1
|
Barranco MM, Zecchinati F, Perdomo VG, Habib MJ, Rico MJ, Rozados VR, Salazar M, Fusini ME, Scharovsky OG, Villanueva SSM, Mainetti LE, García F. Intestinal ABC transporters: Influence on the metronomic cyclophosphamide-induced toxic effect in an obese mouse mammary cancer model. Toxicol Appl Pharmacol 2024; 492:117130. [PMID: 39426530 DOI: 10.1016/j.taap.2024.117130] [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: 01/18/2024] [Revised: 09/26/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
Metronomic chemotherapy (MCT) is a cancer therapeutic approach characterized by low dose drug chronic administration and limited or null toxicity. Obesity-induced metabolic alterations worsen cancer prognosis and influence the intestinal biochemical barrier, altering the Multidrug resistance-associated protein 2 (Mrp2) and Multidrug resistance protein-1 (Mdr-1), efflux pumps that transport chemotherapeutic drugs. Obesity and cancer are frequent co-morbidities; thus, our aim was to evaluate the effectiveness and toxicity of MCT with cyclophosphamide (Cy) in obese mice with metabolic alterations bearing a mammary adenocarcinoma. Simultaneously, the expression and activities of intestinal Mrp2 and Mdr-1 were assessed. CBi male mice, were fed with chow diet (C) or diet with 40 % of fat (HFD). After 16 weeks, metabolic alterations were confirmed by biochemical and morphological parameters. At that time-point, HFD group showed decreased expressions of Mrp2 mRNA (53 %) as well as Mdr-1a and Mdr-1b (42 % and 59 %, respectively), compared to C (P < 0.05). This result correlated with decreased intestinal Mrp2 and Mdr-1 efflux activities (64 % and 45 %, respectively), compared to C (P < 0.05). Ultimately, mice were challenged with M-406 mammary adenocarcinoma; when the tumor was palpable, mice were distributed into 4 groups. The % inhibition of tumor growth with Cy (30 mg/kg/day) in C + Cy was higher than that of HFD + Cy (P = 0.052). Besides, it was observed a 21 % diminution in body weight and leukopenia in the HFD + Cy group. Conclusion: Obesity-induced metabolic alterations impair intestinal Mrp2 and Mdr-1 functions, bringing about increments in Cy absorption, leading to toxicity; in addition, the antitumor effectiveness of MCT decreased in obese animals.
Collapse
MESH Headings
- Animals
- Cyclophosphamide/toxicity
- Mice
- Obesity/metabolism
- Male
- Female
- Administration, Metronomic
- Multidrug Resistance-Associated Protein 2
- Antineoplastic Agents, Alkylating/toxicity
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/chemically induced
- Mice, Obese
- Multidrug Resistance-Associated Proteins/metabolism
- Multidrug Resistance-Associated Proteins/genetics
- Intestinal Mucosa/drug effects
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/pathology
- Adenocarcinoma/pathology
- Adenocarcinoma/drug therapy
- Adenocarcinoma/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP-Binding Cassette Transporters/metabolism
- ATP-Binding Cassette Transporters/genetics
- Diet, High-Fat
Collapse
Affiliation(s)
- María Manuela Barranco
- Laboratorio de Fisiología Metabólica, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina.; CONICET-Rosario. Rosario, Santa Fe, Argentina
| | - Felipe Zecchinati
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Fisiología Experimental-CONICET. Rosario, Santa Fe, Argentina
| | - Virginia Gabriela Perdomo
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Facultad de Ciencias Bioquímicas y Farmacéuticas-Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Martín José Habib
- Laboratorio de Fisiología Metabólica, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - María José Rico
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Viviana Rosa Rozados
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Mario Salazar
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Laboratorio de Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Matías Ezequiel Fusini
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Olga Graciela Scharovsky
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina.; CIC-UNR, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | | | - Leandro Ernesto Mainetti
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Fabiana García
- Laboratorio de Fisiología Metabólica, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina.; CONICET-Rosario. Rosario, Santa Fe, Argentina..
| |
Collapse
|
2
|
Cai Z, Gao L, Hu K, Wang QM. Parthenolide enhances the metronomic chemotherapy effect of cyclophosphamide in lung cancer by inhibiting the NF-kB signaling pathway. World J Clin Oncol 2024; 15:895-907. [PMID: 39071467 PMCID: PMC11271733 DOI: 10.5306/wjco.v15.i7.895] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/11/2024] [Accepted: 06/06/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND Parthenolide (PTL), a sesquiterpene lactone derived from the medicinal herb Chrysanthemum parthenium, exhibits various biological effects by targeting NF-kB, STAT3, and other pathways. It has emerged as a promising adjunct therapy for multiple malignancies. AIM To evaluate the in vitro and in vivo effect of PTL on cyclophosphamide (CTX) metronomic chemotherapy. METHODS The cytotoxicity of PTL and CTX on Lewis lung cancer cells (LLC cells) was assessed by measuring cell activity and apoptosis. The anti-tumor efficiency was evaluated using a tumor xenograft mice model, and the survival of mice and tumor volume were monitored. Additionally, the collected tumor tissues were analyzed for tumor microenvironment indicators and inflammatory factors. RESULTS In vitro, PTL demonstrated a synergistic effect with CTX in inhibiting the growth of LLC cells and promoting apoptosis. In vivo, metronomic chemotherapy combined with PTL and CTX improved the survival rate of tumor-bearing mice and reduced tumor growth rate. Furthermore, metronomic chemotherapy combined with PTL and CTX reduced NF-κB activation and improved the tumor immune microenvironment by decreasing tumor angiogenesis, reducing Transforming growth factor β, and α-SMA positive cells. CONCLUSION PTL is an efficient compound that enhances the metronomic chemotherapy effects of CTX both in vitro and in vivo, suggesting its potential as a supplementary therapeutic strategy in metronomic chemotherapy to improve the chemotherapy effects.
