51
|
Paoloni M, Mazcko C, Selting K, Lana S, Barber L, Phillips J, Skorupski K, Vail D, Wilson H, Biller B, Avery A, Kiupel M, LeBlanc A, Bernhardt A, Brunkhorst B, Tighe R, Khanna C. Defining the Pharmacodynamic Profile and Therapeutic Index of NHS-IL12 Immunocytokine in Dogs with Malignant Melanoma. PLoS One 2015; 10:e0129954. [PMID: 26091536 PMCID: PMC4474860 DOI: 10.1371/journal.pone.0129954] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 05/14/2015] [Indexed: 11/18/2022] Open
Abstract
Background Interleukin (IL)-12 is a pro-inflammatory cytokine that mediates T-helper type 1 responses and cytotoxic T-cell activation, contributing to its utility as anti-cancer agent. Systemic administration of IL-12 often results in unacceptable toxicity; therefore, strategies to direct delivery of IL-12 to tumors are under investigation. The objective of this study was to assist the preclinical development of NHS-IL12, an immunocytokine consisting of an antibody, which targets necrotic tumor regions, linked to IL-12. Specifically this study sought to evaluate the safety, serum pharmacokinetics, anti-tumor activity, and immune modulation of NHS-IL12 in dogs with naturally occurring cancers. Methodology/Principal Findings A rapid dose-escalation study of NHS-IL12 administered subcutaneously to dogs with melanoma was conducted through the Comparative Oncology Trials Consortium (COTC). Eleven dogs were enrolled in four dose-escalation cohorts; thereafter, an additional seven dogs were treated at the defined tolerable dose of 0.8 mg/m2. The expanded cohort at this fixed dose (ten dogs in total) was accrued for further pharmacokinetics and pharmacodynamics assessment. NHS-IL12 levels, serum cytokine concentrations, and peripheral blood mononuclear cell characterization (post-treatment) and draining lymph node immune profiling, and tumor biopsies (pre- and post-treatment) were collected. Adverse events included thrombocytopenia, liver enzymopathies, fever, and vasculitis. Correlation between interferon (IFN)-γ induction, adverse events, and NHS-IL12 exposure (maximum concentration and area under the concentration-time curve) were dose-dependent. Serum IL-10 levels and intratumoral CD8+ populations increased after treatment. Partial responses, according to Response Evaluation Criteria in Solid Tumors (RECIST) criteria, were observed in two dogs treated with NHS-IL12 0.8 mg/m2 and 1.6 mg/m2. Conclusions/Significance NHS-IL12 was administered safely to dogs with melanoma and both immunologic and clinical activity was observed. This study successfully defined a narrow therapeutic window for systemic delivery of NHS-IL12 via the subcutaneous route. Results will inform the design and implementation of first-in-human clinical trials of NHS-IL12 in cancer patients.
Collapse
Affiliation(s)
- Melissa Paoloni
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Christina Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Kimberly Selting
- College of Veterinary Medicine, University of Missouri-Columbia, Columbia, Missouri, United States of America
| | - Susan Lana
- College of Veterinary Medicine and Biological Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Lisa Barber
- School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Jeffrey Phillips
- College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Katherine Skorupski
- School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - David Vail
- School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Heather Wilson
- College of Veterinary Medicine, Texas A&M University, College Station, Texas, United States of America
| | - Barbara Biller
- College of Veterinary Medicine and Biological Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Anne Avery
- College of Veterinary Medicine and Biological Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Matti Kiupel
- College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, United States of America
| | - Amy LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Anna Bernhardt
- EMD-Serono Research and Development Institute, Billerica, Massachusetts, United States of America
| | - Beatrice Brunkhorst
- EMD-Serono Research and Development Institute, Billerica, Massachusetts, United States of America
| | - Robert Tighe
- EMD-Serono Research and Development Institute, Billerica, Massachusetts, United States of America
| | - Chand Khanna
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
52
|
Yata T, Lee ELQ, Suwan K, Syed N, Asavarut P, Hajitou A. Modulation of extracellular matrix in cancer is associated with enhanced tumor cell targeting by bacteriophage vectors. Mol Cancer 2015; 14:110. [PMID: 26037383 PMCID: PMC4451735 DOI: 10.1186/s12943-015-0383-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/11/2015] [Indexed: 12/20/2022] Open
Abstract
Background Gene therapy has been an attractive paradigm for cancer treatment. However, cancer gene therapy has been challenged by the inherent limitation of vectors that are able to deliver therapeutic genes to tumors specifically and efficiently following systemic administration. Bacteriophage (phage) are viruses that have shown promise for targeted systemic gene delivery. Yet, they are considered poor vectors for gene transfer. Recently, we generated a tumor-targeted phage named adeno-associated virus/phage (AAVP), which is a filamentous phage particle whose genome contains the adeno-associated virus genome. Its effectiveness in delivering therapeutic genes to tumors specifically both in vitro and in vivo has been shown in numerous studies. Despite being a clinically useful vector, a multitude of barriers impede gene transduction to tumor cells. We hypothesized that one such factor is the tumor extracellular matrix (ECM). Methods We used a number of tumor cell lines from different species and histological types in 2D monolayers or 3D multicellular tumor spheroid (MCTS) models. To assess whether the ECM is a barrier to tumor cell targeting by AAVP, we depleted the ECM using collagenase, hyaluronidase, or combination of both. We employed multiple techniques to investigate and quantify the effect of ECM depletion on ECM composition (including collagen type I, hyaluronic acid, fibronectin and laminin), and how AAVP adsorption, internalisation, gene expression and therapeutic efficacy are subsequently affected. Data were analyzed using a student’s t test when comparing two groups or one-way ANOVA and post hoc Tukey tests when using more than two groups. Results We demonstrate that collagenase and hyaluronidase-mediated degradation of tumor ECM affects the composition of collagen, hyaluronic acid and fibronectin. Consequently, AAVP diffusion, internalisation, gene expression and tumor cell killing were enhanced after enzymatic treatment. Our data suggest that enhancement of gene transfer by the AAVP is solely attributed to ECM depletion. We provide substantial evidence that ECM modulation is relevant in clinically applicable settings by using 3D MCTS, which simulates in vivo environments more accurately. Conclusion Our findings suggest that ECM depletion is an effective strategy to enhance the efficiency of viral vector-guided gene therapy.
Collapse
Affiliation(s)
- Teerapong Yata
- Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, London, W12 0NN, UK. .,National Nanotechnology Center, National Science and Technology Development Agency, 111 Thailand Science Park, Pathumthani, 12120, Thailand.
| | - Eugene L Q Lee
- Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, London, W12 0NN, UK.
| | - Keittisak Suwan
- Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, London, W12 0NN, UK.
| | - Nelofer Syed
- The John Fulcher Molecular Neuro-Oncology Laboratory, Division of Brain sciences, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, London, W12 0NN, UK.
| | - Paladd Asavarut
- Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, London, W12 0NN, UK.
| | - Amin Hajitou
- Phage Therapy Group, Division of Brain Sciences, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, Burlington Danes Building, London, W12 0NN, UK. .,Amin Hajitou, Burlington Danes Building, Hammersmith Hospital Campus, 160 Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
53
|
Tang L, Tong R, Coyle VJ, Yin Q, Pondenis H, Borst LB, Cheng J, Fan TM. Targeting tumor vasculature with aptamer-functionalized doxorubicin-polylactide nanoconjugates for enhanced cancer therapy. ACS NANO 2015; 9:5072-5081. [PMID: 25938427 DOI: 10.1021/acsnano.5b00166] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
An A10 aptamer (Apt)-functionalized, sub-100 nm doxorubicin-polylactide (Doxo-PLA) nanoconjugate (NC) with controlled release profile was developed as an intravenous therapeutic strategy to effectively target and cytoreduce canine hemangiosarcoma (cHSA), a naturally occurring solid tumor malignancy composed solely of tumor-associated endothelium. cHSA consists of a pure population of malignant endothelial cells expressing prostate-specific membrane antigen (PSMA) and is an ideal comparative tumor model system for evaluating the specificity and feasibility of tumor-associated endothelial cell targeting by A10 Apt-functionalized NC (A10 NC). In vitro, A10 NCs were selectively internalized across a panel of PSMA-expressing cancer cell lines, and when incorporating Doxo, A10 Doxo-PLA NCs exerted greater cytotoxic effects compared to nonfunctionalized Doxo-PLA NCs and free Doxo. Importantly, intravenously delivered A10 NCs selectively targeted PSMA-expressing tumor-associated endothelial cells at a cellular level in tumor-bearing mice and dramatically increased the uptake of NCs by endothelial cells within the local tumor microenvironment. By virtue of controlled drug release kinetics and selective tumor-associated endothelial cell targeting, A10 Doxo-PLA NCs possess a desirable safety profile in vivo, being well-tolerated following high-dose intravenous infusion in mice, as supported by the absence of any histologic organ toxicity. In cHSA-implanted mice, two consecutive intravenous infusions of A10 Doxo-PLA NCs exerted rapid and substantial cytoreductive activities within a period of 7 days, resulting in greater than 70% reduction in macroscopic tumor-associated endothelial cell burden as a consequence of enhanced cell death and necrosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Luke B Borst
- §Department of Population Health and Pathobiology, North Carolina State University, Raleigh, North Carolina 27607, United States
| | | | | |
Collapse
|
54
|
Dobroff AS, Rangel R, Guzman-Roja L, Salmeron CC, Gelovani JG, Sidman RL, Bologa CG, Oprea TI, Brinker CJ, Pasqualini R, Arap W. Ligand-directed profiling of organelles with internalizing phage libraries. ACTA ACUST UNITED AC 2015; 79:30.4.1-30.4.30. [PMID: 25640897 DOI: 10.1002/0471140864.ps3004s79] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Phage display is a resourceful tool to, in an unbiased manner, discover and characterize functional protein-protein interactions, create vaccines, and engineer peptides, antibodies, and other proteins as targeted diagnostic and/or therapeutic agents. Recently, our group has developed a new class of internalizing phage (iPhage) for ligand-directed targeting of organelles and to identify molecular pathways within live cells. This unique technology is suitable for applications ranging from fundamental cell biology to drug development. This unit describes the methods for generating and screening the iPhage display system, and explains how to select and validate candidate internalizing homing peptide.
