Co-Targeting Luminal B Breast Cancer with S-Adenosylmethionine and Immune Checkpoint Inhibitor Reduces Primary Tumor Growth and Progression, and Metastasis to Lungs and Bone
Abstract
:Simple Summary
Abstract
1. Introduction
2. Results
2.1. SAM Decreases Proliferation, Colony Formation, and Invasion of BCa Cell Lines
2.2. Blocking Programmed Death Ligand 1 (PD-L1) Intrinsic Signalling Has No Effect on Cell Proliferation of BCa Cell Lines
2.3. The SAM and Anti-PD-1 Antibody Combination Has a Superior Effect in Reducing Primary Breast Tumor Growth Compared to Monotherapies
2.4. The SAM and Anti-PD-1 Antibody Combination Decreases Lung Metastasis
2.5. The SAM and Anti-PD-1 Antibody Combination Blocks Bone Metastasis and Protects Bone from Tumor Osteolytic Damage
2.6. The SAM and Anti-PD-1 Antibody Combination Reduces Expression of Oncogenes While Elevating Expression of Immunostimulatory Genes, as well as CD8+ T Cell Infiltration and Activity
3. Discussion
4. Materials and Methods
4.1. Cell Lines
4.2. Proliferation, Soft Agar Colony Formation, and Invasion Assays
4.3. Animal Studies
4.4. RNA Extraction and Reverse Transcriptase Quantitative Real-Time PCR (RT-qPCR)
4.5. RNA-Sequencing (RNA-Seq) and Analysis
4.6. Intratibial Model for Skeletal Metastasis
4.7. Immunohistochemistry (IHC)
4.8. Statistical Analysis
5. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Brenner, D.R.; Weir, H.K.; Demers, A.A.; Ellison, L.F.; Louzado, C.; Shaw, A.; Turner, D.; Woods, R.R.; Smith, L.M.; Canadian Cancer Statistics Advisory Committee. Projected estimates of cancer in Canada in 2020. CMAJ 2020, 192, E199–E205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2017. Cancer J. Clin. 2018, 67, 7–30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seyfried, T.N.; Huysentruyt, L.C. On the origin of cancer metastasis. Crit. Rev. Oncog. 2013, 18, 43–73. [Google Scholar] [CrossRef] [Green Version]
- Chaffer, C.L.; Weinberg, R.A. A perspective on cancer cell metastasis. Science 2011, 331, 1559–1564. [Google Scholar] [CrossRef] [PubMed]
- Yersal, O.; Barutca, S. Biological subtypes of breast cancer: Prognostic and therapeutic implications. World J. Clin. Oncol. 2014, 5, 412–424. [Google Scholar] [CrossRef] [PubMed]
- Cardoso, F.; Kyriakides, S.; Ohno, S.; Penault-Llorca, F.; Poortmans, P.; Rubio, I.T.; Zackrisson, S.; Senkus, E.; on behalf of the ESMO Guidelines Committee. Early breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-updagger. Ann. Oncol. 2019, 30, 1194–1220. [Google Scholar] [CrossRef] [Green Version]
- Viale, G.; Hanlon Newell, A.E.; Walker, E.; Harlow, G.; Bai, I.; Russo, L.; Dell’Orto, P.; Maisonneuve, P. Ki-67 (30-9) scoring and differentiation of Luminal A- and Luminal B-like breast cancer subtypes. Breast Cancer Res. Treat. 2019, 178, 451–458. [Google Scholar] [CrossRef] [Green Version]
- Wu, Q.; Li, J.; Zhu, S.; Wu, J.; Chen, C.; Liu, Q.; Wei, W.; Zhang, Y.; Sun, S. Breast cancer subtypes predict the preferential site of distant metastases: A SEER based study. Oncotarget 2017, 8, 27990–27996. [Google Scholar] [CrossRef] [Green Version]
- Yang, M.; Liu, C.; Yu, X. Skeletal-related adverse events during bone metastasis of breast cancer: Current status. Discov. Med. 2019, 27, 211–220. [Google Scholar]