Collapse
Affiliation(s)
- Zheng Cai
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, Henan Province, China
- Department of Oncology, The No. 1 Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Kunming 650021, Yunnan Province, China
| | - Lang Gao
- Department of Oncology, The No. 1 Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Kunming 650021, Yunnan Province, China
| | - Kai Hu
- Department of Oncology, The No. 1 Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Kunming 650021, Yunnan Province, China
| | - Qi-Ming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, Henan Province, China
| |
Collapse
|
3
|
Perkins DW, Steiner I, Haider S, Robertson D, Buus R, O'Leary L, Isacke CM. Therapy-induced normal tissue damage promotes breast cancer metastasis. iScience 2024; 27:108503. [PMID: 38161426 PMCID: PMC10755366 DOI: 10.1016/j.isci.2023.108503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/02/2023] [Accepted: 11/17/2023] [Indexed: 01/03/2024] Open
Abstract
Disseminated tumor cells frequently exhibit a period of dormancy, rendering them chemotherapy insensitive; conversely, the systemic delivery of chemotherapies can result in normal tissue damage. Using multiple mouse and human breast cancer models, we demonstrate that prior chemotherapy administration enhances metastatic colonization and outgrowth. In vitro, chemotherapy-treated fibroblasts display a pro-tumorigenic senescence-associated secretory phenotype (SASP) and are effectively eliminated by targeting the anti-apoptotic protein BCL-xL. In vivo, chemotherapy treatment induces SASP expression in normal tissues; however, the accumulation of senescent cells is limited, and BCL-xL inhibitors are unable to reduce chemotherapy-enhanced metastasis. This likely reflects that chemotherapy-exposed stromal cells do not enter a BCL-xL-dependent phenotype or switch their dependency to other anti-apoptotic BCL-2 family members. This study highlights the role of the metastatic microenvironment in controlling outgrowth of disseminated tumor cells and the need to identify additional approaches to limit the pro-tumorigenic effects of therapy-induced normal tissue damage.
Collapse
Affiliation(s)
- Douglas W. Perkins
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, 237 Fulham Road, SW3 6JB London, UK
| | - Ivana Steiner
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, 237 Fulham Road, SW3 6JB London, UK
| | - Syed Haider
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, 237 Fulham Road, SW3 6JB London, UK
| | - David Robertson
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, 237 Fulham Road, SW3 6JB London, UK
| | - Richard Buus
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, 237 Fulham Road, SW3 6JB London, UK
| | - Lynda O'Leary
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, 237 Fulham Road, SW3 6JB London, UK
| | - Clare M. Isacke
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, 237 Fulham Road, SW3 6JB London, UK
| |
Collapse
|
4
|
Altorki NK, Walsh ZH, Melms JC, Port JL, Lee BE, Nasar A, Spinelli C, Caprio L, Rogava M, Ho P, Christos PJ, Saxena A, Elemento O, Bhinder B, Ager C, Amin AD, Sanfilippo NJ, Mittal V, Borczuk AC, Formenti SC, Izar B, McGraw TE. Neoadjuvant durvalumab plus radiation versus durvalumab alone in stages I-III non-small cell lung cancer: survival outcomes and molecular correlates of a randomized phase II trial. Nat Commun 2023; 14:8435. [PMID: 38114518 PMCID: PMC10730562 DOI: 10.1038/s41467-023-44195-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/04/2023] [Indexed: 12/21/2023] Open
Abstract
We previously reported the results of a randomized phase II trial (NCT02904954) in patients with early-stage non-small cell lung cancer (NSCLC) who were treated with either two preoperative cycles of the anti-PD-L1 antibody durvalumab alone or combined with immunomodulatory doses of stereotactic radiation (DRT). The trial met its primary endpoint of major pathological response, which was significantly higher following DRT with no new safety signals. Here, we report on the prespecified secondary endpoint of disease-free survival (DFS) regardless of treatment assignment and the prespecified exploratory analysis of DFS in each arm of the trial. DFS at 2 and 3 years across patients in both arms of the trial were 73% (95% CI: 62.1-84.5) and 65% (95% CI: 52.5-76.9) respectively. For the exploratory endpoint of DFS in each arm of the trial, three-year DFS was 63% (95% CI: 46.0-80.4) in the durvalumab monotherapy arm compared to 67% (95% CI: 49.6-83.4) in the dual therapy arm. In addition, we report post hoc exploratory analysis of progression-free survival as well as molecular correlates of response and recurrence through high-plex immunophenotyping of sequentially collected peripheral blood and gene expression profiles from resected tumors in both treatment arms. Together, our results contribute to the evolving landscape of neoadjuvant treatment regimens for NSCLC and identify easily measurable potential biomarkers of response and recurrence.
Collapse
Affiliation(s)
- Nasser K Altorki
- Weill Cornell Medicine, Department of Cardiothoracic Surgery, New York, New York, USA.
| | - Zachary H Walsh
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, New York, USA
| | - Johannes C Melms
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, New York, USA
| | - Jeffery L Port
- Weill Cornell Medicine, Department of Cardiothoracic Surgery, New York, New York, USA
| | - Benjamin E Lee
- Weill Cornell Medicine, Department of Cardiothoracic Surgery, New York, New York, USA
| | - Abu Nasar
- Weill Cornell Medicine, Department of Cardiothoracic Surgery, New York, New York, USA
| | - Cathy Spinelli
- Weill Cornell Medicine, Department of Cardiothoracic Surgery, New York, New York, USA
| | - Lindsay Caprio
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, New York, USA
| | - Meri Rogava
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, New York, USA
| | - Patricia Ho
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, New York, USA
| | - Paul J Christos
- Department of Population Health Sciences, Weill Cornell Medicine, New York, New York, USA
| | - Ashish Saxena
- Weill Cornell Medicine, Division of Hematology and Oncology, New York, New York, USA
| | - Olivier Elemento
- Weill Cornell Medicine, Caryl and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Department of Physiology and Biophysics, New York, New York, USA
| | - Bhavneet Bhinder
- Weill Cornell Medicine, Caryl and Israel Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Department of Physiology and Biophysics, New York, New York, USA
| | - Casey Ager
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, New York, USA
| | - Amit Dipak Amin
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, New York, USA
| | | | - Vivek Mittal
- Weill Cornell Medicine, Department of Cardiothoracic Surgery, New York, New York, USA
| | - Alain C Borczuk
- Department of Pathology, Northwell Health, Greenvale, New York, New York, USA
| | - Silvia C Formenti
- Weill Cornell Medicine, Department of Radiation Oncology, New York, New York, USA
| | - Benjamin Izar
- Department of Medicine, Division of Hematology and Oncology, Columbia University Irving Medical Center, Vagelos College of Physicians & Surgeons, New York, New York, USA.
- Deparmtent of Systems Biology, Program for Mathematical Genomics, Columbia University, New York, New York, USA.
