101
|
Brandão J, Blair R, Kelly A, Fowlkes N, Shiomitsu K, Espinheira Gomes F, Rich G, Tully TN. Amelanotic Melanoma in the Rabbit: A Case Report With an Overview of Immunohistochemical Characterization. J Exot Pet Med 2015. [DOI: 10.1053/j.jepm.2015.04.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
102
|
Boston SE, Lu X, Culp WTN, Montinaro V, Romanelli G, Dudley RM, Liptak JM, Mestrinho LA, Buracco P. Efficacy of systemic adjuvant therapies administered to dogs after excision of oral malignant melanomas: 151 cases (2001-2012). J Am Vet Med Assoc 2015; 245:401-7. [PMID: 25075823 DOI: 10.2460/javma.245.4.401] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To determine prognostic factors for and compare outcome among dogs with oral malignant melanoma following excision with or without various systemic adjuvant therapies. DESIGN Retrospective case series. ANIMALS 151 dogs with naturally occurring oral malignant melanomas treated by excision with or without adjuvant therapies from 2001 to 2012. PROCEDURES Case accrual was solicited from Veterinary Society of Surgical Oncology members via an email list service. Information collected from case records included signalment, tumor staging, tumor characteristics, type of surgical excision, histologic diagnosis, adjuvant therapy, and survival time. RESULTS The overall median survival time was 346 days. Results of multivariate analysis indicated that tumor size, patient age, and intralesional excision (vs marginal, wide, or radical excision) were considered poor prognostic indicators. All other demographic and clinical variables were not significantly associated with survival time after adjusting for the aforementioned 3 variables. A clear survival benefit was not evident with any systemic adjuvant therapy, including vaccination against melanoma or chemotherapy; however, the number of dogs in each treatment group was small. Ninety-eight dogs received no postoperative adjuvant therapy, and there was no difference in survival time between dogs that did (335 days) and did not (352 days) receive systemic adjuvant therapy. CONCLUSIONS AND CLINICAL RELEVANCE For dogs with oral malignant melanoma, increasing tumor size and age were negative prognostic factors. Complete excision of all macroscopic tumor burden improved survival time. Long-term survival was possible following surgery alone. Although systemic adjuvant therapy was not found to improve survival time, this could have been due to type II error.
Collapse
Affiliation(s)
- Sarah E Boston
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608
| | | | | | | | | | | | | | | | | |
Collapse
|
103
|
Killick DR, Stell AJ, Catchpole B. Immunotherapy for canine cancer--is it time to go back to the future? J Small Anim Pract 2015; 56:229-41. [PMID: 25704119 DOI: 10.1111/jsap.12336] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 12/04/2014] [Accepted: 12/17/2014] [Indexed: 12/19/2022]
Abstract
Over the last 50 years, the significance of the immune system in the development and control of cancer has been much debated. However, recent discoveries provide evidence for a role of immunological mechanisms in the detection and destruction of cancer cells. Forty years ago veterinary oncologists were already investigating the feasibility of treating neoplasia by enhancing anticancer immunity. Unfortunately, this research was hindered by lack of a detailed understanding of cancer immunology, this limited the specificity and success of these early approaches. The great forward strides made in our understanding of onco-immunology in recent years have provided the impetus for a resurgence of interest in anticancer immunotherapy for canine patients. In this article both these initial trials and the exciting novel immunotherapeutics currently in development are reviewed.
Collapse
Affiliation(s)
- D R Killick
- School of Veterinary Science, University of Liverpool, Neston, CH64 7TE
| | | | | |
Collapse
|
104
|
Gibson HM, Veenstra JJ, Jones R, Vaishampayan U, Sauerbrey M, Bepler G, Lum L, Reyes J, Weise A, Wei WZ. Induction of HER2 Immunity in Outbred Domestic Cats by DNA Electrovaccination. Cancer Immunol Res 2015; 3:777-86. [PMID: 25711535 DOI: 10.1158/2326-6066.cir-14-0175] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 02/14/2015] [Indexed: 12/20/2022]
Abstract
Domestic cats share human living environments and genetic traits. They develop spontaneous feline mammary carcinoma (FMC) with similar histopathology to human breast cancer. HER2 and AKT phosphorylation was demonstrated in primary FMC by immunoblot analysis, indicating HER2 as a therapeutic target. FMC lines K12 and K248 expressing HER1, HER2, and HER3 were sensitive to receptor tyrosine kinase (RTK) inhibitors gefitinib and lapatinib. To test HER2 vaccine response in cats, purpose-bred, healthy cats were electrovaccinated with heterologous (xenogeneic) or point-mutated feline HER2 DNA. T-cell reactivity to feline self-HER2 was detected in 4 of 10 cats that received bear HER2, human-rat fusion HER2 (E2Neu) or mutant feline HER2 (feHER2-K), which contains a single amino acid substitution. The variable T-cell responses may resemble that in the genetically heterogeneous human population. All immune sera to heterologous HER2 recognized feline HER2 expressed in 3T3 cells (3T3/HER2), but not that in FMC K12 or K248. Immune sera to mutant pfeHER2-K bound 3T3/HER2 cells weakly, but they showed better recognition of K12 and K248 cells that also express HER1 and HER3, suggesting distinct HER2 epitopes displayed by FMC that may be simulated by feHER2-K. In summary, HER2 DNA electroporation overcomes T-cell immune tolerance in approximately 40% of healthy cats and induces antibodies with distinct specificity. Vaccination studies in domestic cats can expedite vaccine iteration to guide human vaccine design and better predict outcome, with the added benefit of helping feline mammary tumor patients.
Collapse
Affiliation(s)
- Heather M Gibson
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Jesse J Veenstra
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Richard Jones
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Ulka Vaishampayan
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | | | - Gerold Bepler
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Lawrence Lum
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Joyce Reyes
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Amy Weise
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Wei-Zen Wei
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan.
| |
Collapse
|
105
|
Hartley AN, Tarleton RL. Chemokine receptor 7 (CCR7)-expression and IFNγ production define vaccine-specific canine T-cell subsets. Vet Immunol Immunopathol 2015; 164:127-36. [PMID: 25758065 DOI: 10.1016/j.vetimm.2015.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 01/22/2015] [Accepted: 02/02/2015] [Indexed: 12/31/2022]
Abstract
Canines suffer from and serve as strong translational animals models for many immunological disorders and infectious diseases. Routine vaccination has been a mainstay of protecting dogs through the stimulation of robust antibody responses and expansion of memory T-cell populations. Commercially available reagents and described techniques are limited for identifying and characterizing canine T-cell subsets and evaluating T-cell-specific effector function. To define reagents for delineating naïve versus activated T-cells and identify antigen-specific T-cells, we tested anti-human and anti-bovine T-cell specific cell surface marker reagents for cross-reactivity with canine peripheral blood mononuclear cells (PBMCs. Both CD4(+) and CD8(+) T-cells from healthy canine donors showed reactivity to CCL19-Ig, a CCR7 ligand, and coexpression with CD62L. An in vitro stimulation with concanavalin A validated downregulation of CCR7 and CD62L expression on stimulated healthy control PBMCs, consistent with an activated T-cell phenotype. Anti-IFNγ antibodies identified antigen-specific IFNγ-producing CD4(+) and CD8(+) T-cells upon in vitro vaccine antigen PBMC stimulation. PBMC isolation within 24h of sample collection allowed for efficienT-cell recovery and accurate T-cell effector function characterization. These data provide a reagent and techniques platform via flow cytometry for identifying canine T-cell subsets and characterizing circulating antigen-specific canine T-cells for potential use in diagnostic and field settings.
