1
|
Serpolamatsang T, Arrington K, Gardeen S, Scherman JA. Angiosarcoma Mimicking Allergic Contact Dermatitis and Response to Timolol. Dermatitis 2024. [PMID: 38669093 DOI: 10.1089/derm.2023.0129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
|
2
|
Moirano S, Turek M, Sanchez D, Vail D, Van Asselt N, Lawrence J, Forrest L. Intensity-modulated radiotherapy and chemotherapy for canine right atrial tumors: A retrospective study of seven dogs. Vet Radiol Ultrasound 2023; 64:1099-1102. [PMID: 37800663 DOI: 10.1111/vru.13303] [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: 02/05/2023] [Revised: 07/30/2023] [Accepted: 08/29/2023] [Indexed: 10/07/2023] Open
Abstract
Most primary cardiac tumors in dogs are located in the right atrium/atrial appendage, with hemangiosarcoma being the most common. The aims of this retrospective, case series were to describe outcomes for seven dogs with right atrial tumors treated with hypofractionated intensity-modulated radiotherapy and concurrent vinblastine and propranolol. One dog had a complete response, four dogs had partial responses and two dogs had stable disease after treatment. Effusions resolved in all dogs. Median progression-free survival was 290 days. Five dogs died from metastatic disease, one dog from unrelated neoplasia, and one dog is alive. Median overall survival was 326 days. Three dogs with confirmed hemangiosarcoma survived 244, 326, and 445 days. Two dogs developed clinically significant, but nonfatal, cardiac arrhythmias. One dog that received three courses of radiation had subclinical myocardial and arterial fibrosis at necropsy. Hypofractionated chemoradiotherapy was well tolerated and may provide clinical benefit in dogs with right atrial tumors.
Collapse
Affiliation(s)
- Steven Moirano
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Michelle Turek
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Diana Sanchez
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David Vail
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Nate Van Asselt
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jessica Lawrence
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Lisa Forrest
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
3
|
Kumar U, Aich J, Devarajan S. Exploring the repurposing potential of telmisartan drug in breast cancer: an in-silico and in-vitro approach. Anticancer Drugs 2023; 34:1094-1103. [PMID: 36847075 DOI: 10.1097/cad.0000000000001509] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
Anticancer drug resistance is one of the biggest hurdles in the treatment of breast cancer. Drug repurposing is a viable option fordeveloping novel medical treatment strategies since this method is more cost-efficient and rapid. Antihypertensive medicines have recently been found to have pharmacological features that could be used to treat cancer, making them effective candidates for therapeutic repurposing. The goal of our research is to find a potent antihypertensive drug that can be repurposed as adjuvant therapy for breast cancer. In this study, virtual screening was performed using a set of Food and Drug Administration (FDA)-approved antihypertensive drugs as ligands with selected receptor proteins (EGFR, KRAS, P53, AGTR1, AGTR2, and ACE) assuming these proteins are regarded to have a significant role in hypertension as well as breast cancer. Further, our in-silico results were further confirmed by an in-vitro experiment (cytotoxicity assay). All the compounds (enalapril, atenolol, acebutolol, propranolol, amlodipine, verapamil, doxazosin, prazosin, hydralazine, irbesartan, telmisartan, candesartan, and aliskiren) showed remarkable affinity towards the target receptor proteins. However, maximum affinity was displayed by telmisartan. Cell-based cytotoxicity study of telmisartan in MCF7 (breast cancer cell line) confirmed the anticancer effect of telmisartan. IC50 of the drug was calculated to be 7.75 µM and at this concentration, remarkable morphological alterations were observed in the MCF7 cells confirming its cytotoxicity in breast cancer cells. Based on both in-silico and in-vitro studies, we can conclude that telmisartan appears to be a promising drug repurposing candidate for the therapeutic treatment of breast cancer.
Collapse
Affiliation(s)
- Urwashi Kumar
- School of Biotechnology and Bioinformatics, D.Y. Patil Deemed to Be University, CBD Belapur, Navi Mumbai, Maharashtra, India
| | | | | |
Collapse
|
4
|
Kapturska KM, Pawlak A. New molecular targets in canine hemangiosarcoma-Comparative review and future of the precision medicine. Vet Comp Oncol 2023; 21:357-377. [PMID: 37308243 DOI: 10.1111/vco.12917] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 05/10/2023] [Accepted: 05/30/2023] [Indexed: 06/14/2023]
Abstract
Human angiosarcoma and canine hemangiosarcoma reveal similarities not only in their aggressive clinical behaviour, but especially in molecular landscape and genetic alterations involved in tumorigenesis and metastasis formation. Currently, no satisfying treatment that allows for achieving long overall survival or even prolonged time to progression does not exist. Due to the progress that has been made in targeted therapies and precision medicine the basis for a new treatment design is to uncover mutations and their functions as possible targets to provide tailored drugs for individual cases. Whole exome or genome sequencing studies and immunohistochemistry brought in the last few years important discoveries and identified the most common mutations with probably crucial role in this tumour development. Also, despite a lack of mutation in some of the culprit genes, the cancerogenesis cause may be buried in main cellular pathways connected with proteins encoded by those genes and involving, for example, pathological angiogenesis. The aim of this review is to highlight the most promising molecular targets for precision oncology treatment from the veterinary perspective aided by the principles of comparative science. Some of the drugs are only undergoing laboratory in vitro studies and others entered the clinic in the management of other cancer types in humans, but those used in dogs with promising responses have been mentioned as priorities.
Collapse
Affiliation(s)
- Karolina Małgorzata Kapturska
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
- Veterinary Clinic NEOVET s.c. Hildebrand, Jelonek, Michalek-Salt, Wroclaw, Poland
| | - Aleksandra Pawlak
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| |
Collapse
|
5
|
Jackson DB, Racz R, Kim S, Brock S, Burkhart K. Rewiring Drug Research and Development through Human Data-Driven Discovery (HD 3). Pharmaceutics 2023; 15:1673. [PMID: 37376121 PMCID: PMC10303279 DOI: 10.3390/pharmaceutics15061673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 05/29/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
In an era of unparalleled technical advancement, the pharmaceutical industry is struggling to transform data into increased research and development efficiency, and, as a corollary, new drugs for patients. Here, we briefly review some of the commonly discussed issues around this counterintuitive innovation crisis. Looking at both industry- and science-related factors, we posit that traditional preclinical research is front-loading the development pipeline with data and drug candidates that are unlikely to succeed in patients. Applying a first principles analysis, we highlight the critical culprits and provide suggestions as to how these issues can be rectified through the pursuit of a Human Data-driven Discovery (HD3) paradigm. Consistent with other examples of disruptive innovation, we propose that new levels of success are not dependent on new inventions, but rather on the strategic integration of existing data and technology assets. In support of these suggestions, we highlight the power of HD3, through recently published proof-of-concept applications in the areas of drug safety analysis and prediction, drug repositioning, the rational design of combination therapies and the global response to the COVID-19 pandemic. We conclude that innovators must play a key role in expediting the path to a largely human-focused, systems-based approach to drug discovery and research.
Collapse
Affiliation(s)
| | - Rebecca Racz
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA; (R.R.); (K.B.)
| | - Sarah Kim
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL 32827, USA;
| | | | - Keith Burkhart
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA; (R.R.); (K.B.)