- Kane, C.M.; Hoskin, P.; Bennett, M.I. Cancer induced bone pain. BMJ 2015, 350, h315. [Google Scholar] [CrossRef] [PubMed]
- Harris, S.R. Differentiating the Causes of Spontaneous Rib Fracture After Breast Cancer. Clin. Breast Cancer 2016, 16, 431–436. [Google Scholar] [CrossRef] [PubMed]
- Alsaab, H.O.; Sau, S.; Alzhrani, R.; Tatiparti, K.; Bhise, K.; Kashaw, S.K.; Iyer, A.K. PD-1 and PD-L1 Checkpoint Signaling Inhibition for Cancer Immunotherapy: Mechanism, Combinations, and Clinical Outcome. Front. Pharmacol. 2017, 8, 561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schadendorf, D.; van Akkooi, A.C.J.; Berking, C.; Griewank, K.G.; Gutzmer, R.; Hauschild, A.; Stang, A.; Roesch, A.; Ugurel, S. Melanoma. Lancet 2018, 392, 971–984. [Google Scholar] [CrossRef] [PubMed]
- Kalbasi, A.; Ribas, A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat. Rev. Immunol. 2020, 20, 25–39. [Google Scholar] [CrossRef]
- Barrueto, L.; Caminero, F.; Cash, L.; Makris, C.; Lamichhane, P.; Deshmukh, R.R. Resistance to Checkpoint Inhibition in Cancer Immunotherapy. Transl. Oncol. 2020, 13, 100738. [Google Scholar] [CrossRef]
- Alexandrov, L.B.; Nik-Zainal, S.; Wedge, D.C.; Aparicio, S.A.; Behjati, S.; Biankin, A.V.; Bignell, G.R.; Bolli, N.; Borg, A.; Borresen-Dale, A.L.; et al. Signatures of Mutational Processes in Human Cancer. Nature 2013, 500, 415–421. [Google Scholar] [CrossRef] [Green Version]
- Yarchoan, M.; Hopkins, A.; Jaffee, E.M. Tumor Mutational Burden and Response Rate to PD-1 Inhibition. N. Engl. J. Med. 2017, 377, 2500–2501. [Google Scholar] [CrossRef]
- Planes-Laine, G.; Rochigneux, P.; Bertucci, F.; Chretien, A.S.; Viens, P.; Sabatier, R.; Goncalves, A. PD-1/PD-L1 Targeting in Breast Cancer: The First Clinical Evidences Are Emerging. A Literature Review. Cancers 2019, 11, 1033. [Google Scholar] [CrossRef] [Green Version]
- Plitas, G.; Konopacki, C.; Wu, K.; Bos, P.D.; Morrow, M.; Putintseva, E.V.; Chudakov, D.M.; Rudensky, A.Y. Regulatory T Cells Exhibit Distinct Features in Human Breast Cancer. Immunity 2016, 45, 1122–1134. [Google Scholar] [CrossRef] [PubMed]
- Dieci, M.V.; Griguolo, G.; Miglietta, F.; Guarneri, V. The immune system and hormone-receptor positive breast cancer: Is it really a dead end? Cancer Treat. Rev. 2016, 46, 9–19. [Google Scholar] [CrossRef]
- Dieci, M.V.; Guarneri, V.; Tosi, A.; Bisagni, G.; Musolino, A.; Spazzapan, S.; Moretti, G.; Vernaci, G.M.; Griguolo, G.; Giarratano, T.; et al. Neoadjuvant Chemotherapy and Immunotherapy in Luminal B-like Breast Cancer: Results of the Phase II GIADA Trial. Clin. Cancer Res. 2022, 28, 308–317. [Google Scholar] [CrossRef]
- Miller, L.D.; Chou, J.A.; Black, M.A.; Print, C.; Chifman, J.; Alistar, A.; Putti, T.; Zhou, X.; Bedognetti, D.; Hendrickx, W.; et al. Immunogenic Subtypes of Breast Cancer Delineated by Gene Classifiers of Immune Responsiveness. Cancer Immunol. Res. 2016, 4, 600–610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jung, H.; Kim, H.S.; Kim, J.Y.; Sun, J.M.; Ahn, J.S.; Ahn, M.J.; Park, K.; Esteller, M.; Lee, S.H.; Choi, J.K. DNA methylation loss promotes immune evasion of tumours with high mutation and copy number load. Nat. Commun. 