1
|
Liu X, Zhang J, Yi T, Li H, Tang X, Liu D, Wu D, Li Y. Decoding tumor angiogenesis: pathways, mechanisms, and future directions in anti-cancer strategies. Biomark Res 2025; 13:62. [PMID: 40251641 PMCID: PMC12007322 DOI: 10.1186/s40364-025-00779-x] [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: 11/26/2024] [Accepted: 04/13/2025] [Indexed: 04/20/2025] Open
Abstract
Angiogenesis, a crucial process in tumor growth and metastasis, necessitates targeted therapeutic intervention. This review reviews the latest knowledge of anti-angiogenesis targets in tumors, with emphasis on the molecular mechanisms and signaling pathways that regulate this process. We emphasize the tumor microenvironment's role in angiogenesis, examine endothelial cell metabolic changes, and evaluated potential therapeutic strategies targeting the tumor vascular system. At the same time, we analyzed the signaling pathway and molecular mechanism of tumor angiogenesis in detail. In addition, this paper also looks at the development trend of tumor anti-angiogenesis drugs, including their future development direction and challenges, aiming to provide prospective insight into the development of this field. Despite their potential, anti-angiogenic therapies encounter challenges like drug resistance and side effects, necessitating ongoing research to enhance cancer treatment strategies and the efficacy of these therapies.
Collapse
Affiliation(s)
- Xueru Liu
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Juan Zhang
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Ting Yi
- Department of Trauma Center, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Hui Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Xing Tang
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Dan Liu
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Daichao Wu
- Laboratory of Structural Immunology, Department of Hepatopancreatobiliary Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China.
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China.
| |
Collapse
|
2
|
Hongu T, Sarenqiqige, Shandan, Kusunoki H, Ishimura A, Suzuki T, Oskarsson T, Gotoh N. Permeable Lung Vasculature Creates Chemoresistant Endothelial Niche by Producing SERPINE1 at Breast Cancer Metastatic Sites. Cancer Sci 2025. [PMID: 40217581 DOI: 10.1111/cas.70050] [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: 11/09/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 04/14/2025] Open
Abstract
Chemotherapy resistance remains a major obstacle for eradicating metastatic cancer cells in distant organs. We identified that endothelial cells (ECs) in the lungs, where breast cancer cells often metastasize, form a chemoresistant perivascular niche for disseminated breast cancer cells. By investigating the lung EC secretome activated by metastasis, we found that serine protease inhibitor family E member 1 (SERPINE1), encoded by Serpine1, is upregulated in metastasis-associated lung ECs. This upregulation shields cancer cells from paclitaxel-induced apoptosis and promotes cancer stem cell properties. Serpine1 expression appears to be driven by YAP-TEAD activation in lung ECs that lose cell-cell contact, a phenomenon associated with increased vascular permeability in lungs affected by metastasis. Crucially, pharmacological inhibition of SERPINE1 enhances the chemotherapy sensitivity of metastatic breast cancer cells in the lung. Overall, our findings underscore the pivotal role of the vascular niche, which produces SERPINE1, in conferring chemoresistance to breast cancer cells during metastatic progression in the lungs.
Collapse
Affiliation(s)
- Tsunaki Hongu
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa City, Japan
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa City, Japan
| | - Sarenqiqige
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa City, Japan
| | - Shandan
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa City, Japan
| | - Hirokazu Kusunoki
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa City, Japan
| | - Akihiko Ishimura
- Division of Functional Genomics, Cancer Research Institute, Kanazawa University, Kanazawa City, Japan
| | - Takeshi Suzuki
- Division of Functional Genomics, Cancer Research Institute, Kanazawa University, Kanazawa City, Japan
| | - Thordur Oskarsson
- Department of Molecular Oncology, H. Lee Moffit Cancer Center & Research Institute, Tampa, Florida, USA
| | - Noriko Gotoh
- Division of Cancer Cell Biology, Cancer Research Institute, Kanazawa University, Kanazawa City, Japan
- Institute for Frontier Science Initiative, Kanazawa University, Kanazawa City, Japan
| |
Collapse
|
3
|
Wang X, Bai Z, Deng W, Wang X. Efficacy and safety of fruquintinib plus capecitabine as first-line treatment in patients with metastatic colorectal cancer ineligible for intravenous chemotherapy: a two-stage, single-armed, phase II study. Invest New Drugs 2025; 43:214-222. [PMID: 39945972 DOI: 10.1007/s10637-025-01510-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 01/28/2025] [Indexed: 05/03/2025]
Abstract
Fruquintinib has been recommended for treating refractory metastatic colorectal cancer. This single-arm, phase II study explored for the first time whether fruquintinib combined with capecitabine could be used as a first-line treatment for patients with metastatic colorectal cancer who are intolerant to intravenous chemotherapy. From December 8, 2021, to December 31, 2024, 17 patients were included in the effect analysis who respectively received capecitabine and fruquintinib at a starting dose of 825 mg/m2 twice a day and 4 mg every day (2 weeks followed by 1-week rest) and recorded changes in safety and quality of life; the dosage can be appropriately adjusted according to the protocol to make it tolerable for the patients. The median age was 76 years old; the study achieved a disease control rate of 88.2%, an overall response rate of 17.6%, and a median progression-free survival of 16.3 months (95% CI 9.7-22.9); the overall survival had not been reached. The median quality of life scores and self-assessment of health scores change, respectively, from 42 (IQR 34, 47) to 45 (IQR 41, 57) and from 5 (IQR 4.25, 6.75) to 4 (IQR 3.00, 6.00). There were only 3 events of grade ≥ 3 TRAEs, including one rare case of aortic dissection. Fruquintinib combined with capecitabine has initially shown ideal disease control, safety, and convenience, especially as a first-line treatment for elderly frail patients with metastatic colorectal cancer. Further phase III study is planned to refine this combination. Clinical Trial Number: NCT04866108.
Collapse
Affiliation(s)
- Xin Wang
- Department of General Surgery, Medical Oncology Group, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-Cheng District, 100050, Beijing, China
| | - Zhigang Bai
- Department of General Surgery, Medical Oncology Group, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-Cheng District, 100050, Beijing, China.
| | - Wei Deng
- Department of General Surgery, Medical Oncology Group, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-Cheng District, 100050, Beijing, China
| | - Xinfeng Wang
- Department of General Surgery, Medical Oncology Group, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong-an Road, Xi-Cheng District, 100050, Beijing, China
| |
Collapse
|
4
|
Lv Y, Pu L, Ran B, Xiang B. Targeting tumor angiogenesis and metabolism with photodynamic nanomedicine. Front Cell Dev Biol 2025; 13:1558393. [PMID: 40235732 PMCID: PMC11996804 DOI: 10.3389/fcell.2025.1558393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/24/2025] [Indexed: 04/17/2025] Open
Abstract
Photodynamic therapy (PDT) holds considerable promise as a tumor treatment modality, characterized by its targeted action, compatibility with other therapeutic approaches, and non - invasive features. PDT can achieve remarkable spatiotemporal precision in tumor ablation through the generation of reactive oxygen species (ROS). Nevertheless, despite its potential in tumor treatment, PDT encounters multiple challenges in practical applications. PDT is highly oxygen - dependent, and thus the effectiveness of PDT can be markedly influenced by tumor hypoxia. The co-existence of abnormal vasculature and metabolic deregulation gives rise to a hypoxic microenvironment, which not only sustains tumor survival but also undermines the therapeutic efficacy of PDT. Consequently, targeting tumor angiogenesis and metabolism is essential for revitalizing PDT. This review emphasizes the mechanisms and strategies for revitalizing PDT in tumor treatment, predominantly concentrating on interfering with tumor angiogenesis and reprogramming tumor cell metabolism. Lastly, the outlining future perspectives and current limitations of PDT are also summarized. This could provide new insights and methodologies for overcoming the challenges associated with PDT in tumor treatment, ultimately advancing the field of PDT.
Collapse
Affiliation(s)
- Yong Lv
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lihui Pu
- Department of Critical Care, West China Hospital, Sichuan University, Chengdu, China
| | - Bei Ran
- School of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Bo Xiang
- Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Ha H, Choi Y, Kim NH, Kim J, Jang J, Niepa THR, Tanaka M, Lee HY, Choi J. Lipid Nanoparticle Delivery System for Normalization of Tumor Microenvironment and Tumor Vascular Structure. Biomater Res 2025; 29:0144. [PMID: 39935791 PMCID: PMC11811622 DOI: 10.34133/bmr.0144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/07/2025] [Accepted: 01/18/2025] [Indexed: 02/13/2025] Open
Abstract
Tumors grow by receiving oxygen and nutrients from the surrounding blood vessels, leading to rapid angiogenesis. This results in functionally and structurally abnormal vasculature characterized by high permeability and irregular blood flow, causing hypoxia within the tumor microenvironment (TME). Hypoxia exacerbates the secretion of pro-angiogenic factors such as vascular endothelial growth factor (VEGF), further perpetuating abnormal vessel formation. This environment compromises the efficacy of radiotherapy, immunotherapy, and chemotherapy. In this study, we developed a pH-sensitive liposome (PSL) system, termed OD_PSL@AKB, to co-deliver oxygen (OD) and razuprotafib (AKB-9778) to tumors. This system rapidly responds to the acidic TME to alleviate hypoxia and inhibit VEGF secretion, restoring VE-cadherin expression in hypoxic endothelial cell/cancer cell cocultures. Our findings highlight the potential of OD_PSL@AKB in normalizing tumor vasculature and improving therapeutic efficacy.
Collapse
Affiliation(s)
- Heejin Ha
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Yonghyun Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
- Department of Chemical Science and Engineering,
Institute of Science Tokyo, Kanagawa 226-8503, Japan
| | - Na-Hyeon Kim
- Department of Chemical Engineering,
Kumoh National Institute of Technology, Gumi 39177, Korea
| | - Jiwon Kim
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Jaehee Jang
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Tagbo H. R. Niepa
- Department of Chemical Engineering,
Carnegie Mellon University, Pittsburgh, PA, USA
- Department of Biomedical Engineering,
Carnegie Mellon University, Pittsburgh, PA, USA
| | - Masayoshi Tanaka
- Department of Chemical Science and Engineering,
Institute of Science Tokyo, Kanagawa 226-8503, Japan
| | - Hee-Young Lee
- Department of Chemical Engineering,
Kumoh National Institute of Technology, Gumi 39177, Korea
| | - Jonghoon Choi
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
- Feynman Institute of Technology, Nanomedicine Corporation, Seoul 06974, Republic of Korea
| |
Collapse
|
6
|
Magdalena JB, Justyna C, Joanna C, Ryszard S, Alina D, Dorota SL, Ewelina P, Sybilla M, Tomasz C. Normalization of tumor vasculature by imiquimod: proposal for a new anticancer therapeutic indication for a TLR7 agonist. Cancer Immunol Immunother 2025; 74:90. [PMID: 39891776 PMCID: PMC11787066 DOI: 10.1007/s00262-025-03943-2] [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: 10/24/2024] [Accepted: 01/08/2025] [Indexed: 02/03/2025]
Abstract
Imiquimod (IMQ), a drug from aminoquinoline group, is the toll-like receptor 7 (TLR7) agonist. It acts as an immunostimulant and radio-sensitizing agent. IMQ stimulates both innate and adaptive immune response. Despite studies conducted, there are no unambiguous data showing how IMQ affects the condition of tumor blood vessels. Tumor vasculature plays the main role in tumor progression. Formation of abnormal blood vessels increases area of hypoxia which recruits suppressor cells, blocks tumor infiltration by CD8+ T lymphocytes, inhibits efficacy of chemoterapeutic drug and leads to cancer relapse. Normalization is a type of therapy targeted at abnormal tumor blood vessels. Here, we demonstrated that 50 µg of IMQ inhibits the growth of melanoma tumors more efficiently, compared to other tested doses and the control group. Dose escalation did not improve the therapeutic antitumor potential of TLR7 agonist. A dose of 50 µg of IMQ most effectively reduced tumor blood vessel density. Imiquimod normalized tumor vasculature both structurally (by reducing vessel tortuosity and increasing pericyte coverage) and functionally (by improving tumor perfusion) in a dose-dependent manner. Hypoxia regions in tumors of treated mice were significantly reduced after IMQ administration. A dose of 50 µg of IMQ had also the greatest impact on the changes in tumor-infiltrating T lymphocytes levels. TLR7 agonist inhibited angiogenesis in treated mice. Functional vascular normalization by IMQ increases the effectiveness of low dose of doxorubicin. Higher dose of IMQ showed worse effects than lower doses including decreased tumor perfusion, increased tumor hypoxia and immunosuppression. This knowledge may help to optimize the combination of the selected IMQ dose with e.g. chemotherapy or radiotherapy to elicit synergistic effect in cancer treatment. To conclude, we outline IMQ repurposing as a vascular normalizing agent. Our research results may contribute to expanding the therapeutic indications for the use of IMQ in anticancer therapy in the future.
