1
|
Kosharek AT, Zuleger CL, Glass WS, Eickhoff J, Sondel PM, Vail DM, Albertini MR. In vitro detection of canine anti-human antibodies following intratumoral injection of the hu14.18-IL2 immunocytokine in spontaneous canine melanoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.21.644578. [PMID: 40196525 PMCID: PMC11974718 DOI: 10.1101/2025.03.21.644578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Background Canine and human malignant melanoma are naturally occurring cancers with many similarities, making the dog an important parallel patient population to study both diseases. However, development of canine anti-human antibodies (CAHA) needs to be considered when evaluating humanized biotherapeutics in dogs. Objectives Characterize CAHA in sera from dogs with spontaneous melanoma receiving radiotherapy and intratumoral immunocytokine (IT-IC) with humanized 14.18-IL2. Methods Serum samples were obtained pre-treatment and at several post-treatment times from 12 dogs with locally advanced or metastatic melanoma treated with radiotherapy to the primary site and regional lymph nodes (when clinically involved) followed by IT-IC of humanized 14.18-IL2. Two CAHA assays were developed. A sandwich enzyme-linked immunosorbent assay (ELISA) was developed to detect antibodies against the humanized IgG component of hu14.18-IL2. A flow cytometry assay was developed to determine the ability of CAHA to inhibit binding of a mouse anti-GD2 monoclonal antibody to its target. Results Post-treatment sera from 7 of 12 dogs developed CAHA levels over pre-treatment that were identified by ELISA as significant increases at Day 30 and/or Day 60. Day 10, Day 30, and Day 60 post-treatment sera from 10 of 12 dogs significantly inhibited the binding of anti-GD2 monoclonal antibody to its target compared to pre-treatment. Significant binding inhibition was also detected in 2 of 12 dogs after local RT but before IT-IC (Day 1). Normal canine sera did not mediate binding inhibition. Conclusions This study advances CAHA detection strategies and reports the kinetics of CAHA following IT-IC in dogs with spontaneous melanoma.
Collapse
Affiliation(s)
- Andrew T. Kosharek
- University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America
| | - Cindy L. Zuleger
- University of Wisconsin School of Medicine and Public Health, Department of Medicine, Madison, WI, United States of America
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States of America
| | - William S. Glass
- University of Wisconsin-Madison, Madison, WI, United States of America
| | - Jens Eickhoff
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States of America
- University of Wisconsin School of Medicine and Public Health, Department of Biostatistics & Medical Informatics, Madison, WI, United States of America
| | - Paul M. Sondel
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States of America
- University of Wisconsin School of Medicine and Public Health, Department of Human Oncology, Madison, WI, United States of America
- University of Wisconsin School of Medicine and Public Health, Department of Pediatrics, Madison, WI, United States of America
| | - David M. Vail
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States of America
- University of Wisconsin School of Veterinary Medicine, Department of Medical Sciences, Madison, WI, United States of America
| | - Mark R. Albertini
- University of Wisconsin School of Medicine and Public Health, Department of Medicine, Madison, WI, United States of America
- University of Wisconsin Carbone Cancer Center, Madison, WI, United States of America
- University of Wisconsin School of Medicine and Public Health, Department of Dermatology, Madison, WI, United States of America
- The Medical Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States of America
| |
Collapse
|
2
|
Wang J, Zhou X, Elazab ST, Huang J, Hsu WH. Current Review of Monoclonal Antibody Therapeutics in Small Animal Medicine. Animals (Basel) 2025; 15:472. [PMID: 40002954 PMCID: PMC11852019 DOI: 10.3390/ani15040472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/30/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Monoclonal antibody therapy has been a cornerstone of human healthcare for nearly four decades, effectively treating a wide range of diseases including cancers, autoimmune disorders, and inflammatory conditions. However, its application in veterinary medicine is a relatively recent development, offering a promising therapeutic approach for managing chronic diseases in small animals. Dogs and cats, like humans, suffer from chronic conditions such as cancer, arthritis, allergies, and chronic pain, which mAb therapy could potentially address. This review aims to explore the therapeutic potential of mAb therapy in small animal medicine, focusing on currently authorized products, including their mechanisms of action, clinical efficacy, and safety concerns. A comprehensive review of the literature was conducted to evaluate the use of mAbs in veterinary medicine, specifically in the treatment of chronic disorders. While mAb therapy has shown significant benefits in human healthcare, challenges remain in its application to veterinary practice, including safety concerns and the limited availability of approved products. Despite these challenges, mAb therapy holds great promise for improving the management of chronic diseases in animals, with future research and development potentially expanding its clinical use.
Collapse
Affiliation(s)
- Jianzhong Wang
- Shanxi Key Laboratory for Modernization of TCVM, College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, China;
| | - Xueying Zhou
- Department of Veterinary Clinical Science, College of Veterinary Medicine, China Agricultural University, Beijing 100107, China;
| | - Sara T. Elazab
- Department of Pharmacology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Jian Huang
- Institute of Qinhai-Tibetan Plateau, College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu 610041, China;
| | - Walter H. Hsu
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
3
|
Bryan JN, Maitz CA. Translational History and Hope of Immunotherapy of Canine Tumors. Clin Cancer Res 2024; 30:4272-4285. [PMID: 39042399 PMCID: PMC11444889 DOI: 10.1158/1078-0432.ccr-23-2266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/13/2024] [Accepted: 07/01/2024] [Indexed: 07/24/2024]
Abstract
Companion dogs have served an important role in cancer immunotherapy research. Sharing similar environments and diets with humans, dogs naturally develop many of the same cancers. These shared exposures, coupled with dogs' diverse genetic makeup, make them ideal subjects for studying cancer therapies. Tumors like osteosarcoma, hemangiosarcoma, soft-tissue sarcoma, and non-Hodgkin lymphoma occur with greater frequency than their counterpart disease in humans. Canine brain tumors allow the study of therapy strategies with imaging, surgery, and radiotherapy equipment in veterinary patients with near-human geometry. Nonspecific immunostimulants, autologous and allogeneic vaccines, immune checkpoint inhibitors, and cellular therapies used in treating canine cancers have been tested in veterinary clinical trials. These treatments have not only improved outcomes for dogs but have also provided valuable insights for human cancer treatment. Advancements in radiation technology and the development of tools to characterize canine immune responses have further facilitated the ability to translate veterinary clinical trial results to human applications. Advancements in immunotherapy of canine tumors have directly supported translation to human clinical trials leading to approved therapies for patients with cancer around the world. The study of immunotherapy in dogs has been and will continue to be a promising avenue for advancing human cancer treatment.