Collapse
Affiliation(s)
- Andrey S Dobroff
- Division of Hematology/Oncology, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico.,Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico.,These authors contributed equally to this work
| | - Roberto Rangel
- Cancer Research Program, Houston Methodist Research Institute, Houston, Texas.,These authors contributed equally to this work
| | - Liliana Guzman-Roja
- Cancer Research Program, Houston Methodist Research Institute, Houston, Texas.,These authors contributed equally to this work
| | - Carolina C Salmeron
- Division of Hematology/Oncology, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico.,Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Juri G Gelovani
- Department of Biomedical Engineering, Wayne State University, Detroit, Michigan
| | - Richard L Sidman
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Cristian G Bologa
- Translational Informatics Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Tudor I Oprea
- Translational Informatics Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - C Jeffrey Brinker
- Department of Chemical and Nuclear Engineering, The University of New Mexico Cancer Center, Albuquerque, New Mexico
| | - Renata Pasqualini
- Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico.,These authors contributed equally as senior authors to this work
| | - Wadih Arap
- Division of Hematology/Oncology, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico.,These authors contributed equally as senior authors to this work
| |
Collapse
|
55
|
Pranjol MZI, Hajitou A. Bacteriophage-derived vectors for targeted cancer gene therapy. Viruses 2015; 7:268-84. [PMID: 25606974 PMCID: PMC4306838 DOI: 10.3390/v7010268] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 01/13/2015] [Indexed: 01/04/2023] Open
Abstract
Cancer gene therapy expanded and reached its pinnacle in research in the last decade. Both viral and non-viral vectors have entered clinical trials, and significant successes have been achieved. However, a systemic administration of a vector, illustrating safe, efficient, and targeted gene delivery to solid tumors has proven to be a major challenge. In this review, we summarize the current progress and challenges in the targeted gene therapy of cancer. Moreover, we highlight the recent developments of bacteriophage-derived vectors and their contributions in targeting cancer with therapeutic genes following systemic administration.
Collapse
Affiliation(s)
- Md Zahidul Islam Pranjol
- Institute of Clinical and Biomedical Science, University of Exeter Medical School, Exeter, Devon EX1 2LU, UK.
| | - Amin Hajitou
- Phage Therapy Group, Department of Medicine, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK.
| |
Collapse
|
56
|
Veterinary oncology clinical trials: design and implementation. Vet J 2014; 205:226-32. [PMID: 25582798 DOI: 10.1016/j.tvjl.2014.12.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 11/20/2014] [Accepted: 12/14/2014] [Indexed: 11/20/2022]
Abstract
There has been a recent increase in interest among veterinarians and the larger biomedical community in the evaluation of novel cancer therapies in client-owned (pet) animals with spontaneous cancer. This includes novel drugs designed to be veterinary therapeutics, as well as agents for which data generated in animals with tumors may inform human clinical trial design and implementation. An understanding of the process involved in moving a therapeutic agent through the stages of clinical evaluation is critical to the successful implementation of clinical investigations, as well as interpretation of the veterinary oncology literature. This review outlines considerations in the design and conduct of the various phases of oncology clinical trials, along with recent adaptations/modifications of these basic designs that can enhance the generation of timely and meaningful clinical data.
Collapse
|
57
|
|
58
|
Barnard RA, Wittenburg LA, Amaravadi RK, Gustafson DL, Thorburn A, Thamm DH. Phase I clinical trial and pharmacodynamic evaluation of combination hydroxychloroquine and doxorubicin treatment in pet dogs treated for spontaneously occurring lymphoma. Autophagy 2014; 10:1415-25. [PMID: 24991836 DOI: 10.4161/auto.29165] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a lysosomal degradation process that may act as a mechanism of survival in a variety of cancers. While pharmacologic inhibition of autophagy with hydroxychloroquine (HCQ) is currently being explored in human clinical trials, it has never been evaluated in canine cancers. Non-Hodgkin lymphoma (NHL) is one of the most prevalent tumor types in dogs and has similar pathogenesis and response to treatment as human NHL. Clinical trials in canine patients are conducted in the same way as in human patients, thus, to determine a maximum dose of HCQ that can be combined with a standard chemotherapy, a Phase I, single arm, dose escalation trial was conducted in dogs with spontaneous NHL presenting as patients to an academic, tertiary-care veterinary teaching hospital. HCQ was administered daily by mouth throughout the trial, beginning 72 h prior to doxorubicin (DOX), which was given intravenously on a 21-d cycle. Peripheral blood mononuclear cells and biopsies were collected before and 3 d after HCQ treatment and assessed for autophagy inhibition and HCQ concentration. A total of 30 patients were enrolled in the trial. HCQ alone was well tolerated with only mild lethargy and gastrointestinal-related adverse events. The overall response rate (ORR) for dogs with lymphoma was 93.3%, with median progression-free interval (PFI) of 5 mo. Pharmacokinetic analysis revealed a 100-fold increase in HCQ in tumors compared with plasma. There was a trend that supported therapy-induced increase in LC3-II (the cleaved and lipidated form of microtubule-associated protein 1 light chain 3/LC3, which serves as a maker for autophagosomes) and SQSTM1/p62 (sequestosome 1) after treatment. The superior ORR and comparable PFI to single-agent DOX provide strong support for further evaluation via randomized, placebo-controlled trials in canine and human NHL.
Collapse
Affiliation(s)
- Rebecca A Barnard
- Department of Clinical Sciences; Colorado State University; Fort Collins, CO USA
| | - Luke A Wittenburg
- Department of Clinical Sciences; Colorado State University; Fort Collins, CO USA
| | - Ravi K Amaravadi
- Department of Medicine Hematology Oncology Division; Pearlman School of Medicine; University of Pennsylvania; Philadelphia, PA USA
| | - Daniel L Gustafson
- Department of Clinical Sciences; Colorado State University; Fort Collins, CO USA
| | - Andrew Thorburn
- Department of Pharmacology; University of Colorado School of Medicine; Aurora, CO USA
| | - Douglas H Thamm
- Department of Clinical Sciences; Colorado State University; Fort Collins, CO USA
| |
Collapse
|
59
|
Khanna C, Fan TM, Gorlick R, Helman LJ, Kleinerman ES, Adamson PC, Houghton PJ, Tap WD, Welch DR, Steeg PS, Merlino G, Sorensen PHB, Meltzer P, Kirsch DG, Janeway KA, Weigel B, Randall L, Withrow SJ, Paoloni M, Kaplan R, Teicher BA, Seibel NL, Smith M, Uren A, Patel SR, Trent J, Savage SA, Mirabello L, Reinke D, Barkaukas DA, Krailo M, Bernstein M. Toward a drug development path that targets metastatic progression in osteosarcoma. Clin Cancer Res 2014; 20:4200-9. [PMID: 24803583 DOI: 10.1158/1078-0432.ccr-13-2574] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Despite successful primary tumor treatment, the development of pulmonary metastasis continues to be the most common cause of mortality in patients with osteosarcoma. A conventional drug development path requiring drugs to induce regression of established lesions has not led to improvements for patients with osteosarcoma in more than 30 years. On the basis of our growing understanding of metastasis biology, it is now reasonable and essential that we focus on developing therapeutics that target metastatic progression. To advance this agenda, a meeting of key opinion leaders and experts in the metastasis and osteosarcoma communities was convened in Bethesda, Maryland. The goal of this meeting was to provide a "Perspective" that would establish a preclinical translational path that could support the early evaluation of potential therapeutic agents that uniquely target the metastatic phenotype. Although focused on osteosarcoma, the need for this perspective is shared among many cancer types. The consensus achieved from the meeting included the following: the biology of metastatic progression is associated with metastasis-specific targets/processes that may not influence grossly detectable lesions; targeting of metastasis-specific processes is feasible; rigorous preclinical data are needed to support translation of metastasis-specific agents into human trials where regression of measurable disease is not an expected outcome; preclinical data should include an understanding of mechanism of action, validation of pharmacodynamic markers of effective exposure and response, the use of several murine models of effectiveness, and where feasible the inclusion of the dog with naturally occurring osteosarcoma to define the activity of new drugs in the micrometastatic disease setting.