Collapse
Affiliation(s)
- Jarosz-Biej Magdalena
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland.
| | - Czapla Justyna
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland
| | - Ciepła Joanna
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland
| | - Smolarczyk Ryszard
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland
| | - Drzyzga Alina
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland
| | - Sprus-Lipka Dorota
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland
| | - Pilny Ewelina
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland
| | - Matuszczak Sybilla
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland
| | - Cichoń Tomasz
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland
| |
Collapse
|
7
|
Kinsey E, Morse MA. Systemic Therapy for Hepatocellular Carcinoma. Clin Liver Dis 2025; 29:105-124. [PMID: 39608951 DOI: 10.1016/j.cld.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Systemic therapy for hepatocellular carcinoma has evolved from sorafenib to now include immune checkpoint blockade, either atezolizumab/bevacizumab or durvalumab/tremelimumab, and soon to include camrelizumab/rivoceranib and nivolumab/ipilimumab. Second-line therapy remains predominantly either a multikinase inhibitor or ramucirumab. Areas of development include testing immune checkpoint-based regimens in the adjuvant setting after surgery, ablation, or transarterial embolization. Also of interest are studies for patients with Child-Pugh B liver function and adding new checkpoint molecules to the current standard platforms.
Collapse
Affiliation(s)
- Emily Kinsey
- Department of Medicine, Division of Hematology, Oncology & Palliative Care, VCU Health, Richmond, VA, USA
| | - Michael A Morse
- Department of Medicine, Division of Medical Oncology, Duke University Health System, Durham, NC, USA.
| |
Collapse
|
8
|
Sweeney A, Langley A, Xavierselvan M, Shethia RT, Solomon P, Arora A, Mallidi S. Vascular regional analysis unveils differential responses to anti-angiogenic therapy in pancreatic xenografts through macroscopic photoacoustic imaging. Theranostics 2025; 15:2649-2671. [PMID: 39990229 PMCID: PMC11840746 DOI: 10.7150/thno.99361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 12/17/2024] [Indexed: 02/25/2025] Open
Abstract
Background: Amongst the various imaging techniques that provide surrogate tumor radiographic indications to aid in planning, monitoring, and predicting outcomes of therapy, ultrasound-guided photoacoustic imaging (US-PAI) is a promising non-ionizing modality based on endogenous blood (hemoglobin) and blood oxygen saturation (StO₂) contrast. Adaptation of US-PAI to the clinical realm requires macroscopic system configurations for adequate depth visualization. Methods: Here we present a vascular regional analysis (VRA) methodology of obtaining areas of low and high vessel density regions within the tumor (LVD and HVD respectively) by frequency domain filtering of macroscopic PA images. In this work, we evaluated the various vascular and oxygenation profiles of different murine xenografts of pancreatic cancer (AsPC-1, MIA PaCa-2, and BxPC-3) that have varying levels of angiogenic potentials and investigated the effects of receptor tyrosine kinase inhibitor (sunitinib) on the tumor microvessel density and StO₂. Results: The administration of sunitinib resulted in transient deoxygenation and reduction in vessel density within 72 h in two (AsPC-1 and MIA PaCa-2) of the three tumor types. Utilizing VRA, the regional change in StO2 (∆StO2) revealed the preferential targeting of sunitinib in LVD regions in only the AsPC-1 tumors. We also identified the presence of vascular normalization (validated through immunohistochemistry) in the sunitinib treated AsPC-1 tumors at day 8 post-treatment where a significant increases in HVD ∆StO2 (~20%) were seen following the 72-hour time point, indicative of improved vessel flow and functionality. Treated AsPC-1 vasculature displayed increased maturity and functionality compared to non-treated tumors on day 8, while these same metrics showed no conclusive evidence of vascular normalization in MIA PaCa-2 or BxPC-3 tumors. Conclusion: Overall, VRA as a tool to monitor treatment response allowed us to identify time points of vascular remodeling, highlighting its ability to provide insights into the tumor microenvironment for sunitinib treatment and other anti-angiogenic therapies.
Collapse
Affiliation(s)
- Allison Sweeney
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Andrew Langley
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Marvin Xavierselvan
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Ronak T. Shethia
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Patrick Solomon
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Aayush Arora
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
| | - Srivalleesha Mallidi
- Department of Biomedical Engineering, Tufts University, Medford, MA, United States
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
9
|
Rachunek-Medved K, Krauß S, Daigeler A, Adams C, Eckert F, Ganser K, Gonzalez-Menendez I, Quintanilla-Martinez L, Kolbenschlag J. Acute remote ischemic conditioning enhances (CD3+)- but not (FoxP3+)-T-cell invasion in the tumor center and increases IL 17 and TNF-alpha expression in a murine melanoma model. Front Immunol 2024; 15:1501885. [PMID: 39650654 PMCID: PMC11621216 DOI: 10.3389/fimmu.2024.1501885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/29/2024] [Indexed: 12/11/2024] Open
Abstract
Introduction Hypoxia can drive tumor progression, suppress anti-tumor immunity, and reduce the effectiveness of radiotherapy and chemotherapy. This study aimed to assess the impact of remote ischemic conditioning (RIC) on tumor oxygenation (sO2) and the anti-tumor immune response. Material and methods Fourteen B16-Ova tumor-bearing C57BL/6N mice received six 5-minute RIC cycles, while another fourteen underwent anesthesia only. Pimonidazole was administered 1.5 hours before sacrifice. Blood flow, sO2, and hemoglobin levels were measured in the non-ischemic hind limb and tumor. Tumor hypoxia was assessed using pimonidazole and CA IX immunohistochemistry, and T cell infiltration by CD3 and FoxP3 staining. Serum levels of 23 cytokines were analyzed using a multiplex immunoassay. Results Isoflurane anesthesia caused a slight intraindividual increase in blood flow (p = 0.07) and sO2 (p = 0.06) of the hind limb and a sole increase in tumor sO2 (p = 0.035), whereas RIC improved sO2 of the tumor in relation to the hind limb (p=0.03). The median of the tumor oxygen saturation reached 51.4% in the control group and 62.7% in the RIC group (p = 0.09), exhibiting a slight tendency towards better oxygenation in the RIC group. Pimonidazole (p=0.24) and CA IX hypoxia score (p=0.48) did not reveal statistically significant differences between the two groups. In RIC-treated tumors, the number of CD3 (p=0.006), but not FoxP3- positive cells (p = 0.84), in the tumor core was significantly higher compared to the control group. In the RIC group, the mean fluorescence intensity (MFI) of IL-17 was significantly higher (p=0.035), and TNF-α was trend-wise higher (p=0.063) compared to the control group. Conclusion Both isoflurane anesthesia and RIC have an impact on microcirculation. The application of RIC counteracted some of the effects of isoflurane, primarily in healthy tissue, and led to a significant improvement in relative tumor tissue oxygenation compared to the non-ischemic hind limb. RIC selectively enhanced immune infiltration within the tumor center, probably by previously activated tumor infiltrating T cells, while having no significant impact on T-regulatory cells. RIC appears to impact the cytokine profile, as indicated by elevated MFIs of TNF-α and IL-17.
Collapse
Affiliation(s)
- Katarzyna Rachunek-Medved
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Sabrina Krauß
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Adrien Daigeler
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Constantin Adams
- Department of Paediatrics, University Hospital Tuebingen, Tuebingen, Germany
| | - Franziska Eckert
- Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany
| | - Katrin Ganser
- Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany
| | - Irene Gonzalez-Menendez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, University Hospital Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, Tuebingen, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, University Hospital Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, Tuebingen, Germany
| | - Jonas Kolbenschlag
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
10
|
Debnath SK, Debnath M, Ghosh A, Srivastava R, Omri A. Targeting Tumor Hypoxia with Nanoparticle-Based Therapies: Challenges, Opportunities, and Clinical Implications. Pharmaceuticals (Basel) 2024; 17:1389. [PMID: 39459028 PMCID: PMC11510357 DOI: 10.3390/ph17101389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Hypoxia is a crucial factor in tumor biology, affecting various solid tumors to different extents. Its influence spans both early and advanced stages of cancer, altering cellular functions and promoting resistance to therapy. Hypoxia reduces the effectiveness of radiotherapy, chemotherapy, and immunotherapy, making it a target for improving therapeutic outcomes. Despite extensive research, gaps persist, necessitating the exploration of new chemical and pharmacological interventions to modulate hypoxia-related pathways. This review discusses the complex pathways involved in hypoxia and the associated pharmacotherapies, highlighting the limitations of current treatments. It emphasizes the potential of nanoparticle-based platforms for delivering anti-hypoxic agents, particularly oxygen (O2), to the tumor microenvironment. Combining anti-hypoxic drugs with conventional cancer therapies shows promise in enhancing remission rates. The intricate relationship between hypoxia and tumor progression necessitates novel therapeutic strategies. Nanoparticle-based delivery systems can significantly improve cancer treatment efficacy by targeting hypoxia-associated pathways. The synergistic effects of combined therapies underscore the importance of multimodal approaches in overcoming hypoxia-mediated resistance. Continued research and innovation in this area hold great potential for advancing cancer therapy and improving patient outcomes.
Collapse
Affiliation(s)
- Sujit Kumar Debnath
- NanoBios Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India; (S.K.D.); (M.D.)
| | - Monalisha Debnath
- NanoBios Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India; (S.K.D.); (M.D.)
| | - Arnab Ghosh
- NanoBios Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India; (S.K.D.); (M.D.)
| | - Rohit Srivastava
- NanoBios Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India; (S.K.D.); (M.D.)
| | - Abdelwahab Omri
- Department of Chemistry and Biochemistry, The Novel Drug and Vaccine Delivery Systems Facility, Laurentian University, Sudbury, ON P3E 2C6, Canada
| |
Collapse
|
11
|
Chen PH, Lee CH, Liaw CC, Liang RT, Khan MAR, Tsai JN, Huang SY, Liu W, Tsai WC. Metachromin C, a marine-derived natural compound, shows potential in antitumor activity. Int J Med Sci 2024; 21:2578-2594. [PMID: 39439453 PMCID: PMC11492879 DOI: 10.7150/ijms.101037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 09/14/2024] [Indexed: 10/25/2024] Open
Abstract
Metachromin C was first isolated from the marine sponge Hippospongia metachromia and has been reported to possess potent cytotoxicity against leukemia cells. However, its antitumor activity and possible mechanisms in pancreatic cancer remain unclear. The effects of Metachromin C on cell viability were estimated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. The compound demonstrated a cytotoxic effect on four pancreatic cancer cell lines (PANC-1, BxPC-3, MiaPaCa-2, and AsPC-1). The significant S phase arrest observed with Metachromin C treatment suggests its impact on DNA replication machinery. Metachromin C might interfere with the binding of Topoisomerase I (TOPO I) to DNA, inhibit TOPO I activity, prevent DNA relaxation, cause DNA damage, and consequently activate the DNA repair pathway. Additionally, anti-migration and anti-invasion abilities of Metachromin C were confirmed using the transwell assay. It also inhibited angiogenesis in human endothelial cells by reducing cell proliferation, migration, and disrupting tube formation. Moreover, Metachromin C dose-dependently inhibited the growth of intersegmental vessels, subintestinal vessels, and the caudal vein plexus in a zebrafish embryo model, confirming its inhibitory effect on new vessel formation in vivo. Taken together, Metachromin C could not only inhibit the growth of pancreatic cancer cells but also act as an anti-angiogenic compound simultaneously.
Collapse
Affiliation(s)
- Pei-Hsuan Chen
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Che-Hsin Lee
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Aerosol Science Research Center, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- College of Semiconductor and Advanced Technology Research, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
| | - Chih-Chuang Liaw
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Rei-Ting Liang
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Mo Aqib Raza Khan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Jen-Ning Tsai
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung 402, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Shin-Yi Huang
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Wangta Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Wan-Chi Tsai
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| |
Collapse
|
12
|
Coursier D, Calvo F. CAFs vs. TECs: when blood feuds fuel cancer progression, dissemination and therapeutic resistance. Cell Oncol (Dordr) 2024; 47:1091-1112. [PMID: 38453816 PMCID: PMC11322395 DOI: 10.1007/s13402-024-00931-z] [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] [Accepted: 02/23/2024] [Indexed: 03/09/2024] Open
Abstract
Neoplastic progression involves complex interactions between cancer cells and the surrounding stromal milieu, fostering microenvironments that crucially drive tumor progression and dissemination. Of these stromal constituents, cancer-associated fibroblasts (CAFs) emerge as predominant inhabitants within the tumor microenvironment (TME), actively shaping multiple facets of tumorigenesis, including cancer cell proliferation, invasiveness, and immune evasion. Notably, CAFs also orchestrate the production of pro-angiogenic factors, fueling neovascularization to sustain the metabolic demands of proliferating cancer cells. Moreover, CAFs may also directly or indirectly affect endothelial cell behavior and vascular architecture, which may impact in tumor progression and responses to anti-cancer interventions. Conversely, tumor endothelial cells (TECs) exhibit a corrupted state that has been shown to affect cancer cell growth and inflammation. Both CAFs and TECs are emerging as pivotal regulators of the TME, engaging in multifaceted biological processes that significantly impact cancer progression, dissemination, and therapeutic responses. Yet, the intricate interplay between these stromal components and the orchestrated functions of each cell type remains incompletely elucidated. In this review, we summarize the current understanding of the dynamic interrelationships between CAFs and TECs, discussing the challenges and prospects for leveraging their interactions towards therapeutic advancements in cancer.
