1
|
Fujimoto H, Yoshihara M, Ricciardelli C, Tano S, Iyoshi S, Miyamoto E, Mogi K, Hayashi M, Hayakawa S, Nomura S, Kitami K, Uno K, Yoshikawa N, Emoto R, Matsui S, Kajiyama H. Aging affects regrowth of stealthperitoneal dissemination of advanced ovarian cancer: a multicenter retrospective cohort study. Sci Rep 2024; 14:23537. [PMID: 39384823 PMCID: PMC11479624 DOI: 10.1038/s41598-024-66419-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 07/01/2024] [Indexed: 10/11/2024] Open
Abstract
Ovarian cancer (OvCa) is one of the most lethal gynecological malignancies, and most patients are diagnosed at advanced stage with peritoneal dissemination. Although age at diagnosis is considered an independent prognostic factor, its impact on peritoneal recurrence after combined cytoreductive surgery and chemotherapy is not clear. The objective of this study was to investigate the impact of aging on peritoneal recurrence from stealth dissemination and gain insight of the pathophysiology of OvCa in elderly patients. A total of 243 patients with pT2b-pT3 epithelial ovarian who achieved complete surgery, no-residual tumor at first surgery, were selected to be analyzed the risk of peritoneal seeding and recurrence. We found that age over 65 years was independently associated with an increased risk of peritoneum-specific (PS) recurrence (. Furthermore, pT3 stages and positive ascites cytology also worsen the PS-relapse-free survival. Collectively, our findings suggest that age, especially over 65 years, predicts reduced peritoneum-specific tumor recurrence in patients with advanced ovarian cancer after complete cytoreduction surgery, particularly those with pT3 tumors and positive ascites cytology.
Collapse
Affiliation(s)
- Hiroki Fujimoto
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Masato Yoshihara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan.
- Center for Medical Education, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Carmela Ricciardelli
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, Australia
| | - Sho Tano
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Shohei Iyoshi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Emiri Miyamoto
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Kazumasa Mogi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Maia Hayashi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Sae Hayakawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Satoshi Nomura
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Kazuhisa Kitami
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
- Department of Obstetrics and Gynecology, Kitasato University Hospital, Sagamihara, Japan
| | - Kaname Uno
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University Graduate School of Medicine, Lund, Sweden
| | - Nobuhisa Yoshikawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Ryo Emoto
- Department of Biostatistics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shigeyuki Matsui
- Department of Biostatistics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| |
Collapse
|
2
|
Fox A, Leonard GD, Adzibolosu N, Wong T, Tedja R, Sharma S, Gogoi R, Morris R, Mor G, Fehl C, Alvero AB. Adipose microenvironment promotes hypersialylation of ovarian cancer cells. Front Oncol 2024; 14:1432333. [PMID: 39104719 PMCID: PMC11299042 DOI: 10.3389/fonc.2024.1432333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 06/28/2024] [Indexed: 08/07/2024] Open
Abstract
Introduction Ovarian and other peritoneal cancers have a strong tendency to metastasize into the surrounding adipose tissue. This study describes an effect of the adipose microenvironment on upregulation of sialic acid-containing glycans in ovarian cancer (OC). Heterogeneous populations of glycosylated OC tumors converged to a highly sialylated cell state that regulates tumorigenesis in an immune-dependent manner. Methods We modeled the adipose microenvironment by conditioning growth media with human patient-derived adipose tissue. OC cell lines grown in the presence vs. absence of adipose conditioned media (ACM) were characterized by transcriptomics, western blotting, and chemical biology glycan labeling methods. Fluorescence-activated cell sorting was used to separate adipose-driven upregulation of hypersialylated ("SNA-high") vs. hyposialylated ("SNA-low") OC subpopulations. The two subpopulations were characterized by further transcriptomic and quantitative polymerase chain reaction analyses, then injected into a syngeneic mouse model. Immune system involvement was implicated using wild type and athymic nude mice with a primary endpoint of overall survival. Results Adipose conditioning resulted in upregulation of sialyltransferases ST3GAL1, ST6GAL1, ST6GALNAC3, and ST8Sia1. In culture, OC cells displayed two distinct sialylated subpopulations that were stable for up to 9 passages, suggesting inherent heterogeneity in sialylation that is maintained throughout cell division and media changes. OC tumors that implanted in the omental adipose tissue exclusively reprogrammed to the highly sialylated subpopulation. In wild type C57BL/6 mice, only the hypersialylated SNA-high subpopulation implanted in the adipose, whereas the hyposialylated SNA-low subpopulation failed to be tumorigenic (p=0.023, n=5). In the single case where SNA-low established a tumor, post-mortem analysis revealed reprogramming of the tumor to the SNA-high state in vivo. In athymic nude mice, both subpopulations rapidly formed tumors, implicating a role of the adaptive immune system. Conclusions These findings suggest a model of glycan-dependent tumor evolution wherein the adipose microenvironment reprograms OC to a tumorigenic state that resists the adaptive immune system. Mechanistically, adipose factors upregulate sialyltransferases. To our knowledge, this is the first demonstration of the effect of adipose microenvironment on OC tumor sialylation. Our results set the stage for translational applications targeting sialic acid pathways in OC and other peritoneal cancer tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Alexandra Fox
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| | - Garry D. Leonard
- Department of Chemistry, Wayne State University, Detroit, MI, United States
| | - Nicholas Adzibolosu
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| | - Terrence Wong
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Detroit, MI, United States
| | - Roslyn Tedja
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Detroit, MI, United States
| | - Sapna Sharma
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| | - Radhika Gogoi
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Detroit, MI, United States
| | - Robert Morris
- Karmanos Cancer Institute, Detroit, MI, United States
| | - Gil Mor
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Detroit, MI, United States
| | - Charlie Fehl
- Department of Chemistry, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Detroit, MI, United States
| | - Ayesha B. Alvero
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
- Karmanos Cancer Institute, Detroit, MI, United States
| |
Collapse
|
3
|
Fox A, Leonard GD, Adzibolosu N, Wong T, Tedja R, Sharma S, Gogoi R, Morris R, Mor G, Fehl C, Alvero AB. Adipose microenvironment promotes hypersialylation of ovarian cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.593990. [PMID: 38798490 PMCID: PMC11118282 DOI: 10.1101/2024.05.13.593990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Sialylation, the addition of negatively charged sialic acid sugars to terminal ends of glycans, is upregulated in most cancers. Hypersialylation supports multiple pro-tumor mechanisms such as enhanced migration and invasion, resistance to apoptosis and immune evasion. A current gap in knowledge is the lack of understanding on how the tumor microenvironment regulates cancer cell sialylation. The adipose niche is a main component of most peritoneal cancers' microenvironment. This includes ovarian cancer (OC), which causes most deaths from all gynecologic cancers. In this report, we demonstrate that the adipose microenvironment is a critical regulator of OC cell sialylation. In vitro adipose conditioning led to an increase in both ⍺2,3- and ⍺2,6-linked cell surface sialic acids in both human and mouse models of OC. Adipose-induced sialylation reprogramming was also observed in vivo from intra-peritoneal OC tumors seeded in the adipose-rich omentum. Mechanistically, we observed upregulation of at least three sialyltransferases, ST3GAL1, ST6GAL1 and ST3GALNAC3. Hypersialylated OC cells consistently formed intra-peritoneal tumors in both immune-competent mice and immune-compromised athymic nude mice. In contrast, hyposiaylated OC cells persistently formed tumors only in athymic nude mice demonstrating that sialylation impacts OC tumor formation in an immune dependent manner. To our knowledge, this is the first demonstration of the effect of adipose microenvironment on OC tumor sialylation. Our results set the stage for translational applications targeting sialic acid pathways in OC and other peritoneal cancers.
Collapse
|
4
|
Alvero AB, Fox A, Madina BR, Krady MM, Gogoi R, Chehade H, Nakaar V, Almassian B, Yarovinsky TO, Rutherford T, Mor G. Immune Modulation of Innate and Adaptive Responses Restores Immune Surveillance and Establishes Antitumor Immunologic Memory. Cancer Immunol Res 2024; 12:261-274. [PMID: 38078853 PMCID: PMC11027955 DOI: 10.1158/2326-6066.cir-23-0127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/02/2023] [Accepted: 12/05/2023] [Indexed: 12/26/2023]
Abstract
Current immunotherapies have proven effective in strengthening antitumor immune responses, but constant opposing signals from tumor cells and the surrounding microenvironment eventually lead to immune escape. We hypothesized that in situ release of antigens and regulation of both the innate and adaptive arms of the immune system would provide a robust and long-term antitumor effect by creating immunologic memory against tumors. To achieve this, we developed CARG-2020, a genetically modified virus-like vesicle (VLV) that is a self-amplifying RNA with oncolytic capacity and encodes immune regulatory genes. CARG-2020 carries three immune modulators: (i) the pleiotropic antitumor cytokine IL12, in which the subunits (p35 and p40) are tethered together; (ii) the extracellular domain (ECD) of the protumor IL17RA, which serves as a dominant-negative antagonist; and (iii) a shRNA targeting PD-L1. Using a mouse model of ovarian cancer, we demonstrated the oncolytic effect and immune-modulatory capacities of CARG-2020. By enhancing IL12 and blocking IL17 and PD-L1, CARG-2020 successfully reactivated immune surveillance by promoting M1, instead of M2, macrophage differentiation, inhibiting MDSC expansion and establishing a potent CD8+ T cell-mediated antitumoral response. Furthermore, we demonstrated that this therapeutic approach provided tumor-specific and long-term protection against the establishment of new tumors. Our results provide a rationale for the further development of this platform as a therapeutic modality for ovarian cancer patients to enhance antitumor responses and prevent a recurrence.
Collapse
Affiliation(s)
- Ayesha B. Alvero
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Alexandra Fox
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | | | | | - Radhika Gogoi
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Hussein Chehade
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | | | | | | | - Thomas Rutherford
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| |
Collapse
|
5
|
Zhang Y, Tedja R, Millman M, Wong T, Fox A, Chehade H, Gershater M, Adzibolosu N, Gogoi R, Anderson M, Rutherford T, Zhang Z, Chopp M, Mor G, Alvero AB. Adipose-derived exosomal miR-421 targets CBX7 and promotes metastatic potential in ovarian cancer cells. J Ovarian Res 2023; 16:233. [PMID: 38037081 PMCID: PMC10688490 DOI: 10.1186/s13048-023-01312-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 11/09/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Chromobox protein homolog 7 (CBX7), a member of the Polycomb repressor complex, is a potent epigenetic regulator and gene silencer. Our group has previously reported that CBX7 functions as a tumor suppressor in ovarian cancer cells and its loss accelerated formation of carcinomatosis and drove tumor progression in an ovarian cancer mouse model. The goal of this study is to identify specific signaling pathways in the ovarian tumor microenvironment that down-regulate CBX7. Given that adipocytes are an integral component of the peritoneal cavity and the ovarian tumor microenvironment, we hypothesize that the adipose microenvironment is an important regulator of CBX7 expression. RESULTS Using conditioned media from human omental explants, we found that adipose-derived exosomes mediate CBX7 downregulation and enhance migratory potential of human ovarian cancer cells. Further, we identified adipose-derived exosomal miR-421 as a novel regulator of CBX7 expression and the main effector that downregulates CBX7. CONCLUSION In this study, we identified miR-421 as a specific signaling pathway in the ovarian tumor microenvironment that can downregulate CBX7 to induce epigenetic change in OC cells, which can drive disease progression. These findings suggest that targeting exosomal miR-421 may curtail ovarian cancer progression.
Collapse
Affiliation(s)
- Yi Zhang
- Neurology, Henry Ford Health System, 2799 W Grand Blvd., Detroit, MI, 48202, USA.
| | - Roslyn Tedja
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, 275 E. Hancock St., Detroit, MI, 48201, USA
| | - Michael Millman
- Neurology, Henry Ford Health System, 2799 W Grand Blvd., Detroit, MI, 48202, USA
| | - Terrence Wong
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, 275 E. Hancock St., Detroit, MI, 48201, USA
| | - Alexandra Fox
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, 275 E. Hancock St., Detroit, MI, 48201, USA
| | - Hussein Chehade
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, 275 E. Hancock St., Detroit, MI, 48201, USA
| | - Meyer Gershater
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, 275 E. Hancock St., Detroit, MI, 48201, USA
| | - Nicholas Adzibolosu
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, 275 E. Hancock St., Detroit, MI, 48201, USA
| | - Radhika Gogoi
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, 275 E. Hancock St., Detroit, MI, 48201, USA
| | - Matthew Anderson
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Thomas Rutherford
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Zhenggang Zhang
- Neurology, Henry Ford Health System, 2799 W Grand Blvd., Detroit, MI, 48202, USA
| | - Michael Chopp
- Neurology, Henry Ford Health System, 2799 W Grand Blvd., Detroit, MI, 48202, USA
- Department of Physics, Oakland University, Rochester, MI, USA
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, 275 E. Hancock St., Detroit, MI, 48201, USA
| | - Ayesha B Alvero
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, 275 E. Hancock St., Detroit, MI, 48201, USA.
| |
Collapse
|
6
|
Zhang Y, Tedja R, Millman M, Wong T, Fox A, Chehade H, Gershater M, Adzibolosu N, Gogoi R, Anderson M, Rutherford T, Zhang Z, Chopp M, Mor G, Alvero AB. Adipose-derived exosomal miR-421 targets CBX7 and promotes metastatic potential in ovarian cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.07.566022. [PMID: 37986971 PMCID: PMC10659572 DOI: 10.1101/2023.11.07.566022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Background Chromobox protein homolog 7 (CBX7), a member of the Polycomb repressor complex, is a potent epigenetic regulator and gene silencer. Our group has previously reported that CBX7 functions as a tumor suppressor in ovarian cancer cells and its loss accelerated formation of carcinomatosis and drove tumor progression in an ovarian cancer mouse model. The goal of this study is to identify specific signaling pathways in the ovarian tumor microenvironment that down-regulate CBX7. Given that adipocytes are an integral component of the peritoneal cavity and the ovarian tumor microenvironment, we hypothesize that the adipose microenvironment is an important regulator of CBX7 expression. Results Using conditioned media from human omental explants, we found that adipose-derived exosomes mediate CBX7 downregulation and enhance migratory potential of human ovarian cancer cells. Further, we identified adipose-derived exosomal miR-421 as a novel regulator of CBX7 expression and the main effector that downregulates CBX7. Conclusion In this study, we identified miR-421 as a specific signaling pathway in the ovarian tumor microenvironment that can downregulate CBX7 to induce epigenetic change in OC cells, which can drive disease progression. These findings suggest that targeting exosomal miR-421 may curtail ovarian cancer progression.
Collapse
Affiliation(s)
- Yi Zhang
- Neurology, Henry Ford Health, Detroit, MI
| | - Roslyn Tedja
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | | | - Terrence Wong
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Alexandra Fox
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Hussein Chehade
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Meyer Gershater
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Nicholas Adzibolosu
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Radhika Gogoi
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Matthew Anderson
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL
| | - Thomas Rutherford
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL
| | | | - Michael Chopp
- Neurology, Henry Ford Health, Detroit, MI
- Department of Physics, Oakland University, Rochester, MI
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Ayesha B. Alvero
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| |
Collapse
|
7
|
Gogoi RP, Galoforo S, Fox A, Morris C, Ramos H, Gogoi VK, Chehade H, Adzibolosu NK, Shi C, Zhang J, Tedja R, Morris R, Alvero AB, Mor G. A Novel Role of Connective Tissue Growth Factor in the Regulation of the Epithelial Phenotype. Cancers (Basel) 2023; 15:4834. [PMID: 37835529 PMCID: PMC10571845 DOI: 10.3390/cancers15194834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/21/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) is a biological process where epithelial cells lose their adhesive properties and gain invasive, metastatic, and mesenchymal properties. Maintaining the balance between the epithelial and mesenchymal stage is essential for tissue homeostasis. Many of the genes promoting mesenchymal transformation have been identified; however, our understanding of the genes responsible for maintaining the epithelial phenotype is limited. Our objective was to identify the genes responsible for maintaining the epithelial phenotype and inhibiting EMT. METHODS RNA seq was performed using an vitro model of EMT. CTGF expression was determined via qPCR and Western blot analysis. The knockout of CTGF was completed using the CTGF sgRNA CRISPR/CAS9. The tumorigenic potential was determined using NCG mice. RESULTS The knockout of CTGF in epithelial ovarian cancer cells leads to the acquisition of functional characteristics associated with the mesenchymal phenotype such as anoikis resistance, cytoskeleton remodeling, increased cell stiffness, and the acquisition of invasion and tumorigenic capacity. CONCLUSIONS We identified CTGF is an important regulator of the epithelial phenotype, and its loss is associated with the early cellular modifications required for EMT. We describe a novel role for CTGF, regulating cytoskeleton and the extracellular matrix interactions necessary for the conservation of epithelial structure and function. These findings provide a new window into understanding the early stages of mesenchymal transformation.
Collapse
Affiliation(s)
- Radhika P. Gogoi
- Karmanos Cancer Institute, Wayne State University, 4100 John R St, Detroit, MI 48202, USA;
| | - Sandra Galoforo
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Alexandra Fox
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Colton Morris
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Harry Ramos
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Vir K. Gogoi
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Hussein Chehade
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Nicholas K. Adzibolosu
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Chenjun Shi
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA; (C.S.); (J.Z.)
| | - Jitao Zhang
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA; (C.S.); (J.Z.)
| | - Roslyn Tedja
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Robert Morris
- Karmanos Cancer Institute, Wayne State University, 4100 John R St, Detroit, MI 48202, USA;
| | - Ayesha B. Alvero
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48202, USA; (S.G.); (A.F.); (C.M.); (H.R.); (V.K.G.); (H.C.); (N.K.A.); (R.T.); (A.B.A.)
| |
Collapse
|
8
|
Alvero AB, Fox A, Madina B, Krady M, Gogoi R, Chehade H, Nakaar V, Almassian B, Yarovinsky T, Rutherford T, Mor G. Immune modulation of innate and adaptive responses restores immune surveillance and establishes anti-tumor immunological memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559828. [PMID: 37808682 PMCID: PMC10557730 DOI: 10.1101/2023.09.27.559828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Current immunotherapies have proven effective in strengthening anti-tumor immune responses but constant opposing signals from tumor cells and surrounding microenvironment eventually lead to immune escape. We hypothesize that in situ release of antigens and regulation of both the innate and adaptive arms of the immune system will provide a robust and long-term anti-tumor effect by creating immunological memory against the tumor. To achieve this, we developed CARG-2020, a virus-like-vesicle (VLV). It is a genetically modified and self-amplifying RNA with oncolytic capacity and encodes immune regulatory genes. CARG-2020 carries three transgenes: 1 ) the pleiotropic antitumor cytokine IL-12 in which the subunits (p35 and p40) are tethered together; 2) the extracellular domain (ECD) of the pro- tumor IL-17RA, which can serve as a dominant negative antagonist; and 3) shRNA for PD-L1. Using a mouse model of ovarian cancer, we demonstrate the oncolytic effect and immune modulatory capacities of CARG-2020. By enhancing IL-12 and blocking IL-17 and PD-L1, CARG-2020 successfully reactivates immune surveillance by promoting M1 instead of M2 macrophage differentiation, inhibiting MDSC expansion, and establishing a potent CD8+ T cell mediated anti-tumoral response. Furthermore, we demonstrate that this therapeutic approach provides tumor-specific and long-term protection preventing the establishment of new tumors. Our results provide rationale for the further development of this platform as a therapeutic modality for ovarian cancer patients to enhance the anti-tumor response and to prevent recurrence.
Collapse
Affiliation(s)
- Ayesha B. Alvero
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Alexandra Fox
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | | | | | - Radhika Gogoi
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Hussein Chehade
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | | | | | | | - Thomas Rutherford
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| |
Collapse
|
9
|
Chehade H, Purandare N, Fox A, Adzibolosu N, Jayee S, Singh A, Tedja R, Gogoi R, Aras S, Grossman LI, Mor G, Alvero AB. MNRR1 is a driver of ovarian cancer progression. Transl Oncol 2023; 29:101623. [PMID: 36641875 PMCID: PMC9860385 DOI: 10.1016/j.tranon.2023.101623] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/30/2022] [Accepted: 01/06/2023] [Indexed: 01/14/2023] Open
Abstract
Cancer progression requires the acquisition of mechanisms that support proliferative potential and metastatic capacity. MNRR1 (also CHCHD2, PARK22, AAG10) is a bi-organellar protein that in the mitochondria can bind to Bcl-xL to enhance its anti-apoptotic function, or to respiratory chain complex IV (COX IV) to increase mitochondrial respiration. In the nucleus, it can act as a transcription factor and promote the expression of genes involved in mitochondrial biogenesis, migration, and cellular stress response. Given that MNRR1 can regulate both apoptosis and mitochondrial respiration, as well as migration, we hypothesize that it can modulate metastatic spread. Using ovarian cancer models, we show heterogeneous protein expression levels of MNRR1 across samples tested and cell-dependent control of its stability and binding partners. In addition to its anti-apoptotic and bioenergetic functions, MNRR1 is both necessary and sufficient for a focal adhesion and ECM repertoire that can support spheroid formation. Its ectopic expression is sufficient to induce the adhesive glycoprotein THBS4 and the type 1 collagen, COL1A1. Conversely, its deletion leads to significant downregulation of these genes. Furthermore, loss of MNRR1 leads to delay in tumor growth, curtailed carcinomatosis, and improved survival in a syngeneic ovarian cancer mouse model. These results suggest targeting MNRR1 may improve survival in ovarian cancer patients.
Collapse
Affiliation(s)
- Hussein Chehade
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States,C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| | - Neeraja Purandare
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
| | - Alexandra Fox
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| | - Nicholas Adzibolosu
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| | - Shawn Jayee
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| | - Aryan Singh
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| | - Roslyn Tedja
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| | - Radhika Gogoi
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| | - Siddhesh Aras
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
| | - Lawrence I. Grossman
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States
| | - Ayesha B. Alvero
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, United States,Corresponding author at: 275 E. Hancock St., Detroit, MI, 48201, United States.
| |
Collapse
|
10
|
Iyoshi S, Sumi A, Yoshihara M, Kitami K, Mogi K, Uno K, Fujimoto H, Miyamoto E, Tano S, Yoshikawa N, Emoto R, Matsui S, Kajiyama H. Obesity contributes to the stealth peritoneal dissemination of ovarian cancer: a multi-institutional retrospective cohort study. Obesity (Silver Spring) 2022; 30:1599-1607. [PMID: 35851756 DOI: 10.1002/oby.23497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/06/2022] [Accepted: 04/28/2022] [Indexed: 11/08/2022]
Abstract
OBJECTIVE The clinical significance of a higher BMI on the prognosis of ovarian cancer remains controversial; therefore, a more detailed analysis is demanded. This study investigated the impact of BMI on peritoneum-specific recurrence to clarify the involvement of adipose tissue in the proliferation of cancer cells at sites of peritoneal dissemination. METHODS Among 4,730 patients with malignant ovarian tumors, 280 diagnosed with International Federation of Gynecology and Obstetrics (FIGO) stage IIB to IIIC epithelial ovarian cancer and who underwent complete resection in the primary surgery were included in the present study. RESULTS There were 42, 201, and 37 women in the low, normal, and high BMI groups, respectively. Peritoneum-specific recurrence-free survival and overall survival were both significantly shorter in patients with a high BMI than in those with a normal BMI (p = 0.028 and 0.018, respectively). No significant differences were observed in the distribution of sites of recurrence between these two groups. A multivariate analysis identified obesity as an independent prognostic factor in addition to pT3 tumor staging and positive ascites cytology. CONCLUSIONS Patients with a high BMI had a significantly worse prognosis than those with a normal BMI, and peritoneal adipose tissue may have contributed to this difference.
Collapse
Affiliation(s)
- Shohei Iyoshi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Asami Sumi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Obstetrics and Gynecology, Tosei General Hospital, Seto, Japan
| | - Masato Yoshihara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuhisa Kitami
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Obstetrics and Gynecology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kazumasa Mogi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kaname Uno
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University Graduate School of Medicine, Lund, Sweden
| | - Hiroki Fujimoto
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Discipline of Obstetrics and Gynaecology, Adelaide Medical School, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Emiri Miyamoto
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Sho Tano
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobuhisa Yoshikawa
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryo Emoto
- Department of Biostatistics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shigeyuki Matsui
- Department of Biostatistics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
11
|
Shu S, Liu X, Xu M, Lin Y, Li R. The inhibitory role of si-UBB delivered by degradable dendrimers-based lipid nanoparticles in ovarian cancer. Cancer Nanotechnol 2022. [DOI: 10.1186/s12645-022-00112-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Abstract
Objective
RNA interference holds tremendous potential in the treatment of malignant tumors. However, efficient and biocompatible delivery methods are needed for systemic delivery of small interfering RNA (siRNA). In this study, we explored the delivery efficiency and therapy effect of si-UBB-5A2SC8 in ovarian cancer.
Methods and materials
Si-UBB-5A2SC8 nanoparticles were successfully prepared according to the established procedure and the characteristic of nanoparticles was determined by digital laser scanning. Flow cytometry and confocal analysis demonstrated si-UBB was efficiently transfected to cell by the delivery of 5A2SC8 complexes. The in vitro gene knockdown efficiency of ubiquitin B was demonstrated by RT-qPCR and Western blot analysis, which was further verified by the inhibition of proliferation and migration of ovarian cancer cells. Accumulative efficiency of si-UBB-5A2SC8 nanoparticles was investigated in BALB/c mice bearing SKOV3-GFP tumor xenograft. In vivo imaging was adopted to test the accurate location of the nanoparticle in the tumor. The feature of the tumor and pivotal organ was determined. TUNEL and caspase-3 expression was used to analyze the underling mechanism of the inhibition effect.
Results
The average size and the zeta potential of the si-UBB-5A2SC8 was (150 ± 11) nm and − (20 ± 4) mV, respectively. Transmission electronic microscopy showed the nanoparticle was near-spherical with the mean size of (100 ± 15) nm. Flow cytometry and confocal microscopic images demonstrated 5A2SC8 complex could successfully deliver Cy5.5-siRNA to the cytoplasm of ovarian cancer cells. qRT-PCR and western blot demonstrated the mRNA and protein expression of ubiquitin B was decreased to 62.5% and 36.5% of the control group, which was accompanied with the decreased proliferation and migration ability in si-UBB-5A2SC8-transfected cells. Ex vivo fluorescence imaging demonstrated 5A2SC8 complex could successfully carry siRNA to the tumor lesion and exert the inhibition effect, which was verified by the decreased tumor weight and increased apoptosis and caspase-3 expression in mice treated with si-UBB-5A2SC8.
Conclusions
5A2SC8 complex was a safe and efficient gene delivery vehicle and ubiquitin B was a potential target for the ovarian cancer targeted therapy.
Collapse
|
12
|
Mendoza-Martinez AK, Loessner D, Mata A, Azevedo HS. Modeling the Tumor Microenvironment of Ovarian Cancer: The Application of Self-Assembling Biomaterials. Cancers (Basel) 2021; 13:5745. [PMID: 34830897 PMCID: PMC8616551 DOI: 10.3390/cancers13225745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/07/2021] [Accepted: 11/11/2021] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer (OvCa) is one of the leading causes of gynecologic malignancies. Despite treatment with surgery and chemotherapy, OvCa disseminates and recurs frequently, reducing the survival rate for patients. There is an urgent need to develop more effective treatment options for women diagnosed with OvCa. The tumor microenvironment (TME) is a key driver of disease progression, metastasis and resistance to treatment. For this reason, 3D models have been designed to represent this specific niche and allow more realistic cell behaviors compared to conventional 2D approaches. In particular, self-assembling peptides represent a promising biomaterial platform to study tumor biology. They form nanofiber networks that resemble the architecture of the extracellular matrix and can be designed to display mechanical properties and biochemical motifs representative of the TME. In this review, we highlight the properties and benefits of emerging 3D platforms used to model the ovarian TME. We also outline the challenges associated with using these 3D systems and provide suggestions for future studies and developments. We conclude that our understanding of OvCa and advances in materials science will progress the engineering of novel 3D approaches, which will enable the development of more effective therapies.
Collapse
Affiliation(s)
- Ana Karen Mendoza-Martinez
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK;
- Institute of Bioengineering, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| | - Daniela Loessner
- Department of Chemical Engineering, Faculty of Engineering, Monash University, Melbourne, VIC 3800, Australia;
- Department of Materials Science and Engineering, Faculty of Engineering, Monash University, Melbourne, VIC 3800, Australia
- Department of Anatomy and Developmental Biology, Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC 3800, Australia
- Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden e.V., 01069 Dresden, Germany
| | - Alvaro Mata
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK;
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham NG7 2RD, UK
- Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Helena S. Azevedo
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK;
- Institute of Bioengineering, Queen Mary University of London, Mile End Road, London E1 4NS, UK
| |
Collapse
|
13
|
Lassa-VSV chimeric virus targets and destroys human and mouse ovarian cancer by direct oncolytic action and by initiating an anti-tumor response. Virology 2020; 555:44-55. [PMID: 33453650 DOI: 10.1016/j.virol.2020.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/18/2020] [Accepted: 10/28/2020] [Indexed: 11/24/2022]
Abstract
Ovarian cancer is the third most common female cancer, with poor survival in later stages of metastatic spread. We test a chimeric virus consisting of genes from Lassa and vesicular stomatitis viruses, LASV-VSV; the native VSV glycoprotein is replaced by the Lassa glycoprotein, greatly reducing neurotropism. Human ovarian cancer cells in immunocompromised nude mice were lethal in controls. Chemotherapeutic paclitaxel and cisplatin showed modest cancer inhibition and survival extension. In contrast, a single intraperitoneal injection of LASV-VSV selectively infected and killed ovarian cancer cells, generating long-term survival. Mice with human ovarian cancer cells in brain showed rapid deterioration; LASV-VSV microinjection into brain blocked cancer growth, and generated long-term survival. Treatment of immunocompetent mice with infected mouse ovarian cancer cells blocked growth of non-infected ovarian cancer cells peritoneally and in brain. These results suggest LASV-VSV is a viable candidate for further study and may be of use in the treatment of ovarian cancer.
Collapse
|
14
|
Alvero AB, Hanlon D, Pitruzzello M, Filler R, Robinson E, Sobolev O, Tedja R, Ventura A, Bosenberg M, Han P, Edelson RL, Mor G. Transimmunization restores immune surveillance and prevents recurrence in a syngeneic mouse model of ovarian cancer. Oncoimmunology 2020; 9:1758869. [PMID: 32566387 PMCID: PMC7302442 DOI: 10.1080/2162402x.2020.1758869] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ovarian cancer accounts for most deaths from gynecologic malignancies. Although more than 80% of patients respond to first-line standard of care, most of these responders present with recurrence and eventually succumb to carcinomatosis and chemotherapy-resistant disease. To improve patient survival, new modalities must, therefore, target or prevent recurrent disease. Here we describe for the first time a novel syngeneic mouse model of recurrent high-grade serous ovarian cancer (HGSOC), which allows immunotherapeutic interventions in a time course relevant to human carcinomatosis and disease course. Using this model, we demonstrate the efficacy of Transimmunization (TI), a dendritic cell (DC) vaccination strategy that uses autologous and physiologically derived DC loaded with autologous whole tumor antigens. TI has been proven successful in the treatment of human cutaneous T cell lymphoma and we report for the first time its in vivo efficacy against an intra-peritoneal solid tumor. Given as a single therapy, TI is able to elicit an effective anti-tumor immune response and inhibit immune-suppressive crosstalks with sufficient power to curtail tumor progression and establishment of carcinomatosis and recurrent disease. Specifically, TI is able to inhibit the expansion of tumor-associated macrophages as well as myeloid-derived suppressive cells consequently restoring T cell immune-surveillance. These results demonstrate the possible value of TI in the management of ovarian cancer and other intra-peritoneal tumors.
Collapse
Affiliation(s)
- Ayesha B Alvero
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Douglas Hanlon
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Mary Pitruzzello
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Renata Filler
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Eve Robinson
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Olga Sobolev
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Roslyn Tedja
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Alessandra Ventura
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Marcus Bosenberg
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Patrick Han
- Department of Chemical & Environmental Engineering, Yale University School of Engineering and Applied Science, New Haven, CT, USA
| | - Richard L Edelson
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Gil Mor
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA.,C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
15
|
Li J, Alvero AB, Nuti S, Tedja R, Roberts CM, Pitruzzello M, Li Y, Xiao Q, Zhang S, Gan Y, Wu X, Mor G, Yin G. CBX7 binds the E-box to inhibit TWIST-1 function and inhibit tumorigenicity and metastatic potential. Oncogene 2020; 39:3965-3979. [PMID: 32205869 PMCID: PMC8343988 DOI: 10.1038/s41388-020-1269-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 03/05/2020] [Accepted: 03/11/2020] [Indexed: 12/16/2022]
Abstract
Deaths from ovarian cancer usually occur when patients succumb to overwhelmingly numerous and widespread micrometastasis. Whereas epithelial-mesenchymal transition is required for epithelial ovarian cancer cells to acquire metastatic potential, the cellular phenotype at secondary sites and the mechanisms required for the establishment of metastatic tumors are not fully determined. Using in vitro and in vivo models we show that secondary epithelial ovarian cancer cells (sEOC) do not fully reacquire the molecular signature of the primary epithelial ovarian cancer cells from which they are derived. Despite displaying an epithelial morphology, sEOC maintains a high expression of the mesenchymal effector, TWIST-1. TWIST-1 is however transcriptionally nonfunctional in these cells as it is precluded from binding its E-box by the PcG protein, CBX7. Deletion of CBX7 in sEOC was sufficient to reactivate TWIST-1-induced transcription, prompt mesenchymal transformation, and enhanced tumorigenicity in vivo. This regulation allows secondary tumors to achieve an epithelial morphology while conferring the advantage of prompt reversal to a mesenchymal phenotype upon perturbation of CBX7. We also describe a subclassification of ovarian tumors based on CBX7 and TWIST-1 expression, which predicts clinical outcomes and patient prognosis.
Collapse
Affiliation(s)
- Juanni Li
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, Hunan Province, China
| | - Ayesha B Alvero
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Sudhakar Nuti
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Roslyn Tedja
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Cai M Roberts
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Mary Pitruzzello
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Yimin Li
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, Hunan Province, China
| | - Qing Xiao
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, Hunan Province, China
| | - Sai Zhang
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, Hunan Province, China
| | - Yaqi Gan
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, Hunan Province, China
| | - Xiaoying Wu
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, Hunan Province, China
| | - Gil Mor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA.
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA.
| | - Gang Yin
- Department of Pathology, Xiangya Hospital, School of Basic Medical Sciences, Central South University, Changsha, Hunan Province, China.
| |
Collapse
|
16
|
He J, Li C, Ding L, Huang Y, Yin X, Zhang J, Zhang J, Yao C, Liang M, Pirraco RP, Chen J, Lu Q, Baldridge R, Zhang Y, Wu M, Reis RL, Wang Y. Tumor Targeting Strategies of Smart Fluorescent Nanoparticles and Their Applications in Cancer Diagnosis and Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1902409. [PMID: 31369176 DOI: 10.1002/adma.201902409] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/30/2019] [Indexed: 06/10/2023]
Abstract
Advantages such as strong signal strength, resistance to photobleaching, tunable fluorescence emissions, high sensitivity, and biocompatibility are the driving forces for the application of fluorescent nanoparticles (FNPs) in cancer diagnosis and therapy. In addition, the large surface area and easy modification of FNPs provide a platform for the design of multifunctional nanoparticles (MFNPs) for tumor targeting, diagnosis, and treatment. In order to obtain better targeting and therapeutic effects, it is necessary to understand the properties and targeting mechanisms of FNPs, which are the foundation and play a key role in the targeting design of nanoparticles (NPs). Widely accepted and applied targeting mechanisms such as enhanced permeability and retention (EPR) effect, active targeting, and tumor microenvironment (TME) targeting are summarized here. Additionally, a freshly discovered targeting mechanism is introduced, termed cell membrane permeability targeting (CMPT), which improves the tumor-targeting rate from less than 5% of the EPR effect to more than 50%. A new design strategy is also summarized, which is promising for future clinical targeting NPs/nanomedicines design. The targeting mechanism and design strategy will inspire new insights and thoughts on targeting design and will speed up precision medicine and contribute to cancer therapy and early diagnosis.
Collapse
Affiliation(s)
- Jiuyang He
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Chenchen Li
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Lin Ding
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Department of Biological Chemistry, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yanan Huang
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Xuelian Yin
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Junfeng Zhang
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Jian Zhang
- Universal Medical Imaging Diagnostic Research Center, Shanghai, 200233, P. R. China
| | - Chenjie Yao
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, 02115, USA
| | - Minmin Liang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, P. R. China
| | - Rogério P Pirraco
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's PT Government Associate Lab, 4805, Braga/Guimarães, Portugal
| | - Jie Chen
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Quan Lu
- Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, 02115, USA
| | - Ryan Baldridge
- Department of Biological Chemistry, The University of Michigan, Ann Arbor, MI, 48109, USA
| | - Yong Zhang
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Department of Biomedical Engineering, National University of Singapore, Singapore, 119077, Singapore
| | - Minghong Wu
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017, Barco, Guimarães, Portugal
- ICVS/3B's PT Government Associate Lab, 4805, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017, Barco, Guimarães, Portugal
| | - Yanli Wang
- Tumor Precision Targeting Research Center, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Institute of Nanochemistry and Nanobiology, School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, P. R. China
- Harvard T. H. Chan School of Public Health, Harvard University, Boston, MA, 02115, USA
| |
Collapse
|
17
|
Samani EN, Mamillapalli R, Li F, Mutlu L, Hufnagel D, Krikun G, Taylor HS. Micrometastasis of endometriosis to distant organs in a murine model. Oncotarget 2019; 10:2282-2291. [PMID: 31040919 PMCID: PMC6481344 DOI: 10.18632/oncotarget.16889] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/30/2017] [Indexed: 11/25/2022] Open
Abstract
Endometriosis is an inflammatory gynecological disorder among reproductive-aged women caused by the engraftment and proliferation of endometrial cells outside the uterus, most commonly in the pelvis. It is thought that the disease arises primarily from retrograde menstruation where cells from the endometrium travel through the fallopian tubes to the peritoneal cavity. However, migration of endometriosis-derived cells to distant organs outside of the peritoneal cavity have not been explored. In the present study, we developed and validated a mouse model of disseminated endometriosis using syngeneic DsRed endometrial tissue introduced into the peritoneum of immunocompetent mice. Flow cytometry and immunofluorescence analysis, demonstrated the presence of endometriosis-derived cells in multiple organs (including lung, spleen, liver and brain) in the murine endometriosis model. Immunostaining revealed the presence of DsRed+/CD45- cells in brain, liver and lung. Engraftment occurred in all experimental animals examined. Cells from endometriotic lesions are capable of migration to and engraftment of multiple organs outside of the peritoneal cavity. Micrometastasis of endometriosis is a novel and frequent phenomenon. These data suggest that widespread dissemination of endometriosis may be common, clinically unrecognized and contribute to the diffuse clinical manifestations of this disease.