| |
Collapse
|
6
|
De Nardi AB, de Oliveira Massoco Salles Gomes C, Fonseca-Alves CE, de Paiva FN, Linhares LCM, Carra GJU, dos Santos Horta R, Ruiz Sueiro FA, Jark PC, Nishiya AT, de Carvalho Vasconcellos CH, Ubukata R, Batschinski K, Sobral RA, Fernandes SC, Biondi LR, De Francisco Strefezzi R, Matera JM, Rangel MMM, dos Anjos DS, Brunner CHM, Laufer-Amorim R, Cadrobbi KG, Cirillo JV, Martins MC, de Paula Reis Filho N, Silva Lessa DF, Portela R, Scarpa Carneiro C, Ricci Lucas SR, Fukumasu H, Feliciano MAR, Gomes Quitzan J, Dagli MLZ. Diagnosis, Prognosis, and Treatment of Canine Hemangiosarcoma: A Review Based on a Consensus Organized by the Brazilian Association of Veterinary Oncology, ABROVET. Cancers (Basel) 2023; 15:cancers15072025. [PMID: 37046686 PMCID: PMC10093745 DOI: 10.3390/cancers15072025] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023] Open
Abstract
Hemangiosarcoma is a mesenchymal neoplasm originating in the endothelial cells of blood vessels; they can be classified as non-visceral and visceral types. Non-visceral hemangiosarcomas can affect the skin, subcutaneous tissues, and muscle tissues; visceral hemangiosarcomas can affect the spleen, liver, heart, lungs, kidneys, oral cavity, bones, bladder, uterus, tongue, and retroperitoneum. Among domestic species, dogs are most affected by cutaneous HSA. Cutaneous HSA represents approximately 14% of all HSA diagnosed in this species and less than 5% of dermal tumors, according to North American studies. However, Brazilian epidemiological data demonstrate a higher prevalence, which may represent 27 to 80% of all canine HSAs and 13.9% of all skin neoplasms diagnosed in this species. Cutaneous HSA most commonly affects middle-aged to elderly dogs (between 8 and 15 years old), with no gender predisposition for either the actinic or non-actinic forms. The higher prevalence of cutaneous HSA in some canine breeds is related to lower protection from solar radiation, as low skin pigmentation and hair coverage lead to greater sun exposure. Actinic changes, such as solar dermatosis, are frequent in these patients, confirming the influence of solar radiation on the development of this neoplasm. There are multiple clinical manifestations of hemangiosarcoma in canines. The diagnostic approach and staging classification of cutaneous HSAs are similar between the different subtypes. The definitive diagnosis is obtained through histopathological analysis of incisional or excisional biopsies. Cytology can be used as a presurgical screening test; however, it has little diagnostic utility in cases of HSA because there is a high risk of blood contamination and sample hemodilution. Surgery is generally the treatment of choice for dogs with localized non-visceral HSA without evidence of metastatic disease. Recently, electrochemotherapy (ECT) has emerged as an alternative therapy for the local ablative treatment of different neoplastic types; the use of radiotherapy for the treatment of dogs with cutaneous HSA is uncommon. There is greater consensus in the literature regarding the indications for adjuvant chemotherapy in subcutaneous and muscular HSA; doxorubicin is the most frequently used antineoplastic agent for subcutaneous and muscular subtypes and can be administered alone or in combination with other drugs. Other therapies include antiangiogenic therapy, photodynamic therapy, the association of chemotherapy with the metronomic dose, targeted therapies, and natural products. The benefits of these therapies are presented and discussed. In general, the prognosis of splenic and cardiac HSA is unfavorable. As a challenging neoplasm, studies of new protocols and treatment modalities are necessary to control this aggressive disease.
Collapse
|
7
|
Wołowiec Ł, Grześk G, Osiak J, Wijata A, Mędlewska M, Gaborek P, Banach J, Wołowiec A, Głowacka M. Beta-blockers in cardiac arrhythmias-Clinical pharmacologist's point of view. Front Pharmacol 2023; 13:1043714. [PMID: 36699057 PMCID: PMC9868422 DOI: 10.3389/fphar.2022.1043714] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/30/2022] [Indexed: 01/11/2023] Open
Abstract
β-blockers is a vast group of antiarrhythmic drugs which differ in their pharmacokinetic and chemical properties. Some of them block β-adrenergic receptors selectively while the others work non-selectively. Consequently, they reduce the influence of the sympathetic nervous system on the heart, acting negatively inotropic, chronotropic, bathmotropic and dromotropic. Although they have been present in medicine since the beginning of the 1960s, they still play a crucial role in the treatment of cardiac arrhythmias. They are also first-line group of drugs used to control the ventricular rate in patients with the most common arrhythmia-atrial fibrillation. Previous reports indicate that infection with SARS-CoV-2 virus may constitute an additional risk factor for arrhythmia. Due to the aging of the population in developed countries and the increase in the number of patients with cardiac burden, the number of people suffering from cardiac arrhythmias will increase in the upcoming years. As a result the role of above-mentioned beta-blockers will remain significant. Particularly noteworthy is propranolol-the oldest beta adrenergic antagonist, which in recent years has found additional applications due to its unique properties. In this article, we reviewed the accessible literature and summarized the current guidelines on the use of beta-blockers in the treatment of cardiac arrhythmias.
Collapse
Affiliation(s)
- Łukasz Wołowiec
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Grzegorz Grześk
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Joanna Osiak
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Aleksandra Wijata
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Martyna Mędlewska
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Patryk Gaborek
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Joanna Banach
- Department of Cardiology and Clinical Pharmacology, Faculty of Health Sciences, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | - Anna Wołowiec
- Department of Geriatrics, Division of Biochemistry and Biogerontology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Toruń, Poland
| | | |
Collapse
|
8
|
Ammons DT, Guth A, Rozental AJ, Kurihara J, Marolf AJ, Chow L, Griffin JF, Makii R, MacQuiddy B, Boss MK, Regan DP, Frank C, McGrath S, Packer RA, Dow S. Reprogramming the Canine Glioma Microenvironment with Tumor Vaccination plus Oral Losartan and Propranolol Induces Objective Responses. CANCER RESEARCH COMMUNICATIONS 2022; 2:1657-1667. [PMID: 36644324 PMCID: PMC9835010 DOI: 10.1158/2767-9764.crc-22-0388] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/09/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022]
Abstract
Purpose Malignant gliomas have a highly immune suppressive tumor microenvironment (TME) which renders them largely unresponsive to conventional therapeutics. Therefore, the present study evaluated a therapeutic protocol designed overcome the immune barrier by combining myeloid cell targeted immunotherapy with tumor vaccination. Experimental Design We utilized a spontaneously occurring canine glioma model to investigate an oral TME modifying immunotherapy in conjunction with cancer stem cell (CSC) vaccination. Dogs were treated daily with losartan (monocyte migration inhibitor) and propranolol (myeloid-derived suppressor cell depleting agent) plus anti-CSC vaccination on a bi-weekly then monthly schedule. Tumor volume was monitored by MRI and correlated with patient immune responses. Results Ten dogs with histologically confirmed gliomas were enrolled into a prospective, open-label clinical trial to evaluate the immunotherapy protocol. Partial tumor regression was observed in 2 dogs, while 6 dogs experienced stable disease, for an overall clinical benefit rate of 80%. Overall survival times (median = 351 days) and progression-free intervals (median = 163 days) were comparable to prior studies evaluating surgical debulking followed by immunotherapy. Dogs with detectable anti-CSC antibody responses had an increased overall survival time relative to dogs that did not generate antibody responses (vaccine responder MST = 500 days; vaccine non-responder MST = 218 days; p = 0.02). Conclusions These findings suggest that combining myeloid cell targeted oral immunotherapy with tumor vaccination can generate objective tumor responses, even in the absence of conventional therapy. Overall, this approach has promise as a readily implemented therapeutic strategy for use in brain cancer patients.
Collapse
Affiliation(s)
- Dylan T. Ammons
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado
| | - Amanda Guth
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado
| | - Aaron J. Rozental
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado
| | - Jade Kurihara
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado
| | - Angela J. Marolf
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| | - Lyndah Chow
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado
| | - John F. Griffin
- Department of Large Animal Clinical Sciences, Texas A&M University, College Station, Texas
| | - Rebecca Makii
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado
| | - Brittany MacQuiddy
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado
| | - Mary-Keara Boss
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| | - Daniel P. Regan
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado
| | - Chad Frank
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado
| | - Stephanie McGrath
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado
| | - Rebecca A. Packer
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado
| | - Steven Dow
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado
- Department of Clinical Sciences, Colorado State University, Fort Collins, Colorado
| |
Collapse
|
9
|
Embaby A, van Merendonk L, Steeghs N, Beijnen J, Huitema A. Beta-adrenergic receptor blockade in angiosarcoma: Which beta-blocker to choose? Front Oncol 2022; 12:940582. [PMID: 36185303 PMCID: PMC9520289 DOI: 10.3389/fonc.2022.940582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/30/2022] [Indexed: 11/28/2022] Open
Abstract
Beta-blockers are currently studied to improve therapeutic options for patients with angiosarcoma. However, most of these patients have no cardiovascular co-morbidity and it is therefore crucial to discuss the most optimal pharmacological properties of beta-blockers for this population. To maximize the possible effectiveness in angiosarcoma, the use of a non-selective beta-blocker is preferred based on in vitro data. To minimize the risk of cardiovascular adverse events a beta-blocker should ideally have intrinsic sympathomimetic activity or vasodilator effects, e.g. labetalol, pindolol or carvedilol. However, except for one case of carvedilol, only efficacy data of propranolol is available. In potential follow-up studies labetalol, pindolol or carvedilol can be considered to reduce the risk of cardiovascular adverse events.