Collapse
Affiliation(s)
- Diane Coursier
- Instituto de Biomedicina y Biotecnología de Cantabria (Consejo Superior de Investigaciones Científicas, Universidad de Cantabria), Santander, Spain
| | - Fernando Calvo
- Instituto de Biomedicina y Biotecnología de Cantabria (Consejo Superior de Investigaciones Científicas, Universidad de Cantabria), Santander, Spain.
| |
Collapse
|
13
|
Llorente A, Brokāne A, Mlynska A, Puurand M, Sagini K, Folkmane S, Hjorth M, Martin‐Gracia B, Romero S, Skorinkina D, Čampa M, Cešeiko R, Romanchikova N, Kļaviņa A, Käämbre T, Linē A. From sweat to hope: The role of exercise-induced extracellular vesicles in cancer prevention and treatment. J Extracell Vesicles 2024; 13:e12500. [PMID: 39183543 PMCID: PMC11345496 DOI: 10.1002/jev2.12500] [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: 04/29/2024] [Revised: 07/03/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024] Open
Abstract
The benefits of regular physical exercise on cancer prevention, as well as reducing fatigue, treatment side effects and recurrence, and improving quality of life and overall survival of cancer patients, are increasingly recognised. Initial studies showed that the concentration of extracellular vesicles (EVs) increases during physical activity and that EVs carry biologically active cargo. These EVs are released by blood cells, skeletal muscle and other organs involved in exercise, thus suggesting that EVs may mediate tissue crosstalk during exercise. This possibility triggered a great interest in the study of the roles of EVs in systemic adaptation to exercise and in their potential applications in the prevention and treatment of various diseases, including cancer. This review presents studies exploring the concentration and molecular cargo of EVs released during exercise. Furthermore, we discuss putative stimuli that may trigger EV release from various cell types, the biological functions and the impact of exercise-induced EVs on cancer development and progression. Understanding the interplay between exercise, EVs, and cancer biology may offer insights into novel therapeutic strategies and preventive measures for cancer.
Collapse
Affiliation(s)
- Alicia Llorente
- Department of Molecular Cell Biology, Institute for Cancer ResearchOslo University HospitalOsloNorway
- Centre for Cancer Cell Reprogramming, Faculty of MedicineUniversity of OsloOsloNorway
- Department for Mechanical, Electronics and Chemical EngineeringOslo Metropolitan UniversityOsloNorway
| | - Agnese Brokāne
- Cancer Biomarker groupLatvian Biomedical Research and Study CentreRigaLatvia
| | - Agata Mlynska
- Laboratory of ImmunologyNational Cancer InstituteVilniusLithuania
- Department of Chemistry and BioengineeringVilnius Gediminas Technical UniversityVilniusLithuania
| | - Marju Puurand
- Laboratory of Chemical BiologyNational Institute of Chemical Physics and BiophysicsTallinnEstonia
| | - Krizia Sagini
- Department of Molecular Cell Biology, Institute for Cancer ResearchOslo University HospitalOsloNorway
- Centre for Cancer Cell Reprogramming, Faculty of MedicineUniversity of OsloOsloNorway
| | - Signe Folkmane
- Cancer Biomarker groupLatvian Biomedical Research and Study CentreRigaLatvia
| | - Marit Hjorth
- Department of Nutrition, Institute of Basic Medical SciencesUniversity of OsloOsloNorway
| | - Beatriz Martin‐Gracia
- Department of Molecular Cell Biology, Institute for Cancer ResearchOslo University HospitalOsloNorway
- Centre for Cancer Cell Reprogramming, Faculty of MedicineUniversity of OsloOsloNorway
| | - Silvana Romero
- Department of Molecular Cell Biology, Institute for Cancer ResearchOslo University HospitalOsloNorway
- Centre for Cancer Cell Reprogramming, Faculty of MedicineUniversity of OsloOsloNorway
| | - Diana Skorinkina
- Cancer Biomarker groupLatvian Biomedical Research and Study CentreRigaLatvia
| | - Mārtiņš Čampa
- Latvian Academy of Sport Education, Riga Stradins UniversityRigaLatvia
| | - Rūdolfs Cešeiko
- Latvian Academy of Sport Education, Riga Stradins UniversityRigaLatvia
| | | | - Aija Kļaviņa
- Latvian Academy of Sport Education, Riga Stradins UniversityRigaLatvia
- Department of Health Promotion and RehabilitationLithuanian Sports UniversityKaunasLithuania
| | - Tuuli Käämbre
- Laboratory of Chemical BiologyNational Institute of Chemical Physics and BiophysicsTallinnEstonia
| | - Aija Linē
- Cancer Biomarker groupLatvian Biomedical Research and Study CentreRigaLatvia
| |
Collapse
|
14
|
Chen Y, Zhang Z, Pan F, Li P, Yao W, Chen Y, Xiong L, Wang T, Li Y, Huang G. Pericytes recruited by CCL28 promote vascular normalization after anti-angiogenesis therapy through RA/RXRA/ANGPT1 pathway in lung adenocarcinoma. J Exp Clin Cancer Res 2024; 43:210. [PMID: 39075504 PMCID: PMC11285179 DOI: 10.1186/s13046-024-03135-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/22/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND It has been proposed that anti-angiogenesis therapy could induce tumor "vascular normalization" and further enhance the efficacy of chemotherapy, radiotherapy, target therapy, and immunotherapy for nearly twenty years. However, the detailed molecular mechanism of this phenomenon is still obscure. METHOD Overexpression and knockout of CCL28 in human lung adenocarcinoma cell line A549 and murine lung adenocarcinoma cell line LLC, respectively, were utilized to establish mouse models. Single-cell sequencing was performed to analyze the proportion of different cell clusters and metabolic changes in the tumor microenvironment (TME). Immunofluorescence and multiplex immunohistochemistry were conducted in murine tumor tissues and clinical biopsy samples to assess the percentage of pericytes coverage. Primary pericytes were isolated from lung adenocarcinoma tumor tissues using magnetic-activated cell sorting (MACS). These pericytes were then treated with recombinant human CCL28 protein, followed by transwell migration assays and RNA sequencing analysis. Changes in the secretome and metabolome were examined, and verification of retinoic acid metabolism alterations in pericytes was conducted using quantitative real-time PCR, western blotting, and LC-MS technology. Chromatin immunoprecipitation followed by quantitative PCR (ChIP-qPCR) was employed to validate the transcriptional regulatory ability and affinity of RXRα to specific sites at the ANGPT1 promoter. RESULTS Our study showed that after undergoing anti-angiogenesis treatment, the tumor exhibited a state of ischemia and hypoxia, leading to an upregulation in the expression of CCL28 in hypoxic lung adenocarcinoma cells by the hypoxia-sensitive transcription factor CEBPB. Increased CCL28 could promote tumor vascular normalization through recruiting and metabolic reprogramming pericytes in the tumor microenvironment. Mechanistically, CCL28 modified the retinoic acid (RA) metabolism and increased ANGPT1 expression via RXRα in pericytes, thereby enhancing the stability of endothelial cells. CONCLUSION We reported the details of the molecular mechanisms of "vascular normalization" after anti-angiogenesis therapy for the first time. Our work might provide a prospective molecular marker for guiding the clinical arrangement of combination therapy between anti-angiogenesis treatment and other therapies.
Collapse
Affiliation(s)
- Ying Chen
- The State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
- Medical Schoolof, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Zhiyong Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
- Medical Schoolof, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Fan Pan
- Medical Schoolof, Nanjing University, Nanjing, Jiangsu, 210093, China
- Department of Medical Oncology, Affiliated Hospital of Medical School, Jinling Hospital, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Pengfei Li
- The State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
- Medical Schoolof, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Weiping Yao
- Medical Schoolof, Nanjing University, Nanjing, Jiangsu, 210093, China
- Department of Medical Oncology, Affiliated Hospital of Medical School, Jinling Hospital, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Yuxi Chen
- The State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China
- Medical Schoolof, Nanjing University, Nanjing, Jiangsu, 210093, China
| | - Lei Xiong
- Department of Cardio-Thoracic Surgery, Affiliated Hospital of Medical School, Jinling Hospital, Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Tingting Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Chemistry and Biomedicine Innovation Center (ChemBIC), Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, China.
- Medical Schoolof, Nanjing University, Nanjing, Jiangsu, 210093, China.
| | - Yan Li
- Department of Respiratory Critical Care Medicine, Affiliated Hospital of Medical School, Nanjing Drum Tower Hospital, Nanjing University, Nanjing, Jiangsu, 210008, China.
| | - Guichun Huang
- Department of Medical Oncology, Affiliated Hospital of Medical School, Jinling Hospital, Nanjing University, Nanjing, Jiangsu, 210008, China.
- Department of Oncology, Medical School, Zhongda Hospital, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
15
|
Moro M, Balestrero FC, Grolla AA. Pericytes: jack-of-all-trades in cancer-related inflammation. Front Pharmacol 2024; 15:1426033. [PMID: 39086395 PMCID: PMC11288921 DOI: 10.3389/fphar.2024.1426033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/25/2024] [Indexed: 08/02/2024] Open
Abstract
Pericytes, recognized as mural cells, have long been described as components involved in blood vessel formation, playing a mere supporting role for endothelial cells (ECs). Emerging evidence strongly suggests their multifaceted roles in tissues and organs. Indeed, pericytes exhibit a remarkable ability to anticipate endothelial cell behavior and adapt their functions based on the specific cells they interact with. Pericytes can be activated by pro-inflammatory stimuli and crosstalk with immune cells, actively participating in their transmigration into blood vessels. Moreover, they can influence the immune response, often sustaining an immunosuppressive phenotype in most of the cancer types studied. In this review, we concentrate on the intricate crosstalk between pericytes and immune cells in cancer, highlighting the primary evidence regarding pericyte involvement in primary tumor mass dynamics, their contributions to tumor reprogramming for invasion and migration of malignant cells, and their role in the formation of pre-metastatic niches. Finally, we explored recent and emerging pharmacological approaches aimed at vascular normalization, including novel strategies to enhance the efficacy of immunotherapy through combined use with anti-angiogenic drugs.
Collapse
Affiliation(s)
| | | | - Ambra A. Grolla
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Novara, Italy
| |
Collapse
|
16
|
Sweeney A, Xavierselvan M, Langley A, Solomon P, Arora A, Mallidi S. Vascular regional analysis unveils differential responses to anti-angiogenic therapy in pancreatic xenografts through macroscopic photoacoustic imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.27.595784. [PMID: 38854042 PMCID: PMC11160648 DOI: 10.1101/2024.05.27.595784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Pancreatic cancer (PC) is a highly lethal malignancy and the third leading cause of cancer deaths in the U.S. Despite major innovations in imaging technologies, there are limited surrogate radiographic indicators to aid in therapy planning and monitoring. Amongst the various imaging techniques Ultrasound-guided photoacoustic imaging (US-PAI) is a promising modality based on endogenous blood (hemoglobin) and blood oxygen saturation (StO 2 ) contrast to monitor response to anti-angiogenic therapies. Adaptation of US-PAI to the clinical realm requires macroscopic configurations for adequate depth visualization, illuminating the need for surrogate radiographic markers, including the tumoral microvessel density (MVD). In this work, subcutaneous xenografts with PC cell lines AsPC-1 and MIA-PaCa-2 were used to investigate the effects of receptor tyrosine kinase inhibitor (sunitinib) treatment on MVD and StO 2 . Through histological correlation, we have shown that regions of high and low vascular density (HVD and LVD) can be identified through frequency domain filtering of macroscopic PA images which could not be garnered from purely global analysis. We utilized vascular regional analysis (VRA) of treatment-induced StO 2 and total hemoglobin (HbT) changes. VRA as a tool to monitor treatment response allowed us to identify potential timepoints of vascular remodeling, highlighting its ability to provide insights into the TME not only for sunitinib treatment but also other anti-angiogenic therapies.
Collapse
|
17
|
Zuo T, Li X, Ma X, Zhang Y, Li X, Fan X, Gao M, Xia D, Cheng H. Engineering tumor-oxygenated nanomaterials: advancing photodynamic therapy for cancer treatment. Front Bioeng Biotechnol 2024; 12:1383930. [PMID: 38544975 PMCID: PMC10965730 DOI: 10.3389/fbioe.2024.1383930] [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: 02/08/2024] [Accepted: 02/27/2024] [Indexed: 11/11/2024] Open
Abstract
Photodynamic therapy (PDT), a promising treatment modality, employs photosensitizers to generate cytotoxic reactive oxygen species (ROS) within localized tumor regions. This technique involves administering a photosensitizer followed by light activation in the presence of oxygen (O2), resulting in cytotoxic ROS production. PDT's spatiotemporal selectivity, minimally invasive nature, and compatibility with other treatment modalities make it a compelling therapeutic approach. However, hypoxic tumor microenvironment (TME) poses a significant challenge to conventional PDT. To overcome this hurdle, various strategies have been devised, including in-situ O2 generation, targeted O2 delivery, tumor vasculature normalization, modulation of mitochondrial respiration, and photocatalytic O2 generation. This review aims to provide a comprehensive overview of recent developments in designing tumor-oxygenated nanomaterials to enhance PDT efficacy. Furthermore, we delineate ongoing challenges and propose strategies to improve PDT's clinical impact in cancer treatment.
