huang M, Zolnoori M, Balls-Berry JE, Brockman TA, Patten CA, Yao L. Technological Innovations in Disease Management: Text Mining US Patent Data From 1995 to 2017. J Med Internet Res 2019; 21:e13316. [PMID: 31038462 PMCID: PMC6611693 DOI: 10.2196/13316] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 04/12/2019] [Accepted: 04/13/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Patents are important intellectual property protecting technological innovations that inspire efficient research and development in biomedicine. The number of awarded patents serves as an important indicator of economic growth and technological innovation. Researchers have mined patents to characterize the focuses and trends of technological innovations in many fields. OBJECTIVE To expand patent mining to biomedicine and facilitate future resource allocation in biomedical research for the United States, we analyzed US patent documents to determine the focuses and trends of protected technological innovations across the entire disease landscape. METHODS We analyzed more than 5 million US patent documents between 1995 and 2017, using summary statistics and dynamic topic modeling. More specifically, we investigated the disease coverage and latent topics in patent documents over time. We also incorporated the patent data into the calculation of our recently developed Research Opportunity Index (ROI) and Public Health Index (PHI), to recalibrate the resource allocation in biomedical research. RESULTS Our analysis showed that protected technological innovations have been primarily focused on socioeconomically critical diseases such as "other cancers" (malignant neoplasm of head, face, neck, abdomen, pelvis, or limb; disseminated malignant neoplasm; Merkel cell carcinoma; and malignant neoplasm, malignant carcinoid tumors, neuroendocrine tumor, and carcinoma in situ of an unspecified site), diabetes mellitus, and obesity. The United States has significantly improved resource allocation to biomedical research and development over the past 17 years, as illustrated by the decreasing PHI. Diseases with positive ROI, such as ankle and foot fracture, indicate potential research opportunities for the future. Development of novel chemical or biological drugs and electrical devices for diagnosis and disease management is the dominating topic in patented inventions. CONCLUSIONS This multifaceted analysis of patent documents provides a deep understanding of the focuses and trends of technological innovations in disease management in patents. Our findings offer insights into future research and innovation opportunities and provide actionable information to facilitate policy makers, payers, and investors to make better evidence-based decisions regarding resource allocation in biomedicine.
Collapse
Affiliation(s)
- Ming Huang
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States
| | - Maryam Zolnoori
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States
| | - Joyce E Balls-Berry
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States
| | - Tabetha A Brockman
- Center for Clinical and Translational Science, Commuity Engagement Program, Mayo Clinic, Rochester, MN, United States.,Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
| | - Christi A Patten
- Center for Clinical and Translational Science, Commuity Engagement Program, Mayo Clinic, Rochester, MN, United States.,Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, United States
| | - Lixia Yao
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
12
|
Targeted human cytolytic fusion proteins at the cutting edge: harnessing the apoptosis-inducing properties of human enzymes for the selective elimination of tumor cells. Oncotarget 2019; 10:897-915. [PMID: 30783518 PMCID: PMC6368230 DOI: 10.18632/oncotarget.26618] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/10/2019] [Indexed: 01/01/2023] Open
Abstract
Patient-specific targeted therapy represents the holy grail of anti-cancer therapeutics, allowing potent tumor depletion without detrimental off-target toxicities. Disease-specific monoclonal antibodies have been employed to bind to oncogenic cell-surface receptors, representing the earliest form of immunotherapy. Targeted drug delivery was first achieved by means of antibody-drug conjugates, which exploit the differential expression of tumor-associated antigens as a guiding mechanism for the specific delivery of chemically-conjugated chemotherapeutic agents to diseased target cells. Biotechnological advances have expanded the repertoire of immunology-based tumor-targeting strategies, also paving the way for the next intuitive step in targeted drug delivery: the construction of recombinant protein drugs consisting of an antibody-based targeting domain genetically fused with a cytotoxic peptide, known as an immunotoxin. However, the most potent protein toxins have typically been derived from bacterial or plant virulence factors and commonly feature both off-target toxicity and immunogenicity in human patients. Further refinement of immunotoxin technology thus led to the replacement of monoclonal antibodies with humanized antibody derivatives, including the substitution of non-human toxic peptides with human cytolytic proteins. Preclinically tested human cytolytic fusion proteins (hCFPs) have proven promising as non-immunogenic combinatory anti-cancer agents, however they still require further enhancement to achieve convincing candidacy as a single-mode therapeutic. To date, a portfolio of highly potent human toxins has been established; ranging from microtubule-associated protein tau (MAP tau), RNases, granzyme B (GrB) and death-associated protein kinase (DAPk). In this review, we discuss the most recent findings on the use of these apoptosis-inducing hCFPs for the treatment of various cancers.
Collapse
|
13
|
Ismael MA, Khan JM, Malik A, Alsenaidy MA, Hidayathulla S, Khan RH, Sen P, Irfan M, Alsenaidy AM. Unraveling the molecular mechanism of the effects of sodium dodecyl sulfate, salts, and sugars on amyloid fibril formation in camel IgG. Colloids Surf B Biointerfaces 2018; 170:430-437. [DOI: 10.1016/j.colsurfb.2018.06.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 05/14/2018] [Accepted: 06/17/2018] [Indexed: 10/28/2022]
|
14
|
Frontera ED, Khansa RM, Schalk DL, Leakan LE, Guerin-Edbauer TJ, Ratnam M, Gorski DH, Speyer CL. IgA Fc-folate conjugate activates and recruits neutrophils to directly target triple-negative breast cancer cells. Breast Cancer Res Treat 2018; 172:551-560. [PMID: 30155754 DOI: 10.1007/s10549-018-4941-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/25/2018] [Indexed: 10/28/2022]
Abstract
PURPOSE According to the American Cancer Society, 1 in 8 women in the U.S. will develop breast cancer, with triple-negative breast cancer (TNBC) comprising 15-20% of all breast cancer cases. TNBC is an aggressive subtype due to its high metastatic potential and lack of targeted therapy. Recently, folate receptor alpha (FRA) is found to be expressed on 80% of TNBC with high expression correlating with poor prognosis. In this study, we examined whether binding IgA Fc-folate molecules to FRA receptors on TNBC cells can elicit and induce neutrophils (PMNs), by binding their FcαR1 receptors, to destroy TNBC cells. METHODS FRA was analyzed on TNBC cells and binding assays were performed using 3H-folate. Fc-folate was synthesized by linking Fc fragments of IgA via amine groups to folate. Binding specificity and antibody-dependent cellular cytotoxicity (ADCC) potential of Fc-folate to FcαR1 were confirmed by measuring PMN adhesion and myeloperoxidase (MPO) release in a cell-based ELISA. Fc-folate binding to FRA-expressing TNBC cells inducing PMNs to destroy these cells was determined using 51Cr-release and calcein-labeling assays. RESULTS Our results demonstrate expression of FRA on TNBC cells at levels consistent with folate binding. Fc-folate binds with high affinity to FRA compared to whole IgA-folate and induces MPO release from PMN when bound to FcαR1. Fc-folate inhibited binding of 3H-folate to TNBC cells and induced significant cell lysis of TNBC cells when incubated in the presence of PMNs. CONCLUSION These findings support the hypothesis that an IgA Fc-folate conjugate can destroy TNBC cells by eliciting PMN-mediated ADCC.
Collapse
Affiliation(s)
- Eric D Frontera
- College of Osteopathic Medicine, Michigan State University, 4707 St. Antoine, Box 402, Detroit, MI, 48201, USA
| | - Rafa M Khansa
- College of Osteopathic Medicine, Michigan State University, 4707 St. Antoine, Box 402, Detroit, MI, 48201, USA
| | - Dana L Schalk
- Department of Hematology/Oncology, University of Virginia Cancer Center, 1300 Jefferson Park Ave., I MSB 7191 West Complex I, Charlottesville, VA, 22903, USA
| | - Lauren E Leakan
- Wayne State University, 42 West Warren Avenue, Detroit, MI, 48202, USA
| | - Tracey J Guerin-Edbauer
- Michael and Marian Ilitch Department of Surgery, Wayne State University School of Medicine, 4100 John R St., Mailcode HW08AO, Detroit, MI, 48201, USA
| | - Manohar Ratnam
- Department of Oncology, Wayne State University School of Medicine, 4100 John R St., Detroit, MI, 48201, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, 4100 John R St., Detroit, MI, 48201, USA
| | - David H Gorski
- Michael and Marian Ilitch Department of Surgery, Wayne State University School of Medicine, 4100 John R St., Mailcode HW08AO, Detroit, MI, 48201, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, 4100 John R St., Detroit, MI, 48201, USA
| | - Cecilia L Speyer
- Michael and Marian Ilitch Department of Surgery, Wayne State University School of Medicine, 4100 John R St., Mailcode HW08AO, Detroit, MI, 48201, USA. .,Tumor Microenvironment Program, Barbara Ann Karmanos Cancer Institute, 4100 John R St., Detroit, MI, 48201, USA.
| |
Collapse
|
15
|
Patel J, Amrutiya J, Bhatt P, Javia A, Jain M, Misra A. Targeted delivery of monoclonal antibody conjugated docetaxel loaded PLGA nanoparticles into EGFR overexpressed lung tumour cells. J Microencapsul 2018. [DOI: 10.1080/02652048.2018.1453560] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
| | - Jitendra Amrutiya
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Priyanka Bhatt
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Ankit Javia
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Mukul Jain
- Zydus Research Centre, Cadila Healthcare Limited, Ahmedabad, India
| | - Ambikanandan Misra
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, India
| |
Collapse
|
16
|
Abotaleb M, Kubatka P, Caprnda M, Varghese E, Zolakova B, Zubor P, Opatrilova R, Kruzliak P, Stefanicka P, Büsselberg D. Chemotherapeutic agents for the treatment of metastatic breast cancer: An update. Biomed Pharmacother 2018; 101:458-477. [PMID: 29501768 DOI: 10.1016/j.biopha.2018.02.108] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 02/22/2018] [Accepted: 02/23/2018] [Indexed: 12/17/2022] Open
Abstract
Breast cancer is the second greatest cause of death among women worldwide; it comprises a group of heterogeneous diseases that evolves due to uncontrolled cellular growth and differentiation and the loss of normal programmed cell death. There are different molecular sub-types of breast cancer; therefore, various options are selected for treatment of different forms of metastatic breast cancer. However, the use of chemotherapeutic drugs is usually accompanied by deleterious side effects and the development of drug resistance when applied for a longer period. This review offers a classification of these chemotherapeutic agents according to their modes of action and therefore improves the understanding of molecular targets that are affected during treatment. Overall, it will allow the clinician to identify more specific targets to increase the effectiveness of a drug and to reduce general toxicity, resistance and other side effects.
Collapse
Affiliation(s)
- Mariam Abotaleb
- Weill Cornell Medicine in Qatar, Qatar Foundation-Education City, Doha, Qatar
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia; Department of Experimental Carcinogenesis, Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Martin Caprnda
- 1st Department of Internal Medicine, Medical Faculty, Comenius University in Bratislava, Bratislava, Slovakia
| | - Elizabeth Varghese
- Weill Cornell Medicine in Qatar, Qatar Foundation-Education City, Doha, Qatar
| | - Barbora Zolakova
- Department of Experimental Carcinogenesis, Division of Oncology, Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Pavol Zubor
- Clinic of Gynecology and Obsterics, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovakia
| | - Radka Opatrilova
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| | - Peter Kruzliak
- Department of Internal Medicine, Brothers of Mercy Hospital, Brno, Czech Republic; 2nd Department of Surgery, Faculty of Medicine, Masaryk University and St. Anne´s University Hospital, Brno, Czech Republic.
| | - Patrik Stefanicka
- Department of Otorhinolaryngology, Head and Neck Surgery, Faculty of Medicine, Comenius University and University Hospital, Antolska 11, 851 07, Bratislava, Slovakia.
| | - Dietrich Büsselberg
- Weill Cornell Medicine in Qatar, Qatar Foundation-Education City, Doha, Qatar.
| |
Collapse
|
17
|
Ma YV, Lam C, Dalmia S, Gao P, Young J, Middleton K, Liu C, Xu H, You L. Mechanical regulation of breast cancer migration and apoptosis via direct and indirect osteocyte signaling. J Cell Biochem 2018; 119:5665-5675. [DOI: 10.1002/jcb.26745] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 01/29/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Yu‐Heng V. Ma
- Institute of Biomaterials and Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
| | - Candy Lam
- Institute of Biomaterials and Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
| | - Shreyash Dalmia
- Institute of Biomaterials and Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
| | - Peter Gao
- Institute of Biomaterials and Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
| | - Jacob Young
- Institute of Biomaterials and Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
| | - Kevin Middleton
- Institute of Biomaterials and Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
| | - Chao Liu
- Institute of Biomaterials and Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
| | - Henry Xu
- Institute of Biomaterials and Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
| | - Lidan You
- Institute of Biomaterials and Biomedical EngineeringUniversity of TorontoTorontoOntarioCanada
- Department of Mechanical and Industrial EngineeringUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
18
|
Medication-associated gastrointestinal tract injury. Virchows Arch 2017; 470:245-266. [PMID: 28133700 DOI: 10.1007/s00428-017-2077-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 11/05/2016] [Accepted: 01/19/2017] [Indexed: 12/18/2022]
Abstract
Medication-associated gastrointestinal (GI) tract injury has been known for centuries for some medications. The more recently introduced biologicals are a class of drugs that constantly increases, and as such, the spectrum of GI tract side effects is steadily growing. This review covers not only long-known GI tract side effects of drugs but also those more recently described. A comprehensive but concise list of medications used in daily practice and associated with GI tract injury is presented.
Collapse
|
19
|
Kurogochi M, Mori M, Osumi K, Tojino M, Sugawara SI, Takashima S, Hirose Y, Tsukimura W, Mizuno M, Amano J, Matsuda A, Tomita M, Takayanagi A, Shoda SI, Shirai T. Glycoengineered Monoclonal Antibodies with Homogeneous Glycan (M3, G0, G2, and A2) Using a Chemoenzymatic Approach Have Different Affinities for FcγRIIIa and Variable Antibody-Dependent Cellular Cytotoxicity Activities. PLoS One 2015. [PMID: 26200113 PMCID: PMC4511734 DOI: 10.1371/journal.pone.0132848] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Many therapeutic antibodies have been developed, and IgG antibodies have been extensively generated in various cell expression systems. IgG antibodies contain N-glycans at the constant region of the heavy chain (Fc domain), and their N-glycosylation patterns differ during various processes or among cell expression systems. The Fc N-glycan can modulate the effector functions of IgG antibodies, such as antibody-dependent cellular cytotoxicity (ADCC) and complement dependent cytotoxicity (CDC). To control Fc N-glycans, we performed a rearrangement of Fc N-glycans from a heterogeneous N-glycosylation pattern to homogeneous N-glycans using chemoenzymatic approaches with two types of endo-β-N-acetyl glucosaminidases (ENG'ases), one that works as a hydrolase to cleave all heterogeneous N-glycans, another that is used as a glycosynthase to generate homogeneous N-glycans. As starting materials, we used an anti-Her2 antibody produced in transgenic silkworm cocoon, which consists of non-fucosylated pauci-mannose type (Man2-3GlcNAc2), high-mannose type (Man4-9GlcNAc2), and complex type (Man3GlcNAc3-4) N-glycans. As a result of the cleavage of several ENG'ases (endoS, endoM, endoD, endoH, and endoLL), the heterogeneous glycans on antibodies were fully transformed into homogeneous-GlcNAc by a combination of endoS, endoD, and endoLL. Next, the desired N-glycans (M3; Man3GlcNAc1, G0; GlcNAc2Man3GlcNAc1, G2; Gal2GlcNAc2Man3GlcNAc1, A2; NeuAc2Gal2GlcNAc2Man3GlcNAc1) were transferred from the corresponding oxazolines to the GlcNAc residue on the intact anti-Her2 antibody with an ENG'ase mutant (endoS-D233Q), and the glycoengineered anti-Her2 antibody was obtained. The binding assay of anti-Her2 antibody with homogenous N-glycans with FcγRIIIa-V158 showed that the glycoform influenced the affinity for FcγRIIIa-V158. In addition, the ADCC assay for the glycoengineered anti-Her2 antibody (mAb-M3, mAb-G0, mAb-G2, and mAb-A2) was performed using SKBR-3 and BT-474 as target cells, and revealed that the glycoform influenced ADCC activity.
Collapse
Affiliation(s)
- Masaki Kurogochi
- Laboratory of Glycobiology, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Masako Mori
- Laboratory of Glyco-Bioengineering, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Kenji Osumi
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Mami Tojino
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Shu-ichi Sugawara
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Shou Takashima
- Laboratory of Glycobiology, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Yuriko Hirose
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Wataru Tsukimura
- Laboratory of Glyco-Bioengineering, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Mamoru Mizuno
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Junko Amano
- Laboratory of Glycobiology, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Akio Matsuda
- Laboratory of Glyco-Bioengineering, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
| | - Masahiro Tomita
- Immuno-Biological Laboratories Co., Ltd., 1091-1 Naka, Fujioka-shi, Gunma, Japan
| | - Atsushi Takayanagi
- Department of Molecular Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Shin-Ichiro Shoda
- Graduate School of Engineering, Tohoku University, Aoba-ku, Sendai, Japan
| | - Takashi Shirai
- Laboratory of Glycobiology, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
- Laboratory of Glyco-Bioengineering, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
- Laboratory of Glyco-organic Chemistry, The Noguchi Institute, 1-8-1 Kaga, Itabashi-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
20
|
Peters C, Brown S. Antibody-drug conjugates as novel anti-cancer chemotherapeutics. Biosci Rep 2015; 35:e00225. [PMID: 26182432 PMCID: PMC4613712 DOI: 10.1042/bsr20150089] [Citation(s) in RCA: 316] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/18/2015] [Accepted: 05/29/2015] [Indexed: 12/19/2022] Open
Abstract
Over the past couple of decades, antibody-drug conjugates (ADCs) have revolutionized the field of cancer chemotherapy. Unlike conventional treatments that damage healthy tissues upon dose escalation, ADCs utilize monoclonal antibodies (mAbs) to specifically bind tumour-associated target antigens and deliver a highly potent cytotoxic agent. The synergistic combination of mAbs conjugated to small-molecule chemotherapeutics, via a stable linker, has given rise to an extremely efficacious class of anti-cancer drugs with an already large and rapidly growing clinical pipeline. The primary objective of this paper is to review current knowledge and latest developments in the field of ADCs. Upon intravenous administration, ADCs bind to their target antigens and are internalized through receptor-mediated endocytosis. This facilitates the subsequent release of the cytotoxin, which eventually leads to apoptotic cell death of the cancer cell. The three components of ADCs (mAb, linker and cytotoxin) affect the efficacy and toxicity of the conjugate. Optimizing each one, while enhancing the functionality of the ADC as a whole, has been one of the major considerations of ADC design and development. In addition to these, the choice of clinically relevant targets and the position and number of linkages have also been the key determinants of ADC efficacy. The only marketed ADCs, brentuximab vedotin and trastuzumab emtansine (T-DM1), have demonstrated their use against both haematological and solid malignancies respectively. The success of future ADCs relies on improving target selection, increasing cytotoxin potency, developing innovative linkers and overcoming drug resistance. As more research is conducted to tackle these issues, ADCs are likely to become part of the future of targeted cancer therapeutics.
Collapse
Affiliation(s)
- Christina Peters
- School of Life Sciences, Nottingham Medical School, Queen's Medical Centre, Nottingham NG7 2UH, U.K
| | - Stuart Brown
- School of Life Sciences, Nottingham Medical School, Queen's Medical Centre, Nottingham NG7 2UH, U.K.
| |
Collapse
|
21
|
Respaud R, Vecellio L, Diot P, Heuzé-Vourc’h N. Nebulization as a delivery method for mAbs in respiratory diseases. Expert Opin Drug Deliv 2015; 12:1027-39. [DOI: 10.1517/17425247.2015.999039] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
22
|
Atreya I, Neurath MF. Immune cells in colorectal cancer: prognostic relevance and therapeutic strategies. Expert Rev Anticancer Ther 2014; 8:561-72. [DOI: 10.1586/14737140.8.4.561] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
23
|
Laheru D, Biedrzycki B, Jaffee EM. Development of a cytokine-modified allogeneic whole cell pancreatic cancer vaccine. Methods Mol Biol 2013; 980:175-203. [PMID: 23359154 DOI: 10.1007/978-1-62703-287-2_9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Management of patients with pancreatic cancer is a multidisciplinary approach that presents enormous challenges to the clinician. Overall 5-year survival for all patients remains <3%. Symptoms of early pancreas cancer are nonspecific. As such, only a fraction of patients are candidates for surgery. While surgical resection provides the only curative option, most patients will develop tumor recurrence and die of their disease. To date, the clinical benefits of chemotherapy and radiation therapy have been important but have led to modest improvements. Tumor vaccines have the potential to specifically target the needle of pancreas cancer cells amidst the haystack of normal tissue. The discovery of pancreas tumor-specific antigens and the subsequent ability to harness this technology has become an area of intense interest for tumor immunologists and clinicians alike. Without knowledge of specific antigen targets, the whole tumor cell represents the best source of immunizing antigens. This chapter will focus on the development of whole tumor cell vaccine strategies for pancreas cancer.
Collapse
Affiliation(s)
- Dan Laheru
- The Sidney Kimmel Cancer Center, the Skip Viragh Clinical Pancreatic Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| | | | | |
Collapse
|
24
|
Sabbatino F, Ferrone S. Can the “Right” EGFR-Specific mAb Dramatically Improve EGFR-Targeted Therapy? Clin Cancer Res 2013; 19:958-60. [DOI: 10.1158/1078-0432.ccr-12-3586] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
25
|
Gerdes CA, Nicolini VG, Herter S, van Puijenbroek E, Lang S, Roemmele M, Moessner E, Freytag O, Friess T, Ries CH, Bossenmaier B, Mueller HJ, Umaña P. GA201 (RG7160): A Novel, Humanized, Glycoengineered Anti-EGFR Antibody with Enhanced ADCC and Superior In Vivo Efficacy Compared with Cetuximab. Clin Cancer Res 2012. [DOI: 10.1158/1078-0432.ccr-12-0989] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
26
|
Wang YQ, Zhang HH, Liu CL, Xia Q, Wu H, Yu XH, Kong W. Correlation Between Auto-antibodies to Survivin and MUC1 Variable Number Tandem Repeats in Colorectal Cancer. Asian Pac J Cancer Prev 2012; 13:5557-62. [DOI: 10.7314/apjcp.2012.13.11.5557] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
27
|
Teo PZ, Utz PJ, Mollick JA. Using the allergic immune system to target cancer: activity of IgE antibodies specific for human CD20 and MUC1. Cancer Immunol Immunother 2012; 61:2295-309. [PMID: 22692757 DOI: 10.1007/s00262-012-1299-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 05/29/2012] [Indexed: 01/21/2023]
Abstract
Monoclonal antibodies are widely used in the treatment of many B cell lymphomas and certain solid tumors. All currently approved therapeutic monoclonal antibodies are of the immunoglobulin G (IgG) isotype. We hypothesized that tumor-specific monoclonal antibodies of the IgE isotype may serve as effective cancer therapeutics. To test this hypothesis, we produced mouse-human chimeric IgE antibodies specific for the human B cell antigen CD20 and the epithelial antigen MUC1. We demonstrate here that anti-hCD20 IgE antibodies have in vitro cytotoxic activity when used with purified allergic effector cells derived from umbilical cord blood. At an effector-tumor ratio of 2:1, mast cells and tumor-specific IgE induced a 2.5-fold increase in tumor cell death, as compared to control IgE. Similar results were observed when eosinophils were used as effector cells. In an in vivo murine model of breast carcinoma, administration of anti-hMUC1 IgE reduced the growth of MUC1(+) tumors by 25-30 % in hFcεRI transgenic mice. In contrast, local production of IgE and cytokines chemotactic for macrophages, eosinophils and mast cells led to complete tumor eradication. These results suggest that allergic effector cells activated by IgE and cell surface antigens have the capacity to induce tumor cell death in vitro and in vivo. The use of chimeric antibodies and hFcεRI transgenic mice will greatly enhance investigations in the nascent field of allergo-oncology.
Collapse
Affiliation(s)
- Pearline Zhaoying Teo
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | | | | |
Collapse
|
28
|
Harms BD, Kearns JD, Su SV, Kohli N, Nielsen UB, Schoeberl B. Optimizing properties of antireceptor antibodies using kinetic computational models and experiments. Methods Enzymol 2012; 502:67-87. [PMID: 22208982 DOI: 10.1016/b978-0-12-416039-2.00004-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Monoclonal antibodies are valuable as anticancer therapeutics because of their ability to selectively bind tumor-associated target proteins like receptor tyrosine kinases. Kinetic computational models that capture protein-protein interactions using mass action kinetics are a valuable tool for understanding the binding properties of monoclonal antibodies to their targets. Insights from the models can be used to explore different formats, to set antibody design specifications such as affinity and valence, and to predict potency. Antibody binding to target is driven by both intrinsic monovalent affinity and bivalent avidity. In this chapter, we describe a combined experimental and computational method of assessing the relative importance of these effects on observed drug potency. The method, which we call virtual flow cytometry (VFC), merges experimental measurements of monovalent antibody binding kinetics and affinity curves of antibody-antigen binding into a kinetic computational model of antibody-antigen interaction. The VFC method introduces a parameter χ, the avidity factor, which characterizes the ability of an antibody to cross-link its target through bivalent binding. This simple parameterization of antibody cross-linking allows the model to successfully describe and predict antibody binding curves across a wide variety of experimental conditions, including variations in target expression level and incubation time of antibody with target. We further demonstrate how computational models of antibody binding to cells can be used to predict target inhibition potency. Importantly, we demonstrate computationally that antibodies with high ability to cross-link antigen have significant potency advantages. We also present data suggesting that the parameter χ is a physical, epitope-dependent property of an antibody, and as a result propose that determination of antibody cross-linking and avidity should be incorporated into the screening of antibody panels for therapeutic development. Overall, our results suggest that antibody cross-linking, in addition to monovalent binding affinity, is a key design parameter of antibody performance.
Collapse
Affiliation(s)
- Brian D Harms
- Merrimack Pharmaceuticals, Cambridge, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
29
|
Zheng X, Cheung LSL, Schroeder JA, Jiang L, Zohar Y. A high-performance microsystem for isolating circulating tumor cells. LAB ON A CHIP 2011; 11:3269-76. [PMID: 21837324 PMCID: PMC6765387 DOI: 10.1039/c1lc20331b] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
A unique flow field pattern in a bio-functional microchannel is utilized to significantly enhance the performance of a microsystem developed for selectively isolating circulating tumor cells from cell suspensions. For high performance of such systems, disposal of maximum non-target species is just as important as retention of maximum target species; unfortunately, most studies ignore or fail to report this aspect. Therefore, sensitivity and specificity are introduced as quantitative criteria to evaluate the system performance enabling a direct comparison among systems employing different techniques. The newly proposed fluidic scheme combines a slow flow field, for maximum target-cell attachment, followed by a faster flow field, for maximum detachment of non-target cells. Suspensions of homogeneous or binary mixtures of circulating breast tumor cells, with varying relative concentrations, were driven through antibody-functionalized microchannels. Either EpCAM or cadherin-11 transmembrane receptors were targeted to selectively capture target cells from the suspensions. Cadherin-11-expressing MDA-MB-231 cancer cells were used as target cells, while BT-20 cells were used as non-target cells as they do not express cadherin-11. The attachment and detachment of these two cell lines are characterized, and a two-step attachment/detachment flow field pattern is implemented to enhance the system performance in capturing target cells from binary mixtures. While the system sensitivity remains high, above 0.95, the specificity increases from about 0.85 to 0.95 solely due to the second detachment step even for a 1 : 1000 relative concentration of the target cells.
Collapse
Affiliation(s)
- Xiangjun Zheng
- Department of Aerospace and Mechanical Engineering, the University of Arizona, Tucson, AZ, USA
| | - Luthur Siu Lun Cheung
- Department of Aerospace and Mechanical Engineering, the University of Arizona, Tucson, AZ, USA
| | - Joyce A. Schroeder
- Department of Molecular and Cellular Biology, the University of Arizona, Tucson, AZ, USA
- Arizona Cancer Center, the University of Arizona, Tucson, AZ, USA
- BIO5 Innstitute, the University of Arizona, Tucson, AZ, USA
| | - Linan Jiang
- Department of Aerospace and Mechanical Engineering, the University of Arizona, Tucson, AZ, USA
- College of Optical Science, the University of Arizona, Tucson, AZ, USA
| | - Yitshak Zohar
- Department of Aerospace and Mechanical Engineering, the University of Arizona, Tucson, AZ, USA
- Arizona Cancer Center, the University of Arizona, Tucson, AZ, USA
- BIO5 Innstitute, the University of Arizona, Tucson, AZ, USA
- Department of Biomedical Engineering, the University of Arizona, Tucson, AZ, USA
| |
Collapse
|
30
|
Siddiqui MZ. Monoclonal antibodies as diagnostics; an appraisal. Indian J Pharm Sci 2011; 72:12-7. [PMID: 20582184 PMCID: PMC2883214 DOI: 10.4103/0250-474x.62229] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2009] [Revised: 11/16/2009] [Accepted: 12/27/2009] [Indexed: 11/04/2022] Open
Abstract
Ever since the development of Hybridoma Technology in 1975 by Kohler and Milstein, our vision for antibodies as tools for research for prevention, detection and treatment of diseases, vaccine production, antigenic characterization of pathogens and in the study of genetic regulation of immune responses and disease susceptibility has been revolutionized. The monoclonal antibodies being directed against single epitopes are homogeneous, highly specific and can be produced in unlimited quantities. In animal disease diagnosis, they are very useful for identification and antigenic characterization of pathogens. Monoclonal antibodies have tremendous applications in the field of diagnostics, therapeutics and targeted drug delivery systems, not only for infectious diseases caused by bacteria, viruses and protozoa but also for cancer, metabolic and hormonal disorders. They are also used in the diagnosis of lymphoid and myeloid malignancies, tissue typing, enzyme linked immunosorbent assay, radio immunoassay, serotyping of microorganisms, immunological intervention with passive antibody, antiidiotype inhibition, or magic bullet therapy with cytotoxic agents coupled with anti mouse specific antibody. Recombinant deoxyribonucleic acid technology through genetic engineering has successfully led to the possibility of reconstruction of monoclonal antibodies viz. chimeric antibodies, humanized antibodies and complementarily determining region grafted antibodies and their enormous therapeutic use.
Collapse
Affiliation(s)
- M Z Siddiqui
- Processing and Product Development Division, Indian Institute of Natural Resins & Gums, Namkum, Ranch-834 010, India
| |
Collapse
|
31
|
Abstract
Cancer immunotherapy aims to establish immune-mediated control of tumor growth by priming T-cell responses to target tumor-associated antigens. Three signals are required for T-cell activation: (i) presentation of cognate antigen in self MHC molecules; (ii) costimulation by membrane-bound receptor-ligand pairs; and (iii) soluble factors to direct polarization of the ensuing immune response. The ability of dendritic cells (DCs) to provide all three signals required for T-cell activation makes them an ideal cancer vaccine platform. Several strategies have been developed to enhance and control antigen presentation, costimulation, and cytokine production. In this review, we discuss progress toward developing DC-based cancer vaccines by genetic modification using RNA, DNA, and recombinant viruses. Furthermore, the ability of DC-based vaccines to activate natural killer (NK) and B-cells, and the impact of gene modification strategies on these populations is described. Clinical trials using gene-modified DCs have shown modest results, therefore, further considerations for DC manipulation to enhance their clinical efficacy are also discussed.
Collapse
|
32
|
Nagane M, Shimizu S, Mori E, Kataoka S, Shiokawa Y. Predominant antitumor effects by fully human anti-TRAIL-receptor 2 (DR5) monoclonal antibodies in human glioma cells in vitro and in vivo. Neuro Oncol 2010; 12:687-700. [PMID: 20511188 PMCID: PMC2940669 DOI: 10.1093/neuonc/nop069] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Accepted: 10/29/2009] [Indexed: 11/12/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL/Apo2 L) preferentially induces apoptosis in human tumor cells through its cognate death receptors DR4 or DR5, thereby being investigated as a potential agent for cancer therapy. Here, we applied fully human anti-human TRAIL receptor monoclonal antibodies (mAbs) to specifically target one of death receptors for TRAIL in human glioma cells, which could also reduce potential TRAIL-induced toxicity in humans. Twelve human glioma cell lines treated with several fully human anti-human TRAIL receptor mAbs were sensitive to only anti-DR5 mAbs, whereas they were totally insensitive to anti-DR4 mAb. Treatment with anti-DR5 mAbs exerted rapid cytotoxicity and lead to apoptosis induction. The cellular sensitivity was closely associated with cell-surface expression of DR5. Expression of c-FLIP(L), Akt, and Cyclin D1 significantly correlated with sensitivity to anti-DR5 mAbs. Primary cultures of glioma cells were also relatively resistant to anti-DR5 mAbs, exhibiting both lower DR5 and higher c-FLIP(L) expression. Downregulation of c-FLIP(L) expression resulted in the sensitization of human glioma cells to anti-DR5 mAbs, whereas overexpression of c-FLIP(L) conferred resistance to anti-DR5 mAb. Treatment of tumor-burden nude mice with the direct agonist anti-DR5 mAb KMTR2 significantly suppressed growth of subcutaneous glioma xenografts leading to complete regression. Similarly, treatment of nude mice bearing intracerebral glioma xenografts with KMTR2 significantly elongated lifespan without tumor recurrence. These results suggest that DR5 is the predominant TRAIL receptor mediating apoptotic signals in human glioma cells, and sensitivity to anti-DR5 mAbs was determined at least in part by the expression level of c-FLIP(L) and Akt. Specific targeting of death receptor pathway through DR5 using fully human mAbs might provide a novel therapeutic strategy for intractable malignant gliomas.
Collapse
Affiliation(s)
- Motoo Nagane
- Department of Neurosurgery, Kyorin University Faculty of Medicine, Mitaka, Tokyo 181-8611, Japan.
| | | | | | | | | |
Collapse
|
33
|
Intelligent Polymeric Nanocarriers Responding to Physical or Biological Signals: A New Paradigm of Cytosolic Drug Delivery for Tumor Treatment. Polymers (Basel) 2010. [DOI: 10.3390/polym2020086] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
34
|
Li J, Weng X, Liang Z, Zhong M, Chen X, Lu S, Sun W, Song Y, Wu X, Shen G. Viral specific cytotoxic T cells inhibit the growth of TfR-expressing tumor cells with antibody targeted viral peptide/HLA-A2 complex. Cell Immunol 2010; 263:154-60. [PMID: 20406704 DOI: 10.1016/j.cellimm.2010.03.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Revised: 02/25/2010] [Accepted: 03/09/2010] [Indexed: 11/26/2022]
Abstract
A fusion protein of single chain antibody (scFv) specific for transferrin receptor (TfR, CD71) and viral peptide/HLA-A2 complex was prepared in this study to redirect cytotoxic T cells (CTLs) of viral specificity to tumor cells by attaching the ligand of T cell receptor (TCR) to tumor cells via binding of TfR scFv to TfR. The results demonstrate that the fusion protein can attach the active virus-peptide/HLA-A2 complex to HLA class I-negative, TfR-expressing K562 cells through binding of TfR scFv to TfR, and mediate cytotoxicity of viral peptide-specific CTLs against K562 cells in vitro. In addition, the fusion protein can induce inhibition of solid tumor formation and improve survival time in tumor xenograft nude mouse with the injection of the sorted viral peptide-specific CTLs generated by co-culture of peripheral blood lymphocytes from HLA-A2 positive donors with inactivated T2 cells pulsed with the viral peptide.
Collapse
Affiliation(s)
- Jianan Li
- Department of Immunology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Rosenfeldt H, Kropp T, Benson K, Ricci MS, McGuinn WD, Verbois SL. Regulatory aspects of oncology drug safety evaluation: Past practice, current issues, and the challenge of new drugs. Toxicol Appl Pharmacol 2010; 243:125-33. [DOI: 10.1016/j.taap.2009.12.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Revised: 12/14/2009] [Accepted: 12/14/2009] [Indexed: 11/29/2022]
|
36
|
Argyriou AA, Kalofonos HP. Recent advances relating to the clinical application of naked monoclonal antibodies in solid tumors. Mol Med 2009; 15:183-91. [PMID: 19305491 DOI: 10.2119/molmed.2009.00007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Accepted: 02/12/2009] [Indexed: 12/25/2022] Open
Abstract
This review focuses on the recent advances in clinical data regarding antibody-based therapy in the management of solid tumors. We also discuss perspectives on antibody-based therapy in the future. Thorough understanding of the complex interactions between components of the immunological response has led to interest in the concept of immune-mediated therapy for solid tumors. Over the last few years, several humanized and chimeric monoclonal antibodies (MAbs) targeting human epidermal receptor 2 (HER2), epidermal growth factor receptor (EGFR), and vascular endothelial growth factor (VEGF) have been employed in treating solid tumors, including breast, colorectal, lung, head and neck, and gynecologic cancers. Trastuzumab, bevacizumab, cetuximab, and panitumumab are MAbs that are most widely used in clinical practice with acceptable rates of adverse events. Combination of MAbs with small-molecule inhibitors of the same pathway could potentially increase the efficacy and specificity of antibody-based treatment. Immune-mediated effects may be further exploited with the use of bivalent molecules.
