1
|
An overview of kinin mediated events in cancer progression and therapeutic applications. Biochim Biophys Acta Rev Cancer 2022; 1877:188807. [PMID: 36167271 DOI: 10.1016/j.bbcan.2022.188807] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/12/2022] [Accepted: 09/21/2022] [Indexed: 11/22/2022]
Abstract
Kinins are bioactive peptides generated in the inflammatory milieu of the tissue microenvironment, which is involved in cancer progression and inflammatory response. Kinins signals through activation of two G-protein coupled receptors; inducible Bradykinin Receptor B1 (B1R) and constitutive receptor B2 (B2R). Activation of kinin receptors and its cross-talk with receptor tyrosine kinases activates multiple signaling pathways, including ERK/MAPK, PI3K, PKC, and p38 pathways regulating cancer hallmarks. Perturbations of the kinin-mediated events are implicated in various aspects of cancer invasion, matrix remodeling, and metastasis. In the tumor microenvironment, kinins initiate fibroblast activation, mesenchymal stem cell interactions, and recruitment of immune cells. Albeit the precise nature of kinin function in the metastasis and tumor microenvironment are not completely clear yet, several kinin receptor antagonists show anti-metastatic potential. Here, we showcase an overview of the complex biology of kinins and their role in cancer pathogenesis and therapeutic aspects.
Collapse
|
2
|
Bertozzi MM, Saraiva-Santos T, Zaninelli TH, Pinho-Ribeiro FA, Fattori V, Staurengo-Ferrari L, Ferraz CR, Domiciano TP, Calixto-Campos C, Borghi SM, Zarpelon AC, Cunha TM, Casagrande R, Verri WA. Ehrlich Tumor Induces TRPV1-Dependent Evoked and Non-Evoked Pain-like Behavior in Mice. Brain Sci 2022; 12:brainsci12091247. [PMID: 36138983 PMCID: PMC9496717 DOI: 10.3390/brainsci12091247] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/09/2022] [Accepted: 09/11/2022] [Indexed: 11/16/2022] Open
Abstract
We standardized a model by injecting Ehrlich tumor cells into the paw to evaluate cancer pain mechanisms and pharmacological treatments. Opioid treatment, but not cyclooxygenase inhibitor or tricyclic antidepressant treatments reduces Ehrlich tumor pain. To best use this model for drug screening it is essential to understand its pathophysiological mechanisms. Herein, we investigated the contribution of the transient receptor potential cation channel subfamily V member 1 (TRPV1) in the Ehrlich tumor-induced pain model. Dorsal root ganglia (DRG) neurons from the Ehrlich tumor mice presented higher activity (calcium levels using fluo-4 fluorescent probe) and an increased response to capsaicin (TRPV1 agonist) than the saline-injected animals (p < 0.05). We also observed diminished mechanical (electronic von Frey) and thermal (hot plate) hyperalgesia, paw flinching, and normalization of weight distribution imbalance in TRPV1 deficient mice (p < 0.05). On the other hand, TRPV1 deficiency did not alter paw volume or weight, indicating no significant alteration in tumor growth. Intrathecal injection of AMG9810 (TRPV1 antagonist) reduced ongoing Ehrlich tumor-triggered mechanical and thermal hyperalgesia (p < 0.05). Therefore, the contribution of TRPV1 to Ehrlich tumor pain behavior was revealed by genetic and pharmacological approaches, thus, supporting the use of this model to investigate TRPV1-targeting therapies for the treatment of cancer pain.