Collapse
Affiliation(s)
- Jeffrey N. Bryan
- Comparative Oncology, Radiobiology, and Epigenetics Laboratory, Department of Veterinary Medicine and Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO
| | - Charles A. Maitz
- Comparative Oncology, Radiobiology, and Epigenetics Laboratory, Department of Veterinary Medicine and Surgery, Ellis Fischel Cancer Center, University of Missouri, Columbia, MO
| |
Collapse
|
4
|
Xia YY, Liao AT, Liu RM, Yang SY, Kuo CC, Ke CH, Lin CS, Lee JJ. Immunotherapeutic allogeneic dendritic cell and autologous tumor cell fusion vaccine alone or combined with radiotherapy in canine oral malignant melanoma is safe and potentially effective. Front Vet Sci 2024; 11:1397518. [PMID: 39229600 PMCID: PMC11368852 DOI: 10.3389/fvets.2024.1397518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 08/05/2024] [Indexed: 09/05/2024] Open
Abstract
Introduction Immunotherapy represents a promising breakthrough in cancer management and is being explored in canine melanomas. Dendritic cells (DCs) play a crucial role in priming T-cell-mediated immune reactions through the antigen-presenting function. Combining immunotherapy and radiation therapy may generate more substantial anti-cancer efficacy through immunomodulation. Objectives Our research reported a preliminary result of the safety and outcome of a kind of immunotherapy, the allogeneic dendritic cell and autologous tumor cell fusion vaccine, alone or in combination with hypofractionated radiation therapy, in canine oral malignant melanoma. Methods Two groups of dogs with histopathological diagnoses of oral malignant melanoma were recruited. In group 1 (DCRT), dogs received a combination of DC fusion vaccine and radiotherapy. In group 2 (DC), dogs received DC fusion vaccine alone. DC vaccination was given once every 2 weeks for four doses. Radiotherapy was performed weekly for five fractions. Dogs that received carboplatin were retrospectively collected as a control group (group 3). Results Five dogs were included in group 1 (two stage II, three stage III), 11 in group 2 (three stage I/II, eight stage III/IV), and eight (two stage I/II, six stage III/IV) in the control group. Both DC and DCRT were well-tolerated, with only mild adverse events reported, including mucositis, gastrointestinal discomfort, and injection site reactions. The median progression-free intervals in groups 1, 2, and 3 were 214 (95% CI, NA, due to insufficient data), 100 (95% CI, 27-237), and 42 days (95% CI, NA-170), respectively, which were not significantly different. The 1-year survival rates were 20, 54.5, and 12.5% in groups 1, 2, and 3. Dogs in the DCRT group exhibited significantly higher TGF-β signals than the DC group throughout the treatment course, indicating a possible higher degree of immunosuppression. Conclusion The manuscript demonstrated the safety of dendritic cell/tumor cell fusion vaccine immunotherapy, alone or in combination with radiotherapy. The results support further expansion of this immunotherapy, modification of combination treatment and protocols, and investigation of combining DC vaccine with other treatment modalities. Clinical trial registration Preclinical Trials, PCTE0000475.
Collapse
Affiliation(s)
- Yuan-Yuan Xia
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Veterinary Clinical Science, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Albert TaiChing Liao
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Ru-Min Liu
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Shu-Ya Yang
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Chien-Chun Kuo
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Chiao-Hsu Ke
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Chen-Si Lin
- Department of Veterinary Medicine, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Jih-Jong Lee
- Graduate Institute of Veterinary Clinical Science, School of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
- National Taiwan University Veterinary Hospital, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
5
|
Dell’Anno F, Giugliano R, Listorti V, Razzuoli E. A Review on Canine and Human Soft Tissue Sarcomas: New Insights on Prognosis Factors and Treatment Measures. Vet Sci 2024; 11:362. [PMID: 39195816 PMCID: PMC11358912 DOI: 10.3390/vetsci11080362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/26/2024] [Accepted: 08/03/2024] [Indexed: 08/29/2024] Open
Abstract
Soft tissue sarcomas (STSs) represent a diverse group of tumors arising from mesenchymal cells, affecting both humans and animals, including dogs. Although STSs represent a class of rare tumors, especially in humans, they pose significant clinical challenges due to their potential for local recurrence and distant metastasis. Dogs, as a model for human STSs, offer several advantages, including exposure to similar environmental risk factors, genetic diversity among breeds, and the spontaneous development of tumors. Furthermore, canine tumors closely mimic the heterogeneity and complexity of human tumors, making them valuable for research into disease progression and treatment effectiveness. Current treatment approaches for STSs in both dogs and humans primarily involve surgery, radiation therapy, and chemotherapy, with treatment decisions based on tumor characteristics and patient factors. However, the development of novel therapeutic strategies is essential, given the high failure rate of new drugs in clinical trials. To better design new tailored treatments, comprehension of the tumor microenvironment (TME) is fundamental, since it plays a crucial role in STS initiation and progression by modulating tumor behavior, promoting angiogenesis, and suppressing immune responses. Notably, TME features include cancer-associated fibroblasts (CAFs), extracellular matrix (ECM) alterations, and tumor-associated macrophages (TAMs) that, depending on their polarization state, can affect immune responses and thus the patient's prognosis. In this review, new therapeutical approaches based on immunotherapy will be deeply explored as potential treatment options for both dogs and humans with STSs. In conclusion, this review provides an overview of the current understanding of STSs in dogs and humans, emphasizing the importance of the TME and potential treatment strategies.
Collapse
Affiliation(s)
- Filippo Dell’Anno
- National Reference Center of Veterinary and comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, 16129 Genova, Italy; (F.D.); (V.L.); (E.R.)