Collapse
Affiliation(s)
- Ashley N Hartley
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, 30602, USA; Department of Infectious Diseases, University of Georgia, Athens, GA, 30602, USA
| | - Rick L Tarleton
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, GA, 30602, USA; Department of Cellular Biology, University of Georgia, Athens, GA, 30602, USA.
| |
Collapse
|
106
|
Riccardo F, Aurisicchio L, Impellizeri JA, Cavallo F. The importance of comparative oncology in translational medicine. Cancer Immunol Immunother 2015; 64:137-48. [PMID: 25548094 PMCID: PMC11029667 DOI: 10.1007/s00262-014-1645-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 12/15/2014] [Indexed: 12/20/2022]
Abstract
Human cancer is so complex that in vivo preclinical models are needed if effective therapies are to be developed. Naturally occurring cancers in companion animals are therefore a great resource, as shown by the remarkable growth that comparative oncology has seen over the last 30 years. Cancer has become a leading cause of death in companion animals now that more pets are living long enough to develop the disease. Furthermore, more owners are seeking advanced and novel therapies for their pets as they are very much considered family members. Living in the same environments, pets and humans are often afflicted by the same types of cancer which show similar behavior and, in some species, express the same antigen molecules. The treatment of pet tumors using novel therapies is of compelling translational significance.
Collapse
Affiliation(s)
- Federica Riccardo
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126 Turin, Italy
| | | | | | - Federica Cavallo
- Department of Molecular Biotechnologies and Health Sciences, Molecular Biotechnology Center, University of Turin, Via Nizza, 52, 10126 Turin, Italy
| |
Collapse
|
107
|
Tuohy JL, Selmic LE, Worley DR, Ehrhart NP, Withrow SJ. Outcome following curative-intent surgery for oral melanoma in dogs: 70 cases (1998–2011). J Am Vet Med Assoc 2014; 245:1266-73. [DOI: 10.2460/javma.245.11.1266] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
108
|
Clinical trials of immunogene therapy for spontaneous tumors in companion animals. ScientificWorldJournal 2014; 2014:718520. [PMID: 25506617 PMCID: PMC4251357 DOI: 10.1155/2014/718520] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/02/2014] [Indexed: 12/23/2022] Open
Abstract
Despite the important progress obtained in the treatment of some pets' malignancies, new treatments need to be developed. Being critical in cancer control and progression, the immune system's appropriate modulation may provide effective therapeutic options. In this review we summarize the outcomes of published immunogene therapy veterinary clinical trials reported by many research centers. A variety of tumors such as canine melanoma, soft tissue sarcomas, osteosarcoma and lymphoma, feline fibrosarcoma, and equine melanoma were subjected to different treatment approaches. Both viral and mainly nonviral vectors were used to deliver gene products as cytokines, xenogeneic tumor associated antigens, specific ligands, and proapoptotic regulatory factors. In some cases autologous, allogenic, or xenogeneic transgenic cytokine producing cells were assayed. In general terms, minor or no adverse collateral effects appeared during this kind of therapies and treated patients usually displayed a better course of the disease (longer survival, delayed or suppressed recurrence or metastatic spread, and improvement of the quality of life). This suggests the utility of these methodologies as standard adjuvant treatments. The encouraging outcomes obtained in companion animals support their ready application in veterinary clinical oncology and serve as preclinical proof of concept and safety assay for future human gene therapy trials.
Collapse
|
109
|
Novel and enhanced anti-melanoma DNA vaccine targeting the tyrosinase protein inhibits myeloid-derived suppressor cells and tumor growth in a syngeneic prophylactic and therapeutic murine model. Cancer Gene Ther 2014; 21:507-17. [DOI: 10.1038/cgt.2014.56] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Accepted: 09/30/2014] [Indexed: 12/29/2022]
|
110
|
Cancedda S, Rohrer Bley C, Aresu L, Dacasto M, Leone VF, Pizzoni S, Gracis M, Marconato L. Efficacy and side effects of radiation therapy in comparison with radiation therapy and temozolomide in the treatment of measurable canine malignant melanoma. Vet Comp Oncol 2014; 14:e146-e157. [DOI: 10.1111/vco.12122] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 08/17/2014] [Accepted: 09/16/2014] [Indexed: 01/13/2023]
Affiliation(s)
- S. Cancedda
- Centro Oncologico Veterinario; Sasso Marconi Italy
| | - C. Rohrer Bley
- Division of Radiation Oncology, Vetsuisse-Faculty; University of Zurich; Zurich Switzerland
| | - L. Aresu
- Department of Comparative Biomedicine and Food Science; University of Padua; Legnaro Italy
| | - M. Dacasto
- Department of Comparative Biomedicine and Food Science; University of Padua; Legnaro Italy
| | - V. F. Leone
- Centro Oncologico Veterinario; Sasso Marconi Italy
| | - S. Pizzoni
- Centro Oncologico Veterinario; Sasso Marconi Italy
| | - M. Gracis
- Clinica Veterinaria San Siro; Milan Italy
| | - L. Marconato
- Centro Oncologico Veterinario; Sasso Marconi Italy
| |
Collapse
|
111
|
McCrudden CM, McCarthy HO. Current status of gene therapy for breast cancer: progress and challenges. Appl Clin Genet 2014; 7:209-20. [PMID: 25419154 PMCID: PMC4234158 DOI: 10.2147/tacg.s54992] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Breast cancer is characterized by a series of genetic mutations and is therefore ideally placed for gene therapy intervention. The aim of gene therapy is to deliver a nucleic acid-based drug to either correct or destroy the cells harboring the genetic aberration. More recently, cancer gene therapy has evolved to also encompass delivery of RNA interference technologies, as well as cancer DNA vaccines. However, the bottleneck in creating such nucleic acid pharmaceuticals lies in the delivery. Deliverability of DNA is limited as it is prone to circulating nucleases; therefore, numerous strategies have been employed to aid with biological transport. This review will discuss some of the viral and nonviral approaches to breast cancer gene therapy, and present the findings of clinical trials of these therapies in breast cancer patients. Also detailed are some of the most recent developments in nonviral approaches to targeting in breast cancer gene therapy, including transcriptional control, and the development of recombinant, multifunctional bio-inspired systems. Lastly, DNA vaccines for breast cancer are documented, with comment on requirements for successful pharmaceutical product development.
Collapse
|
112
|
Abstract
Recombinant nucleic acids are considered as promising next-generation vaccines. These vaccines express the native antigen upon delivery into tissue, thus mimicking live attenuated vaccines without having the risk of reversion to pathogenicity. They also stimulate the innate immune system, thus potentiating responses. Nucleic acid vaccines are easy to produce at reasonable cost and are stable. During the past years, focus has been on the use of plasmid DNA for vaccination. Now mRNA and replicon vaccines have come into focus as promising technology platforms for vaccine development. This review discusses self-replicating RNA vaccines developed from alphavirus expression vectors. These replicon vaccines can be delivered as RNA, DNA or as recombinant virus particles. All three platforms have been pre-clinically evaluated as vaccines against a number of infectious diseases and cancer. Results have been very encouraging and propelled the first human clinical trials, the results of which have been promising.
