Skip to main content
Top
Published in: Breast Cancer Research 1/2019

Open Access 01-12-2019 | Breast Cancer | Research article

Combining the oncolytic peptide LTX-315 with doxorubicin demonstrates therapeutic potential in a triple-negative breast cancer model

Authors: Ketil A. Camilio, Meng-Yu Wang, Brynjar Mauseth, Stein Waagene, Gunnar Kvalheim, Øystein Rekdal, Baldur Sveinbjørnsson, Gunhild M. Mælandsmo

Published in: Breast Cancer Research | Issue 1/2019

Login to get access

Abstract

Background

Immunochemotherapy, the combined use of immunotherapy and chemotherapy, has demonstrated great promise in several cancers. LTX-315 is an oncolytic peptide with potent immunomodulatory properties designed for the local treatment of solid tumors. By inducing rapid immunogenic cell death through the release of danger-associated molecular pattern molecules (DAMPs), LTX-315 is capable of reshaping the tumor microenvironment, turning “cold” tumors “hot” through a significant increase in tumor-infiltrating lymphocytes.

Methods

We investigated the potential of LTX-315 to be used in combination with standard-of-care chemotherapy (doxorubicin, brand name CAELYX®) against triple-negative breast cancer in an orthotopic 4 T1 mammary fat pad model. Tumor growth curves were compared using one-way ANOVA analysis of variance and Tukey’s multiple comparisons test, and animal survival curves were compared using the log-rank (Mantel-Cox) test. We considered p values ≤0.05 to indicate statistical significance.

Results

We found that LTX-315 displayed a strong additive antitumoral effect when used in combination with CAELYX®, and induced immune-mediated changes in the tumor microenvironment, followed by complete regression in the majority of animals treated. Furthermore, imaging techniques and histological examination showed that the combination induced strong local necrosis, followed by an increase in the infiltration of CD4+ and CD8+ immune cells into the tumor parenchymal tissue.