Collapse
Affiliation(s)
- Andreas A Argyriou
- Department of Medicine-Division of Clinical Oncology, University Hospital, University of Patras Medical School, Rion-Patras, Greece
| | | |
Collapse
|
37
|
Biosensors for cancer markers diagnosis. Semin Cell Dev Biol 2009; 20:55-62. [DOI: 10.1016/j.semcdb.2009.01.015] [Citation(s) in RCA: 349] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Accepted: 01/23/2009] [Indexed: 11/20/2022]
|
38
|
Yang L, Lang JC, Balasubramanian P, Jatana KR, Schuller D, Agrawal A, Zborowski M, Chalmers JJ. Optimization of an enrichment process for circulating tumor cells from the blood of head and neck cancer patients through depletion of normal cells. Biotechnol Bioeng 2009; 102:521-34. [PMID: 18726961 DOI: 10.1002/bit.22066] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The optimization of a purely negative depletion, enrichment process for circulating tumor cells (CTCs) in the peripheral blood of head and neck cancer patients is presented. The enrichment process uses a red cell lysis step followed by immunomagnetic labeling, and subsequent depletion, of CD45 positive cells. A number of relevant variables are quantified, or attempted to be quantified, which control the performance of the enrichment process. Six different immunomagnetic labeling combinations were evaluated as well as the significant difference in performance with respect to the blood source: buffy coats purchased from the Red Cross, fresh, peripheral blood from normal donors, and fresh peripheral blood from human cancer patients. After optimization, the process is able to reduce the number of normal blood cells in a cancer patient's blood from 4.05 x 10(9) to 8.04 x 10(3) cells/mL and still recover, on average, 2.32 CTC per mL of blood. For all of the cancer patient blood samples tested in which CTC were detected (20 out of 26 patients) the average recovery of CTCs was 21.7 per mL of blood, with a range of 282 to 0.53 CTC. Since the initial number of CTC in a patient's blood is unknown, and most probably varies from patient to patient, the recovery of the CTC is unknown. However, spiking studies of a cancer cell line into normal blood, and subsequent enrichment using the optimized protocol indicated an average recovery of approximately 83%. Unlike a majority of other published studies, this study focused on quantifying as many factors as possible to facilitate both the optimization of the process as well as provide information for current and future performance comparisons. The authors are not aware any other reported study which has achieved the performance reported here (a 5.66 log(10)) in a purely negative enrichment mode of operation. Such a mode of operation of an enrichment process provides significant flexibility in that it has no bias with respect to what attributes define a CTC; thereby allowing the researcher or clinician to use any maker they choose to define whether the final, enrich product contains CTCs or other cell type relevant to the specific question (i.e., does the CTC have predominantly epithelial or mesenchymal characteristics?).
Collapse
Affiliation(s)
- Liying Yang
- Department of Chemical and Biomolecular Engineering, The Ohio State University, 125 Koffolt Laboratories, 140 West 19th Avenue, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Oganesyan V, Gao C, Shirinian L, Wu H, Dall’Acqua WF. Structural characterization of a human Fc fragment engineered for lack of effector functions. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2008; 64:700-4. [PMID: 18560159 PMCID: PMC2467532 DOI: 10.1107/s0907444908007877] [Citation(s) in RCA: 144] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Accepted: 03/22/2008] [Indexed: 11/10/2022]
Abstract
The first three-dimensional structure of a human Fc fragment genetically engineered for the elimination of its ability to mediate antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity is reported. When introduced into the lower hinge and CH2 domain of human IgG1 molecules, the triple mutation L234F/L235E/P331S ('TM') causes a profound decrease in their binding to human CD64, CD32A, CD16 and C1q. Enzymatically produced Fc/TM fragment was crystallized and its structure was solved at a resolution of 2.3 A using molecular replacement. This study revealed that the three-dimensional structure of Fc/TM is very similar to those of other human Fc fragments in the experimentally visible region spanning residues 236-445. Thus, the dramatic broad-ranging effects of TM on IgG binding to several effector molecules cannot be explained in terms of major structural rearrangements in this portion of the Fc.
Collapse
Affiliation(s)
- Vaheh Oganesyan
- Department of Antibody Discovery and Protein Engineering, MedImmune Inc., One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Changshou Gao
- Department of Antibody Discovery and Protein Engineering, MedImmune Inc., One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Lena Shirinian
- Department of Antibody Discovery and Protein Engineering, MedImmune Inc., One MedImmune Way, Gaithersburg, MD 20878, USA
| | - Herren Wu
- Department of Antibody Discovery and Protein Engineering, MedImmune Inc., One MedImmune Way, Gaithersburg, MD 20878, USA
| | - William F. Dall’Acqua
- Department of Antibody Discovery and Protein Engineering, MedImmune Inc., One MedImmune Way, Gaithersburg, MD 20878, USA
| |
Collapse
|
40
|
Khaibullina A, Jang BS, Sun H, Le N, Yu S, Frenkel V, Carrasquillo JA, Pastan I, Li KCP, Paik CH. Pulsed high-intensity focused ultrasound enhances uptake of radiolabeled monoclonal antibody to human epidermoid tumor in nude mice. J Nucl Med 2008; 49:295-302. [PMID: 18199622 DOI: 10.2967/jnumed.107.046888] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED The aim of this study was to determine if pulsed high-intensity focused ultrasound (HIFU) exposures could enhance tumor uptake of (111)In-MX-B3, a murine IgG1kappa monoclonal antibody directed against the Le(y) antigen. METHODS MX-B3 was labeled with (111)In, purified, and confirmed for its binding to the antigen-positive A431 cell line. Groups of nude mice were inoculated subcutaneously with A431 tumor cells on both hind flanks. A tumor on one flank was treated with pulsed-HIFU; the other tumor was used as an untreated control. Within 10 min after the HIFU exposure, the mice received intravenous (111)In-MX-B3 for imaging and biodistribution studies. Mice were euthanized at 1, 24, 48, and 120 h after injection for biodistribution studies. RESULTS The HIFU exposure shortened the peak tumor uptake time (24 vs. 48 h for the control) and increased the peak tumor uptake value (38 vs. 25 %ID/g [percentage injected dose per gram] for the control). The HIFU effect on enhancing tumor uptake was greater at earlier times up to 24 h, but the effect was gradually diminished thereafter. The HIFU effect on enhancing tumor uptake was substantiated by nuclear imaging studies. HIFU also increased the uptake of the antibody in surrounding tissues, but the net increase was marginal compared with the increase in tumor uptake. CONCLUSION This study demonstrates that pulsed-HIFU significantly enhances the delivery of (111)In-MX-B3 in human epidermoid tumors xenografted in nude mice. The results of this pilot study warrant further evaluation of other treatment regimens, such as repeated HIFU exposures for greater delivery enhancement of antibodies labeled with cytotoxic radioisotopes or pulsed-HIFU exposure in addition to a combined therapy of (90)Y-B3 and taxol to enhance the synergistic effect.
Collapse
Affiliation(s)
- Alfia Khaibullina
- Department of Radiology, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Immunomodulators: interleukins, interferons, and IV immunoglobulin. CLINICAL NEPHROTOXINS 2008. [PMCID: PMC7120840 DOI: 10.1007/978-0-387-84843-3_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The outstanding progress in immunology and the development of new technologies have resulted in the introduction of new immunotherapies, the so-called “immunomodulators”, for autoimmune diseases, inflammatory disorders, allograft rejection, and cancer. These immunomodulators comprise recombinant cytokines and specific blocking or depleting antibodies. Many of these therapies achieve their effect by stimulating the release of cytokines. The term cytokines includes interleukins (IL-), chemokines, growth factors, interferons (IFN), colony stimulating factors (CSF), and tumor necrosis factors (TNF). These molecules are involved in inflammation, cell proliferation and apoptosis, tissue injury and repair. These new therapeutic tools can be associated with side effects among which nephrotoxicity. The most common immunomodulators associated with nephrotoxicity are described in Table 1. The nephrotoxic side effects of immunomodulators can be roughly divided into (ischemic) tubular necrosis, thrombotic microangiopathy, serum sickness, and autoimmune disorders.