2019, 10, 4278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pakneshan, P.; Szyf, M.; Farias-Eisner, R.; Rabbani, S.A. Reversal of the hypomethylation status of urokinase (uPA) promoter blocks breast cancer growth and metastasis. J. Biol. Chem. 2004, 279, 31735–31744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shukeir, N.; Pakneshan, P.; Chen, G.; Szyf, M.; Rabbani, S.A. Alteration of the methylation status of tumor-promoting genes decreases prostate cancer cell invasiveness and tumorigenesis in vitro and in vivo. Cancer Res. 2006, 66, 9202–9210. [Google Scholar] [CrossRef] [Green Version]
- Mahmood, N.; Cheishvili, D.; Arakelian, A.; Tanvir, I.; Khan, H.A.; Pepin, A.S.; Szyf, M.; Rabbani, S.A. Methyl donor S-adenosylmethionine (SAM) supplementation attenuates breast cancer growth, invasion, and metastasis in vivo; therapeutic and chemopreventive applications. Oncotarget 2018, 9, 5169–5183. [Google Scholar] [CrossRef] [Green Version]
- Mahmood, N.; Arakelian, A.; Cheishvili, D.; Szyf, M.; Rabbani, S.A. S-adenosylmethionine in combination with decitabine shows enhanced anti-cancer effects in repressing breast cancer growth and metastasis. J. Cell. Mol. Med. 2020, 24, 10322–10337. [Google Scholar] [CrossRef]
- Mahmood, N.; Arakelian, A.; Muller, W.J.; Szyf, M.; Rabbani, S.A. An enhanced chemopreventive effect of methyl donor S-adenosylmethionine in combination with 25-hydroxyvitamin D in blocking mammary tumor growth and metastasis. Bone Res. 2020, 8, 28. [Google Scholar] [CrossRef]
- Mahmood, N.; Rabbani, S.A. Targeting DNA Hypomethylation in Malignancy by Epigenetic Therapies. Adv. Exp. Med. Biol. 2019, 1164, 179–196. [Google Scholar] [CrossRef] [PubMed]
- Hote, P.T.; Sahoo, R.; Jani, T.S.; Ghare, S.S.; Chen, T.; Joshi-Barve, S.; McClain, C.J.; Barve, S.S. Ethanol inhibits methionine adenosyltransferase II activity and S-adenosylmethionine biosynthesis and enhances caspase-3-dependent cell death in T lymphocytes: Relevance to alcohol-induced immunosuppression. J. Nutr. Biochem. 2008, 19, 384–391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tobena, R.; Horikawa, S.; Calvo, V.; Alemany, S. Interleukin-2 induces gamma-S-adenosyl-L-methionine synthetase gene expression during T-lymphocyte activation. Biochem. J. 1996, 319, 929–933. [Google Scholar] [CrossRef] [PubMed]
- LeGros, H.L., Jr.; Geller, A.M.; Kotb, M. Differential regulation of methionine adenosyltransferase in superantigen and mitogen stimulated human T lymphocytes. J. Biol. Chem. 1997, 272, 16040–16047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kotb, M.; Dale, J.B.; Beachey, E.H. Stimulation of S-adenosylmethionine synthetase in human lymphocytes by streptococcal M protein. J. Immunol. 1987, 139, 202–206. [Google Scholar]
- De La Rosa, J.; Geller, A.M.; LeGros, H.L., Jr.; Kotb, M. Induction of interleukin 2 production but not methionine adenosyltransferase activity or S-adenosylmethionine turnover in Jurkat T-cells. Cancer Res. 1992, 52, 3361–3366. [Google Scholar]
- De La Rosa, J.; Kotb, M.; Kredich, N.M. Regulation of S-adenosylmethionine synthetase activity in cultured human lymphocytes. Biochim. Biophys. Acta 1991, 1077, 225–232. [Google Scholar] [CrossRef]