- Department of Public Health, Experimental and Forensic Medicine, Section of Biostatistics and Clinical Epidemiology, University of Pavia, 27100 Pavia, Italy
| | - Roberta Giugliano
- National Reference Center of Veterinary and comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, 16129 Genova, Italy; (F.D.); (V.L.); (E.R.)
| | - Valeria Listorti
- National Reference Center of Veterinary and comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, 16129 Genova, Italy; (F.D.); (V.L.); (E.R.)
| | - Elisabetta Razzuoli
- National Reference Center of Veterinary and comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d’Aosta, 16129 Genova, Italy; (F.D.); (V.L.); (E.R.)
| |
Collapse
|
6
|
Albertini MR, Zuleger CL, Ranheim EA, Shiyanbola O, Sondel PM, Morris ZS, Eickhoff J, Newton MA, Ong IM, Schwartz RW, Hayim R, Kurzman ID, Turek M, Vail DM. Administration of intratumoral GD2-directed interleukin-2 immunocytokine and local radiation therapy to activate immune rejection of spontaneous canine melanoma. Melanoma Res 2024; 34:307-318. [PMID: 38768442 PMCID: PMC11444423 DOI: 10.1097/cmr.0000000000000975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Canine malignant melanoma provides a clinically relevant, large animal parallel patient population to study the GD2-reactive hu14.18-IL-2 immunocytokine as it is similar to human melanoma and expresses GD2. The objectives of this study were to evaluate safety, radiation fractionation, and identify informative biomarkers of an in-situ tumor vaccine involving local radiation therapy plus intratumoral-immunocytokine in melanoma tumor-bearing dogs. Twelve dogs (six dogs/arm) with locally advanced or metastatic melanoma were randomized to receive a single 8 Gy fraction (arm A) or three 8 Gy fractions over 1 week (arm B) to the primary site and regional lymph nodes (when clinically involved) with the single or last fraction 5 days before intratumoral-immunocytokine at 12 mg/m 2 on 3 consecutive days. Serial tumor biopsies were obtained. All 12 dogs completed protocol treatment, and none experienced significant or unexpected adverse events. Evidence of antitumor activity includes one dog with a complete response at day 60, one dog with a partial response at day 60, and four dogs with mixed responses. Histology of serial biopsies shows a variably timed increase in intratumoral lymphocytic inflammation in some dogs. Canine NanoString analyses of serial biopsies identified changes in gene signatures of innate and adaptive cell types versus baseline. There were no significant differences in NanoString results between arm A and arm B. We conclude that intratumoral-immunocytokine in combination with local radiation therapy in canine melanoma is well tolerated and has antitumor activity with the potential to inform clinical development in melanoma patients.
Collapse
Affiliation(s)
- Mark R. Albertini
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Medicine, University of Wisconsin School of Veterinary Medicine
- Department of Dermatology, University of Wisconsin School of Veterinary Medicine
- The Medical Service, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Cindy L. Zuleger
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Medicine, University of Wisconsin School of Veterinary Medicine
| | - Erik A. Ranheim
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Pathology & Laboratory Medicine, University of Wisconsin School of Veterinary Medicine
| | - Oyewale Shiyanbola
- Stanford University School of Medicine, Department of Pathology, Stanford, California
| | - Paul M. Sondel
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Pediatrics, University of Wisconsin School of Veterinary Medicine
- Department of Human Oncology, University of Wisconsin School of Veterinary Medicine
| | - Zachary S. Morris
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Human Oncology, University of Wisconsin School of Veterinary Medicine
| | - Jens Eickhoff
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Biostatistics & Medical Informatics, University of Wisconsin School of Veterinary Medicine
| | - Michael A. Newton
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Biostatistics & Medical Informatics, University of Wisconsin School of Veterinary Medicine
| | - Irene M. Ong
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Biostatistics & Medical Informatics, University of Wisconsin School of Veterinary Medicine
- Department of Obstetrics & Gynecology, University of Wisconsin School of Veterinary Medicine
| | - Rene Welch Schwartz
- Department of Biostatistics & Medical Informatics, University of Wisconsin School of Veterinary Medicine
| | - Rubi Hayim
- Department of Medical Sciences, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Ilene D. Kurzman
- Department of Medical Sciences, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - Michelle Turek
- Department of Surgical Sciences, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| | - David M. Vail
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health
- Department of Medical Sciences, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| |
Collapse
|
7
|
Kerr CP, Sheehan-Klenk J, Grudzinski JJ, Adam DP, Nguyen TPT, Ferreira CA, Bates AM, Jin WJ, Kwon O, Olson AP, Lin W, Hyun M, Jagodinsky JC, Powers M, Sriramaneni RN, Clark PA, Shea AG, Rojas HC, Choi C, Massey CF, Zangl LM, Pinchuk AN, Aluicio-Sarduy E, Kim K, Engle JW, Hernandez R, Bednarz BP, Weichert JP, Morris ZS. Effects of clinically relevant radionuclides on the activation of a type I interferon response by radiopharmaceuticals in syngeneic murine tumor models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602990. [PMID: 39071353 PMCID: PMC11275738 DOI: 10.1101/2024.07.10.602990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Radiopharmaceutical therapies (RPT) activate a type I interferon (IFN1) response in tumor cells. We hypothesized that the timing and amplitude of this response varies by isotope. We compared equal doses delivered by 90 Y, 177 Lu, and 225 Ac in vitro as unbound radionuclides and in vivo when chelated to NM600, a tumor-selective alkylphosphocholine. Response in murine MOC2 head and neck carcinoma and B78 melanoma was evaluated by qPCR and flow cytometry. Therapeutic response to 225 Ac-NM600+anti-CTLA4+anti-PD-L1 immune checkpoint inhibition (ICI) was evaluated in wild-type and stimulator of interferon genes knockout (STING KO) B78. The timing and magnitude of IFN1 response correlated with radionuclide half-life and linear energy transfer. CD8 + /Treg ratios increased in tumors 7 days after 90 Y- and 177 Lu-NM600 and day 21 after 225 Ac-NM600. 225 Ac-NM600+ICI improved survival in mice with WT but not with STING KO tumors, relative to monotherapies. Immunomodulatory effects of RPT vary with radioisotope and promote STING-dependent enhanced response to ICIs in murine models. Teaser This study describes the time course and nature of tumor immunomodulation by radiopharmaceuticals with differing physical properties.
