Inhibition of the PI3K/mTOR Pathway in Breast Cancer to Enhance Response to Immune Checkpoint Inhibitors in Breast Cancer

Objectives: Inhibition of the PI3K/mTOR pathway suppresses breast cancer (BC) growth, enhances anti-tumor immune responses, and works synergistically with immune checkpoint inhibitors (ICI). The objective here was to identify a subclass of PI3K inhibitors that, when combined with paclitaxel, is effe...

Full description

Bibliographic Details
Main Authors: Chi Yan, Jinming Yang, Nabil Saleh, Sheau-Chiann Chen, Gregory D. Ayers, Vandana G. Abramson, Ingrid A. Mayer, Ann Richmond
Format: Article
Language:English
Published: MDPI AG 2021-05-01
Series:International Journal of Molecular Sciences
Subjects:
Online Access:https://www.mdpi.com/1422-0067/22/10/5207
_version_ 1797534078072782848
author Chi Yan
Jinming Yang
Nabil Saleh
Sheau-Chiann Chen
Gregory D. Ayers
Vandana G. Abramson
Ingrid A. Mayer
Ann Richmond
author_facet Chi Yan
Jinming Yang
Nabil Saleh
Sheau-Chiann Chen
Gregory D. Ayers
Vandana G. Abramson
Ingrid A. Mayer
Ann Richmond
author_sort Chi Yan
collection DOAJ
description Objectives: Inhibition of the PI3K/mTOR pathway suppresses breast cancer (BC) growth, enhances anti-tumor immune responses, and works synergistically with immune checkpoint inhibitors (ICI). The objective here was to identify a subclass of PI3K inhibitors that, when combined with paclitaxel, is effective in enhancing response to ICI. Methods: C57BL/6 mice were orthotopically implanted with syngeneic luminal/triple-negative-like PyMT cells exhibiting high endogenous PI3K activity. Tumor growth in response to treatment with anti-PD-1 + anti-CTLA-4 (ICI), paclitaxel (PTX), and either the PI3Kα-specific inhibitor alpelisib, the pan-PI3K inhibitor copanlisib, or the broad spectrum PI3K/mTOR inhibitor gedatolisib was evaluated in reference to monotherapy or combinations of these therapies. Effects of these therapeutics on intratumoral immune populations were determined by multicolor FACS. Results: Treatment with alpelisib + PTX inhibited PyMT tumor growth and increased tumor-infiltrating granulocytes but did not significantly affect the number of tumor-infiltrating CD8<sup>+</sup> T cells and did not synergize with ICI. Copanlisib + PTX + ICI significantly inhibited PyMT growth and increased activation of intratumoral CD8<sup>+</sup> T cells as compared to ICI alone, yet did not inhibit tumor growth more than ICI alone. In contrast, gedatolisib + ICI resulted in significantly greater inhibition of tumor growth compared to ICI alone and induced durable dendritic-cell, CD8<sup>+</sup> T-cell, and NK-cell responses. Adding PTX to this regimen yielded complete regression in 60% of tumors. Conclusion: PI3K/mTOR inhibition plus PTX heightens response to ICI and may provide a viable therapeutic approach for treatment of metastatic BC.
first_indexed 2024-03-10T11:24:32Z
format Article
id doaj.art-0f72cf8c86ae470c82c49f9672274a3e
institution Directory Open Access Journal
issn 1661-6596
1422-0067
language English
last_indexed 2024-03-10T11:24:32Z
publishDate 2021-05-01
publisher MDPI AG
record_format Article
series International Journal of Molecular Sciences
spelling doaj.art-0f72cf8c86ae470c82c49f9672274a3e2023-11-21T19:44:50ZengMDPI AGInternational Journal of Molecular Sciences1661-65961422-00672021-05-012210520710.3390/ijms22105207Inhibition of the PI3K/mTOR Pathway in Breast Cancer to Enhance Response to Immune Checkpoint Inhibitors in Breast CancerChi Yan0Jinming Yang1Nabil Saleh2Sheau-Chiann Chen3Gregory D. Ayers4Vandana G. Abramson5Ingrid A. Mayer6Ann Richmond7Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USADepartment of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USADepartment of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USADepartment of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USADepartment of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37232, USADepartment of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USADepartment of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USADepartment of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USAObjectives: Inhibition of the PI3K/mTOR pathway suppresses breast cancer (BC) growth, enhances anti-tumor immune responses, and works synergistically with immune checkpoint inhibitors (ICI). The objective here was to identify a subclass of PI3K inhibitors that, when combined with paclitaxel, is effective in enhancing response to ICI. Methods: C57BL/6 mice were orthotopically implanted with syngeneic luminal/triple-negative-like PyMT cells exhibiting high endogenous PI3K activity. Tumor growth in response to treatment with anti-PD-1 + anti-CTLA-4 (ICI), paclitaxel (PTX), and either the PI3Kα-specific inhibitor alpelisib, the pan-PI3K inhibitor copanlisib, or the broad spectrum PI3K/mTOR inhibitor gedatolisib was evaluated in reference to monotherapy or combinations of these therapies. Effects of these therapeutics on intratumoral immune populations were determined by multicolor FACS. Results: Treatment with alpelisib + PTX inhibited PyMT tumor growth and increased tumor-infiltrating granulocytes but did not significantly affect the number of tumor-infiltrating CD8<sup>+</sup> T cells and did not synergize with ICI. Copanlisib + PTX + ICI significantly inhibited PyMT growth and increased activation of intratumoral CD8<sup>+</sup> T cells as compared to ICI alone, yet did not inhibit tumor growth more than ICI alone. In contrast, gedatolisib + ICI resulted in significantly greater inhibition of tumor growth compared to ICI alone and induced durable dendritic-cell, CD8<sup>+</sup> T-cell, and NK-cell responses. Adding PTX to this regimen yielded complete regression in 60% of tumors. Conclusion: PI3K/mTOR inhibition plus PTX heightens response to ICI and may provide a viable therapeutic approach for treatment of metastatic BC.https://www.mdpi.com/1422-0067/22/10/5207PI3K/mTORbreast cancerimmune checkpoint inhibitorcytotoxic T cells
spellingShingle Chi Yan
Jinming Yang
Nabil Saleh
Sheau-Chiann Chen
Gregory D. Ayers
Vandana G. Abramson
Ingrid A. Mayer
Ann Richmond
Inhibition of the PI3K/mTOR Pathway in Breast Cancer to Enhance Response to Immune Checkpoint Inhibitors in Breast Cancer
International Journal of Molecular Sciences
PI3K/mTOR
breast cancer
immune checkpoint inhibitor
cytotoxic T cells
title Inhibition of the PI3K/mTOR Pathway in Breast Cancer to Enhance Response to Immune Checkpoint Inhibitors in Breast Cancer
title_full Inhibition of the PI3K/mTOR Pathway in Breast Cancer to Enhance Response to Immune Checkpoint Inhibitors in Breast Cancer
title_fullStr Inhibition of the PI3K/mTOR Pathway in Breast Cancer to Enhance Response to Immune Checkpoint Inhibitors in Breast Cancer
title_full_unstemmed Inhibition of the PI3K/mTOR Pathway in Breast Cancer to Enhance Response to Immune Checkpoint Inhibitors in Breast Cancer
title_short Inhibition of the PI3K/mTOR Pathway in Breast Cancer to Enhance Response to Immune Checkpoint Inhibitors in Breast Cancer
title_sort inhibition of the pi3k mtor pathway in breast cancer to enhance response to immune checkpoint inhibitors in breast cancer
topic PI3K/mTOR
breast cancer
immune checkpoint inhibitor
cytotoxic T cells
url https://www.mdpi.com/1422-0067/22/10/5207
work_keys_str_mv AT chiyan inhibitionofthepi3kmtorpathwayinbreastcancertoenhanceresponsetoimmunecheckpointinhibitorsinbreastcancer
AT jinmingyang inhibitionofthepi3kmtorpathwayinbreastcancertoenhanceresponsetoimmunecheckpointinhibitorsinbreastcancer
AT nabilsaleh inhibitionofthepi3kmtorpathwayinbreastcancertoenhanceresponsetoimmunecheckpointinhibitorsinbreastcancer
AT sheauchiannchen inhibitionofthepi3kmtorpathwayinbreastcancertoenhanceresponsetoimmunecheckpointinhibitorsinbreastcancer
AT gregorydayers inhibitionofthepi3kmtorpathwayinbreastcancertoenhanceresponsetoimmunecheckpointinhibitorsinbreastcancer
AT vandanagabramson inhibitionofthepi3kmtorpathwayinbreastcancertoenhanceresponsetoimmunecheckpointinhibitorsinbreastcancer
AT ingridamayer inhibitionofthepi3kmtorpathwayinbreastcancertoenhanceresponsetoimmunecheckpointinhibitorsinbreastcancer
AT annrichmond inhibitionofthepi3kmtorpathwayinbreastcancertoenhanceresponsetoimmunecheckpointinhibitorsinbreastcancer