Collapse
Affiliation(s)
- Mariana M. Bertozzi
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil
| | - Telma Saraiva-Santos
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil
| | - Tiago H. Zaninelli
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil
| | - Felipe A. Pinho-Ribeiro
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Victor Fattori
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil
| | - Larissa Staurengo-Ferrari
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil
| | - Camila R. Ferraz
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil
| | - Talita P. Domiciano
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil
| | - Cassia Calixto-Campos
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil
| | - Sergio M. Borghi
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil
- Center for Research in Health Sciences, University of Northern Londrina, Londrina 86041-120, PR, Brazil
| | - Ana C. Zarpelon
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil
| | - Thiago M. Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, Ribeirão Preto 14049-900, SP, Brazil
| | - Rubia Casagrande
- Department of Pharmaceutical Sciences, Center of Health Science, Londrina State University, Londrina 86038-440, PR, Brazil
| | - Waldiceu A. Verri
- Laboratory of Pain, Inflammation, Neuropathy, and Cancer, Department of Pathology, Center of Biological Sciences, Londrina State University, Londrina 86057-970, PR, Brazil
- Correspondence: or ; Tel.: +55-43-3371-4979; Fax: +55-43-3371-4387
| |
Collapse
|
3
|
Lau J, Rousseau J, Kwon D, Bénard F, Lin KS. A Systematic Review of Molecular Imaging Agents Targeting Bradykinin B1 and B2 Receptors. Pharmaceuticals (Basel) 2020; 13:ph13080199. [PMID: 32824565 PMCID: PMC7464927 DOI: 10.3390/ph13080199] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/11/2020] [Accepted: 08/13/2020] [Indexed: 12/22/2022] Open
Abstract
Kinins, bradykinin and kallidin are vasoactive peptides that signal through the bradykinin B1 and B2 receptors (B1R and B2R). B2R is constitutively expressed in healthy tissues and mediates responses such as vasodilation, fluid balance and retention, smooth muscle contraction, and algesia, while B1R is absent in normal tissues and is induced by tissue trauma or inflammation. B2R is activated by kinins, while B1R is activated by kinins that lack the C-terminal arginine residue. Perturbations of the kinin system have been implicated in inflammation, chronic pain, vasculopathy, neuropathy, obesity, diabetes, and cancer. In general, excess activation and signaling of the kinin system lead to a pro-inflammatory state. Depending on the disease context, agonism or antagonism of the bradykinin receptors have been considered as therapeutic options. In this review, we summarize molecular imaging agents targeting these G protein-coupled receptors, including optical and radioactive probes that have been used to interrogate B1R/B2R expression at the cellular and anatomical levels, respectively. Several of these preclinical agents, described herein, have the potential to guide therapeutic interventions for these receptors.
Collapse
Affiliation(s)
- Joseph Lau
- Department of Molecular Oncology, BC Cancer, Vancouver, BC V5Z 1L3 Canada
| | - Julie Rousseau
- Department of Molecular Oncology, BC Cancer, Vancouver, BC V5Z 1L3 Canada
| | - Daniel Kwon
- Department of Molecular Oncology, BC Cancer, Vancouver, BC V5Z 1L3 Canada
| | - François Bénard
- Department of Molecular Oncology, BC Cancer, Vancouver, BC V5Z 1L3 Canada
- Department of Radiology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Kuo-Shyan Lin
- Department of Molecular Oncology, BC Cancer, Vancouver, BC V5Z 1L3 Canada
- Department of Radiology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
4
|
Dubuc C, Savard M, Bovenzi V, Lessard A, Côté J, Neugebauer W, Geha S, Chemtob S, Gobeil F. Antitumor activity of cell-penetrant kinin B1 receptor antagonists in human triple-negative breast cancer cells. J Cell Physiol 2018; 234:2851-2865. [PMID: 30132865 DOI: 10.1002/jcp.27103] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 06/28/2018] [Indexed: 12/26/2022]
Abstract