Collapse
Affiliation(s)
- Chand Khanna
- Molecular Oncology Section, Metastasis Biology; Center for Cancer Research; National Cancer Institute, NIH, Bethesda, Maryland
| | - Timothy M Fan
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois;
| | - Richard Gorlick
- Department of Pediatrics and Molecular Pharmacology, The Albert Einstein College of Medicine of Yeshiva University; Division of Hematology/Oncology, Department of Pediatrics, The Children's Hospital at Montefiore, Bronx
| | - Lee J Helman
- Center for Cancer Research; National Cancer Institute, NIH, Bethesda, Maryland
| | | | - Peter C Adamson
- Division of Clinical Pharmacology & Therapeutics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Peter J Houghton
- Center for Childhood Cancer, The Research Institute, Nationwide Children's Hospital, Columbus, Ohio
| | - William D Tap
- Sarcoma Oncology, Melanoma and Sarcoma Service, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medical College, New York, New York; Departments of
| | - Danny R Welch
- Kansas University Medical Center, Kansas City, Kansas
| | - Patricia S Steeg
- Laboratory of Molecular Pharmacology; Center for Cancer Research; National Cancer Institute, NIH, Bethesda, Maryland
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics; Center for Cancer Research; National Cancer Institute, NIH, Bethesda, Maryland
| | - Poul H B Sorensen
- Department of Pathology, University of British Columbia; BC Cancer Research Centre, Vancouver, British Columbia; and
| | - Paul Meltzer
- Genetics Branch; Center for Cancer Research; National Cancer Institute, NIH, Bethesda, Maryland
| | - David G Kirsch
- Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina
| | - Katherine A Janeway
- Department of Pediatrics, Harvard Medical School; Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts
| | - Brenda Weigel
- Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Lor Randall
- Huntsman Cancer Institute & Primary Children's Medical Center, University of Utah, Salt Lake City, Utah
| | - Stephen J Withrow
- Flint Animal Cancer Center, Colorado State University, Fort Collins, Colorado; Departments of
| | - Melissa Paoloni
- Comparative Oncology Program; Center for Cancer Research; National Cancer Institute, NIH, Bethesda, Maryland
| | - Rosandra Kaplan
- Tumor Microenvironment Section, Pediatric Oncology Branch; Center for Cancer Research; National Cancer Institute, NIH, Bethesda, Maryland
| | - Beverly A Teicher
- Molecular Pharmacology Branch; Center for Cancer Research; National Cancer Institute, NIH, Bethesda, Maryland
| | - Nita L Seibel
- Cancer Therapy Evaluations Program; Center for Cancer Research; National Cancer Institute, NIH, Bethesda, Maryland
| | | | - Aykut Uren
- Oncology and Biochemistry and Molecular & Cellular Biology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Shreyaskumar R Patel
- Sarcoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey Trent
- Translational Genomics Research Institute (TGen), Phoenix, Arizona
| | - Sharon A Savage
- Clinical Genetics Branch; Center for Cancer Research; National Cancer Institute, NIH, Bethesda, Maryland
| | - Lisa Mirabello
- Genetic Epidemiology Branch, Division of Cancer Epidemiology and Genetics; Center for Cancer Research; National Cancer Institute, NIH, Bethesda, Maryland
| | - Denise Reinke
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan
| | - Donald A Barkaukas
- Children's Oncology Group, QuadW-COG Childhood Sarcoma Biostatistics and Annotation Office, Monrovia
| | - Mark Krailo
- Department of Preventive Medicine, Keck School of Medicine at the University of Southern California, Los Angeles, California
| | - Mark Bernstein
- Department of Pediatrics, IWK Health Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
60
|
Paoloni M, Webb C, Mazcko C, Cherba D, Hendricks W, Lana S, Ehrhart EJ, Charles B, Fehling H, Kumar L, Vail D, Henson M, Childress M, Kitchell B, Kingsley C, Kim S, Neff M, Davis B, Khanna C, Trent J. Prospective molecular profiling of canine cancers provides a clinically relevant comparative model for evaluating personalized medicine (PMed) trials. PLoS One 2014; 9:e90028. [PMID: 24637659 PMCID: PMC3956546 DOI: 10.1371/journal.pone.0090028] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 01/28/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Molecularly-guided trials (i.e. PMed) now seek to aid clinical decision-making by matching cancer targets with therapeutic options. Progress has been hampered by the lack of cancer models that account for individual-to-individual heterogeneity within and across cancer types. Naturally occurring cancers in pet animals are heterogeneous and thus provide an opportunity to answer questions about these PMed strategies and optimize translation to human patients. In order to realize this opportunity, it is now necessary to demonstrate the feasibility of conducting molecularly-guided analysis of tumors from dogs with naturally occurring cancer in a clinically relevant setting. METHODOLOGY A proof-of-concept study was conducted by the Comparative Oncology Trials Consortium (COTC) to determine if tumor collection, prospective molecular profiling, and PMed report generation within 1 week was feasible in dogs. Thirty-one dogs with cancers of varying histologies were enrolled. Twenty-four of 31 samples (77%) successfully met all predefined QA/QC criteria and were analyzed via Affymetrix gene expression profiling. A subsequent bioinformatics workflow transformed genomic data into a personalized drug report. Average turnaround from biopsy to report generation was 116 hours (4.8 days). Unsupervised clustering of canine tumor expression data clustered by cancer type, but supervised clustering of tumors based on the personalized drug report clustered by drug class rather than cancer type. CONCLUSIONS Collection and turnaround of high quality canine tumor samples, centralized pathology, analyte generation, array hybridization, and bioinformatic analyses matching gene expression to therapeutic options is achievable in a practical clinical window (<1 week). Clustering data show robust signatures by cancer type but also showed patient-to-patient heterogeneity in drug predictions. This lends further support to the inclusion of a heterogeneous population of dogs with cancer into the preclinical modeling of personalized medicine. Future comparative oncology studies optimizing the delivery of PMed strategies may aid cancer drug development.
Collapse
Affiliation(s)
- Melissa Paoloni
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Craig Webb
- Van Andel Research Institute, Grand Rapids, Michigan, United States of America
| | - Christina Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - David Cherba
- Van Andel Research Institute, Grand Rapids, Michigan, United States of America
| | - William Hendricks
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Susan Lana
- Colorado State University, College of Veterinary Medicine, Fort Collins, Colorado, United States of America
| | - E. J. Ehrhart
- Colorado State University, College of Veterinary Medicine, Fort Collins, Colorado, United States of America
| | - Brad Charles
- Colorado State University, College of Veterinary Medicine, Fort Collins, Colorado, United States of America
| | - Heather Fehling
- Clinical Reference Laboratory, Lenexa, Kansas, United States of America
| | - Leena Kumar
- Clinical Reference Laboratory, Lenexa, Kansas, United States of America
| | - David Vail
- University of Wisconsin-Madison, School of Veterinary Medicine, Madison, Wisconsin, United States of America
| | - Michael Henson
- University of Minnesota, College of Veterinary Medicine, St. Paul, Minnesota, United States of America
| | - Michael Childress
- Purdue University, School of Veterinary Medicine, West Lafayette, Indiana, United States of America
| | - Barbara Kitchell
- Michigan State University, College of Veterinary Medicine, East Lansing, Michigan, United States of America
| | - Christopher Kingsley
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Seungchan Kim
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Mark Neff
- Van Andel Research Institute, Grand Rapids, Michigan, United States of America
| | - Barbara Davis
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| | - Chand Khanna
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Jeffrey Trent
- Van Andel Research Institute, Grand Rapids, Michigan, United States of America
- Translational Genomics Research Institute (TGen), Phoenix, Arizona, United States of America
| |
Collapse
|
61
|
Affiliation(s)
- Bethany Powell Gray
- Department of Internal Medicine and The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8807, United States
| | - Kathlynn C. Brown
- Department of Internal Medicine and The Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-8807, United States
| |
Collapse
|
62
|
Yuan Z, Pastoriza J, Quinn T, Libutti SK. Targeting Tumor Vasculature Using Adeno-Associated Virus Phage Vectors Coding Tumor Necrosis Factor-α. GENE THERAPY OF CANCER 2014:19-33. [DOI: 10.1016/b978-0-12-394295-1.00002-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
63
|
Abstract
Antibody-based immunotherapies are important therapy options in human oncology. Although human humoral specific immunity is constituted of five different immunoglobulin classes, currently only IgG-based immunotherapies have proceeded to clinical application. This review, however, discusses the benefits and difficulties of IgE-based immunotherapy of cancer, with special emphasis on how to translate promising preclinical results into clinical studies. Pursuing the “Comparative Oncology” approach, novel drug candidates are investigated in clinical trials with veterinary cancer patients, most often dogs. By this strategy drug development could be speeded up, animal experiments could be reduced and novel therapy options could be introduced benefitting humans as well as man’s best friend.
Collapse
Affiliation(s)
- Josef Singer
- Comparative Medicine, Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, and University Vienna, Vienna, Austria
| | - Erika Jensen-Jarolim
- Comparative Medicine, Messerli Research Institute, University of Veterinary Medicine Vienna, Medical University Vienna, and University Vienna, Vienna, Austria ; Comparative Immunology and Oncology, Institute of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
64
|
Rossmeisl JH, Garcia PA, Daniel GB, Bourland JD, Debinski W, Dervisis N, Klahn S. Invited review--neuroimaging response assessment criteria for brain tumors in veterinary patients. Vet Radiol Ultrasound 2013; 55:115-32. [PMID: 24219161 DOI: 10.1111/vru.12118] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 09/07/2013] [Indexed: 12/28/2022] Open
Abstract
The evaluation of therapeutic response using cross-sectional imaging techniques, particularly gadolinium-enhanced MRI, is an integral part of the clinical management of brain tumors in veterinary patients. Spontaneous canine brain tumors are increasingly recognized and utilized as a translational model for the study of human brain tumors. However, no standardized neuroimaging response assessment criteria have been formulated for use in veterinary clinical trials. Previous studies have found that the pathophysiologic features inherent to brain tumors and the surrounding brain complicate the use of the response evaluation criteria in solid tumors (RECIST) assessment system. Objectives of this review are to describe strengths and limitations of published imaging-based brain tumor response criteria and propose a system for use in veterinary patients. The widely used human Macdonald and response assessment in neuro-oncology (RANO) criteria are reviewed and described as to how they can be applied to veterinary brain tumors. Discussion points will include current challenges associated with the interpretation of brain tumor therapeutic responses such as imaging pseudophenomena and treatment-induced necrosis, and how advancements in perfusion imaging, positron emission tomography, and magnetic resonance spectroscopy have shown promise in differentiating tumor progression from therapy-induced changes. Finally, although objective endpoints such as MR imaging and survival estimates will likely continue to comprise the foundations for outcome measures in veterinary brain tumor clinical trials, we propose that in order to provide a more relevant therapeutic response metric for veterinary patients, composite response systems should be formulated and validated that combine imaging and clinical assessment criteria.