- German, D.C.; Bloch, C.A.; Kredich, N.M. Measurements of S-adenosylmethionine and L-homocysteine metabolism in cultured human lymphoid cells. J. Biol. Chem. 1983, 258, 10997–11003. [Google Scholar] [CrossRef]
- Zeng, Z.; Yang, H.; Huang, Z.Z.; Chen, C.; Wang, J.; Lu, S.C. The role of c-Myb in the up-regulation of methionine adenosyltransferase 2A expression in activated Jurkat cells. Biochem. J. 2001, 353, 163–168. [Google Scholar] [CrossRef]
- Kotb, M.; Kredich, N.M. S-Adenosylmethionine synthetase from human lymphocytes. Purification and characterization. J. Biol. Chem. 1985, 260, 3923–3930. [Google Scholar] [CrossRef]
- Sahin, E.; Sahin, M. Epigenetical Targeting of the FOXP3 Gene by S-Adenosylmethionine Diminishes the Suppressive Capacity of Regulatory T Cells Ex Vivo and Alters the Expression Profiles. J. Immunother. 2019, 42, 11–22. [Google Scholar] [CrossRef] [PubMed]
- Bian, Y.; Li, W.; Kremer, D.M.; Sajjakulnukit, P.; Li, S.; Crespo, J.; Nwosu, Z.C.; Zhang, L.; Czerwonka, A.; Pawlowska, A.; et al. Cancer SLC43A2 alters T cell methionine metabolism and histone methylation. Nature 2020, 585, 277–282. [Google Scholar] [CrossRef] [PubMed]
- Ulanovskaya, O.A.; Zuhl, A.M.; Cravatt, B.F. NNMT promotes epigenetic remodeling in cancer by creating a metabolic methylation sink. Nat. Chem. Biol. 2013, 9, 300–306. [Google Scholar] [CrossRef] [PubMed]
- Larroquette, M.; Domblides, C.; Lefort, F.; Lasserre, M.; Quivy, A.; Sionneau, B.; Bertolaso, P.; Gross-Goupil, M.; Ravaud, A.; Daste, A. Combining immune checkpoint inhibitors with chemotherapy in advanced solid tumours: A review. Eur. J. Cancer 2021, 158, 47–62. [Google Scholar] [CrossRef] [PubMed]
- Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef] [PubMed]
- Escors, D.; Gato-Cañas, M.; Zuazo, M.; Arasanz, H.; García-Granda, M.J.; Vera, R.; Kochan, G. The intracellular signalosome of PD-L1 in cancer cells. Signal Transduct. Target. Ther. 2018, 3, 26. [Google Scholar] [CrossRef] [Green Version]
- Dong, P.; Xiong, Y.; Yue, J.; Hanley, S.J.B.; Watari, H. Tumor-Intrinsic PD-L1 Signaling in Cancer Initiation, Development and Treatment: Beyond Immune Evasion. Front. Oncol. 2018, 8, 386. [Google Scholar] [CrossRef] [Green Version]
- Mehdi, A.; Attias, M.; Mahmood, N.; Arakelian, A.; Mihalcioiu, C.; Piccirillo, C.A.; Szyf, M.; Rabbani, S.A. Enhanced Anticancer Effect of a Combination of S-adenosylmethionine (SAM) and Immune Checkpoint Inhibitor (ICPi) in a Syngeneic Mouse Model of Advanced Melanoma. Front. Oncol. 2020, 10, 1361. [Google Scholar] [CrossRef]
- Le Naour, A.; Rossary, A.; Vasson, M.P. EO771, is it a well-characterized cell line for mouse mammary cancer model? Limit and uncertainty. Cancer Med. 2020, 9, 8074–8085. [Google Scholar] [CrossRef]
- Le Naour, A.; Koffi, Y.; Diab, M.; Le Guennec, D.; Rouge, S.; Aldekwer, S.; Goncalves-Mendes, N.; Talvas, J.; Farges, M.C.; Caldefie-Chezet, F.; et al. EO771, the first luminal B mammary cancer cell line from C57BL/6 mice. Cancer Cell. Int. 2020, 20, 328. [Google Scholar] [CrossRef]
- Gialeli, C.; Theocharis, A.D.; Karamanos, N.K. Roles of matrix metalloproteinases in cancer progression and their pharmacological targeting. FEBS J. 2011, 278, 16–27. [Google Scholar] [CrossRef] [PubMed]