Collapse
Affiliation(s)
- Elham N. Samani
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, Connecticut 06510, New Haven, USA
| | - Ramanaiah Mamillapalli
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, Connecticut 06510, New Haven, USA
| | - Fei Li
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, Connecticut 06510, New Haven, USA
| | - Levent Mutlu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, Connecticut 06510, New Haven, USA
| | - Demetra Hufnagel
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, Connecticut 06510, New Haven, USA
| | - Graciela Krikun
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, Connecticut 06510, New Haven, USA
| | - Hugh S. Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, Connecticut 06510, New Haven, USA
| |
Collapse
|
18
|
Liu J, Lécuyer T, Seguin J, Mignet N, Scherman D, Viana B, Richard C. Imaging and therapeutic applications of persistent luminescence nanomaterials. Adv Drug Deliv Rev 2019; 138:193-210. [PMID: 30414492 DOI: 10.1016/j.addr.2018.10.015] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/26/2018] [Accepted: 10/31/2018] [Indexed: 12/13/2022]
Abstract
The development of probes for biomolecular imaging and diagnostics is a very active research area. Among the different imaging modalities, optics emerged since it is a noninvasive and cheap imaging technique allowing real time imaging. In vitro, this technique is very useful however in vivo, fluorescence suffers from low signal-to-noise ratio due to tissue autofluorescence under constant excitation. To address this limitation, novel types of optical nanoprobes are actually being developed and among them, persistent luminescence nanoparticles (PLNPs), with long lasting near-infrared (NIR) luminescence capability, allows doing optical imaging without constant excitation and so without autofluorescence. This review will begin by introducing the physical phenomenon associated to the long luminescence decay of such nanoprobes, from minutes to hours after ceasing the excitation. Then we will show how this property can be used to develop in vivo imaging probes and also more recently nanotheranostic agents. Finally, preliminary data on their biocompatibility will be mentioned and we will conclude by envisioning on the future applications and improvements of such nanomaterials.
Collapse
|
19
|
Guerrero S, Inostroza-Riquelme M, Contreras-Orellana P, Diaz-Garcia V, Lara P, Vivanco-Palma A, Cárdenas A, Miranda V, Robert P, Leyton L, Kogan MJ, Quest AFG, Oyarzun-Ampuero F. Curcumin-loaded nanoemulsion: a new safe and effective formulation to prevent tumor reincidence and metastasis. NANOSCALE 2018; 10:22612-22622. [PMID: 30484463 DOI: 10.1039/c8nr06173d] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Curcumin is widely considered beneficial to human health, but insolubility and instability greatly hamper reproducible exploitation of the advantageous traits. Here we report on the development, characterization and evaluation of a curcumin-loaded nanoemulsion (CUR-NEM) that is highly effective in preventing post-surgery tumor reincidence and metastasis. The method of fabrication utilized safe excipients and generated particles of 200 nm (PDI ≤ 0.2) with negative zeta potential (-30 mV) and a high yield of curcumin (95%), which can be converted by lyophilization to a dry powder. In vitro assays showed that CUR-NEM is safe in non-cancerous human cells (HEK-293T) and preferentially cytotoxic in gastric (AGS), colon (HT29-ATCC, HT29-US), breast (MDA-MB-231) and melanoma (B16F10) cells. In addition, in melanoma cells the nanoformulation increases intracellular curcumin accumulation and reactive oxygen species (ROS) formation, while preventing cell-migration and invasion. In vivo studies in C57BL/6 mice demonstrated that a single dose, applied topically to the wounded area after surgical excision of primary tumors formed upon subcutaneous injection of syngeneic B16F10 cells, was sufficient to completely prevent reincident tumor growth and spontaneous lung metastasis, while in untreated animals 70% reincidence and metastasis were observed. In vivo experiments also showed that the fluorescence signal due to curcumin was maintained at least 15 days after topical application of CUR-NEM, while when administered in DMSO the curcumin signal disappeared within 4 days. Importantly, the administration of a dose 22 times larger than that applied topically to animals after tumor surgery did not alter biochemical parameters. Due to the safety and efficacy of the formulation, we envisage it as ideal for topical application in cancer patients following surgery, to prevent tumor reincidence and metastasis. In addition, other routes of administration/protocols could also be proposed to treat/prevent malignant tumors in patients.
Collapse
Affiliation(s)
- Simón Guerrero
- Laboratory of Cellular Communication, Program of Cell and Molecular Biology, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Av. Independencia 1027, Santiago 8380453, Chile
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Pitcovski J, Shahar E, Aizenshtein E, Gorodetsky R. Melanoma antigens and related immunological markers. Crit Rev Oncol Hematol 2017; 115:36-49. [DOI: 10.1016/j.critrevonc.2017.05.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 01/12/2023] Open
|