Collapse
Affiliation(s)
- John H Rossmeisl
- Department of Small Animal Clinical Sciences, Virginia-Maryland Regional College of Veterinary Medicine, VA, 24061; Biomechanical Systems and Veterinary and Comparative Neuro-oncology Laboratories, Department of Biomedical Engineering, Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, VA, 24061
| | | | | | | | | | | | | |
Collapse
|
65
|
Angiogenesis in canine mammary tumours: a morphometric and prognostic study. J Comp Pathol 2013; 150:175-83. [PMID: 24231306 DOI: 10.1016/j.jcpa.2013.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 08/07/2013] [Accepted: 09/09/2013] [Indexed: 12/20/2022]
Abstract
Angiogenesis in canine mammary tumours (CMTs) has been described previously; however, only the intratumoural (IT) region has been studied and information on peritumoural (PT) angiogenesis is lacking. In this study, the blood vessel density (BVD), blood vessel perimeter (BVP) and blood vessel area (BVA) in IT and PT regions of 56 benign CMTs, 55 malignant CMTs and 13 samples of normal mammary gland tissue were analyzed. In addition, the blood endothelial cell proliferation (BECP) as an indicator of ongoing angiogenesis was investigated. The prognostic value of each parameter was also examined. Blood vessels and proliferating blood endothelial cells were present in IT and PT regions of both benign and malignant tumours. The vessels in the PT region had a significantly higher area and perimeter compared with those in the IT region. Malignant tumours showed significantly more vessels with a smaller total BVA and a higher BECP compared with benign tumours and control tissue. In the PT regions there was a significantly higher BVD, BVA and BVP compared with the vessels in control tissue. Only the IT and PT BVD and PT BECP in benign tumours allowed prediction of survival. The morphology of blood vessels in CMTs shows similarities with those in human breast cancer, which strengthens the case for the use of dogs with CMTs in comparative oncology trials.
Collapse
|
66
|
Kia A, Yata T, Hajji N, Hajitou A. Inhibition of histone deacetylation and DNA methylation improves gene expression mediated by the adeno-associated virus/phage in cancer cells. Viruses 2013; 5:2561-72. [PMID: 24153059 PMCID: PMC3814604 DOI: 10.3390/v5102561] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 10/14/2013] [Accepted: 10/15/2013] [Indexed: 11/16/2022] Open
Abstract
Bacteriophage (phage), viruses that infect bacteria only, have become promising vectors for targeted systemic delivery of genes to cancer, although, with poor efficiency. We previously designed an improved phage vector by incorporating cis genetic elements of adeno-associated virus (AAV). This novel AAV/phage hybrid (AAVP) specifically targeted systemic delivery of therapeutic genes into tumors. To advance the AAVP vector, we recently introduced the stress-inducible Grp78 tumor specific promoter and found that this dual tumor-targeted AAVP provides persistent gene expression, over time, in cancer cells compared to silenced gene expression from the CMV promoter in the parental AAVP. Herein, we investigated the effect of histone deacetylation and DNA methylation on AAVP-mediated gene expression in cancer cells and explored the effect of cell confluence state on AAVP gene expression efficacy. Using a combination of AAVP expressing the GFP reporter gene, flow cytometry, inhibitors of histone deacetylation, and DNA methylation, we have demonstrated that histone deacetylation and DNA methylation are associated with silencing of gene expression from the CMV promoter in the parental AAVP. Importantly, inhibitors of histone deacetylases boost gene expression in cancer cells from the Grp78 promoter in the dual tumor-targeted AAVP. However, cell confluence had no effect on AAVP-guided gene expression. Our findings prove that combination of histone deacetylase inhibitor drugs with the Grp78 promoter is an effective approach to improve AAVP-mediated gene expression in cancer cells and should be considered for AAVP-based clinical cancer gene therapy.
Collapse
Affiliation(s)
- Azadeh Kia
- Phage Therapy Group, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK; E-Mails: (A.K.); (T.Y.)
| | - Teerapong Yata
- Phage Therapy Group, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK; E-Mails: (A.K.); (T.Y.)
| | - Nabil Hajji
- Epigenetic Group, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK; E-Mail:
| | - Amin Hajitou
- Phage Therapy Group, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK; E-Mails: (A.K.); (T.Y.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +44-207-594-6546; Fax: +44-207-549-6548
| |
Collapse
|
67
|
Rangel R, Dobroff AS, Guzman-Rojas L, Salmeron CC, Gelovani JG, Sidman RL, Pasqualini R, Arap W. Targeting mammalian organelles with internalizing phage (iPhage) libraries. Nat Protoc 2013; 8:1916-39. [PMID: 24030441 PMCID: PMC4309278 DOI: 10.1038/nprot.2013.119] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Techniques that are largely used for protein interaction studies and the discovery of intracellular receptors, such as affinity-capture complex purification and the yeast two-hybrid system, may produce inaccurate data sets owing to protein insolubility, transient or weak protein interactions or irrelevant intracellular context. A versatile tool for overcoming these limitations, as well as for potentially creating vaccines and engineering peptides and antibodies as targeted diagnostic and therapeutic agents, is the phage-display technique. We have recently developed a new technology for screening internalizing phage (iPhage) vectors and libraries using a ligand/receptor-independent mechanism to penetrate eukaryotic cells. iPhage particles provide a unique discovery platform for combinatorial intracellular targeting of organelle ligands along with their corresponding receptors and for fingerprinting functional protein domains in living cells. Here we explain the design, cloning, construction and production of iPhage-based vectors and libraries, along with basic ligand-receptor identification and validation methodologies for organelle receptors. An iPhage library screening can be performed in ∼8 weeks.
Collapse
Affiliation(s)
- Roberto Rangel
- David H. Koch Center, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Andrey S. Dobroff
- David H. Koch Center, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Liliana Guzman-Rojas
- David H. Koch Center, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Carolina C. Salmeron
- David H. Koch Center, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Juri G. Gelovani
- Department of Biomedical Engineering, Wayne State University, Detroit, Michigan 48201, USA
| | - Richard L. Sidman
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Renata Pasqualini
- David H. Koch Center, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Wadih Arap
- David H. Koch Center, The University of Texas M.D. Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
68
|
Henry CJ, Bryan JN. Not lost in translation: how study of diseases in our pets can benefit them and us. MISSOURI MEDICINE 2013; 110:216-219. [PMID: 23829106 PMCID: PMC6179846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Practice-changing medical discovery requires preclinical and clinical assessment be carried out using appropriate disease models. There is growing awareness of companion animals with naturally-occurring disease as such models. They offer significant advantages over more traditional in vivo models of induced disease. This review describes current efforts to promote translation of discoveries between human and veterinary medicine in order to more rapidly and efficiently make progress in improving the health of all human and animal patients.
Collapse
Affiliation(s)
- Carolyn J Henry
- Department of Internal Medicine, Hematology/ Oncology Division, University of Missouri, USA.
| | | |
Collapse
|
69
|
Przystal JM, Umukoro E, Stoneham CA, Yata T, O'Neill K, Syed N, Hajitou A. Proteasome inhibition in cancer is associated with enhanced tumor targeting by the adeno-associated virus/phage. Mol Oncol 2013; 7:55-66. [PMID: 22951279 PMCID: PMC3553581 DOI: 10.1016/j.molonc.2012.08.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 07/31/2012] [Accepted: 08/01/2012] [Indexed: 12/18/2022] Open
Abstract
Bacteriophage (phage), which are viruses that infect bacteria only, have shown promise as vehicles for targeted cancer gene therapy, albeit with poor efficiency. Recently, we generated an improved version of phage vectors by incorporating cis genetic elements of adeno-associated virus (AAV). This novel AAV/phage hybrid (AAVP) efficiently delivered systemically administered therapeutic genes to various tumor targets by displaying an integrin tumor-targeting ligand on the phage capsid. However, inherent limitations in bacteriophage mean that these AAVP vectors still need to be improved. One of the limitations of AAVP in mammalian cells may be its susceptibility to proteasomal degradation. The proteasome is upregulated in cancer and it is known that it constitutes a barrier to gene delivery by certain eukaryotic viruses. We report here that inhibition of proteasome improved targeted reporter gene delivery by AAVP in cancer cells in vitro and in tumors in vivo after intravenous vector administration to tumor-bearing mice. We also show enhanced targeted tumor cell killing by AAVP upon proteasome inhibition. The AAVP particles persisted significantly in cancer cells in vitro and in tumors in vivo after systemic administration, and accumulated polyubiquitinated coat proteins. Our results suggest that the proteasome is indeed a barrier to tumor targeting by AAVP and indicate that a combination of proteasome-inhibiting drugs and AAVP should be considered for clinical anticancer therapy.
Collapse
Affiliation(s)
- Justyna M. Przystal
- Division of Brain Sciences, Hammersmith Hospital Campus, Department of Medicine, Imperial College London, Burlington Danes Building, 160 Du Cane Road, London W12 0NN, United Kingdom
| | - Eloho Umukoro
- Division of Brain Sciences, Hammersmith Hospital Campus, Department of Medicine, Imperial College London, Burlington Danes Building, 160 Du Cane Road, London W12 0NN, United Kingdom
| | - Charlotte A. Stoneham
- Division of Brain Sciences, Hammersmith Hospital Campus, Department of Medicine, Imperial College London, Burlington Danes Building, 160 Du Cane Road, London W12 0NN, United Kingdom
| | - Teerapong Yata
- Division of Brain Sciences, Hammersmith Hospital Campus, Department of Medicine, Imperial College London, Burlington Danes Building, 160 Du Cane Road, London W12 0NN, United Kingdom
| | - Kevin O'Neill
- Division of Brain Sciences, Charing Cross Campus, Department of Medicine, Imperial College London, London, United Kingdom
| | - Nelofer Syed
- Division of Brain Sciences, Charing Cross Campus, Department of Medicine, Imperial College London, London, United Kingdom
| | - Amin Hajitou
- Division of Brain Sciences, Hammersmith Hospital Campus, Department of Medicine, Imperial College London, Burlington Danes Building, 160 Du Cane Road, London W12 0NN, United Kingdom
| |
Collapse
|
70
|
Blockade of inhibitors of apoptosis (IAPs) in combination with tumor-targeted delivery of tumor necrosis factor-α leads to synergistic antitumor activity. Cancer Gene Ther 2012; 20:46-56. [PMID: 23154431 PMCID: PMC3534156 DOI: 10.1038/cgt.2012.83] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In the current study, we examined whether the combination of tumor vasculature-targeted gene therapy with adeno-associated virus bacteriophage-tumor necrosis factor-α (AAVP-TNF-α) and/or the orally administered LCL161, an antagonist of inhibitors of apoptosis proteins (IAPs), enhanced antitumor efficacy without systemic toxicity. M21 human melanoma xenografts were grown subcutaneously in nude mice. Mice were treated according to one of four treatment regimens: AAVP-TNF-α alone (AAVP-TNF-α plus sodium acetate-acetic acid (NaAc) buffer) via tail vein injection; LCL161 alone (phosphate-buffered saline (PBS) plus LCL161) via oral gavage; AAVP-TNF-α plus LCL161; and PBS plus NaAc Buffer as a control group. Tumor volume, survival and toxicity were analyzed. AAVP trafficking and TNF-α production in vivo were detected on days 7 and 21 by real-time PCR, enzyme-linked immunosorbent assay and immunofluorescence. The levels of apoptosis and activation of caspases were assessed on days 7 and 21 by TUNEL (terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling) and immunofluorescence assays. Our results showed that the combination of AAVP-TNF-α and LCL161 significantly inhibited tumor growth and prolonged survival in mice with melanoma xenografts. The combination of AAVP-TNF-α and LCL161 was also significantly more effective than either agent alone, showing a synergistic effect without systemic toxicity.
Collapse
|
71
|
Bentley RT, Mund JA, Pollok KE, Childress MO, Case J. Peripheral blood biomarkers of solid tumor angiogenesis in dogs: a polychromatic flow cytometry pilot study. Vet J 2012; 196:236-40. [PMID: 23063489 DOI: 10.1016/j.tvjl.2012.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 09/03/2012] [Accepted: 09/04/2012] [Indexed: 12/26/2022]
Abstract
A subset of peripheral blood hematopoietic stem and progenitor cells of bone marrow origin is elevated in humans with solid cancers before treatment and declines with therapy. This biomarker of angiogenesis is not specific to tumor type and has great potential in the objective assessment of treatment response in clinical trials. This pilot study was designed to develop a biomarker of neoangiogenesis in dogs for the diagnosis of cancer, the measurement of treatment response, and the provision of objective data in clinical trials. Polychromatic flow cytometry was used to quantify two subsets of circulating hematopoietic stem and progenitor cells in dogs with spontaneous solid tumors before (n = 8) and after (n = 3) treatment, and normal controls (n = 6). Pro-angiogenic peripheral blood cells of bone marrow origin were detected in all eight cases and the six normal controls; however, there was no statistically significant difference between the two groups. Interestingly, an apparent decline in pro-angiogenic cells was observed after treatment. Bone marrow derived hematopoietic cells appear to contribute to tumor angiogenesis in dogs, as has been previously reported in humans. While the methodology for pro-angiogenic cell quantification in a small number of dogs in the current study did not result in a significant difference from normal controls, an optimized canine polychromatic flow cytometry protocol holds great promise in the development of a canine cancer model and for the objective measurements of treatment response in clinical trials.
Collapse
Affiliation(s)
- R Timothy Bentley
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN 47905, USA.
| | | | | | | | | |
Collapse
|
72
|
Kia A, Przystal JM, Nianiaris N, Mazarakis ND, Mintz PJ, Hajitou A. Dual systemic tumor targeting with ligand-directed phage and Grp78 promoter induces tumor regression. Mol Cancer Ther 2012; 11:2566-77. [PMID: 23053496 DOI: 10.1158/1535-7163.mct-12-0587] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The tumor-specific Grp78 promoter is overexpressed in aggressive tumors. Cancer patients would benefit greatly from application of this promoter in gene therapy and molecular imaging; however, clinical benefit is limited by lack of strategies to target the systemic delivery of Grp78-driven transgenes to tumors. This study aims to assess the systemic efficacy of Grp78-guided expression of therapeutic and imaging transgenes relative to the standard cytomegalovirus (CMV) promoter. Combination of ligand and Grp78 transcriptional targeting into a single vector would facilitate systemic applications of the Grp78 promoter. We generated a dual tumor-targeted phage containing the arginine-glycine-aspartic acid tumor homing ligand and Grp78 promoter. Next, we combined flow cytometry, Western blot analysis, bioluminescence imaging of luciferase, and HSVtk/ganciclovir gene therapy and compared efficacy to conventional phage carrying the CMV promoter in vitro and in vivo in subcutaneous models of rat and human glioblastoma. We show that double-targeted phage provides persistent transgene expression in vitro and in tumors in vivo after systemic administration compared with conventional phage. Next, we showed significant tumor killing in vivo using the HSVtk/ganciclovir gene therapy and found a systemic antitumor effect of Grp78-driven HSVtk against therapy-resistant tumors. Finally, we uncovered a novel mechanism of Grp78 promoter activation whereby HSVtk/ganciclovir therapy upregulates Grp78 and transgene expression via the conserved unfolded protein response signaling cascade. These data validate the potential of Grp78 promoter in systemic cancer gene therapy and report the efficacy of a dual tumor targeting phage that may prove useful for translation into gene therapy and molecular imaging applications.
Collapse
Affiliation(s)
- Azadeh Kia
- Centre for Neuroinflammation and Degeneration, Division of Brain Sciences, Department of Medicine, Imperial College London, Hammersmith Hospital Campus, United Kingdom
| | | | | | | | | | | |
Collapse
|
73
|
Yang B, Papoian T. Tyrosine kinase inhibitor (TKI)-induced cardiotoxicity: approaches to narrow the gaps between preclinical safety evaluation and clinical outcome. J Appl Toxicol 2012; 32:945-51. [PMID: 22961481 DOI: 10.1002/jat.2813] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 07/18/2012] [Accepted: 07/18/2012] [Indexed: 12/29/2022]
Abstract
Although therapies targeted to inhibit the activity of certain tyrosine kinases (TK) have helped advance cancer therapy in recent years, reports of cardiac toxicity following treatment with tyrosine kinase inhibitors (TKIs) were unexpected and not well predicted by preclinical studies. Such clinical findings exposed gaps in current preclinical drug testing for predicting the development of cardiac toxicities in humans. These gaps included a lack of a comprehensive TKI mechanism of action determination and appropriate cardiac functional evaluation. New preclinical approaches are suggested to address these issues. In addition to tyrosine kinase inhibition, other factors that may play a role in drug-induced cardiac effects should be assessed, such as unintended secondary targets of TKIs, toxic drug metabolites and drug accumulation in the heart. Both on-target and off-target toxic effects of TKIs on cultured cardiac myocytes have now been shown to be detectable, providing a rationale for using cardiomyocytes as a screening tool to study potential TKI-mediated cardiotoxicity. Incorporating isolated perfused heart methodology to chronic/subchronic rodent studies or including echocardiography in chronic large animal toxicity studies may improve the detection of changes in cardiac function over current methods, and they may eventually become a routine tool for screening drugs with suspected cardiotoxic potential. Further, assessing drug toxicity and efficacy together in an animal model of disease is highly informative for candidate drug selection, and should be encouraged to assess specific safety endpoints, such as cardiovascular function. Together, these approaches will help better close the gaps between preclinical testing and clinical outcomes.
Collapse
Affiliation(s)
- Baichun Yang
- Center for Drug Evaluation and Research, Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA.
| | | |
Collapse
|
74
|
Stoneham CA, Hollinshead M, Hajitou A. Clathrin-mediated endocytosis and subsequent endo-lysosomal trafficking of adeno-associated virus/phage. J Biol Chem 2012; 287:35849-59. [PMID: 22915587 DOI: 10.1074/jbc.m112.369389] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Adeno-associated virus/phage (AAVP) is a gene delivery vector constructed as a hybrid between adeno-associated virus and filamentous phage. Tumor targeting following systemic administration has previously been demonstrated in several in vivo cancer models, with tumor specificity achieved through display of an α(v) integrin-targeting ligand on the capsid. However, high titers of AAVP are required for transduction of large numbers of mammalian cells. This study is the first to investigate the mechanisms involved in entry and intracellular trafficking of AAVP. Using a combination of flow cytometry, confocal, and electron microscopy techniques, together with pharmacological agents, RNAi and dominant negative mutants, we have demonstrated that targeted AAVP endocytosis is both dynamin and clathrin-dependent. Following entry, the majority of AAVP particles are sequestered by the endosomal-lysosomal degradative pathway. Finally, we have demonstrated that disruption of this pathway leads to improved transgene expression by AAVP, thus demonstrating that escape from the late endosomes/lysosomes is a critical step for improving gene delivery by AAVP. These findings have important implications for the rational design of improved AAVP and RGD-targeted vectors.
Collapse
Affiliation(s)
- Charlotte A Stoneham
- Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, 160 Du Cane Road, London W12 0NN, United Kingdom
| | | | | |
Collapse
|
75
|
Fields EL, Robertson ID, Osborne JA, Brown JC. Comparison of abdominal computed tomography and abdominal ultrasound in sedated dogs. Vet Radiol Ultrasound 2012; 53:513-7. [PMID: 22612269 DOI: 10.1111/j.1740-8261.2012.01949.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 04/09/2012] [Indexed: 11/28/2022] Open
Abstract
Abdominal ultrasound (US) is used frequently as a first-line screening tool for abdominal disease. Although computed tomography (CT) is superior to US in the diagnosis of some abdominal diseases, a major impediment is the requirement of general anesthesia to prevent motion and for safe restraint. With multidetector helical CT, faster examinations allow general anesthesia to be avoided, while producing diagnostic-quality images. Abdominal US and CT were compared for lesion detection in 27 sedated dogs, divided into three even groups based on body weight. Lesions were categorized further as to subjective clinical relevance. In dogs less than 25 kg, there is no significant difference in lesion detection between CT and US. In dogs weighing greater than 25 kg, more lesions were detected with CT than with US (P = 0.0001), including clinically relevant lesions (P = 0.0277). From these results, it appears that CT has an advantage in lesion detection in dogs greater than 25 kg, making it a better screening test for abdominal disease in these patients.
Collapse
Affiliation(s)
- Erica L Fields
- Department of Small Animal Clinical Sciences, University of Tennessee, Knoxville, TN, USA
| | | | | | | |
Collapse
|
76
|
Huang SH, Kozak PJ, Kim J, Habineza-Ndikuyeze G, Meade C, Gaurnier-Hausser A, Patel R, Robertson E, Mason NJ. Evidence of an oncogenic gammaherpesvirus in domestic dogs. Virology 2012; 427:107-17. [PMID: 22405628 DOI: 10.1016/j.virol.2012.02.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 01/13/2012] [Accepted: 02/08/2012] [Indexed: 12/12/2022]
Abstract
In humans, chronic infection with the gammaherpesvirus Epstein-Barr virus is usually asymptomatic; however some infected individuals develop hematological and epithelial malignancies. The exact role of EBV in lymphomagenesis is poorly understood partly because of the lack of clinically relevant animal models. Here we report the detection of serological responses against EBV capsid antigens in healthy dogs and dogs with spontaneous lymphoma and that dogs with the highest antibody titers have B cell lymphoma. Moreover, we demonstrate the presence of EBV-like viral DNA and RNA sequences and Latent Membrane Protein-1 in malignant lymph nodes of dogs with lymphoma. Finally, electron microscopy of canine malignant B cells revealed the presence of classic herpesvirus particles. These findings suggest that dogs can be naturally infected with an EBV-like gammaherpesvirus that may contribute to lymphomagenesis and that dogs might represent a spontaneous model to investigate environmental and genetic factors that influence gammaherpesvirus-associated lymphomagenesis in humans.
Collapse
Affiliation(s)
- Shih-Hung Huang
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104-6010, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Garden O, Pinheiro D, Cunningham F. All creatures great and small: regulatory T cells in mice, humans, dogs and other domestic animal species. Int Immunopharmacol 2011; 11:576-88. [DOI: 10.1016/j.intimp.2010.11.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 11/01/2010] [Indexed: 12/12/2022]
|
78
|
Gordon IK, Khanna C. Modeling opportunities in comparative oncology for drug development. ILAR J 2011; 51:214-20. [PMID: 21131722 DOI: 10.1093/ilar.51.3.214] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Successful development of novel cancer drugs depends on well-reasoned scientific drug discovery, rigorous preclinical development, and carefully conceived clinical trials. Failure in any of these steps contributes to poor rates of approval for new drugs to treat cancer. As technological and scientific advances have opened the door to a variety of novel approaches to cancer drug discovery and development, preclinical models that can answer questions about the activity and safety of novel therapies are increasingly necessary. The advance of a drug to clinical trials based on information from preclinical models presupposes that the models convey informative data for future use in human patients with cancer. The study of novel cancer drugs using in vitro models is highly controllable, reproducible, relatively inexpensive, and linked to high throughput. However, these models fail to reproduce many of the complex features of human cancer. Mouse models address some of these limitations but have important biological differences from human cancer. The integration of studies using pet dogs with spontaneously occurring tumors as models in the development path can answer questions not adequately addressed in conventional models and is therefore gaining attention and interest in drug development communities. The study of novel cancer drugs in dogs with naturally occurring tumors allows drug assessment in a cancer that shares many fundamental features with the human cancer condition, and thus provides an opportunity to answer questions that inform the cancer drug development path in ways not possible in more conventional models.
Collapse
Affiliation(s)
- Ira K Gordon
- NCI Center for Cancer Research, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
79
|
Yu H, Zhu GY, Xu RZ, Niu HZ, Lu Q, Li GZ, Wang ZY, Zhang DS, Gu N, Teng GJ. Arterial embolization hyperthermia using As2O3 nanoparticles in VX2 carcinoma-induced liver tumors. PLoS One 2011; 6:e17926. [PMID: 21448278 PMCID: PMC3063167 DOI: 10.1371/journal.pone.0017926] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 02/16/2011] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Combination therapy for arterial embolization hyperthermia (AEH) with arsenic trioxide (As(2)O(3)) nanoparticles (ATONs) is a novel treatment for solid malignancies. This study was performed to evaluate the feasibility and therapeutic effect of AEH with As(2)O(3) nanoparticles in a rabbit liver cancer model. The protocol was approved by our institutional animal use committee. METHODOLOGY/PRINCIPAL FINDINGS In total, 60 VX(2) liver-tumor-bearing rabbits were randomly assigned to five groups (n = 12/group) and received AEH with ATONs (Group 1), hepatic arterial embolization with ATONs (Group 2), lipiodol (Group 3), or saline (Group 4), on day 14 after tumor implantation. Twelve rabbits that received AEH with ATONs were prepared for temperature measurements, and were defined as Group 5. Computed tomography was used to measure the tumors' longest dimension, and evaluation was performed according to the Response Evaluation Criteria in Solid Tumors. Hepatic toxicity, tumor necrosis rate, vascular endothelial growth factor level, and microvessel density were determined. Survival rates were measured using the Kaplan-Meier method. The therapeutic temperature (42.5°C) was obtained in Group 5. Hepatotoxicity reactions occurred but were transient in all groups. Tumor growth was delayed and survival was prolonged in Group 1 (treated with AEH and ATONs). Plasma and tumor vascular endothelial growth factor and microvessel density were significantly inhibited in Group 1, while tumor necrosis rates were markedly enhanced compared with those in the control groups. CONCLUSIONS ATON-based AEH is a safe and effective treatment that can be targeted at liver tumors using the dual effects of hyperthermia and chemotherapy. This therapy can delay tumor growth and noticeably inhibit tumor angiogenesis.