- Jiang, H.; Li, H. Prognostic values of tumoral MMP2 and MMP9 overexpression in breast cancer: A systematic review and meta-analysis. BMC Cancer 2021, 21, 149. [Google Scholar] [CrossRef] [PubMed]
- Goldman, M.J.; Craft, B.; Hastie, M.; Repecka, K.; McDade, F.; Kamath, A.; Banerjee, A.; Luo, Y.; Rogers, D.; Brooks, A.N.; et al. Visualizing and interpreting cancer genomics data via the Xena platform. Nat. Biotechnol. 2020, 38, 675–678. [Google Scholar] [CrossRef] [PubMed]
- Zhang, M.; Fujiwara, K.; Che, X.; Zheng, S.; Zheng, L. DNA methylation in the tumor microenvironment. J. Zhejiang Univ. Sci. B 2017, 18, 365–372. [Google Scholar] [CrossRef] [Green Version]
- Ostrand-Rosenberg, S. Immune Surveillance: A Balance Between Pro- and Anti-tumor Immunity. Curr. Opin. Genet. Dev. 2008, 18, 11–18. [Google Scholar] [CrossRef] [Green Version]
- Sukari, A.; Nagasaka, M.; Al-Hadidi, A.; Lum, L.G. Cancer Immunology and Immunotherapy. Anticancer Res. 2016, 36, 5593–5606. [Google Scholar] [CrossRef]
- Gibney, G.T.; Weiner, L.M.; Atkins, M.B. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016, 17, e542–e551. [Google Scholar] [CrossRef] [Green Version]
- Ji, R.R.; Chasalow, S.D.; Wang, L.; Hamid, O.; Schmidt, H.; Cogswell, J.; Alaparthy, S.; Berman, D.; Jure-Kunkel, M.; Siemers, N.O.; et al. An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer Immunol. Immunother. 2012, 61, 1019–1031. [Google Scholar] [CrossRef]
- Ilisso, C.P.; Sapio, L.; Delle Cave, D.; Illiano, M.; Spina, A.; Cacciapuoti, G.; Naviglio, S.; Porcelli, M. S-Adenosylmethionine Affects ERK1/2 and Stat3 Pathways and Induces Apotosis in Osteosarcoma Cells. J. Cell. Physiol. 2016, 231, 428–435. [Google Scholar] [CrossRef]
- Ilisso, C.P.; Delle Cave, D.; Mosca, L.; Pagano, M.; Coppola, A.; Mele, L.; Caraglia, M.; Cacciapuoti, G.; Porcelli, M. S-Adenosylmethionine regulates apoptosis and autophagy in MCF-7 breast cancer cells through the modulation of specific microRNAs. Cancer Cell. Int. 2018, 18, 197. [Google Scholar] [CrossRef]
- Cave, D.D.; Desiderio, V.; Mosca, L.; Ilisso, C.P.; Mele, L.; Caraglia, M.; Cacciapuoti, G.; Porcelli, M. S-Adenosylmethionine-mediated apoptosis is potentiated by autophagy inhibition induced by chloroquine in human breast cancer cells. J. Cell. Physiol. 2018, 233, 1370–1383. [Google Scholar] [CrossRef] [PubMed]
- Parashar, S.; Cheishvili, D.; Arakelian, A.; Hussain, Z.; Tanvir, I.; Khan, H.A.; Szyf, M.; Rabbani, S.A. S-adenosylmethionine blocks osteosarcoma cells proliferation and invasion in vitro and tumor metastasis in vivo: Therapeutic and diagnostic clinical applications. Cancer Med. 2015, 4, 732–744. [Google Scholar] [CrossRef] [PubMed]
- Shukeir, N.; Stefanska, B.; Parashar, S.; Chik, F.; Arakelian, A.; Szyf, M.; Rabbani, S.A. Pharmacological methyl group donors block skeletal metastasis in vitro and in vivo. Br. J. Pharmacol. 2015, 172, 2769–2781. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Bi, T.; Yuan, F.; Gao, X.; Jia, G.; Tian, Z. S-adenosylmethionine induces apoptosis and cycle arrest of gallbladder carcinoma cells by suppression of JAK2/STAT3 pathways. Naunyn Schmiedebergs Arch. Pharmacol. 2020, 393, 2507–2515. [Google Scholar] [CrossRef] [PubMed]
- Stagg, J.; Divisekera, U.; McLaughlin, N.; Sharkey, J.; Pommey, S.; Denoyer, D.; Dwyer, K.M.; Smyth, M.J. Anti-CD73 antibody therapy inhibits breast tumor growth and metastasis. Proc. Natl. Acad. Sci. USA 2010, 107, 1547–1552. [Google Scholar] [CrossRef] [Green Version]
- Hoover, R.G.; Gullickson, G.; Kornbluth, J. Natural killer lytic-associated molecule plays a role in controlling tumor dissemination and metastasis. Front. Immunol. 2012, 3, 393. [Google Scholar] [CrossRef] [Green Version]
- Singh, G.; Rabbani, A.S. Bone Metastasis; Human Press Inc.: Totowa, NJ, USA, 2005. [Google Scholar]
- Sun, X.; Li, K.; Hase, M.; Zha, R.; Feng, Y.; Li, B.Y.; Yokota, H. Suppression of breast cancer-associated bone loss with osteoblast proteomes via Hsp90ab1/moesin-mediated inhibition of TGFbeta/FN1/CD44 signaling. Theranostics 2022, 12, 929–943. [Google Scholar] [CrossRef]
- Feng, Y.; Liu, S.; Zha, R.; Sun, X.; Li, K.; Robling, A.; Li, B.; Yokota, H. Mechanical Loading-Driven Tumor Suppression Is Mediated by Lrp5-Dependent and Independent Mechanisms. Cancers 2021, 13, 267. [Google Scholar] [CrossRef]
- Pakneshan, P.; Tetu, B.; Rabbani, S.A. Demethylation of urokinase promoter as a prognostic marker in patients with breast carcinoma. Clin. Cancer Res. 2004, 10, 3035–3041. [Google Scholar] [CrossRef] [Green Version]
- Beatty, G.L.; Gladney, W.L. Immune escape mechanisms as a guide for cancer immunotherapy. Clin. Cancer Res. 2015, 21, 687–692. [Google Scholar] [CrossRef] [Green Version]
- Mehdi, A.; Rabbani, S.A. Role of Methylation in Pro- and Anti-Cancer Immunity. Cancers 2021, 13, 545. [Google Scholar] [CrossRef] [PubMed]
- Hung, M.H.; Lee, J.S.; Ma, C.; Diggs, L.P.; Heinrich, S.; Chang, C.W.; Ma, L.; Forgues, M.; Budhu, A.; Chaisaingmongkol, J.; et al. Tumor methionine metabolism drives T-cell exhaustion in hepatocellular carcinoma. Nat. Commun. 2021, 12, 1455. [Google Scholar] [CrossRef] [PubMed]
- Chik, F.; Machnes, Z.; Szyf, M. Synergistic anti-breast cancer effect of a combined treatment with the methyl donor S-adenosyl methionine and the DNA methylation inhibitor 5-aza-2’-deoxycytidine. Carcinogenesis 2014, 35, 138–144. [Google Scholar] [CrossRef] [PubMed]
- Shukeir, N.; Arakelian, A.; Chen, G.; Garde, S.; Ruiz, M.; Panchal, C.; Rabbani, S.A. A synthetic 15-mer peptide (PCK3145) derived from prostate secretory protein can reduce tumor growth, experimental skeletal metastases, and malignancy-associated hypercalcemia. Cancer Res. 2004, 64, 5370–5377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Black, M.; Barsoum, I.B.; Truesdell, P.; Cotechini, T.; Macdonald-Goodfellow, S.K.; Petroff, M.; Siemens, D.R.; Koti, M.; Craig, A.W.; Graham, C.H. Activation of the PD-1/PD-L1 immune checkpoint confers tumor cell chemoresistance associated with increased metastasis. Oncotarget 2016, 7, 10557–10567. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bernardo, M.; Tolstykh, T.; Zhang, Y.A.; Bangari, D.S.; Cao, H.; Heyl, K.A.; Lee, J.S.; Malkova, N.V.; Malley, K.; Marquez, E.; et al. An experimental model of anti-PD-1 resistance exhibits activation of TGFss and Notch pathways and is sensitive to local mRNA immunotherapy. Oncoimmunology 2021, 10, 1881268. [Google Scholar] [CrossRef]