Collapse
Affiliation(s)
- Karl Ljungberg
- Department of Microbiology, Tumor and Cell Biology Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
113
|
Kochetov AV. The alien replicon: Artificial genetic constructs to direct the synthesis of transmissible self-replicating RNAs. Bioessays 2014; 36:1204-12. [DOI: 10.1002/bies.201400111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Alex V. Kochetov
- Institute of Cytology & Genetics, SB RAS; Novosibirsk Russia
- Novosibirsk State University; Novosibirsk Russia
| |
Collapse
|
114
|
Chon E, Flanagan B, de Sá Rodrigues LC, Piskun C, Stein TJ. 6-Bromoindirubin-3'oxime (BIO) decreases proliferation and migration of canine melanoma cell lines. Vet J 2014; 205:305-12. [PMID: 25130776 DOI: 10.1016/j.tvjl.2014.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 07/14/2014] [Accepted: 07/15/2014] [Indexed: 01/06/2023]
Abstract
Despite recent therapeutic advances, malignant melanoma is an aggressive tumor in dogs and is associated with a poor outcome. Novel, targeted agents are necessary to improve survival. In this study, 6-bromoindirubin-3'-oxime (BIO), a serine/threonine kinase inhibitor with reported specificity for glycogen synthase kinase-3 beta (GSK-3β) inhibition, was evaluated in vitro in three canine melanoma cell lines (CML-10C2, UCDK9M2, and UCDK9M3) for β-catenin-mediated transcriptional activity, Axin2 gene and protein expression levels, cell proliferation, chemotoxicity, migration and invasion assays. BIO treatment of canine malignant melanoma cell lines at 5 µM for 72 h enhanced β-catenin-mediated transcriptional activity, suggesting GSK-3β inhibition, and reduced cell proliferation and migration. There were no significant effects on invasion, chemotoxicity, or apoptosis. The results suggest that serine/threonine kinases may be viable therapeutic targets for the treatment of canine malignant melanoma.
Collapse
Affiliation(s)
- Esther Chon
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Brandi Flanagan
- College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Lucas Campos de Sá Rodrigues
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Caroline Piskun
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Timothy J Stein
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA; Institute for Clinical & Translational Research, University of Wisconsin-Madison, Madison, WI, USA; Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
115
|
Abstract
Tumor immunology and immunotherapy is one of the most exciting and rapidly expanding fields. The immune system is divided into 2 primary components: the innate immune response and the highly specific, but more slowly developing, adaptive or acquired immune response. Immune responses are separated by whether they are induced by exposure to a foreign antigen (active response) or transferred through serum or lymphocytes from an immunized individual (passive response). The ideal cancer immunotherapy agent should discriminate between cancer and normal cells (specificity), be potent enough to kill small or large numbers of tumor cells (sensitivity), and prevent recurrence of a tumor (durability).
Collapse
Affiliation(s)
- Philip J Bergman
- Clinical Studies, VCA, 546 Bedford Road, Bedford Hills, New York, NY 10507, USA; Department of Molecular Pharmacology & Chemistry, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
116
|
Breit MN, Kisseberth WC, Bear MD, Landesman Y, Kashyap T, McCauley D, Kauffman MG, Shacham S, London CA. Biologic activity of the novel orally bioavailable selective inhibitor of nuclear export (SINE) KPT-335 against canine melanoma cell lines. BMC Vet Res 2014; 10:160. [PMID: 25022346 PMCID: PMC4105800 DOI: 10.1186/1746-6148-10-160] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 07/01/2014] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Exportin 1 (XPO1, also known as CRM1), is a chaperone protein responsible for the export of over 200 target proteins out of the nucleus. The expression and activity of XPO1 is upregulated in several human cancers and its expression is also linked to the development of chemotherapy resistance. Recent studies using both human and murine cancer cell lines have demonstrated that XPO1 is a relevant target for therapeutic intervention. The present study sought to characterize the biologic activity of an orally bioavailable selective inhibitor of nuclear export (SINE), KPT-335, against canine melanoma cell lines as a prelude to future clinical trials in dogs with melanoma. RESULTS We evaluated the effects of KPT-335 on 4 canine malignant melanoma cell lines and found that KPT-335 inhibited proliferation, blocked colony formation, and induced apoptosis of treated cells at biologically relevant concentrations of drug. Additionally, KPT-335 downregulated XPO1 protein while inducing a concomitant increase in XPO1 messenger RNA. Lastly, KPT-335 treatment of cell lines upregulated the expression of both protein and mRNA for the tumor suppressor proteins p53 and p21, and promoted their nuclear localization. CONCLUSIONS KPT-335 demonstrates biologic activity against canine melanoma cell lines at physiologically relevant doses, suggesting that KPT-335 may represent a viable treatment option for dogs with malignant melanoma.
Collapse
Affiliation(s)
- Megan N Breit
- Departments of Veterinary Biosciences and Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, 1925 Coffey Rd., Columbus, OH 43210, USA
| | - William C Kisseberth
- Departments of Veterinary Biosciences and Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, 1925 Coffey Rd., Columbus, OH 43210, USA
| | - Misty D Bear
- Departments of Veterinary Biosciences and Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, 1925 Coffey Rd., Columbus, OH 43210, USA
| | | | | | | | | | | | - Cheryl A London
- Departments of Veterinary Biosciences and Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, 1925 Coffey Rd., Columbus, OH 43210, USA
| |
Collapse
|
117
|
Cavallo F, Aurisicchio L, Mancini R, Ciliberto G. Xenogene vaccination in the therapy of cancer. Expert Opin Biol Ther 2014; 14:1427-42. [DOI: 10.1517/14712598.2014.927433] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
118
|
Bailey-Kellogg C, Gutiérrez AH, Moise L, Terry F, Martin WD, De Groot AS. CHOPPI: a web tool for the analysis of immunogenicity risk from host cell proteins in CHO-based protein production. Biotechnol Bioeng 2014; 111:2170-82. [PMID: 24888712 PMCID: PMC4282101 DOI: 10.1002/bit.25286] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 04/14/2014] [Accepted: 05/07/2014] [Indexed: 02/04/2023]
Abstract
Despite high quality standards and continual process improvements in manufacturing, host cell protein (HCP) process impurities remain a substantial risk for biological products. Even at low levels, residual HCPs can induce a detrimental immune response compromising the safety and efficacy of a biologic. Consequently, advanced-stage clinical trials have been cancelled due to the identification of antibodies against HCPs. To enable earlier and rapid assessment of the risks in Chinese Hamster Ovary (CHO)-based protein production of residual CHO protein impurities (CHOPs), we have developed a web tool called CHOPPI, for CHO Protein Predicted Immunogenicity. CHOPPI integrates information regarding the possible presence of CHOPs (expression and secretion) with characterizations of their immunogenicity (T cell epitope count and density, and relative conservation with human counterparts). CHOPPI can generate a report for a specified CHO protein (e.g., identified from proteomics or immunoassays) or characterize an entire specified subset of the CHO genome (e.g., filtered based on confidence in transcription and similarity to human proteins). The ability to analyze potential CHOPs at a genomic scale provides a baseline to evaluate relative risk. We show here that CHOPPI can identify clear differences in immunogenicity risk among previously validated CHOPs, as well as identify additional “risky” CHO proteins that may be expressed during production and induce a detrimental immune response upon delivery. We conclude that CHOPPI is a powerful tool that provides a valuable computational complement to existing experimental approaches for CHOP risk assessment and can focus experimental efforts in the most important directions. Biotechnol. Bioeng. 2014;111: 2170–2182.
Collapse
|
119
|
Kallen KJ, Theß A. A development that may evolve into a revolution in medicine: mRNA as the basis for novel, nucleotide-based vaccines and drugs. THERAPEUTIC ADVANCES IN VACCINES 2014; 2:10-31. [PMID: 24757523 DOI: 10.1177/2051013613508729] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recent advances strongly suggest that mRNA rather than DNA will be the nucleotide basis for a new class of vaccines and drugs. Therapeutic cancer vaccines against a variety of targets have been developed on this basis and initial clinical experience suggests that preclinical activity can be successfully translated to human application. Likewise, prophylactic vaccines against viral pathogens and allergens have demonstrated their activity in animal models. These successes could be extended preclinically to mRNA protein and gene replacement therapy as well as the induction of pluripotent stem cells by mRNA encoded transcription factors. The production of mRNA-based vaccines and drugs is highly flexible, scalable and cost competitive, and eliminates the requirement of a cold chain. mRNA-based drugs and vaccines offer all the advantages of a nucleotide-based approach at reduced costs and represent a truly disruptive technology that may start a revolution in medicine.