- Columbia Center for Translational Immunology, New York, New York, USA.
| | - Timothy E McGraw
- Weill Cornell Medicine, Department of Biochemistry, New York, New York, USA.
| |
Collapse
|
5
|
Park M, Kim J, Kim T, Kim S, Park W, Ha KS, Cho SH, Won MH, Lee JH, Kwon YG, Kim YM. REDD1 is a determinant of low-dose metronomic doxorubicin-elicited endothelial cell dysfunction through downregulation of VEGFR-2/3 expression. Exp Mol Med 2021; 53:1612-1622. [PMID: 34697389 PMCID: PMC8568908 DOI: 10.1038/s12276-021-00690-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/21/2021] [Accepted: 06/29/2021] [Indexed: 01/10/2023] Open
Abstract
Low-dose metronomic chemotherapy (LDMC) inhibits tumor angiogenesis and growth by targeting tumor-associated endothelial cells, but the molecular mechanism has not been fully elucidated. Here, we examined the functional role of regulated in development and DNA damage responses 1 (REDD1), an inhibitor of mammalian target of rapamycin complex 1 (mTORC1), in LDMC-mediated endothelial cell dysfunction. Low-dose doxorubicin (DOX) treatment induced REDD1 expression in cultured vascular and lymphatic endothelial cells and subsequently repressed the mRNA expression of mTORC1-dependent translation of vascular endothelial growth factor receptor (Vegfr)-2/3, resulting in the inhibition of VEGF-mediated angiogenesis and lymphangiogenesis. These regulatory effects of DOX-induced REDD1 expression were additionally confirmed by loss- and gain-of-function studies. Furthermore, LDMC with DOX significantly suppressed tumor angiogenesis, lymphangiogenesis, vascular permeability, growth, and metastasis in B16 melanoma-bearing wild-type but not Redd1-deficient mice. Altogether, our findings indicate that REDD1 is a crucial determinant of LDMC-mediated functional dysregulation of tumor vascular and lymphatic endothelial cells by translational repression of Vegfr-2/3 transcripts, supporting the potential therapeutic properties of REDD1 in highly progressive or metastatic tumors.
Collapse
Affiliation(s)
- Minsik Park
- grid.412010.60000 0001 0707 9039Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 24341 Republic of Korea
| | - Joohwan Kim
- grid.412010.60000 0001 0707 9039Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 24341 Republic of Korea
| | - Taesam Kim
- grid.412010.60000 0001 0707 9039Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 24341 Republic of Korea
| | - Suji Kim
- grid.412010.60000 0001 0707 9039Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 24341 Republic of Korea
| | - Wonjin Park
- grid.412010.60000 0001 0707 9039Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 24341 Republic of Korea
| | - Kwon-Soo Ha
- grid.412010.60000 0001 0707 9039Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 24341 Republic of Korea
| | - Sung Hwan Cho
- grid.412010.60000 0001 0707 9039Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, Gangwon-do 24341 Republic of Korea
| | - Moo-Ho Won
- grid.412010.60000 0001 0707 9039Department of Neurobiology, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 24341 Republic of Korea
| | - Jeong-Hyung Lee
- grid.412010.60000 0001 0707 9039Department of Biochemistry, Kangwon National University, Chuncheon, Gangwon-Do 24341 Republic of Korea
| | - Young-Guen Kwon
- grid.15444.300000 0004 0470 5454Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722 Republic of Korea
| | - Young-Myeong Kim
- grid.412010.60000 0001 0707 9039Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon, Gangwon-do 24341 Republic of Korea ,grid.412010.60000 0001 0707 9039Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, Gangwon-do 24341 Republic of Korea
| |
Collapse
|
6
|
Cazzaniga ME, Cordani N, Capici S, Cogliati V, Riva F, Cerrito MG. Metronomic Chemotherapy. Cancers (Basel) 2021; 13:cancers13092236. [PMID: 34066606 PMCID: PMC8125766 DOI: 10.3390/cancers13092236] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/20/2021] [Accepted: 04/28/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary The present article reviews the state of the art of metronomic chemotherapy use to treat the principal types of cancers, namely breast, non-small cell lung cancer and colorectal ones, and of the most recent progresses in understanding the underlying mechanisms of action. Areas of novelty, in terms of new regimens, new types of cancer suitable for Metronomic chemotherapy (mCHT) and the overview of current ongoing trials, along with a critical review of them, are also provided. Abstract Metronomic chemotherapy treatment (mCHT) refers to the chronic administration of low doses chemotherapy that can sustain prolonged, and active plasma levels of drugs, producing favorable tolerability and it is a new promising therapeutic approach in solid and in hematologic tumors. mCHT has not only a direct effect on tumor cells, but also an action on cell microenvironment, by inhibiting tumor angiogenesis, or promoting immune response and for these reasons can be considered a multi-target therapy itself. Here we review the state of the art of mCHT use in some classical tumour types, such as breast and no small cell lung cancer (NSCLC), see what is new regarding most recent data in different cancer types, such as glioblastoma (GBL) and acute myeloid leukemia (AML), and new drugs with potential metronomic administration. Finally, a look at the strategic use of mCHT in the context of health emergencies, or in low –and middle-income countries (LMICs), where access to adequate healthcare is often not easy, is mandatory, as we always need to bear in in mind that equity in care must be a compulsory part of our medical work and research.
Collapse
Affiliation(s)
- Marina Elena Cazzaniga
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza (MB), Italy;
- Phase 1 Research Centre, ASST-Monza (MB), 20900 Monza, Italy; (S.C.); (V.C.)
- Correspondence: (M.E.C.); (M.G.C.); Tel.: +39-0392-339-037 (M.E.C.)
| | - Nicoletta Cordani
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza (MB), Italy;
| | - Serena Capici
- Phase 1 Research Centre, ASST-Monza (MB), 20900 Monza, Italy; (S.C.); (V.C.)
| | - Viola Cogliati
- Phase 1 Research Centre, ASST-Monza (MB), 20900 Monza, Italy; (S.C.); (V.C.)
| | - Francesca Riva
- Unit of Clinic Oncology, ASST-Monza (MB), 20900 Monza, Italy;
| | - Maria Grazia Cerrito
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza (MB), Italy;
- Correspondence: (M.E.C.); (M.G.C.); Tel.: +39-0392-339-037 (M.E.C.)