Collapse
Affiliation(s)
- Tingting Zuo
- College of Biological Sciences and Technology, Yili Normal University, Yining, China
| | - Xiaodie Li
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xuan Ma
- No. 1 Traditional Chinese Medicine Hospital in Changde, Changde, China
| | - Ye Zhang
- College of Biological Sciences and Technology, Yili Normal University, Yining, China
| | - Xueru Li
- College of Biological Sciences and Technology, Yili Normal University, Yining, China
| | - Xuehai Fan
- College of Biological Sciences and Technology, Yili Normal University, Yining, China
| | - Mingze Gao
- No. 1 Traditional Chinese Medicine Hospital in Changde, Changde, China
| | - Donglin Xia
- School of Public Health of Nantong University, Nantong, China
| | - Huijun Cheng
- College of Biological Sciences and Technology, Yili Normal University, Yining, China
- Xinjiang Key Laboratory of Lavender Conservation and Utilization, Yining, China
| |
Collapse
|
18
|
Żak K, Satora M, Skrabalak I, Tarkowski R, Ostrowska-Leśko M, Bobiński M. The Potential Influence of Residual or Recurrent Disease on Bevacizumab Treatment Efficacy in Ovarian Cancer: Current Evidence and Future Perspectives. Cancers (Basel) 2024; 16:1063. [PMID: 38473419 DOI: 10.3390/cancers16051063] [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: 02/03/2024] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
There were high hopes for the new antiangiogenic medicament, bevacizumab, which could inhibit the creation of new blood vessels through binding to isoform A of vascular endothelial growth factor (VEGF). However, it is not only blood vessels that are responsible for tumor cell spread. During the process of tumor growth, lymphangiogenesis is mediated by other members of the VEGF family, specifically VEGF-C and VEGF-D, which act independent to bevacizumab. Therefore, based on the mechanism of bevacizumab action and the processes of angio- and lymphangiogenesis, we formed three hypotheses: (1) if the lymph nodes in primary ovarian cancers are metastatic, the outcome of bevacizumab treatment is worsened; (2) concerning the second-line treatment, bevacizumab will act in a weakened manner if recurrence occurs in lymph nodes as opposed to a local recurrence; (3) patients treated by bevacizumab are more likely to have recurrences in lymph nodes. These hypotheses raise the issue of the existing knowledge gap, which concerns the effect of bevacizumab on metastatic lymph nodes.
Collapse
Affiliation(s)
- Klaudia Żak
- Department of Medical Chemistry, Medical University of Lublin, 20-059 Lublin, Poland
| | - Małgorzata Satora
- I Chair and Department of Oncological Gynaecology and Gynaecology, Student Scientific Association, Medical University of Lublin, 20-059 Lublin, Poland
| | - Ilona Skrabalak
- I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Rafał Tarkowski
- I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Marta Ostrowska-Leśko
- Chair and Department of Toxicology, Medical University of Lublin, 20-090 Lublin, Poland
| | - Marcin Bobiński
- I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-059 Lublin, Poland
| |
Collapse
|
19
|
Fu Y, Sun S, Shi D, Bi J. Construction of endothelial cell signatures for predicting the diagnosis, prognosis and immunotherapy response of bladder cancer via machine learning. J Cell Mol Med 2024; 28:e18155. [PMID: 38429911 PMCID: PMC10907833 DOI: 10.1111/jcmm.18155] [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: 05/11/2023] [Revised: 01/08/2024] [Accepted: 01/16/2024] [Indexed: 03/03/2024] Open
Abstract
We subtyped bladder cancer (BC) patients based on the expression patterns of endothelial cell (EC) -related genes and constructed a diagnostic signature and an endothelial cell prognostic index (ECPI), which are useful for diagnosing BC patients, predicting the prognosis of BC and evaluating drug sensitivity. Differentially expressed genes in ECs were obtained from the Tumour Immune Single-Cell Hub database. Subsequently, a diagnostic signature, a tumour subtyping system and an ECPI were constructed using data from The Cancer Genome Atlas and Gene Expression Omnibus. Associations between the ECPI and the tumour microenvironment, drug sensitivity and biofunctions were assessed. The hub genes in the ECPI were identified as drug candidates by molecular docking. Subtype identification indicated that high EC levels were associated with a worse prognosis and immunosuppressive effect. The diagnostic signature and ECPI were used to effectively diagnose BC and accurately assess the prognosis of BC and drug sensitivity among patients. Three hub genes in the ECPI were extracted, and the three genes had the closest affinity for doxorubicin and curcumin. There was a close relationship between EC and BC. EC-related genes can help clinicians diagnose BC, predict the prognosis of BC and select effective drugs.
Collapse
Affiliation(s)
- Yang Fu
- Department of UrologyThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Shanshan Sun
- Department of PharmacyThe People's Hospital of Liaoning ProvinceShenyangLiaoningChina
| | - Du Shi
- Department of UrologyThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Jianbin Bi
- Department of UrologyThe First Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
20
|
Sarma K, Akther MH, Ahmad I, Afzal O, Altamimi ASA, Alossaimi MA, Jaremko M, Emwas AH, Gautam P. Adjuvant Novel Nanocarrier-Based Targeted Therapy for Lung Cancer. Molecules 2024; 29:1076. [PMID: 38474590 DOI: 10.3390/molecules29051076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 03/14/2024] Open
Abstract
Lung cancer has the lowest survival rate due to its late-stage diagnosis, poor prognosis, and intra-tumoral heterogeneity. These factors decrease the effectiveness of treatment. They release chemokines and cytokines from the tumor microenvironment (TME). To improve the effectiveness of treatment, researchers emphasize personalized adjuvant therapies along with conventional ones. Targeted chemotherapeutic drug delivery systems and specific pathway-blocking agents using nanocarriers are a few of them. This study explored the nanocarrier roles and strategies to improve the treatment profile's effectiveness by striving for TME. A biofunctionalized nanocarrier stimulates biosystem interaction, cellular uptake, immune system escape, and vascular changes for penetration into the TME. Inorganic metal compounds scavenge reactive oxygen species (ROS) through their photothermal effect. Stroma, hypoxia, pH, and immunity-modulating agents conjugated or modified nanocarriers co-administered with pathway-blocking or condition-modulating agents can regulate extracellular matrix (ECM), Cancer-associated fibroblasts (CAF),Tyro3, Axl, and Mertk receptors (TAM) regulation, regulatory T-cell (Treg) inhibition, and myeloid-derived suppressor cells (MDSC) inhibition. Again, biomimetic conjugation or the surface modification of nanocarriers using ligands can enhance active targeting efficacy by bypassing the TME. A carrier system with biofunctionalized inorganic metal compounds and organic compound complex-loaded drugs is convenient for NSCLC-targeted therapy.
Collapse
Affiliation(s)
- Kangkan Sarma
- School of Pharmaceutical and Population Health Informatics (SoPPHI), DIT University, Dehradun 248009, India
| | - Md Habban Akther
- School of Pharmaceutical and Population Health Informatics (SoPPHI), DIT University, Dehradun 248009, India
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 62521, Saudi Arabia
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Manal A Alossaimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mariusz Jaremko
- Smart-Health Initiative (SHI) and Red Sea Research Center (RSRC), Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| | - Abdul-Hamid Emwas
- Core Labs, King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia
| | - Preety Gautam
- School of Pharmaceutical and Population Health Informatics (SoPPHI), DIT University, Dehradun 248009, India
| |
Collapse
|
21
|
Nys N, Khatib AM, Siegfried G. Apela promotes blood vessel regeneration and remodeling in zebrafish. Sci Rep 2024; 14:3718. [PMID: 38355946 PMCID: PMC10867005 DOI: 10.1038/s41598-023-50677-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 12/22/2023] [Indexed: 02/16/2024] Open
Abstract
In contrast to adult mammals, zebrafish display a high capacity to heal injuries and repair damage to various organs. One of the earliest responses to injury in adult zebrafish is revascularization, followed by tissue morphogenesis. Tissue vascularization entails the formation of a blood vessel plexus that remodels into arteries and veins. The mechanisms that coordinate these processes during vessel regeneration are poorly understood. Hence, investigating and identifying the factors that promote revascularization and vessel remodeling have great therapeutic potential. Here, we revealed that fin vessel remodeling critically depends on Apela peptide. We found that Apela selectively accumulated in newly formed zebrafish fin tissue and vessels. The temporal expression of Apela, Apln, and their receptor Aplnr is different during the regenerative process. While morpholino-mediated knockdown of Apela (Mo-Apela) prevented vessel remodeling, exogenous Apela peptide mediated plexus repression and the development of arteries in regenerated fins. In contrast, Apela enhanced subintestinal venous plexus formation (SIVP). The use of sunitinib completely inhibited vascular plexus formation in zebrafish, which was not prevented by exogenous application. Furthermore, Apela regulates the expression of vessel remolding-related genes including VWF, IGFPB3, ESM1, VEGFR2, Apln, and Aplnr, thereby linking Apela to the vascular plexus factor network as generated by the STRING online database. Together, our findings reveal a new role for Apela in vessel regeneration and remodeling in fin zebrafish and provide a framework for further understanding the cellular and molecular mechanisms involved in vessel regeneration.
Collapse
Affiliation(s)
- Nicolas Nys
- RYTME, Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, University of Bordeaux, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615, Pessac, France
| | - Abdel-Majid Khatib
- RYTME, Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, University of Bordeaux, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615, Pessac, France.
- ZebraFish, Research and Technology, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615, Pessac, France.
- Bergonié Institute, Bordeaux, France.
| | - Geraldine Siegfried
- RYTME, Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, University of Bordeaux, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615, Pessac, France.
- ZebraFish, Research and Technology, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615, Pessac, France.
| |
Collapse
|
22
|
Bokhari SZ, Aloss K, Leroy Viana PH, Schvarcz CA, Besztercei B, Giunashvili N, Bócsi D, Koós Z, Balogh A, Benyó Z, Hamar P. Digoxin-Mediated Inhibition of Potential Hypoxia-Related Angiogenic Repair in Modulated Electro-Hyperthermia (mEHT)-Treated Murine Triple-Negative Breast Cancer Model. ACS Pharmacol Transl Sci 2024; 7:456-466. [PMID: 38357275 PMCID: PMC10863435 DOI: 10.1021/acsptsci.3c00296] [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: 10/23/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 02/16/2024]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer type with no targeted therapy and hence limited treatment options. Modulated electrohyperthermia (mEHT) is a novel complementary therapy where a 13.56 MHz radiofrequency current targets cancer cells selectively, inducing tumor damage by thermal and electromagnetic effects. We observed severe vascular damage in mEHT-treated tumors and investigated the potential synergism between mEHT and inhibition of tumor vasculature recovery in our TNBC mouse model. 4T1/4T07 isografts were orthotopically inoculated and treated three to five times with mEHT. mEHT induced vascular damage 4-12 h after treatment, leading to tissue hypoxia detected at 24 h. Hypoxia in treated tumors induced an angiogenic recovery 24 h after the last treatment. Administration of the cardiac glycoside digoxin with the potential hypoxia-inducible factor 1-α (HIF1-α) and angiogenesis inhibitory effects could synergistically augment mEHT-mediated tumor damage and reduce tissue hypoxia signaling and consequent vascular recovery in mEHT-treated TNBC tumors. Conclusively, repeated mEHT induced vascular damage and hypoxic stress in TNBC that promoted vascular recovery. Inhibiting this hypoxic stress signaling enhanced the effectiveness of mEHT and may potentially enhance other forms of cancer treatment.
Collapse
Affiliation(s)
| | - Kenan Aloss
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | | | - Csaba András Schvarcz
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
- Cerebrovascular
and Neurocognitive Disorders Research Group, Eötvös, Loránd Research Network and Semmelweis
University (ELKH-SE), Tűzoltó utca 37-47, Budapest 1094, Hungary
| | - Balázs Besztercei
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Nino Giunashvili
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Dániel Bócsi
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Zoltán Koós
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Andrea Balogh
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Zoltán Benyó
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| | - Péter Hamar
- Institute
of Translational Medicine, Semmelweis University, Üllői út 26, Budapest 1085, Hungary
| |
Collapse
|
23
|
Yin J, Dong F, An J, Guo T, Cheng H, Zhang J, Zhang J. Pattern recognition of microcirculation with super-resolution ultrasound imaging provides markers for early tumor response to anti-angiogenic therapy. Theranostics 2024; 14:1312-1324. [PMID: 38323316 PMCID: PMC10845201 DOI: 10.7150/thno.89306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 12/28/2023] [Indexed: 02/08/2024] Open
Abstract
Rationale: Cancer treatment outcome is traditionally evaluated by tumor volume change in clinics, while tumor microvascular heterogeneity reflecting tumor response has not been fully explored due to technical limitations. Methods: We introduce a new paradigm in super-resolution ultrasound imaging, termed pattern recognition of microcirculation (PARM), which identifies both hemodynamic and morphological patterns of tumor microcirculation hidden in spatio-temporal space trajectories of microbubbles. Results: PARM demonstrates the ability to distinguish different local blood flow velocities separated by a distance of 24 μm. Compared with traditional vascular parameters, PARM-derived heterogeneity parameters prove to be more sensitive to microvascular changes following anti-angiogenic therapy. Particularly, PARM-identified "sentinel" microvasculature, exhibiting evident structural changes as early as 24 hours after treatment initiation, correlates significantly with subsequent tumor volume changes (|r| > 0.9, P < 0.05). This provides prognostic insight into tumor response much earlier than clinical criteria. Conclusions: The ability of PARM to noninvasively quantify tumor vascular heterogeneity at the microvascular level may shed new light on early-stage assessment of cancer therapy.
Collapse
Affiliation(s)
- Jingyi Yin
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Feihong Dong
- College of Future Technology, Peking University, Beijing, China
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Center for Life Sciences, and Institute of Molecular Medicine, Peking University, Beijing, China
- National Biomedical Imaging Center, Peking University, Beijing, China
| | - Jian An
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Tianyu Guo
- College of Future Technology, Peking University, Beijing, China
| | - Heping Cheng
- College of Future Technology, Peking University, Beijing, China
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Center for Life Sciences, and Institute of Molecular Medicine, Peking University, Beijing, China
- National Biomedical Imaging Center, Peking University, Beijing, China
- Research Unit of Mitochondria in Brain Diseases, Chinese Academy of Medical Sciences, PKU-Nanjing Institute of Translational Medicine, Nanjing, China
| | - Jiabin Zhang
- College of Future Technology, Peking University, Beijing, China
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Center for Life Sciences, and Institute of Molecular Medicine, Peking University, Beijing, China
- National Biomedical Imaging Center, Peking University, Beijing, China
| | - Jue Zhang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- National Biomedical Imaging Center, Peking University, Beijing, China
- Research Unit of Mitochondria in Brain Diseases, Chinese Academy of Medical Sciences, PKU-Nanjing Institute of Translational Medicine, Nanjing, China
- College of Engineering, Peking University, Beijing, China
| |
Collapse
|
24
|
Zeng Y, Zhang S, Li S, Song G, Meng T, Yuan H, Hu F. Normalizing Tumor Blood Vessels to Improve Chemotherapy and Inhibit Breast Cancer Metastasis by Multifunctional Nanoparticles. Mol Pharm 2023; 20:5078-5089. [PMID: 37728215 DOI: 10.1021/acs.molpharmaceut.3c00381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
The abnormal tumor blood vessels with high leakage can promote tumor cells to infiltrate into the systemic circulation and increase the risk of tumor metastasis. In addition, chemotherapy may destroy tumor blood vessels and further aggravate metastasis. Normalizing tumor blood vessels can reduce vascular leakage and increase vascular integrity. The simultaneous administration of vascular normalization drugs and chemotherapy drugs may resist the blood vessels' destruction of chemotherapy. Here, multifunctional nanoparticles (CCM@LMSN/DOX&St), which combined chemotherapy with tumor blood vessel normalization, were prepared for the treatment of breast cancer. The results showed that CCM@LMSN/DOX&St-loaded sunitinib (St) promoted the expression of junction proteins Claudin-4 and VE-cadherin of endothelial cells, reversed the destruction of DOX to the endothelial cell layer, protected the integrity of the endothelial cell layer, and inhibited the migration of 4T1 tumor cells across the endothelial cell layer. In vivo experiments showed that CCM@LMSN/DOX&St effectively inhibited tumor growth in situ; what is exciting was that it also inhibited distal metastasis of breast cancer. CCM@LMSN/DOX&St encapsulated with St can normalize tumor blood vessels, reverse the damage of DOX to tumor blood vessels, increase the integrity of blood vessels, and prevent tumor cell invasion into blood vessels, which can inhibit breast cancer spontaneous metastasis and reduce chemotherapy-induced metastasis. This drug delivery platform effectively inhibited the progression of tumors and provided a promising solution for effective tumor treatment.
Collapse
Affiliation(s)
- Yingping Zeng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Shufen Zhang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Sufen Li
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Guangtao Song
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Tingting Meng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| |
Collapse
|
25
|
Mo T, Brandal SHB, Geier OM, Engebråten O, Nilsen LB, Kristensen VN, Hole KH, Hompland T, Fleischer T, Seierstad T. MRI Assessment of Changes in Tumor Vascularization during Neoadjuvant Anti-Angiogenic Treatment in Locally Advanced Breast Cancer Patients. Cancers (Basel) 2023; 15:4662. [PMID: 37760629 PMCID: PMC10526130 DOI: 10.3390/cancers15184662] [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: 08/02/2023] [Revised: 09/16/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023] Open
Abstract
Anti-VEGF (vascular endothelial growth factor) treatment improves response rates, but not progression-free or overall survival in advanced breast cancer. It has been suggested that subgroups of patients may benefit from this treatment; however, the effects of adding anti-VEGF treatment to a standard chemotherapy regimen in breast cancer patients are not well studied. Understanding the effects of the anti-vascular treatment on tumor vasculature may provide a selection of patients that can benefit. The aim of this study was to study the vascular effect of bevacizumab using clinical dynamic contrast-enhanced MRI (DCE-MRI). A total of 70 women were randomized to receive either chemotherapy alone or chemotherapy with bevacizumab for 25 weeks. DCE-MRI was performed at baseline and at 12 and 25 weeks, and in addition 25 of 70 patients agreed to participate in an early MRI after one week. Voxel-wise pharmacokinetic analysis was performed using semi-quantitative methods and the extended Tofts model. Vascular architecture was assessed by calculating the fractal dimension of the contrast-enhanced images. Changes during treatment were compared with baseline and between the treatment groups. There was no significant difference in tumor volume at any point; however, DCE-MRI parameters revealed differences in vascular function and vessel architecture. Adding bevacizumab to chemotherapy led to a pronounced reduction in vascular DCE-MRI parameters, indicating decreased vascularity. At 12 and 25 weeks, the difference between the treatment groups is severely reduced.
Collapse
Affiliation(s)
- Torgeir Mo
- Faculty of Clinical Medicine, University of Oslo, 0316 Oslo, Norway; (T.M.); (S.H.B.B.); (O.E.); (V.N.K.); (K.H.H.)
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 4950 Oslo, Norway;
| | - Siri Helene Bertelsen Brandal
- Faculty of Clinical Medicine, University of Oslo, 0316 Oslo, Norway; (T.M.); (S.H.B.B.); (O.E.); (V.N.K.); (K.H.H.)
- Department of Breast Diagnostic, Oslo University Hospital, 0379 Oslo, Norway
| | - Oliver Marcel Geier
- Department of Diagnostic Physics, Oslo University Hospital, 0379 Oslo, Norway;
| | - Olav Engebråten
- Faculty of Clinical Medicine, University of Oslo, 0316 Oslo, Norway; (T.M.); (S.H.B.B.); (O.E.); (V.N.K.); (K.H.H.)
- Department of Oncology, Oslo University Hospital, 0379 Oslo, Norway
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, 4950 Oslo, Norway
| | | | - Vessela N. Kristensen
- Faculty of Clinical Medicine, University of Oslo, 0316 Oslo, Norway; (T.M.); (S.H.B.B.); (O.E.); (V.N.K.); (K.H.H.)
- Department of Medical Genetics, Oslo University Hospital, 0450 Oslo, Norway
| | - Knut Håkon Hole
- Faculty of Clinical Medicine, University of Oslo, 0316 Oslo, Norway; (T.M.); (S.H.B.B.); (O.E.); (V.N.K.); (K.H.H.)
- Department of Oncologic Radiology, Division of Radiology and Nuclear Medicine, Oslo University Hospital, 0379 Oslo, Norway
| | - Tord Hompland
- Department of Radiation Biology, Oslo University Hospital, 4950 Oslo, Norway;
| | - Thomas Fleischer
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, 4950 Oslo, Norway;
| | - Therese Seierstad
- Department of Research and Development, Division for Radiology and Nuclear Medicine, Oslo University Hospital, 0379 Oslo, Norway
| |
Collapse
|
26
|
Zhong J, Xiao C, Chen Q, Pan X, Xu T, Wang Y, Hou W, Liu L, Cao F, Wang Y, Li X, Zhou L, Yang H, Yang Y, Zhao C. Zebrafish functional xenograft vasculature platform identifies PF-502 as a durable vasculature normalization drug. iScience 2023; 26:107734. [PMID: 37680473 PMCID: PMC10480778 DOI: 10.1016/j.isci.2023.107734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/21/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023] Open
Abstract
Tumor vasculature often exhibits disorder and inefficiency. Vascular normalization offers potential for alleviating hypoxia and optimizing drug delivery in tumors. However, identifying effective agents is hindered by a lack of robust screening. We aimed to establish a comprehensive method using the zebrafish functional xenograft vasculature platform (zFXVP) to visualize and quantify tumor vasculature changes. Employing zFXVP, we systematically screened compounds, identifying PF-502 as a robust vascular normalization agent. Mechanistic studies showed PF-502 induces endothelial cell-cycle arrest, streamlines vasculature, and activates Notch1 signaling, enhancing stability and hemodynamics. In murine models, PF-502 exhibited pronounced vascular normalization and improved drug delivery at a sub-maximum tolerated dose. These findings highlight zFXVP's utility and suggest PF-502 as a promising adjunctive for vascular normalization in clinical settings.
Collapse
Affiliation(s)
- Jian Zhong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Chaoxin Xiao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Qin Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Xiangyu Pan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Tongtong Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Yiyun Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Wanting Hou
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Lu Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Fujun Cao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Yulin Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Xiaoying Li
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Lin Zhou
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanshuo Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| | - Yu Yang
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, Sichuan 610041, China
| | - Chengjian Zhao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, China
| |
Collapse
|
27
|
Kakabadze Z, Paresishvili T. Intravital tumor decellularization as a new approach to cancer treatment. Am J Cancer Res 2023; 13:4192-4207. [PMID: 37818079 PMCID: PMC10560955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/08/2023] [Indexed: 10/12/2023] Open
Abstract
This study demonstrates the possibility of tumor decellularization in living animals. Subcutaneous Ehrlich tumor induced by isolated Ehrlich ascitic carcinoma cells in mice was used as a model. The study also presents methods for ex vivo decellularization of human gastric adenocarcinoma (HGA) and hepatocellular carcinoma (HCC) induced by diethylnitrosamine (DEN) in rat. Sodium dodecyl sulfate (SDS) and Triton X-100 were used as detergents for tumor decellularization. The detergents for HGA and HCC were administered through organ vessels. For intravital decellularization of Ehrlich's subcutaneous tumor, detergents were injected directly into the tumor parenchyma. The results of the study showed that the effectiveness of tumor decellularization using SDS and Triton X-100 depended on the size, structure, stiffness and density of the tumor, as well as on the concentration, route and speed of detergent administration. The study also showed that an hour after the initiation of decellularization, the central part of Ehrlich's tumor changed the color, and after three hours, it completely acquired a translucent white color. Chemical contamination of tissues surrounding the tumor with the detergents was not observed. Histological studies showed the complete absence of all cellular components of Ehrlich's tumor and a slightly deformed extracellular matrix (ECM). There were no loco-regional recurrences or metastases of Ehrlich's tumor within 150 days after decellularization. The developed intravital decellularization method allows the effective removal of the cellular components and the DNA content of Ehrlich's subcutaneous tumor without compromising animal health. Additionally, this method can destroy tumor ECM, which will significantly improve the delivery of anticancer drugs to the tumor cells. However, more detailed and extensive studies are needed to develop an in vivo technique for isolated decellularization of the tumor or a part of the organ with the tumor. It is also necessary to identify less toxic decellularization agents and to develop the most efficient route for their delivery to the tumor cells.
Collapse
Affiliation(s)
- Zurab Kakabadze
- Department of Clinical Anatomy, Tbilisi State Medical University 0186 Tbilisi, Georgia
| | - Teona Paresishvili
- Department of Clinical Anatomy, Tbilisi State Medical University 0186 Tbilisi, Georgia
| |
Collapse
|
28
|
Paresishvili T, Kakabadze Z. Challenges and Opportunities Associated With Drug Delivery for the Treatment of Solid Tumors. Oncol Rev 2023; 17:10577. [PMID: 37711860 PMCID: PMC10497757 DOI: 10.3389/or.2023.10577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/17/2023] [Indexed: 09/16/2023] Open
Abstract
In this review, we discuss the effectiveness of drug delivery system based on metal nanoparticles, and also, describe the problems associated with their delivery to tumor cells. Throughout recent years, more reports have appeared in the literature that demonstrate promising results for the treatment of various types of cancer using metal-based nanoparticles. Due to their unique physical and chemical properties, metal nanoparticles are effectively being used for the delivery of drug to the tumor cells, for cancer diagnosis and treatment. They can also be synthesized allowing the control of size and shape. However, the effectiveness of the metal nanoparticles for cancer treatment largely depends on their stability, biocompatibility, and ability to selectively affect tumor cells after their systemic or local administration. Another major problem associated with metal nanoparticles is their ability to overcome tumor tissue barriers such as atypical blood vessel structure, dense and rigid extracellular matrix, and high pressure of tumor interstitial fluid. The review also describes the design of tumor drug delivery systems that are based on metal nanoparticles. The mechanism of action of metal nanoparticles on cancer cells is also discussed. Considering the therapeutic safety and toxicity of metal nanoparticles, the prospects for their use for future clinical applications are being currently reviewed.
Collapse
Affiliation(s)
- Teona Paresishvili
- Department of Clinical Anatomy, Tbilisi State Medical University, Tbilisi, Georgia
| | | |
Collapse
|
29
|
Sun XX, Nosrati Z, Ko J, Lee CM, Bennewith KL, Bally MB. Induced Vascular Normalization-Can One Force Tumors to Surrender to a Better Microenvironment? Pharmaceutics 2023; 15:2022. [PMID: 37631236 PMCID: PMC10458586 DOI: 10.3390/pharmaceutics15082022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/19/2023] [Accepted: 07/14/2023] [Indexed: 08/27/2023] Open
Abstract
Immunotherapy has changed the way many cancers are being treated. Researchers in the field of immunotherapy and tumor immunology are investigating similar questions: How can the positive benefits achieved with immunotherapies be enhanced? Can this be achieved through combinations with other agents and if so, which ones? In our view, there is an urgent need to improve immunotherapy to make further gains in the overall survival for those patients that should benefit from immunotherapy. While numerous different approaches are being considered, our team believes that drug delivery methods along with appropriately selected small-molecule drugs and drug candidates could help reach the goal of doubling the overall survival rate that is seen in some patients that are given immunotherapeutics. This review article is prepared to address how immunotherapies should be combined with a second treatment using an approach that could realize therapeutic gains 10 years from now. For context, an overview of immunotherapy and cancer angiogenesis is provided. The major targets in angiogenesis that have modulatory effects on the tumor microenvironment and immune cells are highlighted. A combination approach that, for us, has the greatest potential for success involves treatments that will normalize the tumor's blood vessel structure and alter the immune microenvironment to support the action of immunotherapeutics. So, this is reviewed as well. Our focus is to provide an insight into some strategies that will engender vascular normalization that may be better than previously described approaches. The potential for drug delivery systems to promote tumor blood vessel normalization is considered.
Collapse
Affiliation(s)
- Xu Xin Sun
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Interdisciplinary Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- NanoMedicines Innovation Network, Vancouver, BC V6T 1Z3, Canada
- Cuprous Pharmaceuticals, Vancouver, BC V6N 3P8, Canada
| | - Zeynab Nosrati
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Interdisciplinary Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- Cuprous Pharmaceuticals, Vancouver, BC V6N 3P8, Canada
| | - Janell Ko
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
| | - Che-Min Lee
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Kevin L. Bennewith
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Marcel B. Bally
- Experimental Therapeutics, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (Z.N.); (J.K.); (C.-M.L.); (K.L.B.); (M.B.B.)
- Interdisciplinary Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- NanoMedicines Innovation Network, Vancouver, BC V6T 1Z3, Canada
- Cuprous Pharmaceuticals, Vancouver, BC V6N 3P8, Canada
- Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
30
|
Shafqat A, Omer MH, Ahmed EN, Mushtaq A, Ijaz E, Ahmed Z, Alkattan K, Yaqinuddin A. Reprogramming the immunosuppressive tumor microenvironment: exploiting angiogenesis and thrombosis to enhance immunotherapy. Front Immunol 2023; 14:1200941. [PMID: 37520562 PMCID: PMC10374407 DOI: 10.3389/fimmu.2023.1200941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/15/2023] [Indexed: 08/01/2023] Open
Abstract
This review focuses on the immunosuppressive effects of tumor angiogenesis and coagulation on the tumor microenvironment (TME). We summarize previous research efforts leveraging these observations and targeting these processes to enhance immunotherapy outcomes. Clinical trials have documented improved outcomes when combining anti-angiogenic agents and immunotherapy. However, their overall survival benefit over conventional therapy remains limited and certain tumors exhibit poor response to anti-angiogenic therapy. Additionally, whilst preclinical studies have shown several components of the tumor coagulome to curb effective anti-tumor immune responses, the clinical studies reporting combinations of anticoagulants with immunotherapies have demonstrated variable treatment outcomes. By reviewing the current state of the literature on this topic, we address the key questions and future directions in the field, the answers of which are crucial for developing effective strategies to reprogram the TME in order to further the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Mohamed H. Omer
- School of Medicine, Cardiff University, Cardiff, United Kingdom
| | | | - Ali Mushtaq
- Department of Internal Medicine, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Eman Ijaz
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Zara Ahmed
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Khaled Alkattan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | | |
Collapse
|
31
|
Hoffmann E, Gerwing M, Krähling T, Hansen U, Kronenberg K, Masthoff M, Geyer C, Höltke C, Wachsmuth L, Schinner R, Hoerr V, Heindel W, Karst U, Eisenblätter M, Maus B, Helfen A, Faber C, Wildgruber M. Vascular response patterns to targeted therapies in murine breast cancer models with divergent degrees of malignancy. Breast Cancer Res 2023; 25:56. [PMID: 37221619 DOI: 10.1186/s13058-023-01658-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 05/14/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Response assessment of targeted cancer therapies is becoming increasingly challenging, as it is not adequately assessable with conventional morphological and volumetric analyses of tumor lesions. The tumor microenvironment is particularly constituted by tumor vasculature which is altered by various targeted therapies. The aim of this study was to noninvasively assess changes in tumor perfusion and vessel permeability after targeted therapy in murine models of breast cancer with divergent degrees of malignancy. METHODS Low malignant 67NR or highly malignant 4T1 tumor-bearing mice were treated with either the multi-kinase inhibitor sorafenib or immune checkpoint inhibitors (ICI, combination of anti-PD1 and anti-CTLA4). Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) with i.v. injection of albumin-binding gadofosveset was conducted on a 9.4 T small animal MRI. Ex vivo validation of MRI results was achieved by transmission electron microscopy, immunohistochemistry and laser ablation-inductively coupled plasma-mass spectrometry. RESULTS Therapy-induced changes in tumor vasculature differed between low and highly malignant tumors. Sorafenib treatment led to decreased tumor perfusion and endothelial permeability in low malignant 67NR tumors. In contrast, highly malignant 4T1 tumors demonstrated characteristics of a transient window of vascular normalization with an increase in tumor perfusion and permeability early after therapy initiation, followed by decreased perfusion and permeability parameters. In the low malignant 67NR model, ICI treatment also mediated vessel-stabilizing effects with decreased tumor perfusion and permeability, while ICI-treated 4T1 tumors exhibited increasing tumor perfusion with excessive vascular leakage. CONCLUSION DCE-MRI enables noninvasive assessment of early changes in tumor vasculature after targeted therapies, revealing different response patterns between tumors with divergent degrees of malignancy. DCE-derived tumor perfusion and permeability parameters may serve as vascular biomarkers that allow for repetitive examination of response to antiangiogenic treatment or immunotherapy.
Collapse
Grants
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
Collapse
Affiliation(s)
- Emily Hoffmann
- Clinic of Radiology, University of Münster, Münster, Germany.
| | - Mirjam Gerwing
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Tobias Krähling
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Uwe Hansen
- Institute for Musculoskeletal Medicine, University of Münster, Münster, Germany
| | - Katharina Kronenberg
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Max Masthoff
- Clinic of Radiology, University of Münster, Münster, Germany
| | | | - Carsten Höltke
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Lydia Wachsmuth
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Regina Schinner
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Verena Hoerr
- Clinic of Radiology, University of Münster, Münster, Germany
- Heart Center Bonn, Department of Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Walter Heindel
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Michel Eisenblätter
- Clinic of Radiology, University of Münster, Münster, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty OWL, University of Bielefeld, Bielefeld, Germany
| | - Bastian Maus
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Anne Helfen
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Cornelius Faber
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Moritz Wildgruber
- Clinic of Radiology, University of Münster, Münster, Germany
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
32
|
Liu ZL, Chen HH, Zheng LL, Sun LP, Shi L. Angiogenic signaling pathways and anti-angiogenic therapy for cancer. Signal Transduct Target Ther 2023; 8:198. [PMID: 37169756 PMCID: PMC10175505 DOI: 10.1038/s41392-023-01460-1] [Citation(s) in RCA: 404] [Impact Index Per Article: 202.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/20/2023] [Accepted: 04/20/2023] [Indexed: 05/13/2023] Open
Abstract
Angiogenesis, the formation of new blood vessels, is a complex and dynamic process regulated by various pro- and anti-angiogenic molecules, which plays a crucial role in tumor growth, invasion, and metastasis. With the advances in molecular and cellular biology, various biomolecules such as growth factors, chemokines, and adhesion factors involved in tumor angiogenesis has gradually been elucidated. Targeted therapeutic research based on these molecules has driven anti-angiogenic treatment to become a promising strategy in anti-tumor therapy. The most widely used anti-angiogenic agents include monoclonal antibodies and tyrosine kinase inhibitors (TKIs) targeting vascular endothelial growth factor (VEGF) pathway. However, the clinical benefit of this modality has still been limited due to several defects such as adverse events, acquired drug resistance, tumor recurrence, and lack of validated biomarkers, which impel further research on mechanisms of tumor angiogenesis, the development of multiple drugs and the combination therapy to figure out how to improve the therapeutic efficacy. Here, we broadly summarize various signaling pathways in tumor angiogenesis and discuss the development and current challenges of anti-angiogenic therapy. We also propose several new promising approaches to improve anti-angiogenic efficacy and provide a perspective for the development and research of anti-angiogenic therapy.
Collapse
Affiliation(s)
- Zhen-Ling Liu
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Huan-Huan Chen
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Li-Li Zheng
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China
| | - Li-Ping Sun
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China.
| | - Lei Shi
- Department of Medicinal Chemistry, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 210009, Nanjing, China.
| |
Collapse
|
33
|
Pan Y, Liu Y, Wei W, Yang X, Wang Z, Xin W. Extracellular Vesicles as Delivery Shippers for Noncoding RNA-Based Modulation of Angiogenesis: Insights from Ischemic Stroke and Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2205739. [PMID: 36592424 DOI: 10.1002/smll.202205739] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 12/02/2022] [Indexed: 06/17/2023]
Abstract
Ischemic stroke and systemic cancer are two of the leading causes of mortality. Hypoxia is a central pathophysiological component in ischemic stroke and cancer, representing a joint medical function. This function includes angiogenesis regulation. Vascular remodeling coupled with axonal outgrowth following cerebral ischemia is critical in improving poststroke neurological functional recovery. Antiangiogenic strategies can inhibit cancer vascularization and play a vital role in impeding cancer growth, invasion, and metastasis. Although there are significant differences in the cause of angiogenesis across both pathophysiological conditions, emerging evidence states that common signaling structures, such as extracellular vesicles (EVs) and noncoding RNAs (ncRNAs), are involved in this context. EVs, heterogeneous membrane vesicles encapsulating proteomic genetic information from parental cells, act as multifunctional regulators of intercellular communication. Among the multifaceted roles in modulating biological responses, exhaustive evidence shows that ncRNAs are selectively sorted into EVs, modulating common specific aspects of cancer development and stroke prognosis, namely, angiogenesis. This review will discuss recent advancements in the EV-facilitated/inhibited progression of specific elements of angiogenesis with a particular concern about ncRNAs within these vesicles. The review is concluded by underlining the clinical opportunities of EV-derived ncRNAs as diagnostic, prognostic, and therapeutic agents.
Collapse
Affiliation(s)
- Yongli Pan
- Department of Neurology, University Medical Center of Göttingen, Georg-August-University of Göttingen, 37075, Göttingen, Lower Saxony, Germany
- Department of Neurology, Weifang Medical University, Weifang, Shandong, 261053, China
| | - Yuheng Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Neurological Institute, Tianjin, 300052, China
| | - Wei Wei
- Department of Neurology, University Medical Center of Göttingen, Georg-August-University of Göttingen, 37075, Göttingen, Lower Saxony, Germany
- Department of Neurology, Mianyang Central Hospital, Mianyang, Sichuan, 621000, China
| | - Xinyu Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Neurological Institute, Tianjin, 300052, China
| | - Zengguang Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Neurological Institute, Tianjin, 300052, China
| | - Wenqiang Xin
- Department of Neurology, University Medical Center of Göttingen, Georg-August-University of Göttingen, 37075, Göttingen, Lower Saxony, Germany
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
- Tianjin Neurological Institute, Tianjin, 300052, China
| |
Collapse
|
34
|
Melssen MM, Sheybani ND, Leick KM, Slingluff CL. Barriers to immune cell infiltration in tumors. J Immunother Cancer 2023; 11:jitc-2022-006401. [PMID: 37072352 PMCID: PMC10124321 DOI: 10.1136/jitc-2022-006401] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2023] [Indexed: 04/20/2023] Open
Abstract
Increased immune cell infiltration into tumors is associated with improved patient survival and predicts response to immune therapies. Thus, identification of factors that determine the extent of immune infiltration is crucial, so that methods to intervene on these targets can be developed. T cells enter tumor tissues through the vasculature, and under control of interactions between homing receptors on the T cells and homing receptor ligands (HRLs) expressed by tumor vascular endothelium and tumor cell nests. HRLs are often deficient in tumors, and there also may be active barriers to infiltration. These remain understudied but may be crucial for enhancing immune-mediated cancer control. Multiple intratumoral and systemic therapeutic approaches show promise to enhance T cell infiltration, including both approved therapies and experimental therapies. This review highlights the intracellular and extracellular determinants of immune cell infiltration into tumors, barriers to infiltration, and approaches for intervention to enhance infiltration and response to immune therapies.
Collapse
Affiliation(s)
- Marit M Melssen
- Immunology, Genetics & Pathology, Uppsala University, Uppsala, Sweden
| | - Natasha D Sheybani
- Biomedical Engineering, University of Virginia Health System, Charlottesville, Virginia, USA
| | | | | |
Collapse
|
35
|
Brogowska KK, Zajkowska M, Mroczko B. Vascular Endothelial Growth Factor Ligands and Receptors in Breast Cancer. J Clin Med 2023; 12:jcm12062412. [PMID: 36983412 PMCID: PMC10056253 DOI: 10.3390/jcm12062412] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Breast cancer (BC) is the most common malignancy responsible for the largest number of deaths in women worldwide. The risk of developing BC is predisposed by many factors such as age, presence of genetic mutations or body weight. The diagnosis is mostly made relatively late, which is why patients are exposed to radical surgical treatments, long-term chemotherapy and lower survival rates. There are no sufficiently sensitive and specific screening tests; therefore, researchers are still looking for new diagnostic biomarkers that would indicate the appearance of neoplastic changes in the initial stage of neoplasm. The VEGF family of proteins (VEGF-A, VEGF-B, VEGF-C, VEGF-D, EG-VEGF, PlGF) and their receptors are significant factors in the pathogenesis of BC. They play a significant role in the process of angiogenesis and lymphangiogenesis in both physiological and pathological conditions. The usefulness of these proteins as potential diagnostic biomarkers has been initially proven. Moreover, the blockage of VEGF-related pathways seems to be a valid therapeutic target. Recent studies have tried to describe novel strategies, including targeting pericytes, use of miRNAs and extracellular tumor-associated vesicles, immunotherapeutic drugs and nanotechnology. This indicates their possible contribution to the formation of breast cancer and their usefulness as potential biomarkers and therapeutic targets.