Collapse
|
120
|
O'Connor CM, Wilson-Robles H. Developing T Cell Cancer Immunotherapy in the Dog with Lymphoma. ILAR J 2014; 55:169-81. [DOI: 10.1093/ilar/ilu020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
121
|
Denies S, Cicchelero L, Van Audenhove I, Sanders NN. Combination of interleukin-12 gene therapy, metronomic cyclophosphamide and DNA cancer vaccination directs all arms of the immune system towards tumor eradication. J Control Release 2014; 187:175-82. [PMID: 24887014 DOI: 10.1016/j.jconrel.2014.05.045] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 05/15/2014] [Accepted: 05/23/2014] [Indexed: 11/29/2022]
Abstract
In this work a combination therapy that acts upon the immune suppressive, the innate and specific arms of the immune system is proposed. This combination therapy, which consists of intratumoral interleukin-12 (IL-12) gene therapy, human tyrosinase (hTyr) DNA vaccination and metronomic cyclophosphamide (CPX), was evaluated in a B16-F10 mouse model. The following groups were compared: (1) no treatment, (2) control vector, (3) intratumoral IL-12 gene therapy, (4) intratumoral IL-12 gene therapy+metronomic CPX, (5) intratumoral IL-12 gene therapy+metronomic CPX+hTyr DNA vaccination. Next to clinical efficacy and safety, we characterized acute effects of IL-12 and anti-tumor immune response after a second tumor challenge. All treatment groups showed increased survival and higher cure rates than control groups. Survival of non-cured mice was increased when metronomic CPX was combined with IL-12 gene therapy. Furthermore, mice that received metronomic CPX had significantly lower percentages of regulatory T cells. Addition of the hTyr DNA vaccine increased cure rate and resulted in increased survival compared to other treatment groups. We also demonstrated that the manifest necrosis within days after IL-12 gene therapy is at least partly due to IL-12 mediated activation of NK cells. All cured mice were resistant to a second challenge. A humoral memory response against the tumor cells was observed in all groups that received IL-12 gene therapy, while a cellular memory response was observed only in the vaccinated mice. In conclusion, every component of this combination treatment contributed a unique immunologic trait with associated clinical benefits.
Collapse
Affiliation(s)
- Sofie Denies
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium
| | - Laetitia Cicchelero
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium
| | - Isabel Van Audenhove
- Department of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Albert Baertsoenkaai 3, B-9000 Gent, Belgium
| | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium.
| |
Collapse
|
122
|
Delany I, Rappuoli R, De Gregorio E. Vaccines for the 21st century. EMBO Mol Med 2014; 6:708-20. [PMID: 24803000 PMCID: PMC4203350 DOI: 10.1002/emmm.201403876] [Citation(s) in RCA: 274] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/20/2014] [Accepted: 04/07/2014] [Indexed: 12/18/2022] Open
Abstract
In the last century, vaccination has been the most effective medical intervention to reduce death and morbidity caused by infectious diseases. It is believed that vaccines save at least 2-3 million lives per year worldwide. Smallpox has been eradicated and polio has almost disappeared worldwide through global vaccine campaigns. Most of the viral and bacterial infections that traditionally affected children have been drastically reduced thanks to national immunization programs in developed countries. However, many diseases are not yet preventable by vaccination, and vaccines have not been fully exploited for target populations such as elderly and pregnant women. This review focuses on the state of the art of recent clinical trials of vaccines for major unmet medical needs such as HIV, malaria, TB, and cancer. In addition, we describe the innovative technologies currently used in vaccine research and development including adjuvants, vectors, nucleic acid vaccines, and structure-based antigen design. The hope is that thanks to these technologies, more diseases will be addressed in the 21st century by novel preventative and therapeutic vaccines.
Collapse
|
123
|
Riccardo F, Iussich S, Maniscalco L, Lorda Mayayo S, La Rosa G, Arigoni M, De Maria R, Gattino F, Lanzardo S, Lardone E, Martano M, Morello E, Prestigio S, Fiore A, Quaglino E, Zabarino S, Ferrone S, Buracco P, Cavallo F. CSPG4-specific immunity and survival prolongation in dogs with oral malignant melanoma immunized with human CSPG4 DNA. Clin Cancer Res 2014; 20:3753-62. [PMID: 24874834 DOI: 10.1158/1078-0432.ccr-13-3042] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE Due to the many similarities with its human counterpart, canine malignant melanoma (cMM) is a valuable model in which to assess the efficacy of novel therapeutic strategies. The model is herein used to evaluate the immunogenicity, safety, and therapeutic efficacy of a human chondroitin sulfate proteoglycan-4 (hCSPG4) DNA-based vaccine. The fact that homology between hCSPG4 and cCSPG4 amino-acidic sequences stands at more than 80% provides the rationale for using an hCSPG4 DNA vaccine in the cMM model. EXPERIMENTAL DESIGN Dogs with stage II-III surgically resected CSPG4-positive oral MM were subjected to monthly intramuscular plasmid administration, which was followed immediately by electroporation (electrovaccination) for at least 6, and up to 20, months. The immunogenicity, safety, and therapeutic efficacy of the vaccine have been evaluated. RESULTS hCSPG4 electrovaccination caused no clinically relevant local or systemic side effects and resulted in significantly longer overall and disease-free survival times in 14 vaccinated dogs as compared with 13 nonvaccinated controls. All vaccinated dogs developed antibodies against both hCSPG4 and cCSPG4. Seven vaccinated dogs were also tested for a cCSPG4-specific T-cell response and only two gave a detectable interferon (IFN)γ response. CONCLUSION Xenogeneic electrovaccination against CSPG4 is able to overcome host unresponsiveness to the "self" antigen and seems to be effective in treating cMM, laying the foundation for its translation to a human clinical setting.
Collapse
Affiliation(s)
- Federica Riccardo
- Authors' Affiliations: Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino
| | - Selina Iussich
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | - Lorella Maniscalco
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | - Saray Lorda Mayayo
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | | | - Maddalena Arigoni
- Authors' Affiliations: Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino
| | - Raffaella De Maria
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | - Francesca Gattino
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | - Stefania Lanzardo
- Authors' Affiliations: Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino
| | - Elena Lardone
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | - Marina Martano
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | - Emanuela Morello
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | - Simone Prestigio
- Authors' Affiliations: Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino
| | - Alessandra Fiore
- Authors' Affiliations: Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino
| | - Elena Quaglino
- Authors' Affiliations: Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino
| | - Sara Zabarino
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Paolo Buracco
- Department of Veterinary Science, University of Torino, Grugliasco, Torino, Italy;
| | - Federica Cavallo
- Authors' Affiliations: Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino;
| |
Collapse
|
124
|
Strioga MM, Darinskas A, Pasukoniene V, Mlynska A, Ostapenko V, Schijns V. Xenogeneic therapeutic cancer vaccines as breakers of immune tolerance for clinical application: to use or not to use? Vaccine 2014; 32:4015-24. [PMID: 24837511 DOI: 10.1016/j.vaccine.2014.05.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/29/2014] [Accepted: 05/01/2014] [Indexed: 02/06/2023]
Abstract
Accumulation of firm evidence that clinically apparent cancer develops only when malignant cells manage to escape immunosurveillance led to the introduction of tumor immunotherapy strategies aiming to reprogramm the cancer-dysbalanced antitumor immunity and restore its capacity to control tumor growth. There are several immunotherapeutical strategies, among which specific active immunotherapy or therapeutic cancer vaccination is one of the most promising. It targets dendritic cells (DCs) which have a unique ability of inducing naive and central memory T cell-mediated immune response in the most efficient manner. DCs can be therapeutically targeted either in vivo/in situ or by ex vivo manipulations followed by their re-injection back into the same patient. The majority of current DC targeting strategies are based on autologous or allogeneic tumor-associated antigens (TAAs) which possess various degrees of inherent tolerogenic potential. Therefore still limited efficacy of various tumor immunotherapy approaches may be attributed, among various other mechanisms, to the insufficient immunogenicity of self-protein-derived TAAs. Based on such an idea, the use of homologous xenogeneic antigens, derived from different species was suggested to overcome the natural immune tolerance to self TAAs. Xenoantigens are supposed to differ sufficiently from self antigens to a degree that renders them immunogenic, but at the same time preserves an optimal homology range with self proteins still allowing xenoantigens to induce cross-reactive T cells. Here we discuss the concept of xenogeneic vaccination, describe the cons and pros of autologous/allogeneic versus xenogeneic therapeutic cancer vaccines, present the results of various pre-clinical and several clinical studies and highlight the future perspectives of integrating xenovaccination into rapidly developing tumor immunotherapy regimens.