High nuclear expression of G protein-coupled receptors, including kinin B1 receptors (B1R), has been observed in several human cancers, but the clinical significance of this is unknown. We put forward the hypothesis that these "nuclearized" kinin B1R contribute to tumorigenicity and can be a new target in anticancer strategies. Our initial immunostaining and ultrastructural electron microscopy analyses demonstrated high B1R expression predominantly located at internal/nuclear compartments in the MDA-MB-231 triple-negative breast cancer (TNBC) cell line as well as in clinical samples of patients with TNBC. On the basis of these findings, in the present study, we evaluated the anticancer therapeutic potential of newly identified, cell-permeable B1R antagonists in MDA-MB-231 cells (ligand-receptor binding/activity assays and LC-MS/MS analyses). We found that these compounds (SSR240612, NG67, and N2000) were more toxic to MDA-MB-231 cells in comparison with low- or non-B1R expressing MCF-10A normal human mammary epithelial cells and COS-1 cells, respectively (clonogenic, MTT proliferative/cytocidal assays, and fluorescence-activated cell-sorting (FACS)-based apoptosis analyses). By comparison, the peptide B1R antagonist R954 unable to cross cell membrane failed to produce anticancer effects. Furthermore, the putative mechanisms underlying the anticancer activities of cell-penetrant B1R antagonists were assessed by analyzing cell cycle regulation and signaling molecules related to cell survival and apoptosis (FACS and western blot). Finally, drug combination experiments showed that cell-penetrant B1R antagonists can cooperate with suboptimal doses of chemotherapeutic agents (doxorubicin and paclitaxel) to promote TNBC death. This study provides evidence on the potential value of internally acting kinin B1R antagonists in averting growth of breast cancer.
Collapse
Affiliation(s)
- Céléna Dubuc
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Martin Savard
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Veronica Bovenzi
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Andrée Lessard
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jérôme Côté
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Witold Neugebauer
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Sameh Geha
- Department of Pathology, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
| | - Sylvain Chemtob
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, Québec, Canada
| | - Fernand Gobeil
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
5
|
da Costa PLN, Wynne D, Fifis T, Nguyen L, Perini M, Christophi C. The kallikrein-Kinin system modulates the progression of colorectal liver metastases in a mouse model. BMC Cancer 2018; 18:382. [PMID: 29618333 PMCID: PMC5885419 DOI: 10.1186/s12885-018-4260-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 03/20/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The Kallikrein-Kinin System (KKS) has been found to play a role in tumor progression in several cancers. The KKS metabolic cascade depends on signalling through two cross talking receptors; bradykinin receptor 1 (B1R) and bradykinin receptor 2 (B2R). Activation of the Kinin receptor is responsible for multiple pathophysiologic functions including increase of vascular permeability and induction of host inflammatory responses that exert diverse effects on tumor growth. METHODS B1R and B2R expression on mouse and human CRC cell lines was investigated. Changes in tumor growth and progression was assessed in male CBA mice bearing colorectal liver metastases (CRLM) following treatment with B1R or B2R blockers. In vitro cultures of human SW-480 and mouse colorectal cancer (MoCR) cell lines were examined for changes in their proliferation and migration properties following treatment with B1R or B2R blockers. RESULTS Both colorectal cancer cell lines tested strongly positive for B1R and B2R expression. Inhibition of both receptors retarded tumor growth but only B1R blockade significantly reduced tumor load and increased tumor apoptosis. Blockade of either receptor reduced tumor vascularization in vivo and significantly inhibited proliferation and migration of colorectal cancer cells in vitro. CONCLUSION Taken together, the present study demonstrated that kinin receptor blockade inhibited tumor growth and reduced its invading properties suggesting that KKS manipulation could be a novel target in colorectal cancer therapy.
Collapse
Affiliation(s)
- Patricia Luiza Nunes da Costa
- Department of Surgery, University of Melbourne, Austin Health, Lance Townsend Building Level 8, Studley Rd, Heidelberg, VIC, 3084, Australia
- Laboratório de Oncologia Experimental, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| | - David Wynne
- Department of Surgery, University of Melbourne, Austin Health, Lance Townsend Building Level 8, Studley Rd, Heidelberg, VIC, 3084, Australia
| | - Theodora Fifis
- Department of Surgery, University of Melbourne, Austin Health, Lance Townsend Building Level 8, Studley Rd, Heidelberg, VIC, 3084, Australia.