Conclusions

Our data demonstrate that LTX-315 is a promising combination partner with CAELYX® for the treatment of triple-negative breast cancer.
Literature
1.
2.
go back to reference Denkert C, et al. Molecular alterations in triple-negative breast cancer—the road to new treatment strategies. Lancet. 2017;389(10087):2430–42.PubMedCrossRef Denkert C, et al. Molecular alterations in triple-negative breast cancer—the road to new treatment strategies. Lancet. 2017;389(10087):2430–42.PubMedCrossRef
3.
go back to reference Engebraaten O, Vollan HKM, Børresen-Dale A-L. Triple-negative breast cancer and the need for new therapeutic targets. Am J Pathol. 2013;183(4):1064–74.PubMedCrossRef Engebraaten O, Vollan HKM, Børresen-Dale A-L. Triple-negative breast cancer and the need for new therapeutic targets. Am J Pathol. 2013;183(4):1064–74.PubMedCrossRef
5.
go back to reference Mahmoud SMA, et al. Tumor-Infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J Clin Oncol. 2011;29(15):1949–55.CrossRef Mahmoud SMA, et al. Tumor-Infiltrating CD8+ lymphocytes predict clinical outcome in breast cancer. J Clin Oncol. 2011;29(15):1949–55.CrossRef
6.
7.
8.
go back to reference Shai Y. Mechanism of the binding, insertion and destabilization of phospholipid bilayer membranes by α-helical antimicrobial and cell non-selective membrane-lytic peptides. Biochim Biophys Acta. 1999;1462(1–2):55–70.PubMedCrossRef Shai Y. Mechanism of the binding, insertion and destabilization of phospholipid bilayer membranes by α-helical antimicrobial and cell non-selective membrane-lytic peptides. Biochim Biophys Acta. 1999;1462(1–2):55–70.PubMedCrossRef
9.
go back to reference Domalaon R, et al. Ultrashort cationic lipopeptides and lipopeptoids: evaluation and mechanistic insights against epithelial cancer cells. Peptides. 2016;84(Supplement C):58–67.PubMedCrossRef Domalaon R, et al. Ultrashort cationic lipopeptides and lipopeptoids: evaluation and mechanistic insights against epithelial cancer cells. Peptides. 2016;84(Supplement C):58–67.PubMedCrossRef
11.
go back to reference Gaspar D, Veiga AS, Castanho MARB. From antimicrobial to anticancer peptides. Rev Front Microbiol. 2013;4:294. Gaspar D, Veiga AS, Castanho MARB. From antimicrobial to anticancer peptides. Rev Front Microbiol. 2013;4:294.
12.
go back to reference Riedl S, et al. In vitro and in vivo cytotoxic activity of human lactoferricin derived antitumor peptide R-DIM-P-LF11-334 on human malignant melanoma. Oncotarget. 2017;8(42):71817–32.PubMedPubMedCentralCrossRef Riedl S, et al. In vitro and in vivo cytotoxic activity of human lactoferricin derived antitumor peptide R-DIM-P-LF11-334 on human malignant melanoma. Oncotarget. 2017;8(42):71817–32.PubMedPubMedCentralCrossRef
13.
go back to reference Riedl S, Zweytick D, Lohner K. Membrane-active host defense peptides - challenges and perspectives for the development of novel anticancer drugs. Chem Phys Lipids. 2011;164(8):766–81.PubMedPubMedCentralCrossRef Riedl S, Zweytick D, Lohner K. Membrane-active host defense peptides - challenges and perspectives for the development of novel anticancer drugs. Chem Phys Lipids. 2011;164(8):766–81.PubMedPubMedCentralCrossRef
14.
go back to reference Schweizer F. Cationic amphiphilic peptides with cancer-selective toxicity. Eur J Pharmacol. 2009;625(1–3):190–4.PubMedCrossRef Schweizer F. Cationic amphiphilic peptides with cancer-selective toxicity. Eur J Pharmacol. 2009;625(1–3):190–4.PubMedCrossRef
15.
go back to reference Wu D, et al. Peptide-based cancer therapy: opportunity and challenge. Cancer Lett. 2014;351(1):13–22.PubMedCrossRef Wu D, et al. Peptide-based cancer therapy: opportunity and challenge. Cancer Lett. 2014;351(1):13–22.PubMedCrossRef
16.
go back to reference Camilio KA, Rekdal Ø, Sveinbjörnsson B. LTX-315 (Oncopore™): A short synthetic anticancer peptide and novel immunotherapeutic agent. Oncoimmunology. 2014;3:e29181.PubMedPubMedCentralCrossRef Camilio KA, Rekdal Ø, Sveinbjörnsson B. LTX-315 (Oncopore™): A short synthetic anticancer peptide and novel immunotherapeutic agent. Oncoimmunology. 2014;3:e29181.PubMedPubMedCentralCrossRef
17.