Collapse
Affiliation(s)
- Alaa Embaby
- Department of Medical Oncology and Clinical Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, Netherlands
- *Correspondence: Alaa Embaby,
| | - Lisanne van Merendonk
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, Netherlands
| | - Neeltje Steeghs
- Department of Medical Oncology and Clinical Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, Netherlands
| | - Jos Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, Netherlands
| | - Alwin Huitema
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, Netherlands
- Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
10
|
Solernó LM, Sobol NT, Gottardo MF, Capobianco CS, Ferrero MR, Vásquez L, Alonso DF, Garona J. Propranolol blocks osteosarcoma cell cycle progression, inhibits angiogenesis and slows xenograft growth in combination with cisplatin-based chemotherapy. Sci Rep 2022; 12:15058. [PMID: 36075937 PMCID: PMC9458647 DOI: 10.1038/s41598-022-18324-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 08/09/2022] [Indexed: 11/09/2022] Open
Abstract
Osteosarcoma is still associated with limited response to standard-of-care therapy and alarmingly elevated mortality rates, especially in low- and middle-income countries. Despite multiple efforts to repurpose β-blocker propranolol in oncology, its potential application in osteosarcoma management remains largely unexplored. Considering the unsatisfied clinical needs of this aggressive disease, we evaluated the antitumoral activity of propranolol using different in vitro and in vivo osteosarcoma preclinical models, alone or in addition to chemotherapy. Propranolol significantly impaired cellular growth in β2-adrenergic receptor-expressing MG-63 and U-2OS cells, and was capable of blocking growth-stimulating effects triggered by catecholamines. siRNA-mediated ADRB2 knockdown in MG-63 cells was associated with decreased cell survival and a significant attenuation of PPN anti-osteosarcoma activity. Direct cytostatic effects of propranolol were independent of apoptosis induction and were associated with reduced mitosis, G0/G1 cell cycle arrest and a significant down-regulation of cell cycle regulator Cyclin D1. Moreover, colony formation, 3D spheroid growth, cell chemotaxis and capillary-like tube formation were drastically impaired after propranolol treatment. Interestingly, anti-migratory activity of β-blocker was associated with altered actin cytoskeleton dynamics. In vivo, propranolol treatment (10 mg/kg/day i.p.) reduced the early angiogenic response triggered by MG-63 cells in nude mice. Synergistic effects were observed in vitro after combining propranolol with chemotherapeutic agent cisplatin. Sustained administration of propranolol (10 mg/kg/day i.p., five days a week), alone and especially in addition to low-dose metronomic cisplatin (2 mg/kg/day i.p., three times a week), markedly reduced xenograft progression. After histological analysis, propranolol and cisplatin combination resulted in low tumor mitotic index and increased tumor necrosis. β-blockade using propranolol seems to be an achievable and cost-effective therapeutic approach to modulate osteosarcoma aggressiveness. Further translational studies of propranolol repurposing in osteosarcoma are warranted.
Collapse
Affiliation(s)
- Luisina M Solernó
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina.,Center for Translational Medicine, El Cruce "Néstor Kirchner" Hospital, Buenos Aires, Argentina
| | - Natasha T Sobol
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina.,Center for Translational Medicine, El Cruce "Néstor Kirchner" Hospital, Buenos Aires, Argentina
| | - María F Gottardo
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina.,Center for Translational Medicine, El Cruce "Néstor Kirchner" Hospital, Buenos Aires, Argentina
| | - Carla S Capobianco
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina
| | - Maximiliano R Ferrero
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Biomedicine Research Institute of Buenos Aires (IBioBA), Buenos Aires, Argentina
| | - Liliana Vásquez
- Precision Medicine Research Center, School of Medicine, University of San Martín de Porres, Lima, Perú
| | - Daniel F Alonso
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina.,Center for Translational Medicine, El Cruce "Néstor Kirchner" Hospital, Buenos Aires, Argentina.,National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| | - Juan Garona
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina. .,Center for Translational Medicine, El Cruce "Néstor Kirchner" Hospital, Buenos Aires, Argentina. .,National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
11
|
Gales L, Forsea L, Mitrea D, Stefanica I, Stanculescu I, Mitrica R, Georgescu M, Trifanescu O, Anghel R, Serbanescu L. Antidiabetics, Anthelmintics, Statins, and Beta-Blockers as Co-Adjuvant Drugs in Cancer Therapy. Medicina (B Aires) 2022; 58:medicina58091239. [PMID: 36143915 PMCID: PMC9503803 DOI: 10.3390/medicina58091239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/25/2022] Open
Abstract
Over the last years, repurposed agents have provided growing evidence of fast implementation in oncology treatment such as certain antimalarial, anthelmintic, antibiotics, anti-inflammatory, antihypertensive, antihyperlipidemic, antidiabetic agents. In this study, the four agents of choice were present in our patients’ daily treatment for nonmalignant-associated pathology and have known, light toxicity profiles. It is quite common for a given patient’s daily administration schedule to include two or three of these drugs for the duration of their treatment. We chose to review the latest literature concerning metformin, employed as a first-line treatment for type 2 diabetes; mebendazole, as an anthelmintic; atorvastatin, as a cholesterol-lowering drug; propranolol, used in cardiovascular diseases as a nonspecific inhibitor of beta-1 and beta-2 adrenergic receptors. At the same time, certain key action mechanisms make them feasible antitumor agents such as for mitochondrial ETC inhibition, activation of the enzyme adenosine monophosphate-activated protein kinase, amelioration of endogenous hyperinsulinemia, inhibition of selective tyrosine kinases (i.e., VEGFR2, TNIK, and BRAF), and mevalonate pathway inhibition. Despite the abundance of results from in vitro and in vivo studies, the only solid data from randomized clinical trials confirm metformin-related oncological benefits for only a small subset of nondiabetic patients with HER2-positive breast cancer and early-stage colorectal cancer. At the same time, clinical studies confirm metformin-related detrimental/lack of an effect for lung, breast, prostate cancer, and glioblastoma. For atorvastatin we see a clinical oncological benefit in patients and head and neck cancer, with a trend towards radioprotection of critical structures, thus supporting the role of atorvastatin as a promising agent for concomitant association with radiotherapy. Propranolol-related increased outcomes were seen in clinical studies in patients with melanoma, breast cancer, and sarcoma.
Collapse
Affiliation(s)
- Laurentia Gales
- Department of Oncology, “Carol Davila” University of Medicine & Pharmacy, 022328 Bucharest, Romania
- Department of Oncology, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Leyla Forsea
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Diana Mitrea
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Irina Stefanica
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Irina Stanculescu
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Radu Mitrica
- Department of Oncology, “Carol Davila” University of Medicine & Pharmacy, 022328 Bucharest, Romania
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
- Correspondence: ; Tel.: +40-741-964-311
| | - Mihai Georgescu
- Department of Oncology, “Carol Davila” University of Medicine & Pharmacy, 022328 Bucharest, Romania
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Oana Trifanescu
- Department of Oncology, “Carol Davila” University of Medicine & Pharmacy, 022328 Bucharest, Romania
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Rodica Anghel
- Department of Oncology, “Carol Davila” University of Medicine & Pharmacy, 022328 Bucharest, Romania
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Luiza Serbanescu
- Department of Oncology, “Carol Davila” University of Medicine & Pharmacy, 022328 Bucharest, Romania
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| |
Collapse
|
12
|
Rossi M, Talbot J, Piris P, Grand ML, Montero MP, Matteudi M, Agavnian-Couquiaud E, Appay R, Keime C, Williamson D, Buric D, Bourgarel V, Padovani L, Clifford SC, Ayrault O, Pasquier E, André N, Carré M. Beta-blockers disrupt mitochondrial bioenergetics and increase radiotherapy efficacy independently of beta-adrenergic receptors in medulloblastoma. EBioMedicine 2022; 82:104149. [PMID: 35816899 PMCID: PMC9283511 DOI: 10.1016/j.ebiom.2022.104149] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 11/03/2022] Open
Abstract
Background Medulloblastoma is the most frequent brain malignancy of childhood. The current multimodal treatment comes at the expense of serious and often long-lasting side effects. Drug repurposing is a strategy to fast-track anti-cancer therapy with low toxicity. Here, we showed the ability of β-blockers to potentiate radiotherapy in medulloblastoma with bad prognosis. Methods Medulloblastoma cell lines, patient-derived xenograft cells, 3D spheroids and an innovative cerebellar organotypic model were used to identify synergistic interactions between β-blockers and ionising radiations. Gene expression profiles of β-adrenergic receptors were analysed in medulloblastoma samples from 240 patients. Signaling pathways were explored by RT-qPCR, RNA interference, western blotting and RNA sequencing. Medulloblastoma cell bioenergetics were evaluated by measuring the oxygen consumption rate, the extracellular acidification rate and superoxide production. Findings Low concentrations of β-blockers significantly potentiated clinically relevant radiation protocols. Although patient biopsies showed detectable expression of β-adrenergic receptors, the ability of the repurposed drugs to potentiate ionising radiations did not result from the inhibition of the canonical signaling pathway. We highlighted that the efficacy of the combinatorial treatment relied on a metabolic catastrophe that deprives medulloblastoma cells of their adaptive bioenergetics capacities. This led to an overproduction of superoxide radicals and ultimately to an increase in ionising radiations-mediated DNA damages. Interpretation These data provide the evidence of the efficacy of β-blockers as potentiators of radiotherapy in medulloblastoma, which may help improve the treatment and quality of life of children with high-risk brain tumours. Funding This study was funded by institutional grants and charities.