Collapse
|
8
|
Fonseca-Alves CE, Queiroga FL, Massoco CDO. Editorial: New insights in veterinary cancer immunology. Front Vet Sci 2024; 11:1440527. [PMID: 38948673 PMCID: PMC11211622 DOI: 10.3389/fvets.2024.1440527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 07/02/2024] Open
Affiliation(s)
| | - Felisbina Luísa Queiroga
- Animal and Veterinary Research Center (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Cristina de Oliveira Massoco
- Department of Veterinary Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo—USP, São Paulo, Brazil
| |
Collapse
|
9
|
Basran PS, Turek M, Selting KA, Rancilio N. AAPM WGVRTO report 390: A survey of veterinary radiation oncology equipment and infrastructure in 2022. Med Phys 2024; 51:3924-3931. [PMID: 38626571 DOI: 10.1002/mp.17042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/19/2024] [Accepted: 03/10/2024] [Indexed: 04/18/2024] Open
Abstract
Since 2010, there has been little published data on the state of equipment and infrastructure in veterinary radiation oncology clinical practice. These data are important not only to identify the status and use of technology within the veterinary radiation oncology community but also to help identify the extent of medical physics support. The purpose of our study is to report findings from a survey of veterinary radiation oncologists in the USA, Canada, and select centers outside of North America in 2022. A 40-question survey covering topics such as type of radiotherapy equipment, techniques offered, treatment planning systems and dose calculation algorithms, special techniques, board-certified radiation oncologists and residents, and extent of medical physics support was distributed through an online survey tool. Results from 40 veterinary radiation oncology institutions, with equipment explicitly used for veterinary care, suggest that the current state of practice is not dissimilar to what currently exists in human radiation oncology facilities; techniques and technologies commonly employed include flattening filter-free mode megavoltage beams, volumetric arc therapy, daily cone-beam computed tomography, image-guided radiation therapy, and sophisticated dose calculation algorithms. These findings suggest the need for modern radiation oncology acceptance testing, commissioning, and quality assurance programs within the veterinary community. The increase in veterinary radiation oncology residency positions and increasing sophistication of equipment suggests that increased levels of standardized medical physics support would benefit the veterinary radiation oncology community.
Collapse
Affiliation(s)
- Parminder S Basran
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Michelle Turek
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Kimberly A Selting
- Department of Veterinary Clinical Medicine, University of Illinois, Urbana, Illinois, USA
| | - Nicholas Rancilio
- Department Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Iowa, USA
| |
Collapse
|
10
|
Maitz CA, Bryan JN. The role of companion animal models in radiopharmaceutical development and translation. Vet Comp Oncol 2024; 22:165-173. [PMID: 38439693 DOI: 10.1111/vco.12969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/29/2024] [Accepted: 02/14/2024] [Indexed: 03/06/2024]
Abstract
Advancements in molecular imaging and drug targeting have created a renaissance in the development of radiopharmaceuticals for therapy and theranostics. While some radiopharmaceuticals, such as Na[131I]I, have been used clinically for decades, new agents are being approved using small-molecules, peptides, and antibodies for targeting. As these agents are being developed, the need to understand dosimetry and biologic effects of the systemically delivered radiotherapy becomes more important, particularly as highly potent radiopharmaceuticals using targeted alpha therapy become clinically utilized. As the processes being targeted become more complex, and the radiobiology of different particulate radiation becomes more diverse, models that better recapitulate human cancer and geometry are necessary. Companion animals develop many of the same types of cancer, carrying many of the same genetic drivers as those seen in people, and the scale and geometry of tumours in dogs more closely mimics those in humans than murine tumour models. Key translational challenges in oncology, such as alterations in tumour microenvironment, hypoxia, heterogeneity, and geometry are addressed by companion animal models. This review paper will provide background on radiopharmaceutical targeting techniques, review the use of radiopharmaceuticals in companion animal oncology, and explore the translational value of treating these patients in terms of dosimetry, treatment outcomes, and normal tissue complication rates.
Collapse
Affiliation(s)
- Charles A Maitz
- Comparative Oncology Radiobiology and Epigenetics Laboratory, Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri, USA
| | - Jeffrey N Bryan
- Comparative Oncology Radiobiology and Epigenetics Laboratory, Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
11
|
Bergman PJ. Cancer Immunotherapy. Vet Clin North Am Small Anim Pract 2024; 54:441-468. [PMID: 38158304 DOI: 10.1016/j.cvsm.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
The enhanced understanding of immunology experienced over the last 5 decades afforded through the tools of molecular biology has recently translated into cancer immunotherapy becoming one of the most exciting and rapidly expanding fields. Human cancer immunotherapy is now recognized as one of the pillars of treatment alongside surgery, radiation, and chemotherapy. The field of veterinary cancer immunotherapy has also rapidly advanced in the last decade with a handful of commercially available products and a plethora of investigational cancer immunotherapies, which will hopefully expand our veterinary oncology treatment toolkit over time.
Collapse
Affiliation(s)
- Philip J Bergman
- Clinical Studies, VCA; Katonah Bedford Veterinary Center, Bedford Hills, NY, USA; Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
12
|
Bryan JN. Updates in Osteosarcoma. Vet Clin North Am Small Anim Pract 2024; 54:523-539. [PMID: 38158305 DOI: 10.1016/j.cvsm.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Clinical care of osteosarcoma (OSA) in dogs has seen little change during the past 2 decades, relying on amputation and platinum-based chemotherapy for pain control and survival. Recent advancements offer hope for improved outcomes. Genomic research reveals shared genetic abnormalities between canine and human OSA. Multidimensional imaging provides valuable staging and prognostic information. Limb-sparing approaches including stereotactic body radiation therapy are routine. Ablative therapies such as microwave ablation and histotripsy show promise. Immunotherapy including cell therapy and immune checkpoint inhibition are available. Radiopharmaceuticals are tuned to target OSA cells directly. These innovations may enhance treatment and prognosis for dogs with OSA.
