Targeting Tumor Acidosis and Regulatory T Cells Unmasks Anti-Metastatic Potential of Local Tumor Ablation in Triple-Negative Breast Cancer

Triple-negative breast cancer (TNBC) is an immunologically heterogenous disease that lacks clinically actionable targets and is more likely to progress to metastatic disease than other types of breast cancer. Tumor ablation has been used to increase response rates to checkpoint inhibitors, which rem...

Full description

Bibliographic Details
Main Authors: Corrine A. Nief, Alana Gonzales, Erika Chelales, Júlia Sroda Agudogo, Brian T. Crouch, Smita K. Nair, Nirmala Ramanujam
Format: Article
Language:English
Published: MDPI AG 2022-07-01
Series:International Journal of Molecular Sciences
Subjects:
Online Access:https://www.mdpi.com/1422-0067/23/15/8479
_version_ 1797432818419105792
author Corrine A. Nief
Alana Gonzales
Erika Chelales
Júlia Sroda Agudogo
Brian T. Crouch
Smita K. Nair
Nirmala Ramanujam
author_facet Corrine A. Nief
Alana Gonzales
Erika Chelales
Júlia Sroda Agudogo
Brian T. Crouch
Smita K. Nair
Nirmala Ramanujam
author_sort Corrine A. Nief
collection DOAJ
description Triple-negative breast cancer (TNBC) is an immunologically heterogenous disease that lacks clinically actionable targets and is more likely to progress to metastatic disease than other types of breast cancer. Tumor ablation has been used to increase response rates to checkpoint inhibitors, which remain low for TNBC patients. We hypothesized that tumor ablation could produce an anti-tumor response without using checkpoint inhibitors if immunosuppression (i.e., Tregs, tumor acidosis) was subdued. Tumors were primed with sodium bicarbonate (200 mM p.o.) to reduce tumor acidosis and low-dose cyclophosphamide (100–200 mg/kg i.p.) to deplete regulatory T cells, as has been shown independently in previous studies. A novel injectable ablative was then used to necrose the tumor, release tumor antigens, and initiate an immune event that could create an abscopal effect. This combination of bicarbonate, cyclophosphamide, and ablation, called “BiCyclA”, was tested in three syngeneic models of TNBC: E0771 (C57BL/6), 67NR (BALB/c), and 4T1-Luc (BALB/c). In E0771 and 67NR, BiCyclA therapy significantly reduced tumor growth and cured 5/7 and 6/10 mice 50 days after treatment respectively. In the metastatic 4T1-Luc tumors, for which surgery and checkpoint inhibitors fail, BiCyclA cured 5/10 mice of primary tumors and lung metastases. Notably, CD4+ and CD8+ T cells were found to be crucial for the anti-metastatic response, and cured mice were able to resist tumor rechallenge, suggesting production of immune memory. Reduction of tumor acidity and regulatory T cells with ablation is a simple yet effective therapy for local and systemic tumor control with broad applicability as it is not limited by expensive supplies.
first_indexed 2024-03-09T10:08:17Z
format Article
id doaj.art-1cee492a216641638ff1f1200ad3651a
institution Directory Open Access Journal
issn 1661-6596
1422-0067
language English
last_indexed 2024-03-09T10:08:17Z
publishDate 2022-07-01
publisher MDPI AG
record_format Article
series International Journal of Molecular Sciences
spelling doaj.art-1cee492a216641638ff1f1200ad3651a2023-12-01T22:57:43ZengMDPI AGInternational Journal of Molecular Sciences1661-65961422-00672022-07-012315847910.3390/ijms23158479Targeting Tumor Acidosis and Regulatory T Cells Unmasks Anti-Metastatic Potential of Local Tumor Ablation in Triple-Negative Breast CancerCorrine A. Nief0Alana Gonzales1Erika Chelales2Júlia Sroda Agudogo3Brian T. Crouch4Smita K. Nair5Nirmala Ramanujam6Department of Biomedical Engineering, Duke University, Durham, NC 27708, USADepartment of Biomedical Engineering, Duke University, Durham, NC 27708, USADepartment of Biomedical Engineering, Duke University, Durham, NC 27708, USADepartment of Biomedical Engineering, Duke University, Durham, NC 27708, USADepartment of Biomedical Engineering, Duke University, Durham, NC 27708, USADepartment of Surgery, Duke University School of Medicine, Durham, NC 27708, USADepartment of Biomedical Engineering, Duke University, Durham, NC 27708, USATriple-negative breast cancer (TNBC) is an