| | - Linh Nguyen
- Department of Surgery, University of Melbourne, Austin Health, Lance Townsend Building Level 8, Studley Rd, Heidelberg, VIC, 3084, Australia
| | - Marcos Perini
- Department of Surgery, University of Melbourne, Austin Health, Lance Townsend Building Level 8, Studley Rd, Heidelberg, VIC, 3084, Australia
| | - Christopher Christophi
- Department of Surgery, University of Melbourne, Austin Health, Lance Townsend Building Level 8, Studley Rd, Heidelberg, VIC, 3084, Australia
| |
Collapse
|
6
|
Khan M, Huang T, Lin CY, Wu J, Fan BM, Bian ZX. Exploiting cancer's phenotypic guise against itself: targeting ectopically expressed peptide G-protein coupled receptors for lung cancer therapy. Oncotarget 2017; 8:104615-104637. [PMID: 29262666 PMCID: PMC5732832 DOI: 10.18632/oncotarget.18403] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/23/2017] [Indexed: 02/07/2023] Open
Abstract
Lung cancer, claiming millions of lives annually, has the highest mortality rate worldwide. This advocates the development of novel cancer therapies that are highly toxic for cancer cells but negligibly toxic for healthy cells. One of the effective treatments is targeting overexpressed surface receptors of cancer cells with receptor-specific drugs. The receptors-in-focus in the current review are the G-protein coupled receptors (GPCRs), which are often overexpressed in various types of tumors. The peptide subfamily of GPCRs is the pivot of the current article owing to the high affinity and specificity to and of their cognate peptide ligands, and the proven efficacy of peptide-based therapeutics. The article summarizes various ectopically expressed peptide GPCRs in lung cancer, namely, Cholecystokinin-B/Gastrin receptor, the Bombesin receptor family, Bradykinin B1 and B2 receptors, Arginine vasopressin receptors 1a, 1b and 2, and the Somatostatin receptor type 2. The autocrine growth and pro-proliferative pathways they mediate, and the distinct tumor-inhibitory effects of somatostatin receptors are then discussed. The next section covers how these pathways may be influenced or 'corrected' through therapeutics (involving agonists and antagonists) targeting the overexpressed peptide GPCRs. The review proceeds on to Nano-scaled delivery platforms, which enclose chemotherapeutic agents and are decorated with peptide ligands on their external surface, as an effective means of targeting cancer cells. We conclude that targeting these overexpressed peptide GPCRs is potentially evolving as a highly promising form of lung cancer therapy.
Collapse
Affiliation(s)
- Mahjabin Khan
- Laboratory of Brain-Gut Research, School of Chinese Medicine, Hong Kong Baptist University, HKSAR, Kowloon Tong, P.R. China
| | - Tao Huang
- Laboratory of Brain-Gut Research, School of Chinese Medicine, Hong Kong Baptist University, HKSAR, Kowloon Tong, P.R. China
| | - Cheng-Yuan Lin
- Laboratory of Brain-Gut Research, School of Chinese Medicine, Hong Kong Baptist University, HKSAR, Kowloon Tong, P.R. China
- YMU-HKBU Joint Laboratory of Traditional Natural Medicine, Yunnan Minzu University, Kunming, P.R. China
| | - Jiang Wu
- State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, P. R. China
| | - Bao-Min Fan
- YMU-HKBU Joint Laboratory of Traditional Natural Medicine, Yunnan Minzu University, Kunming, P.R. China
| | - Zhao-Xiang Bian
- Laboratory of Brain-Gut Research, School of Chinese Medicine, Hong Kong Baptist University, HKSAR, Kowloon Tong, P.R. China
| |
Collapse
|
7
|
Araújo WF, Naves MA, Ravanini JN, Schor N, Teixeira VPC. Renin-angiotensin system (RAS) blockade attenuates growth and metastatic potential of renal cell carcinoma in mice. Urol Oncol 2015; 33:389.e1-7. [PMID: 25595575 DOI: 10.1016/j.urolonc.2014.11.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 11/27/2014] [Accepted: 11/27/2014] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Renal cell carcinoma (RCC) is the most frequent type of cancer among renal neoplasms in adults and responds poorly to radiotherapy and chemotherapy. There is evidence that blockade of the renin-angiotensin system (RAS) might have antineoplastic effects. The aim of this study was to investigate the effects of RAS blockade on RCC in a murine model. METHODS AND MATERIALS Murine renal cancer cells (Renca) were injected (1 × 10(5)) into the subcapsular space of the left kidney of BALB/c mice (8 wk of age). The animals were divided into 4 groups: a control group (no treatment), angiotensin-receptor blockers group (losartan 100mg/kg/d), angiotensin-converting enzyme inhibitor group (captopril 10mg/kg/d), and angiotensin-receptor blockers +angiotensin-converting enzyme inhibitor group (losartan 100mg/kg/d +captopril 10mg/kg/d). The animals received the drugs by gavage for 21 days after inoculation, beginning 2 days before tumor induction, and were then euthanized. After killing the animals, the kidneys and lungs were removed, weighed, and processed for histopathological and immunohistochemical analyses. Angiogenesis and vascular microvessels were assessed with the antibodies anti-vascular endothelial growth factor and anti-CD34. RESULTS Angiotensin II-inoculated animals developed renal tumors. Treated animals presented smaller tumors, regardless of the therapeutic regimen, and far fewer lung metastases in both quantity and dimension compared with the controls. The expression of vascular endothelial growth factor and CD34 were significantly decreased in renal tumors of treated animals compared with the controls. CONCLUSIONS Our findings suggest that blockade of RAS decreases tumor proliferation and metastatic capacity of RCC in this experimental model.
Collapse
Affiliation(s)
- Wedson F Araújo
- Department of Pathology, Universidade Federal de São Paulo (UNIFESP-EPM), São Paulo, Brazil.
| | - Marcelo A Naves
- Department of Medicine, Universidade Federal de São Paulo (UNIFESP-EPM), São Paulo, Brazil
| | - Juliana N Ravanini
- Department of Pathology, Universidade de São Paulo (USP), São Paulo, Brazil
| | - Nestor Schor
- Department of Medicine, Universidade Federal de São Paulo (UNIFESP-EPM), São Paulo, Brazil
| | - Vicente P C Teixeira
- Department of Pathology, Universidade Federal de São Paulo (UNIFESP-EPM), São Paulo, Brazil; Department of Medicine, Universidade Federal de São Paulo (UNIFESP-EPM), São Paulo, Brazil
| |
Collapse
|
8
|
Gobeil F, Sirois P, Regoli D. Preclinical pharmacology, metabolic stability, pharmacokinetics and toxicology of the peptidic kinin B1 receptor antagonist R-954. Peptides 2014; 52:82-9. [PMID: 24361511 DOI: 10.1016/j.peptides.2013.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 12/06/2013] [Accepted: 12/06/2013] [Indexed: 01/06/2023]
Abstract
We previously showed that R-954 (AcOrn[Oic(2),(αMe)Phe(5),dβNal(7),Ile(8)]desArg(9)-bradykinin) is a potent, selective and stable peptide antagonist of the inducible GPCR kinin B1 receptor. This compound shows potential applications for the treatment of several diseases, including cancer and neurological disturbances of diabetes. To enable clinical translation, more information regarding its pharmacological, pharmacokinetics (PK) and toxicological properties at preclinical stage is warranted. This was the principal objective of the present study. Herein, specificity of R-954 was characterized in binding studies on 133 human molecular targets to reveal minor cross-reactivities against the angiotensin AT2 and the bombesin receptors (110- and 330-fold lower affinity than for B1R, respectively). The pharmacokinetic of R-954 was studied in both normal and streptozotocin-diabetic anaesthetized rats providing half-lives of 1.9-2.7h. R-954 does not appear to be metabolized in the rat circulation and in several rat tissue homogenates, as the kidney, lung and liver. It appears to be excreted as parent drug in the bile (21%) and in urine. A preliminary toxicological profile of R-954 was obtained in rats under various administration routes. R-954 appears to be well tolerated. Overall, these results indicate that R-954 exhibits favorable preclinical pharmacological/PK characteristics and encouraging safety profiles, suitable for early studies in humans.