go back to reference Haug BE, et al. Discovery of a 9-mer cationic peptide (LTX-315) as a potential first in class oncolytic peptide. J Med Chem. 2016;59(7):2918–27.PubMedCrossRef Haug BE, et al. Discovery of a 9-mer cationic peptide (LTX-315) as a potential first in class oncolytic peptide. J Med Chem. 2016;59(7):2918–27.PubMedCrossRef
18.
go back to reference Eike L-M, et al. The oncolytic peptide LTX-315 induces cell death and DAMP release by mitochondria distortion in human melanoma cells. Oncotarget. 2015;6(33):34910–23.PubMedPubMedCentralCrossRef Eike L-M, et al. The oncolytic peptide LTX-315 induces cell death and DAMP release by mitochondria distortion in human melanoma cells. Oncotarget. 2015;6(33):34910–23.PubMedPubMedCentralCrossRef
20.
go back to reference Zhou H, et al. The oncolytic peptide LTX-315 kills cancer cells through Bax/Bak-regulated mitochondrial membrane permeabilization. Oncotarget. 2015;6(29):26599–614.PubMedPubMedCentralCrossRef Zhou H, et al. The oncolytic peptide LTX-315 kills cancer cells through Bax/Bak-regulated mitochondrial membrane permeabilization. Oncotarget. 2015;6(29):26599–614.PubMedPubMedCentralCrossRef
22.
go back to reference Camilio KA, et al. Complete regression and systemic protective immune responses obtained in B16 melanomas after treatment with LTX-315. Cancer Immunol Immunother. 2014;63(6):601–13.PubMedPubMedCentralCrossRef Camilio KA, et al. Complete regression and systemic protective immune responses obtained in B16 melanomas after treatment with LTX-315. Cancer Immunol Immunother. 2014;63(6):601–13.PubMedPubMedCentralCrossRef
23.
24.
go back to reference Yamazaki T, et al. The oncolytic peptide LTX-315 overcomes resistance of cancers to immunotherapy with CTLA4 checkpoint blockade. Cell Death Differ. 2016;23(6):1004–15.PubMedPubMedCentralCrossRef Yamazaki T, et al. The oncolytic peptide LTX-315 overcomes resistance of cancers to immunotherapy with CTLA4 checkpoint blockade. Cell Death Differ. 2016;23(6):1004–15.PubMedPubMedCentralCrossRef
25.
go back to reference Abu Eid R, et al. Old-school chemotherapy in immunotherapeutic combination in cancer, a low-cost drug repurposed. Cancer Immunol Res. 2016;4(5):377–82.PubMedCrossRef Abu Eid R, et al. Old-school chemotherapy in immunotherapeutic combination in cancer, a low-cost drug repurposed. Cancer Immunol Res. 2016;4(5):377–82.PubMedCrossRef
26.
go back to reference Emens LA, Reilly RT, Jaffee EM. Breast cancer vaccines: maximizing cancer treatment by tapping into host immunity. Endocr Relat Cancer. 2005;12(1):1–17.PubMedCrossRef Emens LA, Reilly RT, Jaffee EM. Breast cancer vaccines: maximizing cancer treatment by tapping into host immunity. Endocr Relat Cancer. 2005;12(1):1–17.PubMedCrossRef
27.
go back to reference Sheng Sow H, Mattarollo SR. Combining low-dose or metronomic chemotherapy with anticancer vaccines: a therapeutic opportunity for lymphomas. Oncoimmunology. 2013;2(12):e27058.PubMedPubMedCentralCrossRef Sheng Sow H, Mattarollo SR. Combining low-dose or metronomic chemotherapy with anticancer vaccines: a therapeutic opportunity for lymphomas. Oncoimmunology. 2013;2(12):e27058.PubMedPubMedCentralCrossRef
28.
go back to reference Alizadeh D, et al. Doxorubicin eliminates myeloid-derived suppressor cells and enhances the efficacy of adoptive T-cell transfer in breast cancer. Cancer Res. 2014;74(1):104–18.PubMedCrossRef Alizadeh D, et al. Doxorubicin eliminates myeloid-derived suppressor cells and enhances the efficacy of adoptive T-cell transfer in breast cancer. Cancer Res. 2014;74(1):104–18.PubMedCrossRef
29.
30.
go back to reference Wu J, Waxman DJ. Immunogenic chemotherapy: dose and schedule dependence and combination with immunotherapy. Cancer Lett. 2018;419:210–21.PubMedCrossRef Wu J, Waxman DJ. Immunogenic chemotherapy: dose and schedule dependence and combination with immunotherapy. Cancer Lett. 2018;419:210–21.PubMedCrossRef
32.
go back to reference Morrissey KM, et al. Immunotherapy and novel combinations in oncology: current landscape, challenges, and opportunities. Clin Transl Sci. 2016;9(2):89–104.PubMedPubMedCentralCrossRef Morrissey KM, et al. Immunotherapy and novel combinations in oncology: current landscape, challenges, and opportunities. Clin Transl Sci. 2016;9(2):89–104.PubMedPubMedCentralCrossRef
33.