Collapse
Affiliation(s)
- Marius M Strioga
- Department of Immunology, Center of Oncosurgery, Institute of Oncology, Vilnius University, Vilnius, Lithuania.
| | - Adas Darinskas
- Department of Immunology, Center of Oncosurgery, Institute of Oncology, Vilnius University, Vilnius, Lithuania.
| | - Vita Pasukoniene
- Department of Immunology, Center of Oncosurgery, Institute of Oncology, Vilnius University, Vilnius, Lithuania.
| | - Agata Mlynska
- Department of Immunology, Center of Oncosurgery, Institute of Oncology, Vilnius University, Vilnius, Lithuania.
| | - Valerijus Ostapenko
- Section of Breast Surgery, 3(rd) Department of Surgery, Center of Oncosurgery, Institute of Oncology, Vilnius University, Vilnius, Lithuania.
| | - Virgil Schijns
- Immune Intervention, Cell Biology & Immunology group, Wageningen University, Wageningen, the Netherlands; Epitopoietic Research Corporation (ERC), Namur, Belgium.
| |
Collapse
|
125
|
Borrego JF, Huelsmeyer MK, Pinkerton ME, Muszynski JL, Miller SAK, Kurzman ID, Vail DM. Neurokinin-1 receptor expression and antagonism by the NK-1R antagonist maropitant in canine melanoma cell lines and primary tumour tissues. Vet Comp Oncol 2014; 14:210-24. [PMID: 24751104 DOI: 10.1111/vco.12093] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 03/04/2014] [Accepted: 03/24/2014] [Indexed: 01/13/2023]
Abstract
We interrogated the neurokinin-1 receptor (NK-1R)/substance P (SP) pathway in canine melanoma tumour tissues and cell lines. NK-1R messenger RNA (mRNA) and protein expression were observed in the majority of tumour tissues. Immunohistochemical assessment of archived tissue sections revealed NK-1R immunoreactivity in 11 of 15 tumours, which may have diagnostic, prognostic and therapeutic utility. However, we were unable to identify a preclinical in vitro cell line or in vivo xenograft model that recapitulates NK-1R mRNA and protein expression documented in primary tumours. While maropitant inhibited proliferation and enhanced apoptosis in cell lines, in the absence of documented NK-1R expression, this may represent off-target effects. Furthermore, maropitant failed to suppress tumour growth in a canine mouse xenograft model derived from a cell line expressing mRNA but not protein. While NK-1R represents a novel target, in the absence of preclinical models, in-species clinical trials will be necessary to investigate the therapeutic potential for antagonists such as maropitant.
Collapse
Affiliation(s)
- J F Borrego
- The Barbara Suran Comparative Oncology Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - M K Huelsmeyer
- The Barbara Suran Comparative Oncology Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - M E Pinkerton
- The Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - J L Muszynski
- The Barbara Suran Comparative Oncology Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - S A K Miller
- The Barbara Suran Comparative Oncology Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - I D Kurzman
- The Barbara Suran Comparative Oncology Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - D M Vail
- The Barbara Suran Comparative Oncology Research Institute, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA.,Carbone Comprehensive Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
126
|
Ottnod JM, Smedley RC, Walshaw R, Hauptman JG, Kiupel M, Obradovich JE. A retrospective analysis of the efficacy of Oncept vaccine for the adjunct treatment of canine oral malignant melanoma. Vet Comp Oncol 2014; 11:219-29. [PMID: 23909996 DOI: 10.1111/vco.12057] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 06/25/2013] [Accepted: 06/26/2013] [Indexed: 11/27/2022]
Abstract
Oral malignant melanoma (OMM) in the dog is often locally aggressive with a high metastatic potential and there are few treatment options that have been demonstrated to improve outcome of this disease. The purpose of this study was to determine whether adjunctive treatment with the Oncept melanoma vaccine affected the outcome of dogs with OMM that had achieved loco-regional cancer control. Medical records from 45 dogs that presented to the Animal Cancer and Imaging Center were reviewed, including 30 dogs with stage II and III disease. Dogs that received the vaccine did not achieve a greater progression-free survival, disease-free interval or median survival time than dogs that did not receive the vaccine.
Collapse
Affiliation(s)
- J M Ottnod
- Medical Oncology, Animal Cancer and Imaging Center, Canton, MI, USA.
| | | | | | | | | | | |
Collapse
|
127
|
|
128
|
Smith HA, Rekoske BT, McNeel DG. DNA vaccines encoding altered peptide ligands for SSX2 enhance epitope-specific CD8+ T-cell immune responses. Vaccine 2014; 32:1707-15. [PMID: 24492013 DOI: 10.1016/j.vaccine.2014.01.048] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 12/19/2013] [Accepted: 01/18/2014] [Indexed: 12/22/2022]
Abstract
Plasmid DNA serves as a simple and easily modifiable form of antigen delivery for vaccines. The USDA approval of DNA vaccines for several non-human diseases underscores the potential of this type of antigen delivery method as a cost-effective approach for the treatment or prevention of human diseases, including cancer. However, while DNA vaccines have demonstrated safety and immunological effect in early phase clinical trials, they have not consistently elicited robust anti-tumor responses. Hence many recent efforts have sought to increase the immunological efficacy of DNA vaccines, and we have specifically evaluated several target antigens encoded by DNA vaccine as treatments for human prostate cancer. In particular, we have focused on SSX2 as one potential target antigen, given its frequent expression in metastatic prostate cancer. We have previously identified two peptides, p41-49 and p103-111, as HLA-A2-restricted SSX2-specific epitopes. In the present study we sought to determine whether the efficacy of a DNA vaccine could be enhanced by an altered peptide ligand (APL) strategy wherein modifications were made to anchor residues of these epitopes to enhance or ablate their binding to HLA-A2. A DNA vaccine encoding APL modified to increase epitope binding elicited robust peptide-specific CD8+ T cells producing Th1 cytokines specific for each epitope. Ablation of one epitope in a DNA vaccine did not enhance immune responses to the other epitope. These results demonstrate that APL encoded by a DNA vaccine can be used to elicit increased numbers of antigen-specific T cells specific for multiple epitopes simultaneously, and suggest this could be a general approach to improve the immunogenicity of DNA vaccines encoding tumor antigens.