Collapse
Affiliation(s)
- Jeffrey N Bryan
- Comparative Oncology Radiobiology and Epigenetics Laboratory, University of Missouri Columbia, Ellis Fischel Cancer Center, 900 East Campus Drive, Columbia, MO 65211, USA.
| |
Collapse
|
13
|
Razmara AM, Gingrich AA, Toedebusch CM, Rebhun RB, Murphy WJ, Kent MS, Canter RJ. Improved characterization and translation of NK cells for canine immunotherapy. Front Vet Sci 2024; 11:1336158. [PMID: 38379924 PMCID: PMC10877038 DOI: 10.3389/fvets.2024.1336158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 01/11/2024] [Indexed: 02/22/2024] Open
Abstract
The field of cancer immunology has seen a meteoric rise in interest and application due to the discovery of immunotherapies that target immune cells, often leading to dramatic anti-tumor effects. However, successful cellular immunotherapy for solid tumors remains a challenge, and the application of immunotherapy to dogs with naturally occurring cancers has emerged as a high yield large animal model to bridge the bench-to-bedside challenges of immunotherapies, including those based on natural killer (NK) cells. Here, we review recent developments in the characterization and understanding of canine NK cells, a critical springboard for future translational NK immunotherapy research. The characterization of canine NK cells is exceptionally pertinent given the ongoing challenges in defining them and contextualizing their similarities and differences compared to human and murine NK cells compounded by the limited availability of validated canine specific reagents. Additionally, we summarize the current landscape of the clinical and translational literature employing strategies to capitalize on endogenous and exogenous NK cell immunotherapy in canine cancer patients. The insights regarding efficacy and immune correlates from these trials provide a solid foundation to design and test novel combinational therapies to enhance NK cell activity with the added benefit of motivating comparative work to translate these findings to human cancers with extensive similarities to their canine counterparts. The compilation of knowledge from basic canine NK phenotype and function to applications in first-in-dog clinical trials will support the canine cancer model and enhance translational work to improve cancer outcomes for both dogs and humans.
Collapse
Affiliation(s)
- Aryana M. Razmara
- Department of Surgery, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Alicia A. Gingrich
- MD Anderson Cancer Center, University of Texas, Houston, TX, United States
| | - Christine M. Toedebusch
- Center for Companion Animal Health, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Robert B. Rebhun
- Center for Companion Animal Health, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - William J. Murphy
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Michael S. Kent
- Center for Companion Animal Health, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Robert J. Canter
- Department of Surgery, University of California Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
14
|
Yang YT, Engleberg AI, Yuzbasiyan-Gurkan V. Establishment and Characterization of Cell Lines from Canine Metastatic Osteosarcoma. Cells 2023; 13:25. [PMID: 38201229 PMCID: PMC10778184 DOI: 10.3390/cells13010025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/15/2023] [Accepted: 12/17/2023] [Indexed: 01/12/2024] Open
Abstract
Despite the advancements in treatments for other cancers, the outcomes for osteosarcoma (OSA) patients have not improved in the past forty years, especially in metastatic patients. Moreover, the major cause of death in OSA patients is due to metastatic lesions. In the current study, we report on the establishment of three cell lines derived from metastatic canine OSA patients and their transcriptome as compared to normal canine osteoblasts. All the OSA cell lines displayed significant upregulation of genes in the epithelial mesenchymal transition (EMT) pathway, and upregulation of key cytokines such as CXCL8, CXCL10 and IL6. The two most upregulated genes are MX1 and ISG15. Interestingly, ISG15 has recently been identified as a potential therapeutic target for OSA. In addition, there is notable downregulation of cell cycle control genes, including CDKN2A, CDKN2B and THBS1. At the protein level, p16INK4A, coded by CDKN2A, was undetectable in all the canine OSA cell lines, while expression of the tumor suppressor PTEN was variable, with one cell line showing complete absence and others showing low levels of expression. In addition, the cells express a variety of actionable genes, including KIT, ERBB2, VEGF and immune checkpoint genes. These findings, similar to those reported in human OSA, point to some genes that can be used for prognosis, targeted therapies and novel drug development for both canine and human OSA patients.
Collapse
Affiliation(s)
- Ya-Ting Yang
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA; (Y.-T.Y.); (A.I.E.)
| | - Alexander I. Engleberg
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA; (Y.-T.Y.); (A.I.E.)
| | - Vilma Yuzbasiyan-Gurkan
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA; (Y.-T.Y.); (A.I.E.)
- Department of Microbiology & Molecular Genetics, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
15
|
London CA, Gardner H, Zhao S, Knapp DW, Utturkar SM, Duval DL, Chambers MR, Ostrander E, Trent JM, Kuffel G. Leading the pack: Best practices in comparative canine cancer genomics to inform human oncology. Vet Comp Oncol 2023; 21:565-577. [PMID: 37778398 PMCID: PMC12065084 DOI: 10.1111/vco.12935] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 10/03/2023]
Abstract
Pet dogs develop spontaneous cancers at a rate estimated to be five times higher than that of humans, providing a unique opportunity to study disease biology and evaluate novel therapeutic strategies in a model system that possesses an intact immune system and mirrors key aspects of human cancer biology. Despite decades of interest, effective utilization of pet dog cancers has been hindered by a limited repertoire of necessary cellular and molecular reagents for both in vitro and in vivo studies, as well as a dearth of information regarding the genomic landscape of these cancers. Recently, many of these critical gaps have been addressed through the generation of a highly annotated canine reference genome, the creation of several tools necessary for multi-omic analysis of canine tumours, and the development of a centralized repository for key genomic and associated clinical information from canine cancer patients, the Integrated Canine Data Commons. Together, these advances have catalysed multidisciplinary efforts designed to integrate the study of pet dog cancers more effectively into the translational continuum, with the ultimate goal of improving human outcomes. The current review summarizes this recent progress and provides a guide to resources and tools available for comparative study of pet dog cancers.