Collapse
Affiliation(s)
- Fernand Gobeil
- Department of Pharmacology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Quebec, Canada J1H 5N4; Institute of Pharmacology of Sherbrooke (IPS), Faculty of Medicine and Health Sciences, Université de Sherbrooke, Quebec, Canada J1H 5N4.
| | - Pierre Sirois
- CHUL Research Center, Laval University, Quebec, Canada G1V 4G2
| | - Domenico Regoli
- Department of Pharmacology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Quebec, Canada J1H 5N4
| |
Collapse
|
9
|
da Costa PLN, Sirois P, Tannock IF, Chammas R. The role of kinin receptors in cancer and therapeutic opportunities. Cancer Lett 2013; 345:27-38. [PMID: 24333733 DOI: 10.1016/j.canlet.2013.12.009] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 11/29/2013] [Accepted: 12/02/2013] [Indexed: 12/20/2022]
Abstract
Kinins are generated within inflammatory tissue microenvironments, where they exert diverse functions, including cell proliferation, leukocyte activation, cell migration, endothelial cell activation and nociception. These pleiotropic functions depend on signaling through two cross talking receptors, the constitutively expressed kinin receptor 2 (B2R) and the inducible kinin receptor 1 (B1R). We have reviewed evidence, which supports the concept that kinin receptors, especially kinin receptor 1, are promising targets for cancer therapy, since (1) many tumor cells express aberrantly high levels of these receptors; (2) some cancers produce kinins and use them as autocrine factors to stimulate their growth; (3) activation of kinin receptors leads to activation of macrophages, dendritic cells and other cells from the tumor microenvironment; (4) kinins have pro-angiogenic properties; (5) kinin receptors have been implicated in cancer migration, invasion and metastasis; and (6) selective antagonists for either B1R or B2R have shown anti-proliferative, anti-inflammatory, anti-angiogenic and anti-migratory properties. The multiple cross talks between kinin receptors and renin-angiotensin system (RAS) as well as its implications for targeting KKS or RAS for the treatment of malignancies are also discussed. It is expected that B1R antagonists would interfere less with housekeeping functions and therefore would be attractive compounds to treat selected types of cancer. Reliable clinical studies are needed to establish the translatability of these data to human settings and the usefulness of kinin receptor antagonists.
Collapse
Affiliation(s)
- Patrícia L N da Costa
- Laboratório de Oncologia Experimental, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, Brazil
| | - Pierre Sirois
- CHUL Research Center, Laval University, Quebec City, Canada
| | - Ian F Tannock
- Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada
| | - Roger Chammas
- Laboratório de Oncologia Experimental, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, Brazil.
| |
Collapse
|
10
|
Okwan-Duodu D, Landry J, Shen XZ, Diaz R. Angiotensin-converting enzyme and the tumor microenvironment: mechanisms beyond angiogenesis. Am J Physiol Regul Integr Comp Physiol 2013; 305:R205-15. [DOI: 10.1152/ajpregu.00544.2012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The renin angiotensin system (RAS) is a network of enzymes and peptides that coalesce primarily on the angiotensin II type 1 receptor (AT1R) to induce cell proliferation, angiogenesis, fibrosis, and blood pressure control. Angiotensin-converting enzyme (ACE), the key peptidase of the RAS, is promiscuous in that it cleaves other substrates such as substance P and bradykinin. Accumulating evidence implicates ACE in the pathophysiology of carcinogenesis. While the role of ACE and its peptide network in modulating angiogenesis via the AT1R is well documented, its involvement in shaping other aspects of the tumor microenvironment remains largely unknown. Here, we review the role of ACE in modulating the immune compartment of the tumor microenvironment, which encompasses the immunosuppressive, cancer-promoting myeloid-derived suppressor cells, alternatively activated tumor-associated macrophages, and T regulatory cells. We also discuss the potential roles of peptides that accumulate in the setting of chronic ACE inhibitor use, such as bradykinin, substance P, and N-acetyl-seryl-aspartyl-lysyl-proline (Ac-SDKP), and how they may undercut the gains of anti-angiogenesis from ACE inhibition. These emerging mechanisms may harmonize the often-conflicting results on the role of ACE inhibitors and ACE polymorphisms in various cancers and call for further investigations into the potential benefit of ACE inhibitors in some neoplasms.