Collapse
Affiliation(s)
- Heath A Smith
- Department of Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Brian T Rekoske
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Douglas G McNeel
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
129
|
Lollini PL, Cavallo F, De Giovanni C, Nanni P. Preclinical vaccines against mammary carcinoma. Expert Rev Vaccines 2014; 12:1449-63. [DOI: 10.1586/14760584.2013.845530] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
130
|
Riccardo F, Bolli E, Macagno M, Arigoni M, Cavallo F, Quaglino E. Chimeric DNA Vaccines: An Effective Way to Overcome Immune Tolerance. Curr Top Microbiol Immunol 2014; 405:99-122. [PMID: 25294003 DOI: 10.1007/82_2014_426] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The fact that cancer immunotherapy is considered to be a safe and successful weapon for use in combination with surgery, radiation, and chemotherapy treatments means that it has recently been chosen as Breakthrough of the Year 2013 by Science editors. Anticancer vaccines have been extensively tested, in this field, both in preclinical cancer models and in the clinic. However, tumor-associated antigens (TAAs) are often self-tolerated molecules and cancer patients suffer from strong immunosuppressive effects, meaning that the triggering of an effective anti-tumor immune response is difficult. One possible means to overcome immunological tolerance to self-TAAs is of course the use of vaccines that code for xenogeneic proteins. However, a low-affinity antibody response against the self-homologous protein expressed by cancer cells is generally induced by xenovaccination. This issue becomes extremely limiting when working with tumors in which the contribution of the humoral rather than the cellular immune response is required if tumor growth is to be hampered. A possible way to avoid this problem is to use hybrid vaccines which code for chimeric proteins that include both homologous and xenogeneic moieties. In fact, a superior protective anti-tumor immune response against ErbB2+ transplantable and autochthonous mammary tumors was observed over plasmids that coded for the fully rat or fully human proteins when hybrid plasmids that coded for chimeric rat/human ErbB2 protein were tested in ErbB2 transgenic mice. In principle, these findings may become the basis for a new rational means of designing effective vaccines against TAAs.
Collapse
Affiliation(s)
- Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy
| | - Elisabetta Bolli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy
| | - Marco Macagno
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy
| | - Maddalena Arigoni
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, via Nizza 52, 10126, Torino, Italy.
| |
Collapse
|
131
|
Colluru VT, Johnson LE, Olson BM, McNeel DG. Preclinical and clinical development of DNA vaccines for prostate cancer. Urol Oncol 2013; 34:193-204. [PMID: 24332642 DOI: 10.1016/j.urolonc.2013.09.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 09/12/2013] [Accepted: 09/13/2013] [Indexed: 11/26/2022]
Abstract
Prostate cancer is the most commonly diagnosed cancer in the United States. It is also the second leading cause of cancer-related death in men, making it one of the largest public health concerns today. Prostate cancer is an ideal disease for immunotherapies because of the generally slow progression, the dispensability of the target organ in the patient population, and the availability of several tissue-specific antigens. As such, several therapeutic vaccines have entered clinical trials, with one autologous cellular vaccine (sipuleucel-T) recently gaining Food and Drug Administration approval after demonstrating overall survival benefit in randomized phase III clinical trials. DNA-based vaccines are safe, economical, alternative "off-the-shelf" approaches that have undergone extensive evaluation in preclinical models. In fact, the first vaccine approved in the United States for the treatment of cancer was a DNA vaccine for canine melanoma. Several prostate cancer-specific DNA vaccines have been developed in the last decade and have shown promising results in early phase clinical trials. This review summarizes anticancer human DNA vaccine trials, with a focus on those conducted for prostate cancer. We conclude with an outline of special considerations important for the development and successful translation of DNA vaccines from the laboratory to the clinic.
Collapse
Affiliation(s)
- V T Colluru
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI
| | - Laura E Johnson
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI
| | - Brian M Olson
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI
| | - Douglas G McNeel
- Department of Medicine, University of Wisconsin Carbone Cancer Center, Madison, WI.
| |
Collapse
|
132
|
Ledley FD, McNamee LM, Uzdil V, Morgan IW. Why commercialization of gene therapy stalled; examining the life cycles of gene therapy technologies. Gene Ther 2013; 21:188-94. [DOI: 10.1038/gt.2013.72] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 10/22/2013] [Accepted: 10/24/2013] [Indexed: 01/23/2023]
|
133
|
Marconato L, Frayssinet P, Rouquet N, Comazzi S, Leone VF, Laganga P, Rossi F, Vignoli M, Pezzoli L, Aresu L. Randomized, Placebo-Controlled, Double-Blinded Chemoimmunotherapy Clinical Trial in a Pet Dog Model of Diffuse Large B-cell Lymphoma. Clin Cancer Res 2013; 20:668-77. [DOI: 10.1158/1078-0432.ccr-13-2283] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
134
|
Diagnosis and treatment of a dermal malignant melanoma in an African lion (Panthera leo). J Zoo Wildl Med 2013; 44:721-7. [PMID: 24063101 DOI: 10.1638/2013-0023r2.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A 13-yr-old intact male African lion (Panthera leo) presented with a 4-mo history of left maxillary lip swelling. On physical examination, a 10-cm-diameter, ulcerated, round, firm, and pigmented mass at the level of the left maxillary canine tooth was noticed. All other organ systems examined were within normal limits. Multiple biopsies of the mass were collected and fixed in 10% neutral buffered formalin. Histopathologic evaluation of the biopsies revealed a malignant dermal melanoma. Hematologic and plasma biochemical parameters were within normal reference ranges. Thoracic radiographs taken 3 days following initial presentation showed no evidence of metastasis of the tumor. Computed tomography of the skull and neck was performed to evaluate local tumor invasion and to plan for hypofractionated radiation therapy. Therapy included four weekly treatments of 8 gray external-beam hypofractionated radiation and four bimonthly immunotherapy treatments. Following this treatment regime, the tumor size was reduced by 50%, and surgical excision was performed. No major side effects associated with radiation or immunotherapy were seen. Six months after diagnosis, hematologic and plasma biochemical parameters were within normal limits, thoracic radiographs showed no evidence of metastasis, and the lion showed no clinical signs of disease. The lion will continue to receive immunotherapy every 6 mo for the rest of its life. To the authors' knowledge, this is the first report of a successful treatment of a malignant dermal melanoma with external-beam hypofractionated radiation, immunotherapy, and surgical excision in an African lion.
Collapse
|
135
|
Abstract
Melanomas are among the most common skin tumors in horses, with prevalence rates reaching as high as 80% in adult gray horses. Most melanocytic tumors are benign at initial presentation; however, if left untreated, up to two-thirds can progress to overt malignant behavior. Standard local treatment options can be used to treat solitary early-stage lesions but do not address the underlying risk of recurrent tumor formation or the transformation to a malignant phenotype. An understanding of the specific molecular genetic factors associated with tumor formation should lead to targeted therapies that can be used to treat or ideally prevent disease.
Collapse
Affiliation(s)
- Jeffrey C Phillips
- College of Veterinary Medicine, Lincoln Memorial University, 6965 Cumberland Gap Parkway, Harrogate, TN 37752, USA.
| | | |
Collapse
|
136
|
Cu Y, Broderick KE, Banerjee K, Hickman J, Otten G, Barnett S, Kichaev G, Sardesai NY, Ulmer JB, Geall A. Enhanced Delivery and Potency of Self-Amplifying mRNA Vaccines by Electroporation in Situ. Vaccines (Basel) 2013; 1:367-83. [PMID: 26344119 PMCID: PMC4494232 DOI: 10.3390/vaccines1030367] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 08/12/2013] [Accepted: 08/14/2013] [Indexed: 01/06/2023] Open
Abstract
Nucleic acid-based vaccines such as viral vectors, plasmid DNA (pDNA), and mRNA are being developed as a means to address limitations of both live-attenuated and subunit vaccines. DNA vaccines have been shown to be potent in a wide variety of animal species and several products are now licensed for commercial veterinary but not human use. Electroporation delivery technologies have been shown to improve the generation of T and B cell responses from synthetic DNA vaccines in many animal species and now in humans. However, parallel RNA approaches have lagged due to potential issues of potency and production. Many of the obstacles to mRNA vaccine development have recently been addressed, resulting in a revival in the use of non-amplifying and self-amplifying mRNA for vaccine and gene therapy applications. In this paper, we explore the utility of EP for the in vivo delivery of large, self-amplifying mRNA, as measured by reporter gene expression and immunogenicity of genes encoding HIV envelope protein. These studies demonstrated that EP delivery of self-amplifying mRNA elicited strong and broad immune responses in mice, which were comparable to those induced by EP delivery of pDNA.