Collapse
|
42
|
Immunotoxin and Taxol synergy results from a decrease in shed mesothelin levels in the extracellular space of tumors. Proc Natl Acad Sci U S A 2007; 104:17099-104. [PMID: 17940013 DOI: 10.1073/pnas.0708101104] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Recombinant immunotoxins are chimeric proteins composed of the Fv portion of a tumor-specific antibody fused to a toxin. SS1P (CAT-5001) is an immunotoxin composed of an antimesothelin Fv fused to a 38-kDa portion of Pseudomonas exotoxin A. Immunotoxins have been shown to be active in lymphomas and leukemias, but are much less active against solid tumors. We recently reported that Taxol and other chemotherapeutic agents show striking synergistic antitumor activity in mice when immunotoxin SS1P, which targets the mesothelin antigen on solid tumors, is given with Taxol. Using a pair of Taxol-sensitive and Taxol-resistant KB tumors equally sensitive to immunotoxin SS1P, we examined the mechanism of synergy. We show that synergy is only observed with Taxol-sensitive tumors, ruling out an effect of Taxol on endothelial cells. We also show that the KB tumors have high levels of shed mesothelin in their extracellular space; these levels increase with tumor size and, after Taxol treatment, dramatically fall in the drug-sensitive but not the drug-resistant tumors. Because the mesothelin levels in the tumor exceed the levels of SS1P in the tumor, and because shed mesothelin is being continuously released into the circulation at a high rate, we propose that synergy is due to the Taxol-induced fall in shed antigen levels.
Collapse
|
43
|
Li P, Jiang S, Pero SC, Oligino L, Krag DN, Michejda CJ, Roller PP. Design and synthesis of paclitaxel conjugated with an ErbB2-recognizing peptide, EC-1. Biopolymers 2007; 87:225-30. [PMID: 17879382 DOI: 10.1002/bip.20828] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The selective delivery of therapeutic agents to receptors overexpressed in cancer cells without harming the rest of the body is a major challenge in clinical oncology today. In this study, we report the design and synthesis of paclitaxel (PTX) conjugated with an erbB2-recognizing peptide (EC-1). The cyclic peptide EC-1 specifically binds to the extracellular domain of ErbB2 and selectively inhibits proliferation of breast cancer cells overexpressing ErbB2. PTX is a potent antitumor agent commonly used in the treatment of advanced metastatic breast cancer, yet patients have to suffer some side effects caused by its systemic toxicity. The aim of our conjugate is to specifically deliver antitumor agent PTX to breast cancer cells that overexpress oncogenic ErbB2 with the purpose to reduce toxicity and enhance selective killing of cancer cells. In this study, a concise and efficient synthetic route for the preparation of the PTX-EC-1 conjugate has been developed in 6% overall yield. This synthetic approach provides a general method for conjugating a highly functionalized and disulfide-bridge containing cyclopeptide to Taxol or other antitumor agents.
Collapse
Affiliation(s)
- Peng Li
- Laboratory of Medicinal Chemistry, CCR, NCI-Frederick, NIH, Frederick, MD 21702, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Dall'Acqua WF, Cook KE, Damschroder MM, Woods RM, Wu H. Modulation of the effector functions of a human IgG1 through engineering of its hinge region. THE JOURNAL OF IMMUNOLOGY 2006; 177:1129-38. [PMID: 16818770 DOI: 10.4049/jimmunol.177.2.1129] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We report here the engineering of a humanized anti-human EphA2 mAb (mAb 12G3H11) in an effort to explore the relationship between the hinge of a human IgG1 and its effector functions. mAb 12G3H11, used here as a model, is directed against the human receptor tyrosine kinase EphA2, which is an actively investigated target for cancer therapy due to its up-regulation in many cancer cells. Various rational modifications were introduced into the hinge region of mAb 12G3H11. These mutations were predicted to modulate the hinge's length, flexibility, and/or biochemical properties. We show that the upper and middle hinge both play important, although functionally distinct roles. In particular, middle hinge modifications predicted to decrease its rigidity or length as well as eliminating either one of its two cysteine residues had a strong negative impact on C1q binding and complement-dependent cytotoxicity. Disruption of covalent bonds between both H chains may account in part for these effects. We also describe middle hinge mutants with a significantly decreased ability to bind FcgammaRIIIA and trigger Ab-dependent cell-mediated cytotoxicity. Conversely, we also generated upper hinge mutants exhibiting an increase in C1q binding and complement-dependent cytotoxicity activity. Therefore, this approach represents a novel strategy to fine-tune the biological activity of a given human IgG1. We also define, for the first time in such a systematic fashion, the relationship between various characteristics of the middle and upper hinge and the corresponding effector functions.
Collapse
Affiliation(s)
- William F Dall'Acqua
- Department of Antibody Discovery and Protein Engineering, MedImmune, One MedImmune Way, Gaithersburg, MD 20878, USA.
| | | | | | | | | |
Collapse
|
45
|
Safavy A, Raisch KP, Matusiak D, Bhatnagar S, Helson L. Single-drug multiligand conjugates: synthesis and preliminary cytotoxicity evaluation of a paclitaxel-dipeptide "scorpion" molecule. Bioconjug Chem 2006; 17:565-70. [PMID: 16704191 DOI: 10.1021/bc050224c] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
To improve the targeting properties of receptor-directed drug-peptide conjugates, a multiligand approach was proposed and a model "scorpion" conjugate (6, Figure 1), consisting of two peptide "claws" and a paclitaxel (PTX) "tail", was synthesized. The cell surface receptor-directed peptide used in this single-drug multiligand (SDML) model was a segment of the amphibian peptide bombesin (BBN) which had the Y6Q7W8A9V10G11H12L13M14-NH2 sequence, designated here as BBN[6-14] (2, Figure 2). Due to the lipophilic nature of both PTX and BBN[6-14], compound 6 had a low water solubility. To enhance the solubility, PEG derivatives of this conjugate were prepared with the polymer inserted either in the claws or in the tail regions. In a preliminary random screening, conjugate 6 showed superior cytotoxic activity in several GRPR-positive human cancer cell lines as compared to free PTX and two single-drug single-ligand (SDSL) conjugates. In a receptor blocking experiment, addition of excess unconjugated BBN[6-14] ligand reduced the cytotoxicity of conjugate 6, indicating the receptor-mediated mechanism of drug delivery. The PEG-derived conjugates showed activities which were intermediate between SDSL and the SDML congeners. Also, an increase in the number of the PEG segments lowered cytotoxicity, possibly due to steric hindrance against ligand-receptor binding. Taken together, these results demonstrate the potential of the multiligand approach in the design of receptor-targeting conjugates for tumor-specific drug delivery.