Collapse
Affiliation(s)
| | - Monika Zajkowska
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
36
|
Cunningham C, Bolcaen J, Bisio A, Genis A, Strijdom H, Vandevoorde C. Recombinant Endostatin as a Potential Radiosensitizer in the Treatment of Non-Small Cell Lung Cancer. Pharmaceuticals (Basel) 2023; 16:219. [PMID: 37259367 PMCID: PMC9961924 DOI: 10.3390/ph16020219] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 11/03/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most prevalent type of lung cancer, which is the leading cause of cancer-related deaths worldwide. Over the past decades, tumour angiogenesis has been intensely studied in the treatment of NSCLC due to its fundamental role in cancer progression. Several anti-angiogenic drugs, such as recombinant endostatin (RE), have been evaluated in several preclinical and clinical trials, with mixed and often disappointing results. However, there is currently an emerging interest in RE due to its ability to create a vascular normalization window, which could further improve treatment efficacy of the standard NSCLC treatment. This review provides an overview of preclinical and clinical studies that combined RE and radiotherapy for NSCLC treatment. Furthermore, it highlights the ongoing challenges that have to be overcome in order to maximize the benefit; as well as the potential advantage of combinations with particle therapy and immunotherapy, which are rapidly gaining momentum in the treatment landscape of NSCLC. Different angiogenic and immunosuppressive effects are observed between particle therapy and conventional X-ray radiotherapy. The combination of RE, particle therapy and immunotherapy presents a promising future therapeutic triad for NSCLC.
Collapse
Affiliation(s)
- Charnay Cunningham
- Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Stellenbosch University, Cape Town 7602, South Africa
- Radiation Biophysics Division, SSC Laboratory, NRF Ithemba LABS, Cape Town 7131, South Africa
| | - Julie Bolcaen
- Radiation Biophysics Division, SSC Laboratory, NRF Ithemba LABS, Cape Town 7131, South Africa
| | - Alessandra Bisio
- Department of Cellular, Computational and Integrative Biology—CIBIO, University of Trento, 38123 Trento, Italy
| | - Amanda Genis
- Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Stellenbosch University, Cape Town 7602, South Africa
| | - Hans Strijdom
- Centre for Cardio-Metabolic Research in Africa (CARMA), Division of Medical Physiology, Stellenbosch University, Cape Town 7602, South Africa
| | - Charlot Vandevoorde
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung, Planckstr. 1, 64291 Darmstadt, Germany
| |
Collapse
|
37
|
Qin Y, Xu G. Enhancing CAR T-cell therapies against solid tumors: Mechanisms and reversion of resistance. Front Immunol 2022; 13:1053120. [PMID: 36569859 PMCID: PMC9773088 DOI: 10.3389/fimmu.2022.1053120] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy, belonging to adoptive immune cells therapy, utilizes engineered immunoreceptors to enhance tumor-specific killing. By now new generations of CAR T-cell therapies dramatically promote the effectiveness and robustness in leukemia cases. However, only a few CAR T-cell therapies gain FDA approval till now, which are applied to hematologic cancers. Targeting solid tumors through CAR T-cell therapies still faces many problems, such as tumor heterogeneity, antigen loss, infiltration inability and immunosuppressive micro-environment. Recent advances provide new insights about the mechanisms of CAR T-cell therapy resistance and give rise to potential reversal therapies. In this review, we mainly introduce existing barriers when treating solid tumors with CAR T-cells and discuss the methods to overcome these challenges.
Collapse
Affiliation(s)
- Yue Qin
- National Institute of Biological Sciences, Beijing, China,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Guotai Xu
- National Institute of Biological Sciences, Beijing, China,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China,*Correspondence: Guotai Xu,
| |
Collapse
|
38
|
Xu M, Zhang T, Xia R, Wei Y, Wei X. Targeting the tumor stroma for cancer therapy. Mol Cancer 2022; 21:208. [PMID: 36324128 PMCID: PMC9628074 DOI: 10.1186/s12943-022-01670-1] [Citation(s) in RCA: 128] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/13/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
Tumors are comprised of both cancer cells and surrounding stromal components. As an essential part of the tumor microenvironment, the tumor stroma is highly dynamic, heterogeneous and commonly tumor-type specific, and it mainly includes noncellular compositions such as the extracellular matrix and the unique cancer-associated vascular system as well as a wide variety of cellular components including activated cancer-associated fibroblasts, mesenchymal stromal cells, pericytes. All these elements operate with each other in a coordinated fashion and collectively promote cancer initiation, progression, metastasis and therapeutic resistance. Over the past few decades, numerous studies have been conducted to study the interaction and crosstalk between stromal components and neoplastic cells. Meanwhile, we have also witnessed an exponential increase in the investigation and recognition of the critical roles of tumor stroma in solid tumors. A series of clinical trials targeting the tumor stroma have been launched continually. In this review, we introduce and discuss current advances in the understanding of various stromal elements and their roles in cancers. We also elaborate on potential novel approaches for tumor-stroma-based therapeutic targeting, with the aim to promote the leap from bench to bedside.
Collapse
Affiliation(s)
- Maosen Xu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Tao Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Ruolan Xia
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China.
| |
Collapse
|
39
|
Tian Z, Zeng Y, Peng Y, Liu J, Wu F. Cancer immunotherapy strategies that target the cGAS-STING pathway. Front Immunol 2022; 13:996663. [PMID: 36353640 PMCID: PMC9639746 DOI: 10.3389/fimmu.2022.996663] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/03/2022] [Indexed: 10/22/2023] Open
Abstract
Activation of the cGAS-STING pathway by cytoplasmic DNA induces the production of Type-1 interferons. Recent advances in research suggest that the cGAS-STING pathway is involved in different parts of the cancer-immunity cycle (CIC) to promote or suppress antitumor immune responses. Combination therapy of STING agonists has made certain progress in preclinical as well as clinical trials, but the selection of combination therapy regimens remains a challenge. In this review, we summarize the role of the cGAS-STING in all aspects of CIC, and focus on the combination immunotherapy strategies of STING agonists and current unsolved challenges.
Collapse
Affiliation(s)
- Zhuoying Tian
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yue Zeng
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yurong Peng
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Junqi Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fang Wu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Cancer Mega-Data Intelligent Application and Engineering Research Centre, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
40
|
Swamy K. Vascular normalization and immunotherapy: Spawning a virtuous cycle. Front Oncol 2022; 12:1002957. [PMID: 36276103 PMCID: PMC9582256 DOI: 10.3389/fonc.2022.1002957] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Anti-angiogenics, radiotherapy (especially stereotactic body radiotherapy, SBRT)/chemotherapy, and immunotherapy form a critical trimodal approach in modern cancer therapy. The normalization window, however short, is the beachhead for the strategic initiation of a decipherable disruption of cancer cells. This opening can be the opportunity for designing controlled stepwise cancer cell death (CCD) and immunological augmentation. The next step is to induce immunogenic cell death (ICD) through chemotherapy/radiotherapy concurrently with the facilitation of professional phagocytosis. Immunotherapy at this stage, when interstitial pressure decreases considerably, leads to the improved perfusion of oxygen with solutes and improved immune-friendly pH and is additionally expected to open up the tumor microenvironment (TME) for a “flood” of tumor-infiltrating lymphocytes. Furthermore, there would be enhanced interaction in “hot” nodules and the incorporation of immune reaction in “cold” nodules. Simultaneously, the added adjuvant-assisted neoantigen–immune cell interaction will likely set in a virtuous cycle of CCD induction followed by tumor cell-specific antigenic reaction boosting CCD, in turn promoting the normalization of the vasculature, completing the loop. There should be a conscious concern to protect the extracellular matrix (ECM), which will nurture the long-term immunological cross-talk to discourage dormancy, which is as essential as obtaining a complete response in imaging. The caveat is that the available therapies should be appropriately ranked during the start of the treatment since the initial administration is the most opportune period. A fast-paced development in the nanomedicine field is likely to assist in all the steps enumerated.
Collapse
|
41
|
Oronsky B, Gastman B, Conley AP, Reid C, Caroen S, Reid T. Oncolytic Adenoviruses: The Cold War against Cancer Finally Turns Hot. Cancers (Basel) 2022; 14:4701. [PMID: 36230621 PMCID: PMC9562194 DOI: 10.3390/cancers14194701] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/21/2022] [Accepted: 09/25/2022] [Indexed: 11/27/2022] Open
Abstract
Oncolytic viruses, colloquially referred to as "living drugs", amplify themselves and the therapeutic transgenes that they carry to stimulate an immune response both locally and systemically. Remarkable exceptions aside, such as the recent 14-patient trial with the PD-1 inhibitor, dostarlimab, in mismatch repair (MMR) deficient rectal cancer, where the complete response rate was 100%, checkpoint inhibitors are not cure-alls, which suggests the need for a combination partner like oncolytic viruses to prime and augment their activity. This review focuses on adenoviruses, the most clinically investigated of all the oncolytic viruses. It covers specific design features of clinical adenoviral candidates and highlights their potential both alone and in combination with checkpoint inhibitors in clinical trials to turn immunologically "cold" and unresponsive tumors into "hotter" and more responsive ones through a domino effect. Finally, a "mix-and-match" combination of therapies based on the paradigm of the cancer-immunity cycle is proposed to augment the immune responses of oncolytic adenoviruses.
Collapse
Affiliation(s)
| | | | - Anthony P. Conley
- University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Scott Caroen
- EpicentRx, Torrey Pines, La Jolla, CA 92037, USA
| | - Tony Reid
- EpicentRx, Torrey Pines, La Jolla, CA 92037, USA
| |
Collapse
|
42
|
Kuo CL, Ponneri Babuharisankar A, Lin YC, Lien HW, Lo YK, Chou HY, Tangeda V, Cheng LC, Cheng AN, Lee AYL. Mitochondrial oxidative stress in the tumor microenvironment and cancer immunoescape: foe or friend? J Biomed Sci 2022; 29:74. [PMID: 36154922 PMCID: PMC9511749 DOI: 10.1186/s12929-022-00859-2] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 09/19/2022] [Indexed: 12/07/2022] Open
Abstract
The major concept of "oxidative stress" is an excess elevated level of reactive oxygen species (ROS) which are generated from vigorous metabolism and consumption of oxygen. The precise harmonization of oxidative stresses between mitochondria and other organelles in the cell is absolutely vital to cell survival. Under oxidative stress, ROS produced from mitochondria and are the major mediator for tumorigenesis in different aspects, such as proliferation, migration/invasion, angiogenesis, inflammation, and immunoescape to allow cancer cells to adapt to the rigorous environment. Accordingly, the dynamic balance of oxidative stresses not only orchestrate complex cell signaling events in cancer cells but also affect other components in the tumor microenvironment (TME). Immune cells, such as M2 macrophages, dendritic cells, and T cells are the major components of the immunosuppressive TME from the ROS-induced inflammation. Based on this notion, numerous strategies to mitigate oxidative stresses in tumors have been tested for cancer prevention or therapies; however, these manipulations are devised from different sources and mechanisms without established effectiveness. Herein, we integrate current progress regarding the impact of mitochondrial ROS in the TME, not only in cancer cells but also in immune cells, and discuss the combination of emerging ROS-modulating strategies with immunotherapies to achieve antitumor effects.
Collapse
Affiliation(s)
- Cheng-Liang Kuo
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan
| | - Ananth Ponneri Babuharisankar
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan.,Joint PhD Program in Molecular Medicine, NHRI & NCU, Zhunan, Miaoli, 35053, Taiwan
| | - Ying-Chen Lin
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan
| | - Hui-Wen Lien
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan
| | - Yu Kang Lo
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan
| | - Han-Yu Chou
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan
| | - Vidhya Tangeda
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan.,Joint PhD Program in Molecular Medicine, NHRI & NCU, Zhunan, Miaoli, 35053, Taiwan
| | - Li-Chun Cheng
- Liver Research Center, Linkou Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
| | - An Ning Cheng
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
| | - Alan Yueh-Luen Lee
- National Institute of Cancer Research, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053, Taiwan. .,Joint PhD Program in Molecular Medicine, NHRI & NCU, Zhunan, Miaoli, 35053, Taiwan. .,Department of Life Sciences, College of Health Sciences and Technology, National Central University, Zhongli, Taoyuan, 32001, Taiwan. .,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 40402, Taiwan. .,Department of Biotechnology, College of Life Science, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
43
|
Ileiwat ZE, Tabish TA, Zinovkin DA, Yuzugulen J, Arghiani N, Pranjol MZI. The mechanistic immunosuppressive role of the tumour vasculature and potential nanoparticle-mediated therapeutic strategies. Front Immunol 2022; 13:976677. [PMID: 36045675 PMCID: PMC9423123 DOI: 10.3389/fimmu.2022.976677] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/29/2022] [Indexed: 11/26/2022] Open
Abstract
The tumour vasculature is well-established to display irregular structure and hierarchy that is conducive to promoting tumour growth and metastasis while maintaining immunosuppression. As tumours grow, their metabolic rate increases while their distance from blood vessels furthers, generating a hypoxic and acidic tumour microenvironment. Consequently, cancer cells upregulate the expression of pro-angiogenic factors which propagate aberrant blood vessel formation. This generates atypical vascular features that reduce chemotherapy, radiotherapy, and immunotherapy efficacy. Therefore, the development of therapies aiming to restore the vasculature to a functional state remains a necessary research target. Many anti-angiogenic therapies aim to target this such as bevacizumab or sunitinib but have shown variable efficacy in solid tumours due to intrinsic or acquired resistance. Therefore, novel therapeutic strategies such as combination therapies and nanotechnology-mediated therapies may provide alternatives to overcoming the barriers generated by the tumour vasculature. This review summarises the mechanisms that induce abnormal tumour angiogenesis and how the vasculature’s features elicit immunosuppression. Furthermore, the review explores examples of treatment regiments that target the tumour vasculature.