- Bourgeois-Daigneault, M.C.; Roy, D.G.; Aitken, A.S.; El Sayes, N.; Martin, N.T.; Varette, O.; Falls, T.; St-Germain, L.E.; Pelin, A.; Lichty, B.D.; et al. Neoadjuvant oncolytic virotherapy before surgery sensitizes triple-negative breast cancer to immune checkpoint therapy. Sci. Transl. Med. 2018, 10. [Google Scholar] [CrossRef] [Green Version]
- Gao, M.; Wang, T.; Ji, L.; Bai, S.; Tian, L.; Song, H. Therapy with Carboplatin and Anti-PD-1 Antibodies Before Surgery Demonstrates Sustainable Anti-Tumor Effects for Secondary Cancers in Mice With Triple-Negative Breast Cancer. Front. Immunol. 2020, 11, 366. [Google Scholar] [CrossRef] [Green Version]
- Rastelli, L.; Valentino, M.L.; Minderman, M.C.; Landin, J.; Malyankar, U.M.; Lescoe, M.K.; Kitson, R.; Brunson, K.; Souan, L.; Forenza, S.; et al. A KDR-binding peptide (ST100,059) can block angiogenesis, melanoma tumor growth and metastasis in vitro and in vivo. Int. J. Oncol. 2011, 39, 401–408. [Google Scholar] [CrossRef] [Green Version]
- Rabbani, S.A.; Ateeq, B.; Arakelian, A.; Valentino, M.L.; Shaw, D.E.; Dauffenbach, L.M.; Kerfoot, C.A.; Mazar, A.P. An anti-urokinase plasminogen activator receptor antibody (ATN-658) blocks prostate cancer invasion, migration, growth, and experimental skeletal metastasis in vitro and in vivo. Neoplasia 2010, 12, 778–788. [Google Scholar] [CrossRef] [Green Version]
- Yang, M.; Burton, D.W.; Geller, J.; Hillegonds, D.J.; Hastings, R.H.; Deftos, L.J.; Hoffman, R.M. The bisphosphonate olpadronate inhibits skeletal prostate cancer progression in a green fluorescent protein nude mouse model. Clin. Cancer Res. 2006, 12, 2602–2606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rabbani, S.A.; Arakelian, A.; Farookhi, R. LRP5 knockdown: Effect on prostate cancer invasion growth and skeletal metastasis in vitro and in vivo. Cancer Med. 2013, 2, 625–635. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Mehdi, A.; Attias, M.; Arakelian, A.; Piccirillo, C.A.; Szyf, M.; Rabbani, S.A. Co-Targeting Luminal B Breast Cancer with S-Adenosylmethionine and Immune Checkpoint Inhibitor Reduces Primary Tumor Growth and Progression, and Metastasis to Lungs and Bone. Cancers 2023, 15, 48. https://doi.org/10.3390/cancers15010048
Mehdi A, Attias M, Arakelian A, Piccirillo CA, Szyf M, Rabbani SA. Co-Targeting Luminal B Breast Cancer with S-Adenosylmethionine and Immune Checkpoint Inhibitor Reduces Primary Tumor Growth and Progression, and Metastasis to Lungs and Bone. Cancers. 2023; 15(1):48. https://doi.org/10.3390/cancers15010048
Chicago/Turabian StyleMehdi, Ali, Mikhael Attias, Ani Arakelian, Ciriaco A. Piccirillo, Moshe Szyf, and Shafaat A. Rabbani. 2023. "Co-Targeting Luminal B Breast Cancer with S-Adenosylmethionine and Immune Checkpoint Inhibitor Reduces Primary Tumor Growth and Progression, and Metastasis to Lungs and Bone" Cancers 15, no. 1: 48. https://doi.org/10.3390/cancers15010048
APA StyleMehdi, A., Attias, M., Arakelian, A., Piccirillo, C. A., Szyf, M., & Rabbani, S. A. (2023). Co-Targeting Luminal B Breast Cancer with S-Adenosylmethionine and Immune Checkpoint Inhibitor Reduces Primary Tumor Growth and Progression, and Metastasis to Lungs and Bone. Cancers, 15(1), 48. https://doi.org/10.3390/cancers15010048