Collapse
Affiliation(s)
- Ahmad Safavy
- Department of Radiation Oncology and the Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama 35294-6832, USA.
| | | | | | | | | |
Collapse
|
46
|
Dall'Acqua WF, Kiener PA, Wu H. Properties of human IgG1s engineered for enhanced binding to the neonatal Fc receptor (FcRn). J Biol Chem 2006; 281:23514-24. [PMID: 16793771 DOI: 10.1074/jbc.m604292200] [Citation(s) in RCA: 461] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We describe here the functional implications of an increase in IgG binding to the neonatal Fc receptor. We have defined in a systematic fashion the relationship between enhanced FcRn binding of a humanized anti-respiratory syncytial virus (RSV) monoclonal antibody (MEDI-524) and the corresponding biological consequences in cynomolgus monkeys. The triple mutation M252Y/S254T/T256E (YTE) was introduced into the Fc portion of MEDI-524. Whereas these substitutions did not affect the ability of MEDI-524 to bind to its cognate antigen and inhibit RSV replication, they resulted in a 10-fold increase in its binding to both cynomolgus monkey and human FcRn at pH 6.0. MEDI-524-YTE was efficiently released from FcRn at pH 7.4 in both cases. We show that MEDI-524-YTE consistently exhibited a nearly 4-fold increase in serum half-life in cynomolgus monkeys when compared with MEDI-524. This constituted the largest half-life improvement described to date for an IgG in a primate. For the first time, we demonstrate that these sustained serum levels resulted in an up to 4-fold increase in lung bioavailability. Importantly, we also establish that our non-human primate model is relevant to human. Finally, we report that the YTE triple substitution provided a means to modulate the antibody-dependent cell-mediated cytotoxicity (ADCC) activity of a humanized IgG1 directed against the human integrin alpha(v)beta3. Therefore, the YTE substitutions allow the simultaneous modulation of serum half-life, tissue distribution and activity of a given human IgG1.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/blood
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal, Humanized
- Antibody Diversity
- Antibody-Dependent Cell Cytotoxicity
- Binding Sites, Antibody
- Half-Life
- Histocompatibility Antigens Class I/blood
- Histocompatibility Antigens Class I/metabolism
- Humans
- Immunoglobulin G/administration & dosage
- Immunoglobulin G/blood
- Immunoglobulin G/chemistry
- Immunoglobulin G/metabolism
- Macaca fascicularis
- Organ Specificity
- Protein Binding
- Protein Engineering/methods
- Receptors, Fc/blood
- Receptors, Fc/metabolism
- Receptors, IgG/metabolism
Collapse
Affiliation(s)
- William F Dall'Acqua
- Department of Antibody Discovery and Protein Engineering, MedImmune, Inc., Gaithersburg, Maryland 20878, USA. dall'
| | | | | |
Collapse
|
47
|
Soper SA, Brown K, Ellington A, Frazier B, Garcia-Manero G, Gau V, Gutman SI, Hayes DF, Korte B, Landers JL, Larson D, Ligler F, Majumdar A, Mascini M, Nolte D, Rosenzweig Z, Wang J, Wilson D. Point-of-care biosensor systems for cancer diagnostics/prognostics. Biosens Bioelectron 2006; 21:1932-42. [PMID: 16473506 DOI: 10.1016/j.bios.2006.01.006] [Citation(s) in RCA: 217] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2005] [Revised: 01/10/2006] [Accepted: 01/12/2006] [Indexed: 11/16/2022]
Abstract
With the growing number of fatalities resulting from the 100 or so cancer-related diseases, new enabling tools are required to provide extensive molecular profiles of patients to guide the clinician in making viable diagnosis and prognosis. Unfortunately with cancer-related diseases, there is not one molecular marker that can provide sufficient information to assist the clinician in making effective prognoses or even diagnoses. Indeed, large panels of markers must typically be evaluated that cut across several different classes (mutations in certain gene fragments--DNA; over/under-expression of gene activity as monitored by messenger RNAs; the amount of proteins present in serum or circulating tumor cells). The classical biosensor format (dipstick approach for monitoring the presence of a single element) is viewed as a valuable tool in many bioassays, but possesses numerous limitations in cancer due primarily to the single element nature of these sensing platforms. As such, if biosensors are to become valuable tools in the arsenal of the clinician to manage cancer patients, new formats are required. This review seeks to provide an overview of the current thinking on molecular profiling for diagnosis and prognosis of cancers and also, provide insight into the current state-of-the-art in the biosensor field and new strategies that must be considered to bring this important technology into the cancer field.
Collapse
Affiliation(s)
- Steven A Soper
- Louisiana State University, Baton Rouge, LA 70803, United States.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
Cancer is the second leading cause of death in the industrialized world. Most cancer patients are treated by a combination of surgery, radiation and/or chemotherapy. Whereas the primary tumor can, in most cases, be efficiently treated by a combination of these standard therapies, preventing the metastatic spread of the disease through disseminated tumor cells is often not effective. The eradication of disseminated tumor cells present in the blood circulation and micro-metastases in distant organs therefore represents another promising approach in cancer immunotherapy. Main strategies of cancer immunotherapy aim at exploiting the therapeutic potential of tumor-specific antibodies and cellular immune effector mechanisms. Whereas passive antibody therapy relies on the repeated application of large quantities of tumor antigen-specific antibodies, active immunotherapy aims at the generation of a tumor-specific immune response combining both humoral and cytotoxic T cell effector mechanisms by the host's immune system following vaccination. In the first part of this review, concurrent developments in active and passive cancer immunotherapy are discussed. In the second part, the various approaches for the production of optimized monoclonal antibodies used for anti-cancer vaccination are summarized.
Collapse
Affiliation(s)
- Manfred Schuster
- Apeiron Biologics Forschungs- und Entwicklungs-GmbH, Brunnerstrasse 59, 1230 Vienna, Austria.
| | | | | |
Collapse
|
49
|
Riddle DS, Sanz L, Chong H, Thompson J, Vile RG. Tumor cell surface display of immunoglobulin heavy chain Fc by gene transfer as a means to mimic antibody therapy. Hum Gene Ther 2005; 16:830-44. [PMID: 16000065 DOI: 10.1089/hum.2005.16.830] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We hypothesized that inducing display of the immunoglobulin Fc (IgFc) molecule on the tumor cell surface by gene transfer would promote tumor cell killing by the same mechanisms as antibody-based approaches but would alleviate some of the problems inherent in the use of antibodies for cancer therapy. We expressed the cDNA of the Fc portion of the murine IgG2a heavy chain on the surface of tumor cells such that its C terminus projected away from the tumor cell surface, mimicking a natural antibody-tagging event. In vitro, Fc receptor-positive natural killer (NK) cells specifically recognized and lysed B16 melanoma cells expressing surface IgFc. Macrophages bound to B16-Fc cells significantly more than to parental B16 cells and surface IgFc expression promoted formation of the terminal complement pore complex leading to cell lysis and death. Expression of IgFc dramatically delayed the ability of B16 cells to form tumors in vivo, attributable largely to the effects of NK cells. Furthermore, fluorescence-activated cell-sorting analysis showed that cells from outgrowth B16 IgFc tumors had lost all IgFc expression. When additional immunostimulatory signals were provided at the time of IgFc-mediated tumor cell killing through expression of heat shock protein 70 (hsp70), significant antitumor immunity was generated. Intratumoral delivery of an adenoviral vector expressing IgFc was effective at treating locally accessible tumors but did not impact metastatic disease. However, delivery of adenoviral vectors expressing both IgFc and hsp70 cured both local and metastatic tumors established for 6 days before viral treatment. These data suggest that it is possible to use gene transfer to mimic the beneficial properties of antibody therapy while alleviating some of the associated problems.
Collapse
Affiliation(s)
- David S Riddle
- Molecular Medicine Program and Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | | | |
Collapse
|
50
|
Abstract
The past five years have witnessed the emergence of monoclonal antibodies as important therapeutics for cancer treatment. Lower toxicity for antibodies versus small molecules, the potential for increased efficacy by conjugation to radioisotopes and cellular toxins, or the ability to exploit immune cell functions have led to clinical performances on par or superior to conventional drug therapies. This review outlines the various immunoglobulin design strategies currently available, techniques used to reduce Ig antigenicity and toxicity, and points to consider during the manufacture of antibodies for use in clinical oncology.
Collapse
Affiliation(s)
- Jerome E Tanner
- TanTec Biosystems Inc., Dollard-Des-Ormeaux, Montreal, Quebec, Canada.
| |
Collapse
|