| |
Collapse
|
7
|
Sodium nitrate co-supplementation does not exacerbate low dose metronomic doxorubicin-induced cachexia in healthy mice. Sci Rep 2020; 10:15044. [PMID: 32973229 PMCID: PMC7518269 DOI: 10.1038/s41598-020-71974-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 08/17/2020] [Indexed: 12/18/2022] Open
Abstract
The purpose of this study was to determine whether (1) sodium nitrate (SN) treatment progressed or alleviated doxorubicin (DOX)-induced cachexia and muscle wasting; and (2) if a more-clinically relevant low-dose metronomic (LDM) DOX treatment regimen compared to the high dosage bolus commonly used in animal research, was sufficient to induce cachexia in mice. Six-week old male Balb/C mice (n = 16) were treated with three intraperitoneal injections of either vehicle (0.9% NaCl; VEH) or DOX (4 mg/kg) over one week. To test the hypothesis that sodium nitrate treatment could protect against DOX-induced symptomology, a group of mice (n = 8) were treated with 1 mM NaNO3 in drinking water during DOX (4 mg/kg) treatment (DOX + SN). Body composition indices were assessed using echoMRI scanning, whilst physical and metabolic activity were assessed via indirect calorimetry, before and after the treatment regimen. Skeletal and cardiac muscles were excised to investigate histological and molecular parameters. LDM DOX treatment induced cachexia with significant impacts on both body and lean mass, and fatigue/malaise (i.e. it reduced voluntary wheel running and energy expenditure) that was associated with oxidative/nitrostative stress sufficient to induce the molecular cytotoxic stress regulator, nuclear factor erythroid-2-related factor 2 (NRF-2). SN co-treatment afforded no therapeutic potential, nor did it promote the wasting of lean tissue. Our data re-affirm a cardioprotective effect for SN against DOX-induced collagen deposition. In our mouse model, SN protected against LDM DOX-induced cardiac fibrosis but had no effect on cachexia at the conclusion of the regimen.
Collapse
|
8
|
Mainetti LE, Rico MJ, Kaufman CD, Grillo MC, Guercetti J, Baglioni MV, Del Giúdice A, Capitani MC, Fusini M, Rozados VR, Scharovsky OG. Losartan improves the therapeutic effect of metronomic cyclophosphamide in triple negative mammary cancer models. Oncotarget 2020; 11:3048-3060. [PMID: 32850009 PMCID: PMC7429183 DOI: 10.18632/oncotarget.27694] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/25/2020] [Indexed: 12/24/2022] Open
Abstract
Metronomic chemotherapy refers to the minimum biologically effective doses of a chemotherapy agent given as a continuous regimen without extended rest periods. Drug repurposing is defined as the use of an already known drug for a new medical indication, different from the original one. In oncology the combination of these two therapeutic approaches is called "Metronomics". The aim of this work is to evaluate the therapeutic effect of cyclophosphamide in a metronomic schedule in combination with the repurposed drug losartan in two genetically different mice models of triple negative breast cancer. Our findings showed that adding losartan to metronomic cyclophosphamide significantly improved the therapeutic outcome. In both models the combined treatment increased the mice's survival without sings of toxicity. Moreover, we elucidated some of the mechanisms of action involved, which include a decrease of intratumor hypoxia, stimulation of the immune response and remodeling of the tumor microenvironment. The remarkable therapeutic effect, the lack of toxicity, the low cost of the drugs and its oral administration, strongly suggest its translation to the clinical setting in the near future.
Collapse
Affiliation(s)
- Leandro E. Mainetti
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- These authors contributed equally and are co-first authors
| | - María José Rico
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- These authors contributed equally and are co-first authors
| | - Cintia Daniela Kaufman
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Monica Carolina Grillo
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Julian Guercetti
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - María Virginia Baglioni
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Antonela Del Giúdice
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Maria Celeste Capitani
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Matias Fusini
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Viviana Rosa Rozados
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- These authors contributed equally and are co-senior authors
| | - O. Graciela Scharovsky
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Metronomics Global Health Initiative, Marseille, France
- These authors contributed equally and are co-senior authors
| |
Collapse
|
9
|
Scharovsky OG, Rico MJ, Mainetti LE, Perroud HA, Rozados VR. Achievements and challenges in the use of metronomics for the treatment of breast cancer. Biochem Pharmacol 2020; 175:113909. [DOI: 10.1016/j.bcp.2020.113909] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/09/2020] [Indexed: 12/17/2022]
|
10
|
Kim JY, Kim YM. Tumor endothelial cells as a potential target of metronomic chemotherapy. Arch Pharm Res 2019; 42:1-13. [PMID: 30604201 DOI: 10.1007/s12272-018-01102-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022]
Abstract
Drug resistance and toxic side effects are major therapeutic hurdles affecting cancer patients receiving conventional chemotherapy based on the maximum tolerated dose. Metronomic chemotherapy (MCT), a new therapeutic approach developed to avoid these problems generally, consists of the continuous administration of low-dose cytotoxic agents without extended intervals. This therapy targets the tumor microenvironment, rather than exerting a direct effect on tumor cells. As a result, the MCT regimen functionally impairs tumor endothelial cells and circulating endothelial progenitor cells, leading to tumor dormancy via anti-angiogenesis. Over the past 10 years, several studies have highlighted the impact of MCT on the tumor microenvironment and angiogenesis and demonstrated its potential as a switch from the pro-angiogenic to the anti-angiogenic state. However, the mechanisms of action are still obscure. Here, we systematically review the evidence regarding the anti-angiogenic potential of MCT as a crucial determinant of tumor dormancy and cancer treatment.
Collapse
Affiliation(s)
- Ji Yoon Kim
- Department of Anesthesiology and Pain Medicine, Hanyang University Hospital, Seoul, 04763, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry School of Medicine, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, 24341, South Korea.
| |
Collapse
|
11
|
Natale G, Bocci G. Does metronomic chemotherapy induce tumor angiogenic dormancy? A review of available preclinical and clinical data. Cancer Lett 2018; 432:28-37. [PMID: 29885517 DOI: 10.1016/j.canlet.2018.06.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 05/11/2018] [Accepted: 06/03/2018] [Indexed: 02/08/2023]
Abstract
Tumor dormancy is the ability of cancer cells to survive in a non-proliferating state. This condition can depend on three main mechanisms: cell cycle arrest (quiescence or cell dormancy), immunosurveillance (immunologic dormancy), or lack of functional blood vessels (angiogenic dormancy). In particular, under angiogenic dormancy, cancer cell proliferation is counterbalanced by apoptosis owing to poor vascularization, impeding tumor mass expansion beyond a microscopic size, with an asymptomatic and non-metastatic state. Tumor vasculogenic or non-angiogenic switch is essential to promote escape from tumor dormancy, leading to tumor mass proliferation and metastasis. In avascular lesions angiogenesis process results blocked from the equilibrium between pro- and anti-angiogenic factors, such as vascular endothelial growth factor (VEGF) and thrombospondin-1 (TSP-1), respectively. The angiogenic switch mainly depends on the disruption of this balance, in favor of pro-angiogenic factors, and on the recruitment of circulating endothelial progenitors (CEPs) that promote the formation of new blood vessels. Metronomic chemotherapy, the regular intake of doses able to sustain low but active concentrations of chemotherapeutic drugs during protracted time periods, is an encouraging therapeutic approach that has shown to upregulate anti-angiogenic factors such as TSP-1 and decline pro-angiogenic factors such as VEGF, suppressing the proangiogenic cells such as CEPs. In this perspective, metronomic chemotherapy may be one of the available therapeutic approaches capable to modulate favorably the angiogenic tumor dormancy, but further research is essential to better define this particular characteristic.