immunologically heterogenous disease that lacks clinically actionable targets and is more likely to progress to metastatic disease than other types of breast cancer. Tumor ablation has been used to increase response rates to checkpoint inhibitors, which remain low for TNBC patients. We hypothesized that tumor ablation could produce an anti-tumor response without using checkpoint inhibitors if immunosuppression (i.e., Tregs, tumor acidosis) was subdued. Tumors were primed with sodium bicarbonate (200 mM p.o.) to reduce tumor acidosis and low-dose cyclophosphamide (100–200 mg/kg i.p.) to deplete regulatory T cells, as has been shown independently in previous studies. A novel injectable ablative was then used to necrose the tumor, release tumor antigens, and initiate an immune event that could create an abscopal effect. This combination of bicarbonate, cyclophosphamide, and ablation, called “BiCyclA”, was tested in three syngeneic models of TNBC: E0771 (C57BL/6), 67NR (BALB/c), and 4T1-Luc (BALB/c). In E0771 and 67NR, BiCyclA therapy significantly reduced tumor growth and cured 5/7 and 6/10 mice 50 days after treatment respectively. In the metastatic 4T1-Luc tumors, for which surgery and checkpoint inhibitors fail, BiCyclA cured 5/10 mice of primary tumors and lung metastases. Notably, CD4+ and CD8+ T cells were found to be crucial for the anti-metastatic response, and cured mice were able to resist tumor rechallenge, suggesting production of immune memory. Reduction of tumor acidity and regulatory T cells with ablation is a simple yet effective therapy for local and systemic tumor control with broad applicability as it is not limited by expensive supplies.https://www.mdpi.com/1422-0067/23/15/8479ablationtumor microenvironmentimmunomodulationbreast cancerlow-resource
spellingShingle Corrine A. Nief
Alana Gonzales
Erika Chelales
Júlia Sroda Agudogo
Brian T. Crouch
Smita K. Nair
Nirmala Ramanujam
Targeting Tumor Acidosis and Regulatory T Cells Unmasks Anti-Metastatic Potential of Local Tumor Ablation in Triple-Negative Breast Cancer
International Journal of Molecular Sciences
ablation
tumor microenvironment
immunomodulation
breast cancer
low-resource
title Targeting Tumor Acidosis and Regulatory T Cells Unmasks Anti-Metastatic Potential of Local Tumor Ablation in Triple-Negative Breast Cancer
title_full Targeting Tumor Acidosis and Regulatory T Cells Unmasks Anti-Metastatic Potential of Local Tumor Ablation in Triple-Negative Breast Cancer
title_fullStr Targeting Tumor Acidosis and Regulatory T Cells Unmasks Anti-Metastatic Potential of Local Tumor Ablation in Triple-Negative Breast Cancer
title_full_unstemmed Targeting Tumor Acidosis and Regulatory T Cells Unmasks Anti-Metastatic Potential of Local Tumor Ablation in Triple-Negative Breast Cancer
title_short Targeting Tumor Acidosis and Regulatory T Cells Unmasks Anti-Metastatic Potential of Local Tumor Ablation in Triple-Negative Breast Cancer
title_sort targeting tumor acidosis and regulatory t cells unmasks anti metastatic potential of local tumor ablation in triple negative breast cancer
topic ablation
tumor microenvironment
immunomodulation
breast cancer
low-resource
url https://www.mdpi.com/1422-0067/23/15/8479
work_keys_str_mv AT corrineanief targetingtumoracidosisandregulatorytcellsunmasksantimetastaticpotentialoflocaltumorablationintriplenegativebreastcancer
AT alanagonzales targetingtumoracidosisandregulatorytcellsunmasksantimetastaticpotentialoflocaltumorablationintriplenegativebreastcancer
AT erikachelales targetingtumoracidosisandregulatorytcellsunmasksantimetastaticpotentialoflocaltumorablationintriplenegativebreastcancer
AT juliasrodaagudogo targetingtumoracidosisandregulatorytcellsunmasksantimetastaticpotentialoflocaltumorablationintriplenegativebreastcancer
AT briantcrouch targetingtumoracidosisandregulatorytcellsunmasksantimetastaticpotentialoflocaltumorablationintriplenegativebreastcancer
AT smitaknair targetingtumoracidosisandregulatorytcellsunmasksantimetastaticpotentialoflocaltumorablationintriplenegativebreastcancer
AT nirmalaramanujam targetingtumoracidosisandregulatorytcellsunmasksantimetastaticpotentialoflocaltumorablationintriplenegativebreastcancer