Collapse
Affiliation(s)
- Cheryl A. London
- Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | - Heather Gardner
- Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, USA
| | - Shaying Zhao
- University of Georgia Cancer Center, University of Georgia, Athens, Georgia, USA
| | - Deborah W. Knapp
- College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, USA
| | - Sagar M. Utturkar
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, Indiana, USA
| | - Dawn L. Duval
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | | | - Elaine Ostrander
- Cancer Genetics and Comparative Genomics Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Jeffrey M. Trent
- Translational Genomics Research Institute, Phoenix, Arizona, USA
| | - Gina Kuffel
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
16
|
Allen KJH, Kwon O, Hutcheson MR, Grudzinski JJ, Cain SM, Cruz FA, Vinayakamoorthy RM, Sun YS, Fairley L, Prabaharan CB, Dickinson R, MacDonald-Dickinson V, Uppalapati M, Bednarz BP, Dadachova E. Image-Based Dosimetry in Dogs and Cross-Reactivity with Human Tissues of IGF2R-Targeting Human Antibody. Pharmaceuticals (Basel) 2023; 16:979. [PMID: 37513891 PMCID: PMC10384855 DOI: 10.3390/ph16070979] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 06/30/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Osteosarcoma (OS) represents the most common primary bone tumor in humans and in companion dogs, being practically phenotypically identical. There is a need for effective treatments to extend the survival of patients with OS. Here, we examine the dosimetry in beagle dogs and cross-reactivity with human tissues of a novel human antibody, IF3, that targets the insulin growth factor receptor type 2 (IGF2R), which is overexpressed on OS cells, making it a candidate for radioimmunotherapy of OS. METHODS [89Zr]Zr-DFO-IF3 was injected into three healthy beagle dogs. PET/CT was conducted at 4, 24, 48, and 72 h. RAPID analysis was used to determine the dosimetry of [177Lu]Lu-CHXA"-IF3 for a clinical trial in companion dogs with OS. IF3 antibody was biotinylated, and a multitude of human tissues were assessed with immunohistochemistry. RESULTS PET/CT revealed that only the liver, bone marrow, and adrenal glands had high uptake. Clearance was initially through renal and hepatobiliary excretion in the first 72 h followed by primarily physical decay. RAPID analysis showed bone marrow to be the dose-limiting organ with a therapeutic range for 177Lu calculated to be 0.487-0.583 GBq. Immunohistochemistry demonstrated the absence of IGF2R expression on the surface of healthy human cells, thus suggesting that radioimmunotherapy with [177Lu]Lu-CHXA"-IF3 will be well tolerated. CONCLUSIONS Image-based dosimetry has defined a safe therapeutic range for canine clinical trials, while immunohistochemistry has suggested that the antibody will not cross-react with healthy human tissues.
Collapse
Affiliation(s)
- Kevin J H Allen
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Ohyun Kwon
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | | | - Joseph J Grudzinski
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Stuart M Cain
- adMare BioInnovations, Vancouver, BC V6T 1Z3, Canada
| | | | | | - Ying S Sun
- adMare BioInnovations, Vancouver, BC V6T 1Z3, Canada
| | | | - Chandra B Prabaharan
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Ryan Dickinson
- Department of Veterinary Pathology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Valerie MacDonald-Dickinson
- Department of Small Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada
| | - Maruti Uppalapati
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Bryan P Bednarz
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
17
|
Gregucci F, Spada S, Barcellos-Hoff MH, Bhardwaj N, Chan Wah Hak C, Fiorentino A, Guha C, Guzman ML, Harrington K, Herrera FG, Honeychurch J, Hong T, Iturri L, Jaffee E, Karam SD, Knott SR, Koumenis C, Lyden D, Marciscano AE, Melcher A, Mondini M, Mondino A, Morris ZS, Pitroda S, Quezada SA, Santambrogio L, Shiao S, Stagg J, Telarovic I, Timmerman R, Vozenin MC, Weichselbaum R, Welsh J, Wilkins A, Xu C, Zappasodi R, Zou W, Bobard A, Demaria S, Galluzzi L, Deutsch E, Formenti SC. Updates on radiotherapy-immunotherapy combinations: Proceedings of 6 th annual ImmunoRad conference. Oncoimmunology 2023; 12:2222560. [PMID: 37363104 PMCID: PMC10286673 DOI: 10.1080/2162402x.2023.2222560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023] Open
Abstract
Focal radiation therapy (RT) has attracted considerable attention as a combinatorial partner for immunotherapy (IT), largely reflecting a well-defined, predictable safety profile and at least some potential for immunostimulation. However, only a few RT-IT combinations have been tested successfully in patients with cancer, highlighting the urgent need for an improved understanding of the interaction between RT and IT in both preclinical and clinical scenarios. Every year since 2016, ImmunoRad gathers experts working at the interface between RT and IT to provide a forum for education and discussion, with the ultimate goal of fostering progress in the field at both preclinical and clinical levels. Here, we summarize the key concepts and findings presented at the Sixth Annual ImmunoRad conference.