Collapse
Affiliation(s)
- Derick Okwan-Duodu
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia; and
| | - Jerome Landry
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia; and
| | - Xiao Z. Shen
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Roberto Diaz
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia; and
| |
Collapse
|
11
|
Dillenburg-Pilla P, Maria AG, Reis RI, Floriano EM, Pereira CD, De Lucca FL, Ramos SG, Pesquero JB, Jasiulionis MG, Costa-Neto CM. Activation of the kinin B1 receptor attenuates melanoma tumor growth and metastasis. PLoS One 2013; 8:e64453. [PMID: 23691222 PMCID: PMC3656876 DOI: 10.1371/journal.pone.0064453] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Accepted: 04/15/2013] [Indexed: 01/02/2023] Open
Abstract
Melanoma is a very aggressive tumor that does not respond well to standard therapeutic approaches, such as radio- and chemotherapies. Furthermore, acquiring the ability to metastasize in melanoma and many other tumor types is directly related to incurable disease. The B1 kinin receptor participates in a variety of cancer-related pathophysiological events, such as inflammation and angiogenesis. Therefore, we investigated whether this G protein-coupled receptor plays a role in tumor progression. We used a murine melanoma cell line that expresses the kinin B1 receptor and does not express the kinin B2 receptor to investigate the precise contribution of activation of the B1 receptor in tumor progression and correlated events using various in vitro and in vivo approaches. Activation of the kinin B1 receptor in the absence of B2 receptor inhibits cell migration in vitro and decreases tumor formation in vivo. Moreover, tumors formed from cells stimulated with B1-specific agonist showed several features of decreased aggressiveness, such as smaller size and infiltration of inflammatory cells within the tumor area, higher levels of pro-inflammatory cytokines implicated in the host anti-tumor immune response, lower number of cells undergoing mitosis, a poorer vascular network, no signs of invasion of surrounding tissues or metastasis and increased animal survival. Our findings reveal that activation of the kinin B1 receptor has a host protective role during murine melanoma tumor progression, suggesting that the B1 receptor could be a new anti-tumor GPCR and provide new opportunities for therapeutic targeting.
Collapse
Affiliation(s)
- Patricia Dillenburg-Pilla
- Department of Biochemistry and Immunology, Faculty of Medicine at Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Andrea G. Maria
- Department of Biochemistry and Immunology, Faculty of Medicine at Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Rosana I. Reis
- Department of Biochemistry and Immunology, Faculty of Medicine at Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Elaine Medeiros Floriano
- Departament of Pathology, Faculty of Medicine at Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Cacilda Dias Pereira
- Department of Biochemistry and Immunology, Faculty of Medicine at Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Fernando Luiz De Lucca
- Department of Biochemistry and Immunology, Faculty of Medicine at Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Simone Gusmão Ramos
- Departament of Pathology, Faculty of Medicine at Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - João B. Pesquero
- Department of Biophysics, Federal University of São Paulo, São Paulo, Brazil
| | | | - Claudio M. Costa-Neto
- Department of Biochemistry and Immunology, Faculty of Medicine at Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
- * E-mail:
| |
Collapse
|