Collapse
|
13
|
Clanxet J, Teles M, Hernández-Losa J, Rueda MRE, Benitez-Fusté L, Pastor J. Gene expression profiles of beta-adrenergic receptors in canine vascular tumors: a preliminary study. BMC Vet Res 2022; 18:206. [PMID: 35637463 PMCID: PMC9150297 DOI: 10.1186/s12917-022-03317-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 05/18/2022] [Indexed: 11/10/2022] Open
Abstract
Beta adrenergic receptors (β-AR) play a key role in regulating several hallmark pathways of both benign and malignant human and canine tumors. There is scarce information on the expression of β-AR in canine vascular tumors. Therefore, the purpose of the present research work was to study the mRNA expression levels of the three subtypes of the β-AR genes (ADRB1, ADRB2, ADRB3) in hemangiosarcoma (HSA) and hemangioma (HA), as well as in vascular hamartomas (VH) from dogs.Fifty samples (n = 50) were obtained from 38 dogs. Twenty-three animals had HSA, eight animals HA and seven animals VH. HSA were auricular (n = 8), splenic (n = 5), cutaneous (n = 6), auricular and splenic (n = 2), cutaneous-muscular (n = 1) and disseminated (n = 1). There were seven cases of HSA that were divided into primary tumor and secondary (metastatic) tumor. Skin and muscle samples with a normal histological study were used as control group. ADRB gene expression was determinate in all samples by real-time quantitative PCR. Results showed that ADRB1, ADRB2 and ADRB3 were overexpressed in HSA when compared to the control group. ADRB2 was overexpressed in HA when compared to the control group. HSA express higher values of ADBR1 (p = 0.0178) compared to VH. There was a high inter-individual variability in the expression of the three subtypes of ADBR. No statistically significant difference in the expression of ADBR genes were observed between HSA primary when compared to metastatic or in different anatomical locations. In conclusion, canine HSA overexpress the three β-AR subtypes and canine HA β2-AR. High variability was observed in β-AR mRNA levels amongst HSA cases.
Collapse
Affiliation(s)
- Jordi Clanxet
- Department of Animal Medicine and Surgery, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
| | - Mariana Teles
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain.,Institute of Biotechnology and Biomedicine, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
| | - Javier Hernández-Losa
- Molecular Biology Laboratory, Department of Pathology, Hospital Universitario Vall d'Hebron, Passeig Vall d´Hebron, 119-129, 08035, Barcelona, Spain
| | | | | | - Josep Pastor
- Department of Animal Medicine and Surgery, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain.
| |
Collapse
|
14
|
Goerdt LV, Schneider SW, Booken N. Kutane Angiosarkome: molekulare Pathogenese und neue therapeutische Ansätze. J Dtsch Dermatol Ges 2022; 20:429-444. [PMID: 35446507 DOI: 10.1111/ddg.14694_g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 11/16/2021] [Indexed: 12/27/2022]
Abstract
Das kutane Angiosarkom (CAS) ist ein hochaggressiver maligner Tumor mit schlechter Prognose. Das primäre, spontane CAS (pCAS) und das sekundäre, mit einer Bestrahlung oder einem Lymphödem assoziierte CAS (sCAS) unterscheiden sich klinisch sowie molekular. Die Amplifikation/Überexpression von Myc ist ein charakteristisches, wenn auch nicht ausschließliches Merkmal von sCAS, während der Verlust von TP53 selektiv bei pCAS vorkommt. Detaillierte molekulare Analysen mit modernen Multi-Omics-Ansätzen haben gezeigt, dass sowohl pCAS als auch sCAS eine erhebliche molekulare Heterogenität aufweisen. Die betroffenen Gene und ihre molekularen Regulatoren sind mögliche therapeutische Zielstrukturen. Darüber hinaus kann das pCAS in Cluster mit hoher Mutationsrate und/oder ausgeprägten Entzündungssignaturen eingeteilt werden, die als Grundlage für die künftige Stratifizierung von pCAS-Patienten in immuntherapeutischen klinischen Studien dienen können. Während die Aufklärung der der Erkrankung zugrunde liegenden molekularen Veränderungen zügig voranschreitet, verläuft die Entwicklung daraus abgeleiteter neuer Therapien für das CAS jedoch bisher eher langsam. Dennoch wurden einige über die Standardtherapien wie Operation und Radiochemotherapie hinausgehende klinische Studien zu neuen Behandlungsmöglichkeiten initiiert. Dazu gehören zielgerichtete Therapien gegen VEGF und VEGFR1-3 wie Bevacizumab und Pazopanib, sowie β-Adrenozeptorenblocker wie Propranolol. Derzeit werden auch Immuntherapien entwickelt, unter anderem unter Verwendung der Immuncheckpoint-Inhibitoren Pembrolizumab und Nivolumab sowie des Anti-RANKL-Antikörper Denosumab.
Collapse
Affiliation(s)
- Lea V Goerdt
- Klinik für Dermatologie und Venerologie, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg.,Asklepios Campus Hamburg, medizinische Fakultät, Semmelweis Universität Budapest, Hamburg
| | - Stefan W Schneider
- Klinik für Dermatologie und Venerologie, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg
| | - Nina Booken
- Klinik für Dermatologie und Venerologie, Universitätsklinikum Hamburg-Eppendorf (UKE), Hamburg
| |
Collapse
|
15
|
Goerdt LV, Schneider SW, Booken N. Cutaneous Angiosarcomas: Molecular Pathogenesis Guides Novel Therapeutic Approaches. J Dtsch Dermatol Ges 2022; 20:429-443. [PMID: 35218306 DOI: 10.1111/ddg.14694] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 11/16/2021] [Indexed: 12/12/2022]
Abstract
Cutaneous angiosarcoma (CAS) is a highly aggressive cancer with a poor prognosis. Primary, spontaneous CAS (pCAS) and secondary, post-irradiation- or lymphedema-associated CAS (sCAS) are clinically, but also molecularly distinct. Myc amplification/overexpression is a characteristic, although not exclusive feature of sCAS, while loss of TP53 selectively occurs in pCAS. Detailed molecular analyses with modern multi-omics approaches have revealed that both pCAS and sCAS exhibit considerable molecular heterogeneity. Affected genes and their molecular regulators including a plethora of microRNAs may serve as future drug targets. Furthermore, pCAS could be subdivided into clusters with high tumor mutational burden and/or high tumor inflammation signatures providing a rationale for the stratification of pCAS patients in future immunotherapeutic clinical studies. Development of novel treatment regimens guided by these molecular alterations, however, cannot fully keep up with the pace of their discovery due to the low incidence of the disease. Nevertheless, beyond conventional surgery and chemoradiotherapy, clinical trials investigating novel treatment options have been initiated including targeted therapies against VEGF and VEGFR1-3 such as bevacizumab and pazopanib, and β-adrenoreceptor blockers such as propranolol. Finally, immunotherapies are being developed including immune checkpoint inhibitors pembrolizumab and nivolumab as well as anti-RANKL antibody denosumab.
Collapse
Affiliation(s)
- Lea V Goerdt
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany.,Asklepios Campus Hamburg, Medical Faculty, Semmelweis University Budapest, Hamburg, Germany
| | - Stefan W Schneider
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Nina Booken
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
16
|
Propranolol inhibits cell viability and expression of the pro-tumorigenic proteins Akt, NF-ĸB, and VEGF in oral squamous cell carcinoma. Arch Oral Biol 2022; 136:105383. [DOI: 10.1016/j.archoralbio.2022.105383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 02/08/2022] [Accepted: 02/15/2022] [Indexed: 12/11/2022]
|
17
|
Fjæstad KY, Rømer AMA, Goitea V, Johansen AZ, Thorseth ML, Carretta M, Engelholm LH, Grøntved L, Junker N, Madsen DH. Blockade of beta-adrenergic receptors reduces cancer growth and enhances the response to anti-CTLA4 therapy by modulating the tumor microenvironment. Oncogene 2022; 41:1364-1375. [PMID: 35017664 PMCID: PMC8881216 DOI: 10.1038/s41388-021-02170-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 12/08/2021] [Accepted: 12/23/2021] [Indexed: 12/21/2022]
Abstract
The development of immune checkpoint inhibitors (ICI) marks an important breakthrough of cancer therapies in the past years. However, only a limited fraction of patients benefit from such treatments, prompting the search for immune modulating agents that can improve the therapeutic efficacy. The nonselective beta blocker, propranolol, which for decades has been prescribed for the treatment of cardiovascular conditions, has recently been used successfully to treat metastatic angiosarcoma. These results have led to an orphan drug designation by the European Medicines Agency for the treatment of soft tissue sarcomas. The anti-tumor effects of propranolol are suggested to involve the reduction of cancer cell proliferation as well as angiogenesis. Here, we show that oral administration of propranolol delays tumor progression of MCA205 fibrosarcoma model and MC38 colon cancer model and increases the survival rate of tumor bearing mice. Propranolol works by reducing tumor angiogenesis and facilitating an anti-tumoral microenvironment with increased T cell infiltration and reduced infiltration of myeloid-derived suppressor cells (MDSCs). Using T cell deficient mice, we demonstrate that the full anti-tumor effect of propranolol requires the presence of T cells. Flow cytometry-based analysis and RNA sequencing of FACS-sorted cells show that propranolol treatment leads to an upregulation of PD-L1 on tumor associated macrophages (TAMs) and changes in their chemokine expression profile. Lastly, we observe that the co-administration of propranolol significantly enhances the efficacy of anti-CTLA4 therapy. Our results identify propranolol as an immune modulating agent, which can improve immune checkpoint inhibitor therapies in soft tissue sarcoma patients and potentially in other cancers.
Collapse
Affiliation(s)
- Klaire Yixin Fjæstad
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Anne Mette Askehøj Rømer
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Victor Goitea
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Astrid Zedlitz Johansen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Marie-Louise Thorseth
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Marco Carretta
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Lars Henning Engelholm
- Finsen Laboratory, Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Lars Grøntved
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Niels Junker
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Daniel Hargbøl Madsen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark.
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
18
|
Strainienė S, Jauniškis K, Savlan I, Pamedys J, Stundienė I, Liakina V, Valantinas J. Paraneoplastic Phenomena of Disseminated Intravascular Coagulopathy in Hepatic Angiosarcoma – Rare, Challenging and Fatal. Case Report and Literature Review. Acta Med Litu 2021; 28:330-343. [PMID: 35474934 PMCID: PMC8958659 DOI: 10.15388/amed.2021.28.2.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/18/2021] [Accepted: 05/25/2021] [Indexed: 12/18/2022] Open
Abstract
Background. Hepatic angiosarcoma is an uncommon, malignant, primary liver tumor, comprising 2% of liver cancers and accounting for < 1% of all sarcomas. Patients usually present with nonspecific symptoms, such as fatigue, weight loss, right upper quadrant pain, anemia, which leads to late diagnosis of an advanced stage tumor. The median life expectancy after the diagnosis of hepatic angiosarcoma is about 6 months, with only 3% of patients surviving more than 2 years. Liver failure and hemoperitoneum are the leading causes of death in patients with liver angiosarcoma. In rarer cases, it might cause paraneoplastic syndromes such as disseminated intravascular coagulopathy. The treatment of angiosarcomas is complicated as there are no established and effective treatment guidelines due to the tumor’s low frequency and aggressive nature. Case summary. We present the case of a 68-year old woman who was admitted to the hospital due to fatigue and severe anemia (hemoglobin 65 g/l). Laboratory results also revealed high-grade thrombocytopenia (8 × 109/l). The abdominal ultrasound and computed tomography scan showed multiple lesions throughout the liver, spleen and kidneys. After the histological examination of the liver biopsy, the patient was diagnosed with hepatic angiosarcoma. The treatment with first-line chemotherapy (doxorubicin) was initiated despite ongoing paraneoplastic syndrome – disseminative intravascular coagulopathy. However, the disease was terminal, and the patient died 2 months since diagnosed. Conclusions. Hepatic angiosarcoma is a rare and terminal tumor. Therefore, knowledge about its manifestations and effective treatment methods is lacking. Disseminative intravascular coagulopathy is a unique clinical characteristic of angiosarcoma seen in a subset of patients.
Collapse
|
19
|
Martínez-Trufero J, Cruz Jurado J, Gómez-Mateo MC, Bernabeu D, Floría LJ, Lavernia J, Sebio A, García Del Muro X, Álvarez R, Correa R, Hernández-León CN, Marquina G, Hindi N, Redondo A, Martínez V, Asencio JM, Mata C, Valverde Morales CM, Martin-Broto J. Uncommon and peculiar soft tissue sarcomas: Multidisciplinary review and practical recommendations for diagnosis and treatment. Spanish group for Sarcoma research (GEIS - GROUP). Part I. Cancer Treat Rev 2021; 99:102259. [PMID: 34311246 DOI: 10.1016/j.ctrv.2021.102259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/03/2021] [Accepted: 07/06/2021] [Indexed: 12/22/2022]
Affiliation(s)
| | - Josefina Cruz Jurado
- Hospital Universitario Canarias, Medical Oncology Department, Santa Cruz de Tenerife, Spain
| | | | - Daniel Bernabeu
- Hospital Universitario La Paz, Radiology Department, Madrid, Spain
| | - Luis Javier Floría
- Hospital Universitario Miguel Servet, Orthopedic and Traumatology Department, Zaragoza, Spain
| | - Javier Lavernia
- Instituto Valenciano de Oncología, Medical Oncology Department, Valencia, Spain
| | - Ana Sebio
- Hospital Universitario Santa Creu i Sant Pau, Medical Oncology Department, Barcelona, Spain
| | | | - Rosa Álvarez
- Hospital Universitario Gregorio Marañón, Medical Oncology Department, Madrid, Spain
| | - Raquel Correa
- Hospital Virgen de la Victoria, Radiation Oncology Department, Malaga, Spain
| | | | - Gloria Marquina
- Hospital Universitario Clínico San Carlos, Medical Oncology Department, Madrid, Spain
| | - Nadia Hindi
- University Hospital "Fundacion Jimenez Diaz" Madrid, Medical Oncology Department, Madrid, Research Institute FJD-UAM, Madrid (Spain), TBsarc, CITIUS III, Seville, Spain
| | - Andrés Redondo
- Hospital Universitario La Paz, Medical Oncology Department, Madrid, Spain
| | - Virginia Martínez
- Hospital Universitario La Paz, Medical Oncology Department, Madrid, Spain
| | | | - Cristina Mata
- Hospital Universitario Gregorio Marañón, Pediatric and Adolescent Hemato-oncology Department, Madrid, Spain
| | | | - Javier Martin-Broto
- University Hospital "Fundacion Jimenez Diaz" Madrid, Medical Oncology Department, Madrid, Research Institute FJD-UAM, Madrid (Spain), TBsarc, CITIUS III, Seville, Spain
| |
Collapse
|
20
|
Carlos-Escalante JA, de Jesús-Sánchez M, Rivas-Castro A, Pichardo-Rojas PS, Arce C, Wegman-Ostrosky T. The Use of Antihypertensive Drugs as Coadjuvant Therapy in Cancer. Front Oncol 2021; 11:660943. [PMID: 34094953 PMCID: PMC8173186 DOI: 10.3389/fonc.2021.660943] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/19/2021] [Indexed: 12/23/2022] Open
Abstract
Cancer is a complex group of diseases that constitute the second largest cause of mortality worldwide. The development of new drugs for treating this disease is a long and costly process, from the discovery of the molecule through testing in phase III clinical trials, a process during which most candidate molecules fail. The use of drugs currently employed for the management of other diseases (drug repurposing) represents an alternative for developing new medical treatments. Repurposing existing drugs is, in principle, cheaper and faster than developing new drugs. Antihypertensive drugs, primarily belonging to the pharmacological categories of angiotensin-converting enzyme inhibitors, angiotensin II receptors, direct aldosterone antagonists, β-blockers and calcium channel blockers, are commonly prescribed and have well-known safety profiles. Additionally, some of these drugs have exhibited pharmacological properties useful for the treatment of cancer, rendering them candidates for drug repurposing. In this review, we examine the preclinical and clinical evidence for utilizing antihypertensive agents in the treatment of cancer.
Collapse
Affiliation(s)
- José A. Carlos-Escalante
- Plan de Estudios Combinados En Medicina (PECEM) (MD/PhD), Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Marcela de Jesús-Sánchez
- Facultad de Ciencias Biológicas y Agropecuarias, Universidad Veracruzana, Orizaba-Córdoba, Mexico
| | - Alejandro Rivas-Castro
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | | | - Claudia Arce
- Medical Oncology/Breast Tumors, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Talia Wegman-Ostrosky
- Basic Research Subdirection, Instituto Nacional de Cancerología, Mexico City, Mexico
| |
Collapse
|
21
|
Saha J, Kim JH, Amaya CN, Witcher C, Khammanivong A, Korpela DM, Brown DR, Taylor J, Bryan BA, Dickerson EB. Propranolol Sensitizes Vascular Sarcoma Cells to Doxorubicin by Altering Lysosomal Drug Sequestration and Drug Efflux. Front Oncol 2021; 10:614288. [PMID: 33598432 PMCID: PMC7882688 DOI: 10.3389/fonc.2020.614288] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/14/2020] [Indexed: 01/02/2023] Open
Abstract
Angiosarcoma is a rare cancer of blood vessel-forming cells with a high patient mortality and few treatment options. Although chemotherapy often produces initial clinical responses, outcomes remain poor, largely due to the development of drug resistance. We previously identified a subset of doxorubicin-resistant cells in human angiosarcoma and canine hemangiosarcoma cell lines that exhibit high lysosomal accumulation of doxorubicin. Hydrophobic, weak base chemotherapeutics, like doxorubicin, are known to sequester within lysosomes, promoting resistance by limiting drug accessibility to cellular targets. Drug synergy between the beta adrenergic receptor (β-AR) antagonist, propranolol, and multiple chemotherapeutics has been documented in vitro, and clinical data have corroborated the increased therapeutic potential of propranolol with chemotherapy in angiosarcoma patients. Because propranolol is also a weak base and accumulates in lysosomes, we sought to determine whether propranolol enhanced doxorubicin cytotoxicity via antagonism of β-ARs or by preventing the lysosomal accumulation of doxorubicin. β-AR-like immunoreactivities were confirmed in primary tumor tissues and cell lines; receptor function was verified by monitoring downstream signaling pathways of β-ARs in response to receptor agonists and antagonists. Mechanistically, propranolol increased cytoplasmic doxorubicin concentrations in sarcoma cells by decreasing the lysosomal accumulation and cellular efflux of this chemotherapeutic agent. Equivalent concentrations of the receptor-active S-(-) and -inactive R-(+) enantiomers of propranolol produced similar effects, supporting a β-AR-independent mechanism. Long-term exposure of hemangiosarcoma cells to propranolol expanded both lysosomal size and number, yet cells remained sensitive to doxorubicin in the presence of propranolol. In contrast, removal of propranolol increased cellular resistance to doxorubicin, underscoring lysosomal doxorubicin sequestration as a key mechanism of resistance. Our results support the repurposing of the R-(+) enantiomer of propranolol with weak base chemotherapeutics to increase cytotoxicity and reduce the development of drug-resistant cell populations without the cardiovascular and other side effects associated with antagonism of β-ARs.
Collapse
Affiliation(s)
- Jhuma Saha
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States
| | - Jong Hyuk Kim
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States.,Animal Cancer Care and Research Program, College of Veterinary Medicine University of Minnesota, St. Paul, MN, United States.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| | - Clarissa N Amaya
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX, United States.,Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, United States
| | - Caleb Witcher
- Department of Biology, Stephen F. Austin State University, Nacogdoches, TX, United States
| | - Ali Khammanivong
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States
| | - Derek M Korpela
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States
| | - David R Brown
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States
| | - Josephine Taylor
- Department of Biology, Stephen F. Austin State University, Nacogdoches, TX, United States
| | - Brad A Bryan
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX, United States.,Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, United States
| | - Erin B Dickerson
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN, United States.,Animal Cancer Care and Research Program, College of Veterinary Medicine University of Minnesota, St. Paul, MN, United States.,Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
22
|
Yang R, Zhang Y, Liao X, Yao Y, Huang C, Liu L. The Relationship Between Anti-Hypertensive Drugs and Cancer: Anxiety to be Resolved in Urgent. Front Pharmacol 2020; 11:610157. [PMID: 33381045 PMCID: PMC7768037 DOI: 10.3389/fphar.2020.610157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/18/2020] [Indexed: 02/05/2023] Open
Abstract
Hypertension is the prevailing independent risk factor for cardiovascular disease worldwide. Anti-hypertensive drugs are the common and effective cure for lowering blood pressure in patients with hypertension. However, some large-scale clinical studies have pointed out that long-term ingestion of some oral anti-hypertensive drugs was associated with risks of incident cancer and the survival time. In contrast, other studies argue that anti-hypertensive drugs are not related to the occurrence of cancer, even as a complementary therapy of tumor treatment. To resolve the dispute, numerous recent mechanistic studies using animal models have tried to find the causal link between cancer and different anti-hypertensive drugs. However, the results were often contradictory. Such uncertainties have taken a toll on hypertensive patients. In this review, we will summarize advances of longitudinal studies in the association between anti-hypertensive drugs and related tumor risks that have helped to move the field forward from associative to causative conclusions, in hope of providing a reference for more rigorous and evidence-based clinical research on the topic to guide the clinical decision making.
Collapse
Affiliation(s)
- Rong Yang
- Department of International Medical Center/Ward of General Practice, West China Hospital, Sichuan University, Chengdu, China
| | - Yonggang Zhang
- Department of Periodical Press and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyang Liao
- Department of International Medical Center/Ward of General Practice, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Yao
- Department of International Medical Center/Ward of General Practice, West China Hospital, Sichuan University, Chengdu, China
| | - Chuanying Huang
- Department of International Medical Center/Ward of General Practice, West China Hospital, Sichuan University, Chengdu, China
| | - Lixia Liu
- Health Management Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Bejan-Angoulvant T, Angoulvant D. Mise au point sur les bêtabloquants en 2020. Rev Med Interne 2020; 41:741-747. [DOI: 10.1016/j.revmed.2020.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 04/07/2020] [Indexed: 10/24/2022]
|
24
|
Heinhuis KM, IJzerman NS, Koenen AM, van der Graaf WTA, Haas RL, Beijnen JH, Huitema ADR, van Houdt WJ, Steeghs N. PropAngio study protocol: a neoadjuvant trial on the efficacy of propranolol monotherapy in cutaneous angiosarcoma-a proof of principle study. BMJ Open 2020; 10:e039449. [PMID: 32912994 PMCID: PMC7485254 DOI: 10.1136/bmjopen-2020-039449] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION Angiosarcoma is a rare and aggressive malignancy with a high metastatic potential and recurrence rate. Despite optimal treatment with surgery, with or without radiation, the prognosis remains poor and, therefore, new treatment strategies are warranted. Recently, propranolol has effectively been repurposed for the treatment of infantile haemangioma. Propranolol is a β3-sparing antagonist of the β-adrenergic receptor. In infantile haemangioma, the β1, β2 and β3 receptors are highly expressed. Angiosarcoma has several similarities with haemangioma, including its high β-adrenergic receptor expression and the supposedly important role of vascular endothelial growth factor in malignant growth. As a result, propranolol has been administered small scale in individual angiosarcoma cases with promising results. The precise effect of propranolol, however, is not yet established. METHODS AND ANALYSIS The goal of this neoadjuvant window of opportunity study is to prospectively evaluate the activity of propranolol monotherapy in patients with cutaneous angiosarcoma. The neoadjuvant setting provides a good opportunity to rapidly evaluate both the clinical response and histological response, without a significant delay in standard anticancer treatment. Fourteen patients with primary, recurrent or metastatic cutaneous angiosarcoma will be included. Propranolol will be administered orally in an escalating dose during 3-6 weeks, before the initiation of standard treatment. The primary endpoint is clinical response according to Response Evaluation Criteria in Solid Tumours, as measured on consecutive coloured photographs or CT/MRI. The histological response will be determined as secondary endpoint, comparing the difference in proliferation index before and after propranolol by measuring the change in immunohistochemistry staining of Ki-67. The study will be considered positive when at least three patients have a response to propranolol. ETHICS AND DISSEMINATION Ethical approval was obtained from the Medical Ethical Committee of the Netherlands Cancer Institute. Independent of the outcome, results of this study will be shared and submitted for publication in an international peer-reviewed journal. TRIAL REGISTRATION NUMBER NL8118; registry through the Netherlands Trial Register.
Collapse
Affiliation(s)
- Kimberley M Heinhuis
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Nikki S IJzerman
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Anne Miek Koenen
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Winette T A van der Graaf
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rick L Haas
- Department of Radiotherapy, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Winan J van Houdt
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Neeltje Steeghs
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Abstract
Communication between the nervous and immune systems is required for the body to regulate physiological homeostasis. Beta-adrenergic receptors expressed on immune cells mediate the modulation of immune response by neural activity. Activation of beta-adrenergic signaling results in suppression of antitumor immune response and limits the efficacy of cancer immunotherapy. Beta-adrenergic signaling is also involved in regulation of hematopoietic reconstitution, which is critical to the graft-versus-tumor (GVT) effect and to graft-versus-host disease (GVHD) following allogeneic hematopoietic cell transplantation (HCT). In this review, the function of beta-adrenergic signaling in mediating tumor immunosuppression will be highlighted. We will also discuss the implication of targeting beta-adrenergic signaling to improve the efficacy of cancer immunotherapy including the GVT effect, and to diminish the adverse effects including GVHD.