Collapse
Affiliation(s)
- Gianfranco Natale
- Dipartimento di Ricerca Traslazionale e delle Nuove Tecnologie in Medicina e Chirurgia, and Museo di Anatomia Umana ''Filippo Civinini'', Università di Pisa, Pisa, Italy
| | - Guido Bocci
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy.
| |
Collapse
|
12
|
Rico M, Baglioni M, Bondarenko M, Laluce NC, Rozados V, André N, Carré M, Scharovsky OG, Menacho Márquez M. Metformin and propranolol combination prevents cancer progression and metastasis in different breast cancer models. Oncotarget 2018; 8:2874-2889. [PMID: 27926515 PMCID: PMC5356849 DOI: 10.18632/oncotarget.13760] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 11/22/2016] [Indexed: 01/01/2023] Open
Abstract
Discovery of new drugs for cancer treatment is an expensive and time-consuming process and the percentage of drugs reaching the clinic remains quite low. Drug repositioning refers to the identification and development of new uses for existing drugs and represents an alternative drug development strategy. In this work, we evaluated the antitumor effect of metronomic treatment with a combination of two repositioned drugs, metformin and propranolol, in triple negative breast cancer models. By in vitro studies with five different breast cancer derived cells, we observed that combined treatment decreased proliferation (P < 0.001), mitochondrial activity (P < 0.001), migration (P < 0.001) and invasion (P < 0.001). In vivo studies in immunocompetent mice confirmed the potential of this combination in reducing tumor growth (P < 0.001) and preventing metastasis (P < 0.05). Taken together our results suggest that metformin plus propranolol combined treatment might be beneficial for triple negative breast cancer control, with no symptoms of toxicity.
Collapse
Affiliation(s)
- María Rico
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina.,El Consejo Nacional de Investigaciones Científicas y Técnicas, Argentina
| | - María Baglioni
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Maryna Bondarenko
- Aix-Marseille Université, Inserm UMR_S 911, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Faculté de Pharmacie, Marseille, France
| | - Nahuel Cesatti Laluce
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Viviana Rozados
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina
| | - Nicolas André
- Aix-Marseille Université, Inserm UMR_S 911, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Faculté de Pharmacie, Marseille, France.,Service d'Hématologie and Oncologie Pédiatrique, AP-HM, Marseille, France.,Metronomics Global Health Initiative, Marseille, France
| | - Manon Carré
- Aix-Marseille Université, Inserm UMR_S 911, Centre de Recherche en Oncologie biologique et Oncopharmacologie, Faculté de Pharmacie, Marseille, France
| | - O Graciela Scharovsky
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina.,El Consejo Nacional de Investigaciones Científicas y Técnicas, Argentina.,Metronomics Global Health Initiative, Marseille, France
| | - Mauricio Menacho Márquez
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario, Rosario, Argentina.,El Consejo Nacional de Investigaciones Científicas y Técnicas, Argentina
| |
Collapse
|
13
|
Nanometronomic treatment of 4T1 breast cancer with nanocaged doxorubicin prevents drug resistance and circumvents cardiotoxicity. Oncotarget 2018; 8:8383-8396. [PMID: 28039473 PMCID: PMC5352408 DOI: 10.18632/oncotarget.14204] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/24/2016] [Indexed: 11/25/2022] Open
Abstract
Chemotherapeutic treatment of breast cancer is based on maximum tolerated dose (MTD) approach. However, advanced stage tumors are not effectively eradicated by MTD owing to suboptimal drug targeting, onset of therapeutic resistance and neoangiogenesis. In contrast, “metronomic” chemotherapy is based on frequent drug administrations at lower doses, resulting in neovascularization inhibition and induction of tumor dormancy. Here we show the potential of H-ferritin (HFn)-mediated targeted nanodelivery of metronomic doxorubicin (DOX) in the setting of a highly aggressive and metastatic 4T1 breast cancer mouse model with DOX-inducible expression of chemoresistance. We find that HFn-DOX administered at repeated doses of 1.24 mg kg−1 strongly improves the antitumor potential of DOX chemotherapy arresting the tumor progression. We find that such a potent antitumor effect is attributable to multiple nanodrug actions beyond cell killing, including inhibition of tumor angiogenesis and avoidance of chemoresistance. Multiparametric assessment of heart tissues, including histology, ultrastructural analysis of tissue morphology, and measurement of markers of reactive oxygen species and hepatic/renal conditions, provided evidence that metronomic HFn-DOX allowed us to overcome cardiotoxicity. Our results suggest that HFn-DOX has tremendous potential for the development of “nanometronomic” chemotherapy toward safe and tailored oncological treatments.
Collapse
|
14
|
Kwak G, Jo SD, Kim D, Kim H, Kim MG, Kim K, Kwon IC, Kim SH. Synergistic antitumor effects of combination treatment with metronomic doxorubicin and VEGF-targeting RNAi nanoparticles. J Control Release 2017; 267:203-213. [DOI: 10.1016/j.jconrel.2017.08.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/08/2017] [Accepted: 08/14/2017] [Indexed: 12/12/2022]
|
15
|
Wang M, Gui C, Qiu S, Tang J, Peng Z. Periostin silencing suppresses the aggressive phenotype of thyroid carcinoma cells by suppressing the Akt/thyroid stimulating hormone receptor axis. Cytotechnology 2017; 70:275-284. [PMID: 28965266 DOI: 10.1007/s10616-017-0141-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 09/02/2017] [Indexed: 01/08/2023] Open
Abstract
Clinical evidence indicates that high periostin expression correlates with aggressive phenotype in thyroid carcinoma. However, the biological roles of periostin in thyroid carcinoma development and progression are still unclear. In this study, we explored the effects of periostin silencing on thyroid carcinoma cell growth, invasion, and tumorigenesis. We also studied the impact of periostin on the activation of phosphoinositide 3-kinase (PI3-K)/Akt signaling, which is involved in the pathogenesis of thyroid carcinoma. It was found that downregulation of periostin significantly inhibited the proliferation, colony formation, and invasion in both FTC-133 and BCPAP thyroid carcinoma cells. In vivo tumorigenic studies confirmed that periostin depletion retarded the growth of subcutaneous FTC-133 xenograft tumors, which was coupled with a significant decline in the percentage of Ki-67-positive proliferating cells. Western blot analysis demonstrated that periostin downregulation caused a marked inhibition of thyroid stimulating hormone receptor (TSHR) expression and Akt phosphorylation in FTC-133 and BCPAP cells. Co-expression of constitutively active Akt (CA-Akt) significantly reversed periostin-mediated downregulation of TSHR. Most importantly, overexpression of TSHR or CA-Akt rescued FTC-133 cells from periostin-induced growth and invasion suppression. Collectively, periostin regulates thyroid carcinoma growth and progression via the Akt/TSHR axis and represents a promising therapeutic target for this malignancy.
Collapse
Affiliation(s)
- Min Wang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 100 Haining Road, Shanghai, 200080, China
| | - Chunyi Gui
- Nursing Department, Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Shenglong Qiu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 100 Haining Road, Shanghai, 200080, China
| | - Jingdong Tang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 100 Haining Road, Shanghai, 200080, China
| | - Zhihai Peng
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 100 Haining Road, Shanghai, 200080, China.
| |
Collapse
|
16
|
Becattini S, Littmann ER, Carter RA, Kim SG, Morjaria SM, Ling L, Gyaltshen Y, Fontana E, Taur Y, Leiner IM, Pamer EG. Commensal microbes provide first line defense against Listeria monocytogenes infection. J Exp Med 2017; 214:1973-1989. [PMID: 28588016 PMCID: PMC5502438 DOI: 10.1084/jem.20170495] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 04/06/2017] [Accepted: 04/19/2017] [Indexed: 01/22/2023] Open
Abstract
Becattini et al. provide evidence that a diverse gut microbiota antagonizes the foodborne pathogen Listeria monocytogenes in the intestinal lumen, thereby reducing bloodstream invasion. Microbiota perturbation by antibiotic treatment increases susceptibility to listeriosis, with dramatic effects in immunocompromised hosts. Listeria monocytogenes is a foodborne pathogen that causes septicemia, meningitis and chorioamnionitis and is associated with high mortality. Immunocompetent humans and animals, however, can tolerate high doses of L. monocytogenes without developing systemic disease. The intestinal microbiota provides colonization resistance against many orally acquired pathogens, and antibiotic-mediated depletion of the microbiota reduces host resistance to infection. Here we show that a diverse microbiota markedly reduces Listeria monocytogenes colonization of the gut lumen and prevents systemic dissemination. Antibiotic administration to mice before low dose oral inoculation increases L. monocytogenes growth in the intestine. In immunodeficient or chemotherapy-treated mice, the intestinal microbiota provides nonredundant defense against lethal, disseminated infection. We have assembled a consortium of commensal bacteria belonging to the Clostridiales order, which exerts in vitro antilisterial activity and confers in vivo resistance upon transfer into germ free mice. Thus, we demonstrate a defensive role of the gut microbiota against Listeria monocytogenes infection and identify intestinal commensal species that, by enhancing resistance against this pathogen, represent potential probiotics.
Collapse
Affiliation(s)
- Simone Becattini
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Eric R Littmann
- Lucille Castori Center for Microbes Inflammation and Cancer, Molecular Microbiology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Rebecca A Carter
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Sohn G Kim
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Sejal M Morjaria
- Infectious Diseases Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Lilan Ling
- Lucille Castori Center for Microbes Inflammation and Cancer, Molecular Microbiology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Yangtsho Gyaltshen
- Lucille Castori Center for Microbes Inflammation and Cancer, Molecular Microbiology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Emily Fontana
- Lucille Castori Center for Microbes Inflammation and Cancer, Molecular Microbiology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Ying Taur
- Lucille Castori Center for Microbes Inflammation and Cancer, Molecular Microbiology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY.,Infectious Diseases Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Ingrid M Leiner
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | - Eric G Pamer
- Immunology Program, Sloan Kettering Institute, Memorial Sloan-Kettering Cancer Center, New York, NY .,Lucille Castori Center for Microbes Inflammation and Cancer, Molecular Microbiology Core Facility, Memorial Sloan-Kettering Cancer Center, New York, NY.,Infectious Diseases Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY
| |
Collapse
|
17
|
Rosé A, André N, Rozados VR, Mainetti LE, Menacho Márquez M, Rico MJ, Schaiquevich P, Villarroel M, Gregianin L, Graupera JM, García WG, Epelman S, Alasino C, Alonso D, Chantada G, Scharovsky OG. Highlights from the 1st Latin American meeting on metronomic chemotherapy and drug repositioning in oncology, 27-28 May, 2016, Rosario, Argentina. Ecancermedicalscience 2016; 10:672. [PMID: 27610198 PMCID: PMC5014555 DOI: 10.3332/ecancer.2016.672] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Indexed: 12/22/2022] Open
Abstract
Following previous metronomic meetings in Marseille (2011), Milano (2014), and Mumbai (2016), the first Latin American metronomic meeting was held in the School of Medical Sciences, National University of Rosario, Rosario, Argentina on 27 and 28 of May, 2016. For the first time, clinicians and researchers with experience in the field of metronomics, coming from different countries in Latin America, had the opportunity of presenting and discussing their work. The talks were organised in three main sessions related to experience in the pre-clinical, and clinical (paediatric and adult) areas. The different presentations demonstrated that the fields of metronomic chemotherapy and repurposing drugs in oncology, known as metronomics, constitute a branch of cancer therapy in permanent evolution, which have strong groups working in Latin America, both in the preclinical and the clinical settings including large, adequately designed randomised studies. It was shown that metronomics offers treatments, which, whether they are combined or not with the standard therapeutic approaches, are not only effective but also minimally toxic, with the consequent improvement of the patient’s quality of life, and inexpensive, a feature very important in low resource clinical settings. The potential use of metronomic chemotherapy was proposed as a cost/effective treatment in low-/middle-income countries, for adjuvant therapy in selected tumours. The fundamental role of the governmental agencies and non-governmental alliances, as the Metronomic Global Health Initiative, in supporting this research with public interest was underlined.
Collapse
Affiliation(s)
- Adriana Rosé
- Hospital de Pediatría 'JP Garrahan', Combate de los Pozos 1800, C 1245 AAM, CABA Argentina
| | - Nicolas André
- Inserm UMR_S 911, Centre de Recherche en Oncologie Biologique et Oncopharmacologie, 27 Boulevard Jean Moulin, Faculté de Pharmacie, Aix-Marseille Université; Service d'Hématologie & Oncologie Pédiatrique, AP‑HM, 13005 Marseille, France
| | - Viviana R Rozados
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Santa 3100, 2000 Rosario, Argentina
| | - Leandro E Mainetti
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Santa 3100, 2000 Rosario, Argentina
| | - Mauricio Menacho Márquez
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Santa 3100, 2000 Rosario, Argentina
| | - María José Rico
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Santa 3100, 2000 Rosario, Argentina
| | - Paula Schaiquevich
- Unidad de Farmacocinética Clínica, Hospital de Pediatría 'JP Garrahan', Combate de los Pozos 1800, C 1245 AAM, CABA Argentina
| | - Milena Villarroel
- Av Antonio Varas 360, Santiago, Providencia, Región Metropolitana, Chile
| | - Lauro Gregianin
- Hospital de Clínicas de Porto Alegre, Serviço de Oncologia Pediátrica, Rua Ramiro Barcelos, 2350, Petrópolis, Porto Alegre, RS 90670150, Brazil
| | - Jaume Mora Graupera
- Department of Paediatric Haemato-Oncology, Hospital Sant Joan de Déu, Passeig de Sant Joan de Déu, 2, 08950 Esplugues de Llobregat, Barcelona, Spain
| | - Wendy Gómez García
- Hospital Infantil Dr Robert Reid Cabral, Servicio de Hem-Oncología HIRRC, Ave Abraham Lincoln 2, Casi Esq Ave, Independencia, Santo Domingo, Dominican Republic
| | - Sidnei Epelman
- Paediatric Oncology Department, Santa Marcelina Hospital, R Rio Negro, 48, Itaquaquecetuba, São Paulo, SP 08599-280, Brazil
| | - Carlos Alasino
- Instituto de Oncología de Rosario, Córdoba 2457, S2000KZE Rosario, Argentina
| | - Daniel Alonso
- Laboratorio de Oncología Molecular, Universidad Nacional de Quilmes, Roque Sáenz Peña 352, B1876BXD Bernal, Buenos Aires, Argentina
| | - Guillermo Chantada
- Instituto de Investigaciones, Hospital de Pediatría 'JP Garrahan', Combate de los Pozos 1800, C 1245 AAM, CABA Argentina
| | - O Graciela Scharovsky
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Santa 3100, 2000 Rosario, Argentina
| |
Collapse
|
18
|
Metastatic breast cancer patients treated with low-dose metronomic chemotherapy with cyclophosphamide and celecoxib: clinical outcomes and biomarkers of response. Cancer Chemother Pharmacol 2015; 77:365-74. [PMID: 26721701 DOI: 10.1007/s00280-015-2947-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 12/11/2015] [Indexed: 12/25/2022]
Abstract
BACKGROUND Preclinical results showing therapeutic effect and low toxicity of metronomic chemotherapy with cyclophosphamide (Cy) + celecoxib (Cel) for mammary tumors encouraged its translation to the clinic for treating advanced breast cancer patients (ABCP). PATIENTS AND METHODS A single-arm, mono-institutional, non-randomized, phase II, two-step clinical trial (approved by Bioethics Committee and Argentine Regulatory Authority) was designed. Patients received Cy (50 mg po.d) + Cel (200 mg p.o.bid). Patient eligibility criteria included: ABCP who progressed to anthracyclines, taxanes and capecitabine, ≤4 chemotherapy schemes, with good performance status. Several pro- and anti-angiogenic molecules and cells were determined as biomarkers. Informed consent was signed by all patients. Primary endpoint was clinical benefit (CB). RESULTS Twenty patients were enrolled. Main clinical outcomes were prolonged disease stabilization and partial remission in 10/20 and 1/20 patients, respectively. CB was 55 %, and time to progression (TTP) was 21.1 weeks. Median TTP in patients who achieved CB was 35.6 weeks, and mean overall survival was 44.20 weeks. There were no grade 3/4 toxicities associated with treatment. Circulating endothelial cells (CECs) increased at the time of progression in patients who showed CB (P = 0.014). Baseline CECs and circulating endothelial progenitor cells showed marginal associations with TTP. Serum VEGF decreased (P = 0.050), sVEGFR-2 increased (P = 0.005) and VEGF/sVEGFR-2 ratio decreased during treatment (P = 0.041); baseline VEGF and VEGF/sVEGFR-2 were associated with TTP (P = 0.035 and P = 0.030, respectively), while sVEGFR-2 did not. CONCLUSIONS Treatment was effective, showing low toxicity profile and excellent tolerability. The combination had anti-angiogenic effect. Increased levels of CEC could be useful for detecting progression. Baseline VEGF and VEGF/sVEGFR-2 values could be useful as early predictors of response. TRIAL REGISTRATION ANMAT#4596/09.
Collapse
|
19
|
Yuan F, Shi H, Ji J, Cai Q, Chen X, Yu Y, Liu B, Zhu Z, Zhang J. Capecitabine metronomic chemotherapy inhibits the proliferation of gastric cancer cells through anti-angiogenesis. Oncol Rep 2015; 33:1753-62. [PMID: 25634241 DOI: 10.3892/or.2015.3765] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 01/12/2015] [Indexed: 11/05/2022] Open
Abstract
To evaluate the inhibitory effect and mechanism of capecitabine metronomic chemotherapy on gastric cancer cells. In vitro, the effects of 5-fluorouracil (Fu) metronomic chemotherapy on proliferation, apoptosis, tube formation ability, and angiogenesis were detected. In vivo, Ki-67, CD34 and VEGF were detected by immunohistochemical staining (IHC). Flow cytometry was used to detect the percentage of circulating endothelial progenitors (CEPs), and VEGF and PDGF were detected by ELISA in the peripheral blood of nude mice. The proliferation of the SGC-7901 and AGS gastric cancer cell lines in the metronomic 5-Fu group was decreased compared with the control group in vitro. The total length of the small tubes and tubular junction numbers were significantly lower in the metronomic group than the control group. The VEGF and PDGF levels in the cell culture supernatants were lower in the metronomic group than the control group. Compared with the control group, the CEP percentage was decreased in the peripheral blood of tumor-bearing nude mice following treatment with metronomic 5-Fu or capecitabine chemotherapy. No significant changes were found in the conventional or control group. In the peripheral blood of tumor-bearing nude mice, the VEGF and PDGF levels were decreased in the metronomic groups. Metronomic 5-Fu inhibited the proliferation of gastric cancer cells in vitro and in vivo, and their antitumor effects were non-inferior to those of conventional dose chemotherapy, with mild side effects. Thus, tumor inhibition may be attributed to anti-angiogenesis.
Collapse
Affiliation(s)
- Fei Yuan
- Department of Pathology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Hailong Shi
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Jun Ji
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Qu Cai
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Xuehua Chen
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Yingyan Yu
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Bingya Liu
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Zhenggang Zhu
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| | - Jun Zhang
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P.R. China
| |
Collapse
|
20
|
Hao YB, Yi SY, Ruan J, Zhao L, Nan KJ. New insights into metronomic chemotherapy-induced immunoregulation. Cancer Lett 2014; 354:220-6. [DOI: 10.1016/j.canlet.2014.08.028] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Revised: 08/17/2014] [Accepted: 08/20/2014] [Indexed: 12/15/2022]
|
21
|
Shi H, Jiang J, Ji J, Shi M, Cai Q, Chen X, Yu Y, Liu B, Zhu Z, Zhang J. Anti-angiogenesis participates in antitumor effects of metronomic capecitabine on colon cancer. Cancer Lett 2014; 349:128-35. [PMID: 24746899 DOI: 10.1016/j.canlet.2014.04.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 03/22/2014] [Accepted: 04/06/2014] [Indexed: 12/22/2022]
|
22
|
Rico MJ, Perroud HA, Mainetti LE, Rozados VR, Scharovsky OG. Comparative effectiveness of two metronomic chemotherapy schedules-our experience in the preclinical field. Cancer Invest 2014; 32:92-8. [PMID: 24499110 DOI: 10.3109/07357907.2013.877480] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Metronomic chemotherapy refers to the chronic, equally spaced, delivery of low doses of chemotherapeutic drugs, without extended interruptions. Previously, we developed two combined metronomic schemes for the treatment of murine mammary tumors. The aim of this study was to compare their effects on tumor and metastasis growth, survival, and toxicity. Metronomic chemotherapy with Cyclophosphamide + Celecoxib (Cy + Cel) showed higher antimetastatic power than Cyclophosphamide + Doxorubicin (Cy + Dox), while being similar in other aspects. That difference, plus the advantage that represents its oral administration, suggests that the Cy + Cel combination is more suitable than Cy + Dox for metronomic chemotherapy of mammary tumors and could be proposed to the translation to the clinic.
Collapse
Affiliation(s)
- M J Rico
- Institute of Experimental Genetics, School of Medical Sciences, National University of Rosario , Rosario , Argentina
| | | | | | | | | |
Collapse
|
23
|
Sheng Sow H, Mattarollo SR. Combining low-dose or metronomic chemotherapy with anticancer vaccines: A therapeutic opportunity for lymphomas. Oncoimmunology 2013; 2:e27058. [PMID: 24498564 PMCID: PMC3902116 DOI: 10.4161/onci.27058] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 10/31/2013] [Accepted: 11/03/2013] [Indexed: 01/06/2023] Open
Abstract
Therapeutic vaccination is regarded as a promising strategy against multiple hematological malignancies including lymphoma. However, this approach alone possesses limited potential for the treatment of established tumors. As several anticancer regimens relies on the combination of multiple drugs, it is reasonable to predict that also cancer vaccination will be most effective in the context of multimodal approaches. In particular, low-dose or metronomic chemotherapy could be coupled to anticancer vaccines to improve the efficacy of this immunotherapeutic interventions. This review summarizes recent findings in support of the use of anticancer vaccines combined with low-dose or metronomic chemotherapy for the treatment and management of lymphoid malignancies.
Collapse
Affiliation(s)
- Heng Sheng Sow
- The University of Queensland Diamantina Institute; The University of Queensland; Translational Research Institute; Brisbane, Australia
| | - Stephen R Mattarollo
- The University of Queensland Diamantina Institute; The University of Queensland; Translational Research Institute; Brisbane, Australia
| |
Collapse
|
24
|
Azzi S, Hebda JK, Gavard J. Vascular permeability and drug delivery in cancers. Front Oncol 2013; 3:211. [PMID: 23967403 PMCID: PMC3744053 DOI: 10.3389/fonc.2013.00211] [Citation(s) in RCA: 238] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 08/01/2013] [Indexed: 01/22/2023] Open
Abstract
The endothelial barrier strictly maintains vascular and tissue homeostasis, and therefore modulates many physiological processes such as angiogenesis, immune responses, and dynamic exchanges throughout organs. Consequently, alteration of this finely tuned function may have devastating consequences for the organism. This is particularly obvious in cancers, where a disorganized and leaky blood vessel network irrigates solid tumors. In this context, vascular permeability drives tumor-induced angiogenesis, blood flow disturbances, inflammatory cell infiltration, and tumor cell extravasation. This can directly restrain the efficacy of conventional therapies by limiting intravenous drug delivery. Indeed, for more effective anti-angiogenic therapies, it is now accepted that not only should excessive angiogenesis be alleviated, but also that the tumor vasculature needs to be normalized. Recovery of normal state vasculature requires diminishing hyperpermeability, increasing pericyte coverage, and restoring the basement membrane, to subsequently reduce hypoxia, and interstitial fluid pressure. In this review, we will introduce how vascular permeability accompanies tumor progression and, as a collateral damage, impacts on efficient drug delivery. The molecular mechanisms involved in tumor-driven vascular permeability will next be detailed, with a particular focus on the main factors produced by tumor cells, especially the emblematic vascular endothelial growth factor. Finally, new perspectives in cancer therapy will be presented, centered on the use of anti-permeability factors and normalization agents.
Collapse
Affiliation(s)
- Sandy Azzi
- CNRS, UMR8104 , Paris , France ; INSERM, U1016 , Paris , France ; Sorbonne Paris Cite, Universite Paris Descartes , Paris , France
| | | | | |
Collapse
|