Collapse
Affiliation(s)
- Hui Yu
- Jiangsu Key Laboratory of Molecular Imaging
and Functional Imaging, Department of Radiology, Zhong-Da Hospital, Medical
School of Southeast University, Nanjing, China
| | - Guang-Yu Zhu
- Jiangsu Key Laboratory of Molecular Imaging
and Functional Imaging, Department of Radiology, Zhong-Da Hospital, Medical
School of Southeast University, Nanjing, China
| | - Rui-Zhi Xu
- Jiangsu Laboratory for Biomaterials and
Devices, State Key Laboratory of BioElectronics, School of Biological Science
and Medical Engineering, Southeast University, Nanjing, China
| | - Huan-Zhang Niu
- Jiangsu Key Laboratory of Molecular Imaging
and Functional Imaging, Department of Radiology, Zhong-Da Hospital, Medical
School of Southeast University, Nanjing, China
| | - Qin Lu
- Jiangsu Key Laboratory of Molecular Imaging
and Functional Imaging, Department of Radiology, Zhong-Da Hospital, Medical
School of Southeast University, Nanjing, China
| | - Guo-Zhao Li
- Jiangsu Key Laboratory of Molecular Imaging
and Functional Imaging, Department of Radiology, Zhong-Da Hospital, Medical
School of Southeast University, Nanjing, China
| | - Zi-Yu Wang
- Department of Pathology and Pathophysiology,
Medical School of Southeast University, Nanjing, China
| | - Dong-Sheng Zhang
- Department of Pathology and Pathophysiology,
Medical School of Southeast University, Nanjing, China
| | - Ning Gu
- Jiangsu Laboratory for Biomaterials and
Devices, State Key Laboratory of BioElectronics, School of Biological Science
and Medical Engineering, Southeast University, Nanjing, China
| | - Gao-Jun Teng
- Jiangsu Key Laboratory of Molecular Imaging
and Functional Imaging, Department of Radiology, Zhong-Da Hospital, Medical
School of Southeast University, Nanjing, China
- * E-mail:
| |
Collapse
|
80
|
Weichselbaumer M, Willmann M, Reifinger M, Singer J, Bajna E, Sobanov Y, Mechtcherikova D, Selzer E, Thalhammer JG, Kammerer R, Jensen-Jarolim E. Phylogenetic discordance of human and canine carcinoembryonic antigen (CEA, CEACAM) families, but striking identity of the CEA receptors will impact comparative oncology studies. PLOS CURRENTS 2011; 3:RRN1223. [PMID: 21436956 PMCID: PMC3059814 DOI: 10.1371/currents.rrn1223] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/14/2011] [Indexed: 12/20/2022]
Abstract
Comparative oncology aims at speeding up developments for both, human and companion animal cancer patients. Following this line, carcinoembryonic antigen (CEA, CEACAM5) could be a therapeutic target not only for human but also for canine (Canis lupus familiaris; dog) patients. CEACAM5 interacts with CEA-receptor (CEAR) in the cytoplasm of human cancer cells. Our aim was, therefore, to phylogenetically verify the antigenic relationship of CEACAM molecules and CEAR in human and canine cancer. Anti-human CEACAM5 antibody Col-1, previously being applied for cancer diagnosis in dogs, immunohistochemically reacted to 23 out of 30 canine mammary cancer samples. In immunoblot analyses Col-1 specifically detected human CEACAM5 at 180 kDa in human colon cancer cells HT29, and the canine antigen at 60, 120, or 180 kDa in CF33 and CF41 mammary carcinoma cells as well as in spontaneous mammary tumors. While according to phylogenicity canine CEACAM1 molecules should be most closely related to human CEACAM5, Col-1 did not react with canine CEACAM1, -23, -24, -25, -28 or -30 transfected to canine TLM-1 cells. By flow cytometry the Col-1 target molecule was localized intracellularly in canine CF33 and CF41 cells, in contrast to membranous and cytoplasmic expression of human CEACAM5 in HT29. Col-1 incubation had neither effect on canine nor human cancer cell proliferation. Yet, Col-1 treatment decreased AKT-phosphorylation in canine CF33 cells possibly suggestive of anti-apoptotic function, whereas Col-1 increased AKT-phosphorylation in human HT29 cells. We report further a 99% amino acid similarity of human and canine CEA receptor (CEAR) within the phylogenetic tree. CEAR could be detected in four canine cancer cell lines by immunoblot and intracellularly in 10 out of 10 mammary cancer specimens from dog by immunohistochemistry. Whether the specific canine Col-1 target molecule may as functional analogue to human CEACAM5 act as ligand to canine CEAR, remains to be defined. This study demonstrates the limitations of comparative oncology due to the complex functional evolution of the different CEACAM molecules in humans versus dogs. In contrast, CEAR may be a comprehensive interspecies target for novel cancer therapeutics.
Collapse
Affiliation(s)
- Marlene Weichselbaumer
- Clinic for Internal Medicine & Infectious Diseases, Dept. 4, University of Veterinary Medicine Vienna and Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology & Immunology, Medical University of Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Arendt ML, Nasir L, Morgan IM. The human and canine TERT promoters function equivalently in human and canine cells. Vet Comp Oncol 2010; 8:310-6. [PMID: 21062413 DOI: 10.1111/j.1476-5829.2010.00227.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Telomerase targeted cancer gene therapy is being exploited for treatment of human cancer. The high incidence and many comparative aspects of human and canine cancer and the compliance and dedication of dog owners to treat cancer makes the canine pet population a good clinical model for investigating and developing new cancer therapeutics. Here, we report that the human telomerase promoter operates in canine cells, suggesting that human telomerase promoter-driven cancer therapy can be used to treat cancer in canines. Therefore, the canine pet population can act as a clinical model for new drug development based on telomerase therapeutics.
Collapse
Affiliation(s)
- M L Arendt
- Institute of Comparative Medicine, University of Glasgow, Glasgow, UK.
| | | | | |
Collapse
|
82
|
Wittenburg LA, Gustafson DL, Thamm DH. Phase I pharmacokinetic and pharmacodynamic evaluation of combined valproic acid/doxorubicin treatment in dogs with spontaneous cancer. Clin Cancer Res 2010; 16:4832-42. [PMID: 20705615 DOI: 10.1158/1078-0432.ccr-10-1238] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Histone deacetylase inhibitors (HDACi) are targeted anticancer agents with a well-documented ability to act synergistically with cytotoxic agents. We recently showed that the HDACi valproic acid sensitizes osteosarcoma cells to doxorubicin in vitro and in vivo. As there are no published reports on the clinical utility of HDACi in dogs with spontaneous cancers, we sought to determine a safe and biologically effective dose of valproic acid administered prior to a standard dose of doxorubicin. METHODS Twenty-one dogs were enrolled into eight cohorts in an accelerated dose-escalation trial consisting of pretreatment with oral valproic acid followed by doxorubicin on a three-week cycle. Blood and tumor tissue were collected for determination of serum valproic acid concentration and evaluation of pharmacodynamic effects by immunofluorescence cytochemistry and immunohistochemistry. Serum and complete blood counts were obtained for determination of changes in doxorubicin pharmacokinetics or hematologic effects. RESULTS All doses of valproic acid were well tolerated. Serum valproic acid concentrations increased linearly with dose. Doxorubicin pharmacokinetics were comparable with those in dogs receiving doxorubicin alone. A positive correlation was detected between valproic acid dose and histone hyperacetylation in peripheral blood mononuclear cells. No potentiation of doxorubicin-induced myelosuppression was observed. Histone hyperacetylation was documented in tumor and peripheral blood mononuclear cells. Responses included 2 of 21 complete, 3 of 21 partial, 5 of 21 stable disease, and 11 of 21 progressive disease. CONCLUSIONS Valproic acid can be administered to dogs at doses up to 240 mg/kg/day prior to a standard dose of doxorubicin. In addition, we have developed the pharmacokinetic/pharmacodynamic tools necessary for future studies of novel HDACi in the clinical setting of canine cancer.
Collapse
Affiliation(s)
- Luke A Wittenburg
- Animal Cancer Center, Department of Clinical Sciences, Colorado State University Animal Cancer Center, Fort Collins, USA.
| | | | | |
Collapse
|
83
|
Paoloni MC, Mazcko C, Fox E, Fan T, Lana S, Kisseberth W, Vail DM, Nuckolls K, Osborne T, Yalkowsy S, Gustafson D, Yu Y, Cao L, Khanna C. Rapamycin pharmacokinetic and pharmacodynamic relationships in osteosarcoma: a comparative oncology study in dogs. PLoS One 2010; 5:e11013. [PMID: 20543980 PMCID: PMC2882366 DOI: 10.1371/journal.pone.0011013] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2010] [Accepted: 02/24/2010] [Indexed: 11/18/2022] Open
Abstract
Background Signaling through the mTOR pathway contributes to growth, progression and chemoresistance of several cancers. Accordingly, inhibitors have been developed as potentially valuable therapeutics. Their optimal development requires consideration of dose, regimen, biomarkers and a rationale for their use in combination with other agents. Using the infrastructure of the Comparative Oncology Trials Consortium many of these complex questions were asked within a relevant population of dogs with osteosarcoma to inform the development of mTOR inhibitors for future use in pediatric osteosarcoma patients. Methodology/Principal Findings This prospective dose escalation study of a parenteral formulation of rapamycin sought to define a safe, pharmacokinetically relevant, and pharmacodynamically active dose of rapamycin in dogs with appendicular osteosarcoma. Dogs entered into dose cohorts consisting of 3 dogs/cohort. Dogs underwent a pre-treatment tumor biopsy and collection of baseline PBMC. Dogs received a single intramuscular dose of rapamycin and underwent 48-hour whole blood pharmacokinetic sampling. Additionally, daily intramuscular doses of rapamycin were administered for 7 days with blood rapamycin trough levels collected on Day 8, 9 and 15. At Day 8 post-treatment collection of tumor and PBMC were obtained. No maximally tolerated dose of rapamycin was attained through escalation to the maximal planned dose of 0.08 mg/kg (2.5 mg/30kg dog). Pharmacokinetic analysis revealed a dose-dependent exposure. In all cohorts modulation of the mTOR pathway in tumor and PBMC (pS6RP/S6RP) was demonstrated. No change in pAKT/AKT was seen in tumor samples following rapamycin therapy. Conclusions/Significance Rapamycin may be safely administered to dogs and can yield therapeutic exposures. Modulation pS6RP/S6RP in tumor tissue and PBMCs was not dependent on dose. Results from this study confirm that the dog may be included in the translational development of rapamycin and potentially other mTOR inhibitors. Ongoing studies of rapamycin in dogs will define optimal schedules for their use in cancer and evaluate the role of rapamycin use in the setting of minimal residual disease.
Collapse
Affiliation(s)
- Melissa C. Paoloni
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Christina Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Elizabeth Fox
- Division of Oncology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Timothy Fan
- University of Illinois, Urbana, Illinois, United States of America
| | - Susan Lana
- Colorado State University, Fort Collins, Colorado, United States of America
| | | | - David M. Vail
- University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kaylee Nuckolls
- Tumor Metastasis and Biology Section, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Tanasa Osborne
- Tumor Metastasis and Biology Section, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Samuel Yalkowsy
- University of Arizona College of Pharmacy, Tucson, Arizona, United States of America
| | - Daniel Gustafson
- Colorado State University, Fort Collins, Colorado, United States of America
| | - Yunkai Yu
- Molecular Targets Core, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Liang Cao
- Molecular Targets Core, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Chand Khanna
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
- Tumor Metastasis and Biology Section, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
84
|
Paoloni M, Lana S, Thamm D, Mazcko C, Withrow S. The creation of the Comparative Oncology Trials Consortium Pharmacodynamic Core: Infrastructure for a virtual laboratory. Vet J 2010; 185:88-9. [PMID: 20621714 DOI: 10.1016/j.tvjl.2010.04.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The National Cancer Institute-Comparative Oncology Trials Consortium (NCI-COTC) aims to inform the development path of novel drugs and biologicals for human cancer patients through their evaluation in dogs with neoplasia. The advent of sophisticated clinical trials in veterinary medicine requires additional infrastructure to evaluate tissue and fluid end-points vital to questions relating to drug activity, targeting and toxicity. Pharmacokinetic and pharmacodynamic end-points necessitate a centralized laboratory for quality controlled assay development and execution. Establishing the COTC Pharmacodynamic Core (PD Core) has addressed the need for uniform end-point analysis by serving as a virtual laboratory that capitalizes on the expertise of the COTC community of investigators. Veterinary biomarker validation is a secondary benefit of these efforts. The PD Core exemplifies the construction of a successful infrastructure within the veterinary research community in line with advances in technology and focused on improving the health and quality of life of both human and animal cancer patients.
Collapse
Affiliation(s)
- Melissa Paoloni
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | |
Collapse
|
85
|
Abstract
It has been difficult to identify the molecular features central to the pathogenesis of osteosarcoma owing to a lack of understanding of the cell or origin, the absence of identifiable precursor lesions, and its marked genetic complexity at the time of presentation. Interestingly, several human genetic disorders and familial cancer syndromes, such as Li-Fraumeni syndrome, are linked to an increased risk of osteosarcoma. Association of these same genetic alterations and osteosarcoma risk have been confirmed in murine models. Osteosarcoma is associated with a variety of genetic abnormalities that are among the most commonly observed in human cancer; it remains unclear, however, what events initiate and are necessary to form osteosarcoma. The availability of new resources for studying osteosarcoma and newer research methodologies offer an opportunity and promise to answer these currently unanswered questions. Even in the absence of a more fundamental understanding of osteosarcoma, association studies and preclinical drug testing may yield clinically relevant information.
Collapse
Affiliation(s)
- Richard Gorlick
- Department of Pediatrics and Molecular Pharmacology, The Albert Einstein College of Medicine, Yeshiva University, Department of Pediatrics, Children's Hospital at Montefiore, Bronx, NY 10467, USA.
| | | |
Collapse
|
86
|
Abstract
The various types of cells that comprise the tumor mass all carry molecular markers that are not expressed or are expressed at much lower levels in normal cells. These differentially expressed molecules can be used as docking sites to concentrate drug conjugates and nanoparticles at tumors. Specific markers in tumor vessels are particularly well suited for targeting because molecules at the surface of blood vessels are readily accessible to circulating compounds. The increased concentration of a drug in the site of disease made possible by targeted delivery can be used to increase efficacy, reduce side effects, or achieve some of both. We review the recent advances in this delivery approach with a focus on the use of molecular markers of tumor vasculature as the primary target and nanoparticles as the delivery vehicle.
Collapse
Affiliation(s)
- Erkki Ruoslahti
- Vascular Mapping Center, Sanford-Burnham Medical Research Institute, University of California, Santa Barbara, Santa Barbara, CA 93106, USA.
| | | | | |
Collapse
|
87
|
Sreeramoju P, Libutti SK. Strategies for targeting tumors and tumor vasculature for cancer therapy. ADVANCES IN GENETICS 2010; 69:135-52. [PMID: 20807606 DOI: 10.1016/s0065-2660(10)69015-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Effective cancer therapy remains a challenge despite recent advances in the identification of novel targets. A major limitation of most chemotherapeutic drugs is their systemic toxicity and the efficacy of cancer treatments is, by and large, determined by the ability to balance their benefits against their toxicity. Targeted treatments for cancer, especially those that target the tumor vasculature, have provided promising antitumor results with minimal systemic toxicity. To date significant progress has been made in developing a variety of delivery systems to target cancer and its vasculature ranging from isolated limb and organ perfusion to tumor targeted biological and nonbiological vectors.
Collapse
Affiliation(s)
- Prashanth Sreeramoju
- Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | | |
Collapse
|
88
|
Hajitou A. Targeted systemic gene therapy and molecular imaging of cancer contribution of the vascular-targeted AAVP vector. ADVANCES IN GENETICS 2010; 69:65-82. [PMID: 20807602 DOI: 10.1016/s0065-2660(10)69008-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gene therapy and molecular-genetic imaging have faced a major problem: the lack of an efficient systemic gene delivery vector. Unquestionably, eukaryotic viruses have been the vectors of choice for gene delivery to mammalian cells; however, they have had limited success in systemic gene therapy. This is mainly due to undesired uptake by the liver and reticuloendothelial system, broad tropism for mammalian cells causing toxicity, and their immunogenicity. On the other hand, prokaryotic viruses such as bacteriophage (phage) have no tropism for mammalian cells, but can be engineered to deliver genes to these cells. However, phage-based vectors have inherently been considered poor vectors for mammalian cells. We have reported a new generation of vascular-targeted systemic hybrid prokaryotic-eukaryotic vectors as chimeras between an adeno-associated virus (AAV) and targeted bacteriophage (termed AAV/phage; AAVP). In this hybrid vector, the targeted bacteriophage serves as a shuttle to deliver the AAV transgene cassette inserted in an intergenomic region of the phage DNA genome. As a proof of concept, we assessed the in vivo efficacy of vector in animal models of cancer by displaying on the phage capsid the cyclic Arg-Gly-Asp (RGD-4C) ligand that binds to alphav integrin receptors specifically expressed on the angiogenic blood vessels of tumors. The ligand-directed vector was able to specifically deliver imaging and therapeutic transgenes to tumors in mice, rats, and dogs while sparing the normal organs. This chapter reviews some gene transfer strategies and the potential of the vascular-targeted AAVP vector for enhancing the effectiveness of existing systemic gene delivery and genetic-imaging technologies.
Collapse
Affiliation(s)
- Amin Hajitou
- Department of Gene Therapy, Section/ Division of Infectious Diseases, Faculty of Medicine, Imperial College London, Wright-Fleming Institute, St Mary's Campus, Norfolk Place, London, United Kingdom
| |
Collapse
|
89
|
Abstract
As the tumor vasculature is a key element of the tumor stroma, angiogenesis is the target of many cancer therapies. Recent work published in BMC Cell Biology describes a fusion protein that combines a peptide previously shown to home in on the gastric cancer vasculature with the anti-tumor cytokine TNF-α, and assesses its potential for gastric cancer therapy.
Collapse
Affiliation(s)
- Elizabeth R Burton
- Montefiore Medical Center, Medical Arts Pavilion, 3400 Bainbridge Avenue, MMC-MAP, Bronx, NY 10467, USA
| | | |
Collapse
|
90
|
Gordon I, Paoloni M, Mazcko C, Khanna C. The Comparative Oncology Trials Consortium: using spontaneously occurring cancers in dogs to inform the cancer drug development pathway. PLoS Med 2009; 6:e1000161. [PMID: 19823573 PMCID: PMC2753665 DOI: 10.1371/journal.pmed.1000161] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Chand Khanna and colleagues describe the work of the Comparative Oncology Trials Consortium (COTC), which provides infrastructure and resources to integrate naturally occurring dog cancer models into the development of new human cancer drugs, devices, and imaging techniques.
Collapse
Affiliation(s)
- Ira Gordon
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Melissa Paoloni
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Christina Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Chand Khanna
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
91
|
Driessen WHP, Ozawa MG, Arap W, Pasqualini R. Ligand-directed cancer gene therapy to angiogenic vasculature. ADVANCES IN GENETICS 2009; 67:103-121. [PMID: 19914451 PMCID: PMC7172741 DOI: 10.1016/s0065-2660(09)67004-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Gene therapy strategies in cancer have remained an active area of preclinical and clinical research. One of the current limitations to successful trials is the relative transduction efficiency to produce a therapeutic effect. While intratumoral injections are the mainstay of many treatment regimens to date, this approach is hindered by hydrostatic pressures within the tumor and is not always applicable to all tumor subtypes. Vascular-targeting strategies introduce an alternative method to deliver vectors with higher local concentrations and minimization of systemic toxicity. Moreover, therapeutic targeting of angiogenic vasculature often leads to enhanced bystander effects, improving efficacy. While identification of functional and systemically accessible molecular targets is challenging, approaches, such as in vivo phage display and phage-based viral delivery vectors, provide a platform upon which vascular targeting of vectors may become a viable and translational approach.
Collapse
Affiliation(s)
- Wouter H P Driessen
- David H. Koch Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Michael G Ozawa
- David H. Koch Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Wadih Arap
- David H. Koch Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | - Renata Pasqualini
- David H. Koch Center, The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|