Collapse
Affiliation(s)
- Zakaria Elias Ileiwat
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Tanveer A. Tabish
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Jale Yuzugulen
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, Cyprus
| | - Nahid Arghiani
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
- *Correspondence: Nahid Arghiani, ; Md Zahidul I. Pranjol,
| | - Md Zahidul I. Pranjol
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- *Correspondence: Nahid Arghiani, ; Md Zahidul I. Pranjol,
| |
Collapse
|
44
|
Jinna N, Rida P, Smart M, LaBarge M, Jovanovic-Talisman T, Natarajan R, Seewaldt V. Adaptation to Hypoxia May Promote Therapeutic Resistance to Androgen Receptor Inhibition in Triple-Negative Breast Cancer. Int J Mol Sci 2022; 23:ijms23168844. [PMID: 36012111 PMCID: PMC9408190 DOI: 10.3390/ijms23168844] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/03/2022] [Accepted: 08/06/2022] [Indexed: 12/27/2022] Open
Abstract
Triple-negative breast cancer (TNBC) surpasses other BC subtypes as the most challenging to treat due to its lack of traditional BC biomarkers. Nearly 30% of TNBC patients express the androgen receptor (AR), and the blockade of androgen production and AR signaling have been the cornerstones of therapies for AR-positive TNBC. However, the majority of women are resistant to AR-targeted therapy, which is a major impediment to improving outcomes for the AR-positive TNBC subpopulation. The hypoxia signaling cascade is frequently activated in the tumor microenvironment in response to low oxygen levels; activation of the hypoxia signaling cascade allows tumors to survive despite hypoxia-mediated interference with cellular metabolism. The activation of hypoxia signaling networks in TNBC promotes resistance to most anticancer drugs including AR inhibitors. The activation of hypoxia network signaling occurs more frequently in TNBC compared to other BC subtypes. Herein, we examine the (1) interplay between hypoxia signaling networks and AR and (2) whether hypoxia and hypoxic stress adaptive pathways promote the emergence of resistance to therapies that target AR. We also pose the well-supported question, “Can the efficacy of androgen-/AR-targeted treatments be enhanced by co-targeting hypoxia?” By critically examining the evidence and the complex entwinement of these two oncogenic pathways, we argue that the simultaneous targeting of androgen biosynthesis/AR signaling and hypoxia may enhance the sensitivity of AR-positive TNBCs to AR-targeted treatments, derail the emergence of therapy resistance, and improve patient outcomes.
Collapse
Affiliation(s)
- Nikita Jinna
- Department of Population Science, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | | | - Max Smart
- Rowland Hall, Salt Lake City, UT 84102, USA
| | - Mark LaBarge
- Department of Population Science, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | | | - Rama Natarajan
- Department of Diabetes Complications and Metabolism, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| | - Victoria Seewaldt
- Department of Population Science, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
- Correspondence:
| |
Collapse
|
45
|
Li H, Huang H, Zhang T, Feng H, Wang S, Zhang Y, Ji X, Cheng X, Zhao R. Apatinib: A Novel Antiangiogenic Drug in Monotherapy or Combination Immunotherapy for Digestive System Malignancies. Front Immunol 2022; 13:937307. [PMID: 35844616 PMCID: PMC9276937 DOI: 10.3389/fimmu.2022.937307] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 05/30/2022] [Indexed: 02/03/2023] Open
Abstract
Digestive system malignancies are one of the primary causes of cancer-related death. Meanwhile, angiogenesis has been proved to play an important role in the process of cancer neovascularization. Apatinib, a novel targeted antiangiogenic molecule, could generate highly selective competition in the vascular endothelial growth factor receptor-2, involved in tumor progression and metastasis. It has been implied as a promising cancer treatment agent that can prevent tumor cell proliferation meanwhile inhibit tumor angiogenesis. Furthermore, completed clinical trials demonstrated that apatinib could prolong the progression-free survival and overall survival in advanced gastric cancer and primary liver cancer. Recent studies revealed that apatinib had a synergistic effect with immunotherapy as a second-line and third-line treatment regimen for some other cancers. In this review, we summarize the pharmacological properties of apatinib and the latest clinical application in chemotherapy-refractory patients with advanced digestive system cancer. Based on the comparable survival results, the molecular mechanisms of apatinib are prospective to include the antiangiogenic, apoptosis-inducing, and autophagy-inducing properties in the corresponding signaling pathway. Treatment of apatinib monotherapy or combination immunotherapy remains the optimal option for patients with digestive system malignancies in the future.
Collapse
Affiliation(s)
- Haosheng Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyan Huang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoran Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaodong Wang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaqi Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaopin Ji
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Xiaopin Ji, ; Xi Cheng, ; Ren Zhao,
| | - Xi Cheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Xiaopin Ji, ; Xi Cheng, ; Ren Zhao,
| | - Ren Zhao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Xiaopin Ji, ; Xi Cheng, ; Ren Zhao,
| |
Collapse
|
46
|
Zeng Y, Yu T, Zhang S, Song G, Meng T, Yuan H, Hu F. Combination of tumor vessel normalization and immune checkpoint blockade for breast cancer treatment via multifunctional nanocomplexes. Biomater Sci 2022; 10:4140-4155. [PMID: 35726757 DOI: 10.1039/d2bm00600f] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumor vessel normalization can alleviate hypoxia, reduce the intratumoral infiltration of immunosuppressive cells and increase the intratumoral infiltration of immune effector cells (CD8+ T cells), further reversing the immunosuppressive microenvironment. Here, nanocomplexes (lipo/St@FA-COSA/BMS-202) which can accurately deliver drugs to tumor tissues and release different drugs at different sites with different rates were prepared to combine tumor vessel normalization with immune checkpoint blockade. The results of drug release in vitro showed that in a pH 6.5 release medium, lipo/St@FA-COSA/BMS-202 rapidly released the vascular normalizing drug (sunitinib, St) and slowly released the PD-1/PD-L1-blocking drug (BMS-202). The results of in vivo experiments showed that the rapidly released St normalized tumor vessels and formed an immunosupportive microenvironment which improved the anti-tumor efficacy of BMS-202. In conclusion, the drug delivery strategy significantly inhibited tumor growth and had excellent anti-tumor efficacy, which can provide a potential approach for effective tumor treatment.
Collapse
Affiliation(s)
- Yingping Zeng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China.
| | - Tong Yu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China.
| | - Shufen Zhang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China.
| | - Guangtao Song
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China.
| | - Tingting Meng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China.
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China.
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
47
|
Zhou J, Wang L, Peng C, Peng F. Co-Targeting Tumor Angiogenesis and Immunosuppressive Tumor Microenvironment: A Perspective in Ethnopharmacology. Front Pharmacol 2022; 13:886198. [PMID: 35784750 PMCID: PMC9242535 DOI: 10.3389/fphar.2022.886198] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
Tumor angiogenesis is one of the most important processes of cancer deterioration via nurturing an immunosuppressive tumor environment (TME). Targeting tumor angiogenesis has been widely accepted as a cancer intervention approach, which is also synergistically associated with immune therapy. However, drug resistance is the biggest challenge of anti-angiogenesis therapy, which affects the outcomes of anti-angiogeneic agents, and even combined with immunotherapy. Here, emerging targets and representative candidate molecules from ethnopharmacology (including traditional Chinese medicine, TCM) have been focused, and they have been proved to regulate tumor angiogenesis. Further investigations on derivatives and delivery systems of these molecules will provide a comprehensive landscape in preclinical studies. More importantly, the molecule library of ethnopharmacology meets the viability for targeting angiogenesis and TME simultaneously, which is attributed to the pleiotropy of pro-angiogenic factors (such as VEGF) toward cancer cells, endothelial cells, and immune cells. We primarily shed light on the potentiality of ethnopharmacology against tumor angiogenesis, particularly TCM. More research studies concerning the crosstalk between angiogenesis and TME remodeling from the perspective of botanical medicine are awaited.
Collapse
Affiliation(s)
- Jianbo Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Li Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Fu Peng, ; Cheng Peng,
| | - Fu Peng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
- *Correspondence: Fu Peng, ; Cheng Peng,
| |
Collapse
|
48
|
Schiffmann LM, Bruns CJ, Schmidt T. Resistance Mechanisms of the Metastatic Tumor Microenvironment to Anti-Angiogenic Therapy. Front Oncol 2022; 12:897927. [PMID: 35664794 PMCID: PMC9162757 DOI: 10.3389/fonc.2022.897927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/21/2022] [Indexed: 11/23/2022] Open
Abstract
Angiogenesis describes the formation of blood vessels from an existing vascular network. Anti-angiogenic drugs that target tumor blood vessels have become standard of care in many cancer entities. Though very promising results in preclinical evaluation, anti-angiogenic treatments fell short of expectations in clinical trials. Patients develop resistance over time or are primarily refractory to anti-angiogenic therapies similar to conventional chemotherapy. To further improve efficacy and outcome to these therapies, a deeper understanding of mechanisms that mediate resistance to anti-angiogenic therapies is needed. The field has done tremendous efforts to gain knowledge about how tumors engage tumor cell and microenvironmental mechanisms to do so. This review highlights the current state of knowledge with special focus on the metastatic tumor site and potential therapeutic relevance of this understanding from a translational and clinical perspective.
Collapse
Affiliation(s)
- Lars M. Schiffmann
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | - Thomas Schmidt
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
49
|
Superoxide Dismutase-3 Downregulates Laminin α5 Expression in Tumor Endothelial Cells via the Inhibition of Nuclear Factor Kappa B Signaling. Cancers (Basel) 2022; 14:cancers14051226. [PMID: 35267534 PMCID: PMC8909228 DOI: 10.3390/cancers14051226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/22/2022] [Accepted: 02/24/2022] [Indexed: 02/01/2023] Open
Abstract
The balance between laminin isoforms containing the α5 or the α4 chain in the endothelial basement membrane determines the site of leukocyte diapedesis under inflammatory conditions. Extracellular superoxide dismutase (SOD3) induces laminin α4 expression in tumor blood vessels, which is associated with enhanced intratumor T cell infiltration in primary human cancers. We show now that SOD3 overexpression in neoplastic and endothelial cells (ECs) reduces laminin α5 in tumor blood vessels. SOD3 represses the laminin α5 gene (LAMA5), but LAMA5 expression is not changed in SOD1-overexpressing cells. Transcriptomic analyses revealed SOD3 overexpression to change the transcription of 1682 genes in ECs, with the canonical and non-canonical NF-κB pathways as the major SOD3 targets. Indeed, SOD3 reduced the transcription of well-known NF-κB target genes as well as NF-κB-driven promoter activity in ECs stimulated with tumor necrosis factor (TNF)-α, an NF-κB signaling inducer. SOD3 inhibited the phosphorylation and degradation of IκBα (nuclear factor of the kappa light polypeptide gene enhancer in B-cells inhibitor alpha), an NF-κB inhibitor. Finally, TNF-α was found to be a transcriptional activator of LAMA5 but not of LAMA4; LAMA5 induction was prevented by SOD3. In conclusion, SOD3 is a major regulator of laminin balance in the basement membrane of tumor ECs, with potential implications for immune cell infiltration into tumors.
Collapse
|
50
|
Ayoub NM, Jaradat SK, Al-Shami KM, Alkhalifa AE. Targeting Angiogenesis in Breast Cancer: Current Evidence and Future Perspectives of Novel Anti-Angiogenic Approaches. Front Pharmacol 2022; 13:838133. [PMID: 35281942 PMCID: PMC8913593 DOI: 10.3389/fphar.2022.838133] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/03/2022] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is a vital process for the growth and dissemination of solid cancers. Numerous molecular pathways are known to drive angiogenic switch in cancer cells promoting the growth of new blood vessels and increased incidence of distant metastasis. Several angiogenesis inhibitors are clinically available for the treatment of different types of advanced solid cancers. These inhibitors mostly belong to monoclonal antibodies or small-molecule tyrosine kinase inhibitors targeting the classical vascular endothelial growth factor (VEGF) and its receptors. Nevertheless, breast cancer is one example of solid tumors that had constantly failed to respond to angiogenesis inhibitors in terms of improved survival outcomes of patients. Accordingly, it is of paramount importance to assess the molecular mechanisms driving angiogenic signaling in breast cancer to explore suitable drug targets that can be further investigated in preclinical and clinical settings. This review summarizes the current evidence for the effect of clinically available anti-angiogenic drugs in breast cancer treatment. Further, major mechanisms associated with intrinsic or acquired resistance to anti-VEGF therapy are discussed. The review also describes evidence from preclinical and clinical studies on targeting novel non-VEGF angiogenic pathways in breast cancer and several approaches to the normalization of tumor vasculature by targeting pericytes, utilization of microRNAs and extracellular tumor-associate vesicles, using immunotherapeutic drugs, and nanotechnology.
Collapse
Affiliation(s)
- Nehad M. Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
- *Correspondence: Nehad M. Ayoub,
| | - Sara K. Jaradat
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| | - Kamal M. Al-Shami
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Amer E. Alkhalifa
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| |
Collapse
|