Collapse
Affiliation(s)
- Yen Cu
- Novartis Vaccines & Diagnostics, Inc., 350 Massachusetts Ave, Cambridge, MA 02139, USA
| | | | - Kaustuv Banerjee
- Novartis Vaccines & Diagnostics, Inc., 350 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Julie Hickman
- Novartis Vaccines & Diagnostics, Inc., 350 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Gillis Otten
- Novartis Vaccines & Diagnostics, Inc., 350 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Susan Barnett
- Novartis Vaccines & Diagnostics, Inc., 350 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Gleb Kichaev
- Inovio Pharmaceuticals, Blue Bell, PA 19422, USA
| | | | - Jeffrey B Ulmer
- Novartis Vaccines & Diagnostics, Inc., 350 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Andrew Geall
- Novartis Vaccines & Diagnostics, Inc., 350 Massachusetts Ave, Cambridge, MA 02139, USA.
| |
Collapse
|
137
|
Denies S, Sanders NN. Recent progress in canine tumor vaccination: potential applications for human tumor vaccines. Expert Rev Vaccines 2013; 11:1375-86. [PMID: 23249236 DOI: 10.1586/erv.12.104] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tumor vaccination holds great promise for the treatment of cancer and research concerning tumor vaccination in dogs is of great interest for veterinary as well as human medicine. Indeed, cancer is the leading cause of death in adult dogs and companion animals are acknowledged as excellent preclinical models for human oncology. The license of the veterinary melanoma vaccine (Oncept™) and Provenge® for the treatment of prostate cancer in men established tumor vaccination as a valid treatment modality for cancer. Although the results with this and other vaccines are promising, there are still some hurdles to overcome. In this article, preclinical and clinical trials with tumor vaccines in dogs are discussed, as well as the surplus value of canine cancer patients for human medicine.
Collapse
Affiliation(s)
- Sofie Denies
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium
| | | |
Collapse
|
138
|
Albanese V, Newton JC, Waguespack RW. Malignant melanoma of the third eyelid in a horse. EQUINE VET EDUC 2013. [DOI: 10.1111/eve.12012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- V. Albanese
- Department of Clinical Sciences; College of Veterinary Medicine; Auburn University; Auburn Alabama USA
| | - J. C. Newton
- Department of Pathobiology; College of Veterinary Medicine; Auburn University; Auburn Alabama USA
| | - R. W. Waguespack
- Department of Clinical Sciences; College of Veterinary Medicine; Auburn University; Auburn Alabama USA
| |
Collapse
|
139
|
Aldrich JF, Shearer MH, Lowe DB, Winn RE, Jumper CA, Kennedy RC, Bright RK. The role of gamma interferon in DNA vaccine-induced tumor immunity targeting simian virus 40 large tumor antigen. Cancer Immunol Immunother 2013; 62:371-82. [PMID: 22926061 PMCID: PMC11028630 DOI: 10.1007/s00262-012-1338-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 08/09/2012] [Indexed: 11/28/2022]
Abstract
The central role of CD4+ T lymphocytes in mediating DNA vaccine-induced tumor immunity against the viral oncoprotein simian virus 40 (SV40) large tumor antigen (Tag) has previously been described by our laboratory. In the present study, we extend our previous findings by examining the roles of IFN-γ and Th1-associated effector cells within the context of DNA immunization in a murine model of pulmonary metastasis. Immunization of BALB/c mice with plasmid DNA encoding SV40 Tag (pCMV-Tag) generated IFN-γ-secreting T lymphocytes that produced this cytokine upon in vitro stimulation with mKSA tumor cells. The role of IFN-γ as a mediator of protection against mKSA tumor development was assessed via in vivo IFN-γ neutralization, and these experiments demonstrated a requirement for this cytokine in the induction immune phase. Neutralization of IFN-γ was associated with a reduction in Th1 cytokine-producing CD4+ and CD8+ splenocytes, as assessed by flow cytometry analysis, and provided further evidence for the role of CD4+ T lymphocytes as drivers of the cellular immune response. Depletion of NK cells and CD8+ T lymphocytes demonstrated the expendability of these cell types individually, but showed a requirement for a resident cytotoxic cell population within the immune effector phase. Our findings demonstrate the importance of IFN-γ in the induction of protective immunity stimulated by pCMV-Tag DNA-based vaccine and help to clarify the general mechanisms by which DNA vaccines trigger immunity to tumor cells.
Collapse
Affiliation(s)
- Joel F. Aldrich
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 6591, Lubbock, TX 79430 USA
| | - Michael H. Shearer
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 6591, Lubbock, TX 79430 USA
| | - Devin B. Lowe
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 6591, Lubbock, TX 79430 USA
- Present Address: Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA
| | - Richard E. Winn
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 6591, Lubbock, TX 79430 USA
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430 USA
| | - Cynthia A. Jumper
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 6591, Lubbock, TX 79430 USA
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430 USA
| | - Ronald C. Kennedy
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 6591, Lubbock, TX 79430 USA
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430 USA
| | - Robert K. Bright
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, 3601 4th Street, MS 6591, Lubbock, TX 79430 USA
| |
Collapse
|
140
|
Qeska V, Baumgärtner W, Beineke A. Species-specific properties and translational aspects of canine dendritic cells. Vet Immunol Immunopathol 2013; 151:181-92. [DOI: 10.1016/j.vetimm.2012.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 11/20/2012] [Accepted: 12/06/2012] [Indexed: 12/22/2022]
|
141
|
Becker JT, McNeel DG. Presence of antigen-specific somatic allelic mutations and splice variants do not predict for immunological response to genetic vaccination. J Immunother Cancer 2013; 1:2. [PMID: 24764533 PMCID: PMC3986973 DOI: 10.1186/2051-1426-1-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 03/06/2013] [Indexed: 11/10/2022] Open
Abstract
Background Antigen-specific anti-tumor vaccines have demonstrated clinical efficacy, but immunological and clinical responses appear to be patient-dependent. We hypothesized that naturally-occurring differences in amino acid sequence of a host’s target antigen might predict for immunological outcome from genetic vaccination by presentation of epitopes different from the vaccine. Methods Using peripheral blood cells from 33 patients who had been treated with a DNA vaccine encoding prostatic acid phosphatase (PAP), we sequenced the exons encoding PAP and PSA genes from somatic DNA to identify single nucleotide polymorphisms. In addition, mRNA was collected to detect alternative splice variants of PAP. Results We detected four synonymous coding mutations of PAP among 33 patients; non-synonymous coding mutations were not identified. Alternative splice variants of PAP were detected in 22/27 patients tested. The presence of detectable splice variants was not predictive of immunological outcome from vaccination. Immune responses to peptides encoded by these splice variants were common (16/27) prior to immunization, but not associated with immune responses elicited with vaccination. Conclusions These results suggest that antigen-specific immune responses detectable after treatment with this genetic vaccine are specific for the host-encoded antigen and not due to epitope differences between the vaccine and a particular individual’s somatic coding sequence.