Collapse
Affiliation(s)
- Wei Wang
- Department of Microbiology and Immunology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland
| | - Xuefang Cao
- Department of Microbiology and Immunology, Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Maryland
| |
Collapse
|
26
|
Duckett MM, Phung SK, Nguyen L, Khammanivong A, Dickerson E, Dusenbery K, Lawrence J. The adrenergic receptor antagonists propranolol and carvedilol decrease bone sarcoma cell viability and sustained carvedilol reduces clonogenic survival and increases radiosensitivity in canine osteosarcoma cells. Vet Comp Oncol 2019; 18:128-140. [PMID: 31778284 DOI: 10.1111/vco.12560] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 09/28/2019] [Accepted: 11/19/2019] [Indexed: 11/30/2022]
Abstract
Adrenergic receptor (AR) expression has been demonstrated at several sites of primary and metastatic tumour growth and may influence proliferation, survival, metastasis and angiogenesis. AR antagonists like propranolol and carvedilol inhibit proliferation, induce apoptosis and synergize with chemotherapy agents in some cancers. Radiation resistance is mediated in many cells by upregulation of pro-survival pathways, which may be influenced by ARs. Studies evaluating AR antagonists combined with radiation are limited. The purpose of this study was to determine the effect of propranolol and carvedilol on viability and radiosensitivity in sarcoma cell lines. The hypothesis was that propranolol and carvedilol would increase radiosensitivity in four primary bone sarcoma cell lines. Single agent propranolol or carvedilol inhibited cell viability in all cell lines in a concentration-dependent manner. The mean inhibitory concentrations (IC50 ) for carvedilol were approximately 4-fold lower than propranolol and may be clinically relevant in vivo. Immunoblot analysis confirmed AR expression in both human and canine sarcoma cell lines; however, there was no correlation between baseline AR protein expression and radiosensitivity. Short duration treatment with carvedilol and propranolol did not significantly affect clonogenic survival. Prolonged exposure to propranolol and carvedilol significantly decreased the surviving fraction of canine osteosarcoma cells after 3Gy radiation. Based on our results and possible in vivo activity in dogs, further studies investigating the effects of carvedilol on sarcoma are warranted.
Collapse
Affiliation(s)
- Megan M Duckett
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
| | - Shee Kwan Phung
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
| | - Linh Nguyen
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota
| | - Ali Khammanivong
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota.,Masonic Cancer Center, Masonic Cancer Research Building, University of Minnesota, Minneapolis, Minnesota
| | - Erin Dickerson
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota.,Masonic Cancer Center, Masonic Cancer Research Building, University of Minnesota, Minneapolis, Minnesota
| | - Kathryn Dusenbery
- Department of Radiation Oncology, Medical School, University of Minnesota, Minneapolis, Minnesota
| | - Jessica Lawrence
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St Paul, Minnesota.,Masonic Cancer Center, Masonic Cancer Research Building, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
27
|
Borgatti A, Dickerson EB, Lawrence J. Emerging therapeutic approaches for canine sarcomas: Pushing the boundaries beyond the conventional. Vet Comp Oncol 2019; 18:9-24. [PMID: 31749286 DOI: 10.1111/vco.12554] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 12/21/2022]
Abstract
Sarcomas represent a group of genomically chaotic, highly heterogenous tumours of mesenchymal origin with variable mutational load. Conventional therapy with surgery and radiation therapy is effective for managing small, low-grade sarcomas and remains the standard therapeutic approach. For advanced, high-grade, recurrent, or metastatic sarcomas, systemic chemotherapy provides minimal benefit, therefore, there is a drive to develop novel approaches. The discovery of "Coley's toxins" in the 19th century, and their use to stimulate the immune system supported the application of unconventional therapies for the treatment of sarcomas. While promising, this initial work was abandoned and treatment paradigm and disease course of sarcomas was largely unchanged for several decades. Exciting new therapies are currently changing treatment algorithms for advanced carcinomas and melanomas, and similar approaches are being applied to advance the field of sarcoma research. Recent discoveries in subtype-specific cancer biology and the identification of distinct molecular targets have led to the development of promising targeted strategies with remarkable potential to change the landscape of sarcoma therapy in dogs. The purpose of this review article is to describe the current standard of care and limitations as well as emerging approaches for sarcoma therapy that span many of the most active paradigms in oncologic research, including immunotherapies, checkpoint inhibitors, and drugs capable of cellular metabolic reprogramming.
Collapse
Affiliation(s)
- Antonella Borgatti
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota.,Clinical Investigation Center, College of Veterinary Medicine, St. Paul, Minnesota
| | - Erin B Dickerson
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Jessica Lawrence
- Animal Cancer Care and Research (ACCR) Program, University of Minnesota, St. Paul, Minnesota.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota.,Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
28
|
Fumagalli C, Maurizi N, Marchionni N, Fornasari D. β-blockers: Their new life from hypertension to cancer and migraine. Pharmacol Res 2019; 151:104587. [PMID: 31809852 DOI: 10.1016/j.phrs.2019.104587] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/02/2019] [Accepted: 12/02/2019] [Indexed: 12/28/2022]
Abstract
The pharmacological class of β-blockers includes a plea of molecules with largely different pharmacokinetic and pharmacodynamic characteristics with a protective effect that may span far beyond the cardiovascular system. Although all these compounds share the pharmacological blockade of the adrenergic receptors, each of them is characterized by specific pharmacological properties, including selectivity of action depending on the adrenergic receptors subtypes, intrinsic sympathomimetic activity (ISA), lipid solubility, pharmacokinetic profile, and also other ancillary properties that impact their clinical effect. Their use in the treatment of hypertension has been extensively debated and at the moment a class indication is not present. However, in specific niche of patients, such as in those young individuals in which hypertension is mainly driven by a sympathetic hyperactivation, strong evidence pose β-Blockers as a highly reasonable first-line treatment. Lipophilic β-blockers, specifically propranolol and metoprolol, can cross the Blood Brain Barrier and have a Class A indication for the prophylactic treatment of migraine attacks. Moreover, since β-adrenergic receptors affect the proliferative process of both cancer and immune cells, their blockade has been associated with metastasis reduction in several epithelial and solid organ tumors posing β-Blockers as a new attractive, inexpensive and relatively safe therapeutic strategy in patients with several types of cancer. However, further dedicated prospective, randomized, placebo-controlled studies are needed to determine the real efficacy of these compounds.
Collapse
Affiliation(s)
- Carlo Fumagalli
- Cardiothoracovascular Department, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Italy
| | - Niccolò Maurizi
- Cardiothoracovascular Department, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Italy; Hopital du Valais, Department of Internal Medicine, Sion, Switzerland.
| | - Niccolò Marchionni
- Cardiothoracovascular Department, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Italy
| | - Diego Fornasari
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Italy
| |
Collapse
|
29
|
Cao J, Wang J, He C, Fang M. Angiosarcoma: a review of diagnosis and current treatment. Am J Cancer Res 2019; 9:2303-2313. [PMID: 31815036 PMCID: PMC6895451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 10/03/2019] [Indexed: 06/10/2023] Open
Abstract
Angiosarcoma is a highly malignancy of endothelial tumor and represents 1-2% of all soft tissue sarcomas in humans. The aetiology of angiosarcoma is not clear but there are definite risk factors including chronic lymphoedema, history of radiation, environmental carcinogens and certain familial syndromes. Ultrasound, CT and MR are diagnostic tools, but final diagnosis requires pathological and immunohistochemical confirmation. The conventional options of treatment include surgery, radiotherapy and chemotherapy. Targeted medicines and immunotherapy have been studied as promising treatment of angiosarcoma. The goal of this review is to summarize the current data regarding of angiosarcoma and its clinical presentation and management, providing a useful clinical tool to explore the optimal treatment.
Collapse
Affiliation(s)
- Jun Cao
- Department of Comprehensive Medical Oncology, Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer HospitalHangzhou, Zhejiang, China
| | - Jiale Wang
- Second Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhou, Zhejiang, China
| | - Chiyu He
- Second Clinical Medical College, Zhejiang Chinese Medical UniversityHangzhou, Zhejiang, China
| | - Meiyu Fang
- Department of Comprehensive Medical Oncology, Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer HospitalHangzhou, Zhejiang, China
| |
Collapse
|
30
|
Bönisch N, Langan EA, Terheyden P. [Cutaneous angiosarcoma : Radiochemotherapy with liposomal pegylated doxorubicin]. Hautarzt 2019; 70:700-706. [PMID: 31428802 DOI: 10.1007/s00105-019-4462-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND Whilst cutaneous angiosarcoma is rare tumour which primarily affects elderly patients, its management presents a significant therapeutic challenge. Indeed, complete surgical excision is often not possible due to the location and the diffuse and extensive nature of the tumour. Therefore, current treatment strategies often include chemo- and/or radiotherapy. METHODS We report our experience of combined chemo- and radiotherapy in the clinical course of 6 patients with cutaneous angiosarcoma who were treated between 2007 and 2018. RESULTS All patients presented non-resectable tumours and were treated with radiotherapy in combination with the administration of liposomal, pegylated doxrubicin (25 mg/m2 every 2 weeks). The mean duration of progression-free survival was 8 months (5-14 months), corresponding to an overall survival of 13 months (13-34 months). A partial response was seen in 4 patients and 1 patient developed progressive disease. One patient abandoned therapy after one administration. Two patients developed severe adverse events which led to termination of therapy after 1.5 months and 7 months, i.e. after 4 and 15 cycles respectively. DISCUSSION Combined radio- and chemotherapy with liposomal, pegylated doxorubicin is a useful therapeutic option in the management of cutaneous angiosarcoma. Given the short-lived response rate, new treatment options are urgently required.
Collapse
Affiliation(s)
- N Bönisch
- Klinik für Dermatologie, Universität zu Lübeck, Lübeck, Deutschland
| | - E A Langan
- Klinik für Dermatologie, Universität zu Lübeck, Lübeck, Deutschland.,Dermatological Science, University of Manchester, Manchester, Großbritannien
| | - P Terheyden
- Klinik für Dermatologie, Allergologie und Venerologie, Universität zu Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Deutschland.
| |
Collapse
|
31
|
Roth IM, Wickremesekera AC, Wickremesekera SK, Davis PF, Tan ST. Therapeutic Targeting of Cancer Stem Cells via Modulation of the Renin-Angiotensin System. Front Oncol 2019; 9:745. [PMID: 31440473 PMCID: PMC6694711 DOI: 10.3389/fonc.2019.00745] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 07/24/2019] [Indexed: 12/11/2022] Open
Abstract
Cancer stem cells (CSCs) are proposed to be the cells that initiate tumorigenesis and maintain tumor development due to their self-renewal and multipotency properties. CSCs have been identified in many cancer types and are thought to be responsible for treatment resistance, metastasis, and recurrence. As such, targeting CSCs specifically should result in durable cancer treatment. One potential option for targeting CSCs is by manipulation of the renin-angiotensin system (RAS) and pathways that converge on the RAS with numerous inexpensive medications currently in common clinical use. In addition to its crucial role in cardiovascular and body fluid homeostasis, the RAS is vital for stem cell maintenance and differentiation and plays a role in tumorigenesis and cancer prevention, suggesting that these roles may converge and result in modulation of CSC function by the RAS. In support of this, components of the RAS have been shown to be expressed in many cancer types and have been more recently localized to the CSCs in some tumors. Given these roles of the RAS in tumor development, clinical trials using RAS inhibitors either singly or in combination with other therapies are underway in different cancer types. This review outlines the roles of the RAS, with respect to CSCs, and suggests that the presence of components of the RAS in CSCs could offer an avenue for therapeutic targeting using RAS modulators. Due to the nature of the RAS and its crosstalk with numerous other signaling pathways, a systems approach using traditional RAS inhibitors in combination with inhibitors of bypass loops of the RAS and other signaling pathways that converge on the RAS may offer a novel therapeutic approach to cancer treatment.
Collapse
Affiliation(s)
- Imogen M Roth
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | - Agadha C Wickremesekera
- Gillies McIndoe Research Institute, Wellington, New Zealand.,Department of Neurosurgery, Wellington Regional Hospital, Wellington, New Zealand
| | - Susrutha K Wickremesekera
- Gillies McIndoe Research Institute, Wellington, New Zealand.,Upper Gastrointestinal, Hepatobiliary and Pancreatic Section, Department of General Surgery, Wellington Regional Hospital, Wellington, New Zealand
| | - Paul F Davis
- Gillies McIndoe Research Institute, Wellington, New Zealand
| | - Swee T Tan
- Gillies McIndoe Research Institute, Wellington, New Zealand.,Wellington Regional Plastic, Maxillofacial and Burns Unit, Hutt Hospital, Wellington, New Zealand
| |
Collapse
|
32
|
Fiorentino MD, Monteiro JMC, de Siqueira REB, Kim EIM, Curi AP, Ferrreira CR, Nardo M, de Campos FPF. Primary splenic angiosarcoma: a rare entity often associated with rupture and hemoperitoneum. AUTOPSY AND CASE REPORTS 2019; 9:e2019100. [PMID: 31372360 PMCID: PMC6629266 DOI: 10.4322/acr.2019.100] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 06/06/2019] [Indexed: 12/16/2022] Open
Abstract
Primary splenic angiosarcoma (PSA) is a rare neoplasm of vascular origin associated with aggressive behavior and poor prognosis. The clinical presentation is usually non-specific and is mostly characterized by a wasting disease with anemia and splenomegaly, mimicking a wide range of entities. The authors present the case of an 80-year-old woman with cardiovascular comorbidities with a 6-month history of weight loss, fatigue, weakness, pallor, and abdominal pain. The physical examination showed massive splenomegaly and pallor. After a thorough evaluation that ruled out lymphoproliferative diseases, the working diagnosis was a myelodysplastic disorder. A few days after discharge, she returned to the emergency room with severe abdominal pain, worsening fatigue, and a remarkable pallor. Point-of-care ultrasound showed free intraperitoneal fluid. Spleen rupture was confirmed by abdominal computed tomography (CT) scan, and an emergency laparotomy with splenectomy was performed. The postoperative period was uneventful, and the patient recovered in a few days. The histopathology confirmed the diagnosis of PSA and the patient was referred to an oncological center. Two months later staging CT demonstrated liver and peritoneal metastases, and despite the chemotherapy she died 6 months after the diagnosis.
Collapse
Affiliation(s)
- Matheus Dalben Fiorentino
- Universidade de São Paulo (USP), School of Medicine, Internal Medicine Department. São Paulo, SP, Brazil
| | | | | | - Elizabeth Im Myung Kim
- Universidade de São Paulo (USP), Hospital Universitário, Internal Medicine Department. São Paulo, SP, Brazil
| | - Ana Paula Curi
- Universidade de São Paulo (USP), Hospital Universitário, Internal Medicine Department. São Paulo, SP, Brazil
| | - Cristiane Rubia Ferrreira
- Universidade de São Paulo (USP), Hospital Universitário, Internal Medicine Department. São Paulo, SP, Brazil.,Hospital AC Camargo, Unidade Antonio Prudente, Department of Pathology. São Paulo, SP, Brazil
| | - Mirella Nardo
- Instituto do Câncer do Estado de São Paulo, Oncology Department. São Paulo, SP, Brazil
| | | |
Collapse
|
33
|
Phadke S, Clamon G. Beta blockade as adjunctive breast cancer therapy: A review. Crit Rev Oncol Hematol 2019; 138:173-177. [PMID: 31092374 DOI: 10.1016/j.critrevonc.2019.04.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 04/04/2019] [Accepted: 04/06/2019] [Indexed: 12/16/2022] Open
Abstract
Pre-clinical data has shown that beta adrenergic stimulation can affect the development and progression of many types of cancer. The use of beta blockers as an anti-neoplastic therapy has been studied in retrospective trials and observational trials, but no definitive conclusions about efficacy have been made. Within the realm of breast cancer, significant advances in therapy have led to improved survival outcomes, yet there is room for improvement. Beta adrenergic blockade may prove an effective adjunct to standard breast cancer therapy, with little associated toxicity. This article provides a review of the published literature on beta blockade as an adjunctive cancer therapy, with a focus on breast cancer.
Collapse
Affiliation(s)
- Sneha Phadke
- University of Iowa Carver College of Medicine, Department of Internal Medicine, Division of Hematology, Oncology, and Blood and Marrow Transplantation, 200 Hawkins Drive, Iowa City, IA 52242, United States.
| | - Gerald Clamon
- University of Iowa Carver College of Medicine, Department of Internal Medicine, Division of Hematology, Oncology, and Blood and Marrow Transplantation, 200 Hawkins Drive, Iowa City, IA 52242, United States
| |
Collapse
|
34
|
Galván DC, Ayyappan AP, Bryan BA. Regression of primary cardiac angiosarcoma and metastatic nodules following propranolol as a single agent treatment. Oncoscience 2018; 5:264-268. [PMID: 30460329 PMCID: PMC6231448 DOI: 10.18632/oncoscience.472] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/02/2018] [Indexed: 12/11/2022] Open
Abstract
Angiosarcoma is the most common malignant cardiac tumor. Cardiac angiosarcoma is a highly lethal neoplasm that is largely resistant to conventional anti-cancer therapy. Mean survival of patients with cardiac angiosarcoma is only 4 months, and almost all patients will succumb to the disease within 1 year. The beta blocker propranolol is an emerging therapy against angiosarcoma. When combined with conventional therapies, propranolol increases progression free and overall survival in patients with this tumor type. It is currently unknown if propranolol is capable of showing anti-cancer efficacy as a single agent therapy. We report a case of a 61 year old woman diagnosed with primary cardiac angiosarcoma and liver and lung metastases. This patient chose to decline conventional therapy, and instead was prescribed the beta blocker propranolol as a single agent treatment. After 12 months, the mediastinal mass substantially debulked and decreased in size, and the metastatic nodules stabilized or resolved with no evidence of hyper-metabolic activity on PET-CT. This is the first reported data showing long term efficacy of the beta blocker propranolol as a single agent therapy against angiosarcoma.
Collapse
Affiliation(s)
- Dana C Galván
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Anoop P Ayyappan
- Department of Radiology, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Brad A Bryan
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX, USA
| |
Collapse
|