Collapse
Affiliation(s)
- Fabiana Gregucci
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
- Department of Radiation Oncology, Miulli General Regional Hospital, Acquaviva delle Fonti, Bari, Italy
| | - Sheila Spada
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology, School of Medicine, University of California, San Francisco, CA, USA
| | - Nina Bhardwaj
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Alba Fiorentino
- Department of Radiation Oncology, Miulli General Regional Hospital, Acquaviva delle Fonti, Bari, Italy
- Department of Medicine and Surgery, LUM University, Casamassima, Bari, Italy
| | - Chandan Guha
- Department of Radiation Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Monica L. Guzman
- Division of Hematology/Oncology, Department of Medicine, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Kevin Harrington
- The Institute of Cancer Research/The Royal Marsden NHS Foundation Trust, National Institute for Health Research Biomedical Research Centre, London, UK
| | - Fernanda G. Herrera
- Centre Hospitalier Universitaire Vaudois, University of Lausanne and Ludwig Institute for Cancer Research at the Agora Cancer Research Center, Lausanne, Switzerland
| | - Jamie Honeychurch
- Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Theodore Hong
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lorea Iturri
- Institut Curie, Université PSL, CNRS UMR3347, INSERM U1021, Signalisation Radiobiologie et Cancer, Orsay, France
| | - Elisabeth Jaffee
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Sana D. Karam
- Department of Radiation Oncology, University of Colorado, Aurora, CO, USA
| | - Simon R.V. Knott
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David Lyden
- Children’s Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children’s Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | | | - Alan Melcher
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, UK
| | - Michele Mondini
- Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- Université of Paris-Saclay, Saclay, France
- INSERM U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Villejuif, France
| | - Anna Mondino
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sean Pitroda
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
| | - Sergio A. Quezada
- Cancer Immunology Unit, Research Department of Haematology, University College London Cancer Institute, London, UK
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Stephen Shiao
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l’Universite de Montreal, Faculty of Pharmacy, Montreal, Canada
| | - Irma Telarovic
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Robert Timmerman
- Departments of Radiation Oncology and Neurosurgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marie-Catherine Vozenin
- Laboratory of Radiation Oncology, Radiation Oncology Service, Department of Oncology, CHUV, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ralph Weichselbaum
- Department of Radiation and Cellular Oncology, Ludwig Center for Metastases Research, University of Chicago, IL, USA
| | - James Welsh
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anna Wilkins
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom, Royal Marsden Hospital, Sutton, UK
| | - Chris Xu
- School of Applied and Engineering Physics, Cornell University, Ithaca, NY, USA
| | - Roberta Zappasodi
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Weiping Zou
- Departments of Surgery and Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | | | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Eric Deutsch
- Department of Radiation Oncology, Gustave Roussy Cancer Campus, Villejuif, France
- Université of Paris-Saclay, Saclay, France
- INSERM U1030, Radiothérapie Moléculaire et Innovation Thérapeutique, Villejuif, France
| | - Silvia C. Formenti
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
18
|
Ammons DT, MacDonald CR, Chow L, Repasky EA, Dow S. Chronic adrenergic stress and generation of myeloid-derived suppressor cells: Implications for cancer immunotherapy in dogs. Vet Comp Oncol 2023; 21:159-165. [PMID: 36876492 DOI: 10.1111/vco.12891] [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: 01/23/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/07/2023]
Abstract
Recent studies have highlighted a key role played by the sympathetic nervous system (SNS) and adrenergic stress in mediating immune suppression associated with chronic inflammation in cancer and other diseases. The connection between chronic SNS activation, adrenergic stress and immune suppression is linked in part to the ability of catecholamines to stimulate the bone marrow release and differentiation of myeloid-derived suppressor cells (MDSC). Rodent model studies have revealed an important role for β-adrenergic receptor signalling in suppression of cancer immunity in mice subjected to chronic stresses, including thermal stress. Importantly, therapeutic blockade of beta-adrenergic responses by drugs such as propranolol can partially reverse the generation and differentiation of MDSC, and partly restore tumour immunity. Clinical trials in both humans and dogs with cancer have demonstrated that propranolol blockade can improve responses to radiation therapy, cancer vaccines and immune checkpoint inhibitors. Thus, the SNS stress response has become an important new target to relieve immune suppression in cancer and other chronic inflammatory conditions.
Collapse
Affiliation(s)
- Dylan T Ammons
- Flint Animal Cancer Center, Fort Collins, Colorado, USA
- Department of Microbiology, Immunology, and Pathology, Fort Collins, Colorado, USA
| | - Cameron R MacDonald
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Lyndah Chow
- Flint Animal Cancer Center, Fort Collins, Colorado, USA
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Steven Dow
- Flint Animal Cancer Center, Fort Collins, Colorado, USA
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| |
Collapse
|
19
|
Tarone L, Mareschi K, Tirtei E, Giacobino D, Camerino M, Buracco P, Morello E, Cavallo F, Riccardo F. Improving Osteosarcoma Treatment: Comparative Oncology in Action. LIFE (BASEL, SWITZERLAND) 2022; 12:life12122099. [PMID: 36556464 PMCID: PMC9783386 DOI: 10.3390/life12122099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Osteosarcoma (OSA) is the most common pediatric malignant bone tumor. Although surgery together with neoadjuvant/adjuvant chemotherapy has improved survival for localized OSA, most patients develop recurrent/metastatic disease with a dismally poor outcome. Therapeutic options have not improved for these OSA patients in recent decades. As OSA is a rare and "orphan" tumor, with no distinct targetable driver antigens, the development of new efficient therapies is still an unmet and challenging clinical need. Appropriate animal models are therefore critical for advancement in the field. Despite the undoubted relevance of pre-clinical mouse models in cancer research, they present some intrinsic limitations that may be responsible for the low translational success of novel therapies from the pre-clinical setting to the clinic. From this context emerges the concept of comparative oncology, which has spurred the study of pet dogs as a uniquely valuable model of spontaneous OSA that develops in an immune-competent system with high biological and clinical similarities to corresponding human tumors, including in its metastatic behavior and resistance to conventional therapies. For these reasons, the translational power of studies conducted on OSA-bearing dogs has seen increasing recognition. The most recent and relevant veterinary investigations of novel combinatorial approaches, with a focus on immune-based strategies, that can most likely benefit both canine and human OSA patients have been summarized in this commentary.
Collapse
Affiliation(s)
- Lidia Tarone
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
| | - Katia Mareschi
- Department of Public Health and Paediatrics, University of Torino, Piazza Polonia 94, 10126 Torino, Italy
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Department, Regina Margherita Children’s Hospital, City of Health and Science of Torino, 10126 Torino, Italy
| | - Elisa Tirtei
- Department of Public Health and Paediatrics, University of Torino, Piazza Polonia 94, 10126 Torino, Italy
- Stem Cell Transplantation and Cellular Therapy Laboratory, Paediatric Onco-Haematology Department, Regina Margherita Children’s Hospital, City of Health and Science of Torino, 10126 Torino, Italy
| | - Davide Giacobino
- Department of Veterinary Sciences, University of Torino, Largo Paolo Braccini 2, Grugliasco, 10095 Torino, Italy
| | - Mariateresa Camerino
- Department of Veterinary Sciences, University of Torino, Largo Paolo Braccini 2, Grugliasco, 10095 Torino, Italy
| | - Paolo Buracco
- Department of Veterinary Sciences, University of Torino, Largo Paolo Braccini 2, Grugliasco, 10095 Torino, Italy
| | - Emanuela Morello
- Department of Veterinary Sciences, University of Torino, Largo Paolo Braccini 2, Grugliasco, 10095 Torino, Italy
| | - Federica Cavallo
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
- Correspondence: (F.C.); (F.R.)
| | - Federica Riccardo
- Molecular Biotechnology Center “Guido Tarone”, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy
- Correspondence: (F.C.); (F.R.)
| |
Collapse
|
20
|
Fu Y, Yu J, Liatsou I, Du Y, Josefsson A, Nedrow JR, Rindt H, Bryan JN, Kraitchman DL, Sgouros G. Anti-GD2 antibody for radiopharmaceutical imaging of osteosarcoma. Eur J Nucl Med Mol Imaging 2022; 49:4382-4393. [PMID: 35809088 DOI: 10.1007/s00259-022-05888-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/19/2022] [Indexed: 01/29/2023]
Abstract
PURPOSE Osteosarcoma (OS) is the most frequently diagnosed bone cancer in children with little improvement in overall survival in the past decades. The high surface expression of disialoganglioside GD2 on OS tumors and restricted expression in normal tissues makes it an ideal target for anti-OS radiopharmaceuticals. Since human and canine OS share many biological and molecular features, spontaneously occurring OS in canines has been an ideal model for testing new imaging and treatment modalities for human translation. In this study, we evaluated a humanized anti-GD2 antibody, hu3F8, as a potential delivery vector for targeted radiopharmaceutical imaging of human and canine OS. METHODS The cross-reactivity of hu3F8 with human and canine OS cells and tumors was examined by immunohistochemistry and flow cytometry. The hu3F8 was radiolabeled with indium-111, and the biodistribution of [111In]In-hu3F8 was assessed in tumor xenograft-bearing mice. The targeting ability of [111In]In-hu3F8 to metastatic OS was tested in spontaneous OS canines. RESULTS The hu3F8 cross reacts with human and canine OS cells and canine OS tumors with high binding affinity. Biodistribution studies revealed selective uptake of [111In]In-hu3F8 in tumor tissue. SPECT/CT imaging of spontaneous OS canines demonstrated avid uptake of [111In]In-hu3F8 in all metastatic lesions. Immunohistochemistry confirmed the extensive binding of radiolabeled hu3F8 within both osseous and soft lesions. CONCLUSION This study demonstrates the feasibility of targeting GD2 on OS cells and spontaneous OS canine tumors using hu3F8-based radiopharmaceutical imaging. Its ability to deliver an imaging payload in a targeted manner supports the utility of hu3F8 for precision imaging of OS and potential future use in radiopharmaceutical therapy.
Collapse
Affiliation(s)
- Yingli Fu
- Department of Radiology and Radiological Science, the Johns Hopkins University School of Medicine, MD, Baltimore, USA
| | - Jing Yu
- Department of Radiology and Radiological Science, the Johns Hopkins University School of Medicine, MD, Baltimore, USA
| | - Ioanna Liatsou
- Department of Radiology and Radiological Science, the Johns Hopkins University School of Medicine, MD, Baltimore, USA
| | - Yong Du
- Department of Radiology and Radiological Science, the Johns Hopkins University School of Medicine, MD, Baltimore, USA
| | - Anders Josefsson
- Department of Radiology and Radiological Science, the Johns Hopkins University School of Medicine, MD, Baltimore, USA
| | - Jessie R Nedrow
- Department of Radiology and Radiological Science, the Johns Hopkins University School of Medicine, MD, Baltimore, USA
| | - Hans Rindt
- Department of Veterinary Medicine & Surgery, the University of Missouri, Columbia, MO, USA
| | - Jeffrey N Bryan
- Department of Veterinary Medicine & Surgery, the University of Missouri, Columbia, MO, USA
| | - Dara L Kraitchman
- Department of Radiology and Radiological Science, the Johns Hopkins University School of Medicine, MD, Baltimore, USA
| | - George Sgouros
- Department of Radiology and Radiological Science, the Johns Hopkins University School of Medicine, MD, Baltimore, USA.
| |
Collapse
|
21
|
Tarone L, Giacobino D, Camerino M, Ferrone S, Buracco P, Cavallo F, Riccardo F. Canine Melanoma Immunology and Immunotherapy: Relevance of Translational Research. Front Vet Sci 2022; 9:803093. [PMID: 35224082 PMCID: PMC8873926 DOI: 10.3389/fvets.2022.803093] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/10/2022] [Indexed: 11/17/2022] Open
Abstract
In veterinary oncology, canine melanoma is still a fatal disease for which innovative and long-lasting curative treatments are urgently required. Considering the similarities between canine and human melanoma and the clinical revolution that immunotherapy has instigated in the treatment of human melanoma patients, special attention must be paid to advancements in tumor immunology research in the veterinary field. Herein, we aim to discuss the most relevant knowledge on the immune landscape of canine melanoma and the most promising immunotherapeutic approaches under investigation. Particular attention will be dedicated to anti-cancer vaccination, and, especially, to the encouraging clinical results that we have obtained with DNA vaccines directed against chondroitin sulfate proteoglycan 4 (CSPG4), which is an appealing tumor-associated antigen with a key oncogenic role in both canine and human melanoma. In parallel with advances in therapeutic options, progress in the identification of easily accessible biomarkers to improve the diagnosis and the prognosis of melanoma should be sought, with circulating small extracellular vesicles emerging as strategically relevant players. Translational advances in melanoma management, whether achieved in the human or veterinary fields, may drive improvements with mutual clinical benefits for both human and canine patients; this is where the strength of comparative oncology lies.
Collapse
Affiliation(s)
- Lidia Tarone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Davide Giacobino
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | | | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Paolo Buracco
- Department of Veterinary Sciences, University of Turin, Turin, Italy
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| |
Collapse
|