Collapse
Affiliation(s)
- Jordan T Becker
- Department of Medicine, University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, Madison, WI 53705, USA
| | - Douglas G McNeel
- Department of Medicine, University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, Madison, WI 53705, USA
| |
Collapse
|
142
|
Finocchiaro LME, Glikin GC. Cytokine-enhanced vaccine and suicide gene therapy as surgery adjuvant treatments for spontaneous canine melanoma: 9 years of follow-up. Cancer Gene Ther 2012; 19:852-61. [PMID: 23059870 DOI: 10.1038/cgt.2012.72] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
We present here the updated results after 9 years of the beginning of a trial on canine patients with malignant melanoma. This surgery adjuvant approach combined local suicide gene therapy with a subcutaneous vaccine composed by tumor cells extracts and xenogeneic cells producing human interleukin-2 and granulocyte-macrophage colony-stimulating factor. Toxicity was absent or minimal in all patients (0≤VCOG-CTCAE grade≤1). With respect to surgery-treated controls (ST), the complete surgery (CS) arm of this combined treatment (CT) significantly increased the fraction of local disease-free patients from 13 to 81% and distant metastases free from 32 to 84%. Even though less effective than the CS arm, the partial surgery (PS) arm of this CT was significantly better controlling the disease than only surgery (14% while PS-ST: 0%, P<0.01 and CS-ST: 5%, P<0.05). In addition, CT produced a significant sevenfold (CS) and threefold (PS) increase in overall survival. The CS-CT arm significantly improved both CS-ST metastasis-free- and melanoma overall survival from 99 days (respective ranges: 11-563 and 10-568) to >2848 days (81-2848 and 35-2848). Thus, more of 50% of our CT patients died of melanoma unrelated causes, transforming a lethal disease into a chronic one. Finally, surgery adjuvant CT delayed or prevented post-surgical recurrence and distant metastasis, significantly improved disease-free and overall survival maintaining the quality of life. Long-term safety and efficacy of this treatment are supported by the high number of CT patients (283) and extensive follow-up (>9 years). The successful clinical outcome encourages the further translation of similar approaches to human gene therapy trials.
Collapse
Affiliation(s)
- L M E Finocchiaro
- Unidad de Transferencia Genética, Instituto de Oncología Ángel H. Roffo, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | |
Collapse
|
143
|
Barar J, Omidi Y. Translational Approaches towards Cancer Gene Therapy: Hurdles and Hopes. BIOIMPACTS : BI 2012; 2:127-43. [PMID: 23678451 DOI: 10.5681/bi.2012.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Revised: 09/02/2012] [Accepted: 09/11/2012] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Of the cancer gene therapy approaches, gene silencing, suicide/apoptosis inducing gene therapy, immunogene therapy and targeted gene therapy are deemed to sub-stantially control the biological consequences of genomic changes in cancerous cells. Thus, a large number of clinical trials have been conducted against various malignancies. In this review, we will discuss recent translational progresses of gene and cell therapy of cancer. METHODS Essential information on gene therapy of cancer were reviewed and discussed towards their clinical translations. RESULTS Gene transfer has been rigorously studied in vitro and in vivo, in which some of these gene therapy endeavours have been carried on towards translational investigations and clinical applications. About 65% of gene therapy trials are related to cancer therapy. Some of these trials have been combined with cell therapy to produce personalized medicines such as Sipuleucel-T (Provenge®, marketed by Dendreon, USA) for the treatment of asymptomatic/minimally symptomatic metastatic hormone-refractory prostate cancer. CONCLUSION Translational approach links two diverse boundaries of basic and clinical researches. For successful translation of geno-medicines into clinical applications, it is essential 1) to have the guidelines and standard operating procedures for development and application of the genomedicines specific to clinically relevant biomarker(s); 2) to conduct necessary animal experimental studies to show the "proof of concept" for the proposed genomedicines; 3) to perform an initial clinical investigation; and 4) to initiate extensive clinical trials to address all necessary requirements. In short, translational researches need to be refined to accelerate the geno-medicine development and clinical applications.
Collapse
Affiliation(s)
- Jaleh Barar
- Ovarian Cancer Research Center, Translational Research Center, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
144
|
Gutiérrez AH, Moise L, De Groot AS. Of [Hamsters] and men: a new perspective on host cell proteins. Hum Vaccin Immunother 2012; 8:1172-4. [PMID: 23124469 PMCID: PMC3579895 DOI: 10.4161/hv.22378] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Andres H. Gutiérrez
- Institute for Immunology and Informatics, University of Rhode Island; Providence, RI USA
| | - Leonard Moise
- Institute for Immunology and Informatics, University of Rhode Island; Providence, RI USA
- EpiVax Inc.; Providence, RI USA
| | - Annie S. De Groot
- Institute for Immunology and Informatics, University of Rhode Island; Providence, RI USA
- EpiVax Inc.; Providence, RI USA
| |
Collapse
|
145
|
Brockley LK, Cooper MA, Bennett PF. Malignant melanoma in 63 dogs (2001-2011): the effect of carboplatin chemotherapy on survival. N Z Vet J 2012; 61:25-31. [PMID: 22913610 DOI: 10.1080/00480169.2012.699433] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
AIMS The aim of the study was to compare the effect of carboplatin chemotherapy on the survival of canine patients diagnosed with malignant melanoma after loco-regional control or as a sole therapy. METHODS A retrospective study of 63 dogs with oral, digital or cutaneous malignant melanoma treated with surgery and/or chemotherapy was undertaken. Dogs were grouped based on the anatomical site of melanoma development. For oral melanoma, dogs were subclassified into two groups: loco-regional control and gross disease. All patients in the digital and cutaneous groups had achieved loco-regional control with surgery. Comparisons between survival data for each group at each anatomical site were then made. Within the loco-regional control groups survival time was compared between those treated with and without chemotherapy post surgery. For the oral melanoma patients with gross disease survival was compared between those treated with chemotherapy and palliative therapy. The toxicity of carboplatin chemotherapy was evaluated overall. RESULTS The overall median survival times for patients with oral, digital and cutaneous melanoma were 389, 1,350 days and not reached (with a median follow-up of 776 days) respectively. Median survival time was defined as "not reached" when less than 50% of the subjects died of the disease at the end of the follow-up period, or at the time they were lost to follow-up. The addition of chemotherapy to surgery did not confer a survival benefit in the loco-regional control setting when assessing survival for each anatomical site. For oral melanoma patients with gross disease there was no difference between survival of patients treated with chemotherapy and palliative intent therapy. There was however an improvement in survival in the three dogs that responded to chemotherapy (978 days; p=0.039) compared to the eight non-responders (147 days). On univariate and multivariate analysis, anatomic location was the only variable that was significantly related to survival (p=0.0002 and p=0.009, respectively). CONCLUSIONS The addition of chemotherapy to local treatments for canine melanoma at oral, digital and cutaneous sites did not lead to a significant increase in survival times. Carboplatin was well tolerated and appeared to have activity against oral melanoma in a subset of patients with gross disease that responded to treatment. CLINICAL RELEVANCE Carboplatin with piroxicam could be considered for patients with gross disease when more traditional therapies, such as surgery or radiation therapy, are declined or are not available. In the loco-regional control setting, prospective randomised blinded studies with matched control groups are required to determine if chemotherapy has a role in the treatment of these types of cancer.
Collapse
Affiliation(s)
- L K Brockley
- Victorian Animal Cancer Care, Kensington, Victoria, Australia.
| | | | | |
Collapse
|
146
|
Ulmer JB, Mason PW, Geall A, Mandl CW. RNA-based vaccines. Vaccine 2012; 30:4414-8. [DOI: 10.1016/j.vaccine.2012.04.060] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 04/10/2012] [Accepted: 04/18/2012] [Indexed: 02/07/2023]
|