go back to reference Sveinbjørnsson B, et al. LTX-315: a first-in-class oncolytic peptide that reprograms the tumor microenvironment. Future Med Chem. 2017;9(12):1339–44.PubMedCrossRef Sveinbjørnsson B, et al. LTX-315: a first-in-class oncolytic peptide that reprograms the tumor microenvironment. Future Med Chem. 2017;9(12):1339–44.PubMedCrossRef
34.
go back to reference Altin JG, Sloan EK. The role of CD45 and CD45-associated molecules in T cell activation. Immunol Cell Biol. 1997;75(5):430–45.PubMedCrossRef Altin JG, Sloan EK. The role of CD45 and CD45-associated molecules in T cell activation. Immunol Cell Biol. 1997;75(5):430–45.PubMedCrossRef
36.
go back to reference Ostrand-Rosenberg S. CD4+ T lymphocytes: a critical component of antitumor immunity. Cancer Investig. 2005;23(5):413–9. Ostrand-Rosenberg S. CD4+ T lymphocytes: a critical component of antitumor immunity. Cancer Investig. 2005;23(5):413–9.
37.
go back to reference Church SE, et al. Tumor-specific CD4+ T cells maintain effector and memory tumor-specific CD8+ T cells. Eur J Immunol. 2014;44(1):69–79.PubMedCrossRef Church SE, et al. Tumor-specific CD4+ T cells maintain effector and memory tumor-specific CD8+ T cells. Eur J Immunol. 2014;44(1):69–79.PubMedCrossRef
38.
go back to reference Matsuzaki J, et al. Direct tumor recognition by a human CD4+ T-cell subset potently mediates tumor growth inhibition and orchestrates anti-tumor immune responses. Sci Rep. 2015;5:14896.PubMedPubMedCentralCrossRef Matsuzaki J, et al. Direct tumor recognition by a human CD4+ T-cell subset potently mediates tumor growth inhibition and orchestrates anti-tumor immune responses. Sci Rep. 2015;5:14896.PubMedPubMedCentralCrossRef
39.
go back to reference Galon J, et al. Microenvironnement immunitaire et cancer. Med Sci (Paris). 2014;30(4):439–44.CrossRef Galon J, et al. Microenvironnement immunitaire et cancer. Med Sci (Paris). 2014;30(4):439–44.CrossRef
40.
go back to reference Galluzzi L, et al. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov. 2012;11(3):215–33.PubMedCrossRef Galluzzi L, et al. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov. 2012;11(3):215–33.PubMedCrossRef
41.
42.
go back to reference Aymeric L, et al. Tumor cell death and ATP release prime dendritic cells and efficient anticancer immunity. Cancer Res. 2010;70(3):855–8.PubMedCrossRef Aymeric L, et al. Tumor cell death and ATP release prime dendritic cells and efficient anticancer immunity. Cancer Res. 2010;70(3):855–8.PubMedCrossRef
43.
go back to reference Chan T-S, et al. Metronomic chemotherapy prevents therapy-induced stromal activation and induction of tumor-initiating cells. J Exp Med. 2016;213:2967.PubMedPubMedCentralCrossRef Chan T-S, et al. Metronomic chemotherapy prevents therapy-induced stromal activation and induction of tumor-initiating cells. J Exp Med. 2016;213:2967.PubMedPubMedCentralCrossRef
44.
go back to reference Denkert C, et al. Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer. J Clin Oncol. 2010;28(1):105–13.PubMedCrossRef Denkert C, et al. Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer. J Clin Oncol. 2010;28(1):105–13.PubMedCrossRef
45.
go back to reference Loi S, et al. Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin With doxorubicin-based chemotherapy: BIG 02-98. J Clin Oncol. 2013;31(7):860–7.PubMedCrossRef Loi S, et al. Prognostic and predictive value of tumor-infiltrating lymphocytes in a phase III randomized adjuvant breast cancer trial in node-positive breast cancer comparing the addition of docetaxel to doxorubicin With doxorubicin-based chemotherapy: BIG 02-98. J Clin Oncol. 2013;31(7):860–7.PubMedCrossRef
Metadata
Title
Combining the oncolytic peptide LTX-315 with doxorubicin demonstrates therapeutic potential in a triple-negative breast cancer model
Authors
Ketil A. Camilio
Meng-Yu Wang
Brynjar Mauseth
Stein Waagene
Gunnar Kvalheim
Øystein Rekdal
Baldur Sveinbjørnsson
Gunhild M. Mælandsmo
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Breast Cancer Research / Issue 1/2019
Electronic ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-018-1092-x

Other articles of this Issue 1/2019

Breast Cancer Research 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine