1
|
Battistoni A, Lantier L, di Tommaso A, Ducournau C, Lajoie L, Samimi M, Coënon L, Rivière C, Epardaud M, Hertereau L, Poupée-Beaugé A, Rieu J, Mévélec MN, Lee GS, Moiré N, Germon S, Dimier-Poisson I. Nasal administration of recombinant Neospora caninum secreting IL-15/IL-15Rα inhibits metastatic melanoma development in lung. J Immunother Cancer 2023; 11:jitc-2023-006683. [PMID: 37192784 DOI: 10.1136/jitc-2023-006683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2023] [Indexed: 05/18/2023] Open
Abstract
BACKGROUND Metastases are the leading cause of mortality in many cancer types and lungs are one of the most common sites of metastasis alongside the liver, brain, and bones. In melanoma, 85% of late-stage patients harbor lung metastases. A local administration could enhance the targeting of metastases while limiting the systemic cytotoxicity. Therefore, intranasal administration of immunotherapeutic agents seems to be a promising approach to preferentially target lung metastases and decrease their burden on cancer mortality. From observations that certain microorganisms induce an acute infection of the tumor microenvironment leading to a local reactivating immune response, microbial-mediated immunotherapy is a next-generation field of investigation in which immunotherapies are engineered to overcome immune surveillance and escape from microenvironmental cancer defenses. METHODS The goal of our study is to evaluate the potential of the intranasal administration of Neospora caninum in a syngeneic C57BL6 mouse model of B16F10 melanoma lung metastases. It also compares the antitumoral properties of a wild-type N. caninum versus N. caninum secreting human interleukin (IL)-15 fused to the sushi domain of the IL-15 receptor α chain, a potent activator of cellular immune responses. RESULTS The treatment of murine lung metastases by intranasal administration of an N. caninum engineered to secrete human IL-15 impairs lung metastases from further progression with only 0,08% of lung surface harboring metastases versus 4,4% in wild-type N. caninum treated mice and 36% in untreated mice. The control of tumor development is associated with a strong increase in numbers, within the lung, of natural killer cells, CD8+ T cells and macrophages, up to twofold, fivefold and sixfold, respectively. Analysis of expression levels of CD86 and CD206 on macrophages surface revealed a polarization of these macrophages towards an antitumoral M1 phenotype. CONCLUSION Administration of IL-15/IL-15Rα-secreting N. caninum through intranasal administration, a non-invasive route, lend further support to N. caninum-demonstrated clear potential as an effective and safe immunotherapeutic approach for the treatment of metastatic solid cancers, whose existing therapeutic options are scarce. Combination of this armed protozoa with an intranasal route could reinforce the existing therapeutic arsenal against cancer and narrow the spectrum of incurable cancers.
Collapse
Affiliation(s)
- Arthur Battistoni
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | - Louis Lantier
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
- Kymeris Santé SA, Tours, France
| | - Anne di Tommaso
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | - Céline Ducournau
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | - Laurie Lajoie
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | - Mahtab Samimi
- Department de Dermatologie, CHRU de Tours, Tours, France
| | - Loïs Coënon
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | - Clément Rivière
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | | | - Leslie Hertereau
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | | | - Juliette Rieu
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | | | | | - Nathalie Moiré
- INRAE, Université de Tours, ISP, F-37380, Nouzilly, France
| | - Stephanie Germon
- Université de Tours, INRAE, ISP, F-37000, Faculté de pharmacie, Tours, France
| | | |
Collapse
|
2
|
Kramer ED, Tzetzo SL, Colligan SH, Hensen ML, Brackett CM, Clausen BE, Taketo MM, Abrams SI. β-Catenin signaling in alveolar macrophages enhances lung metastasis through a TNF-dependent mechanism. JCI Insight 2023; 8:e160978. [PMID: 37092550 PMCID: PMC10243816 DOI: 10.1172/jci.insight.160978] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 03/08/2023] [Indexed: 04/25/2023] Open
Abstract
The main cause of malignancy-related mortality is metastasis. Although metastatic progression is driven by diverse tumor-intrinsic mechanisms, there is a growing appreciation for the contribution of tumor-extrinsic elements of the tumor microenvironment, especially macrophages, which correlate with poor clinical outcomes. Macrophages consist of bone marrow-derived and tissue-resident populations. In contrast to bone marrow-derived macrophages, the transcriptional pathways that govern the pro-metastatic activities of tissue-resident macrophages (TRMs) remain less clear. Alveolar macrophages (AMs) are a TRM population with critical roles in tissue homeostasis and metastasis. Wnt/β-catenin signaling is a hallmark of cancer and has been identified as a pathologic regulator of AMs in infection. We tested the hypothesis that β-catenin expression in AMs enhances metastasis in solid tumor models. Using a genetic β-catenin gain-of-function approach, we demonstrated that (a) enhanced β-catenin in AMs heightened lung metastasis; (b) β-catenin activity in AMs drove a dysregulated inflammatory program strongly associated with Tnf expression; and (c) localized TNF-α blockade abrogated this metastatic outcome. Last, β-catenin gene CTNNB1 and TNF expression levels were positively correlated in AMs of patients with lung cancer. Overall, our findings revealed a Wnt/β-catenin/TNF-α pro-metastatic axis in AMs with potential therapeutic implications against tumors refractory to the antineoplastic actions of TNF-α.
Collapse
Affiliation(s)
| | | | | | | | - Craig M. Brackett
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Björn E. Clausen
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Makoto M. Taketo
- Division of Experimental Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | |
Collapse
|
3
|
Kyjacova L, Saup R, Rothley M, Schmaus A, Wagner T, Boßerhoff A, Garvalov BK, Thiele W, Sleeman JP. Quantitative Detection of Disseminated Melanoma Cells by Trp-1 Transcript Analysis Reveals Stochastic Distribution of Pulmonary Metastases. J Clin Med 2021; 10:jcm10225459. [PMID: 34830742 PMCID: PMC8618565 DOI: 10.3390/jcm10225459] [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: 10/15/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 11/29/2022] Open
Abstract
A better understanding of the process of melanoma metastasis is required to underpin the development of novel therapies that will improve patient outcomes. The use of appropriate animal models is indispensable for investigating the mechanisms of melanoma metastasis. However, reliable and practicable quantification of metastases in experimental mice remains a challenge, particularly if the metastatic burden is low. Here, we describe a qRT-PCR-based protocol that employs the melanocytic marker Trp-1 for the sensitive quantification of melanoma metastases in the murine lung. Using this protocol, we were able to detect the presence of as few as 100 disseminated melanoma cells in lung tissue. This allowed us to quantify metastatic burden in a spontaneous syngeneic B16-F10 metastasis model, even in the absence of visible metastases, as well as in the autochthonous Tg(Grm1)/Cyld−/− melanoma model. Importantly, we also observed an uneven distribution of disseminated melanoma cells amongst the five lobes of the murine lung, which varied considerably from animal to animal. Together, our findings demonstrate that the qRT-PCR-based detection of Trp-1 allows the quantification of low pulmonary metastatic burden in both transplantable and autochthonous murine melanoma models, and show that the analysis of lung metastasis in such models needs to take into account the stochastic distribution of metastatic lesions amongst the lung lobes.
Collapse
Affiliation(s)
- Lenka Kyjacova
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany; (L.K.); (R.S.); (M.R.); (A.S.); (T.W.); (B.K.G.); (W.T.)
| | - Rafael Saup
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany; (L.K.); (R.S.); (M.R.); (A.S.); (T.W.); (B.K.G.); (W.T.)
| | - Melanie Rothley
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany; (L.K.); (R.S.); (M.R.); (A.S.); (T.W.); (B.K.G.); (W.T.)
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT)-Campus North, D-76344 Karlsruhe, Germany
| | - Anja Schmaus
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany; (L.K.); (R.S.); (M.R.); (A.S.); (T.W.); (B.K.G.); (W.T.)
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT)-Campus North, D-76344 Karlsruhe, Germany
| | - Tabea Wagner
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany; (L.K.); (R.S.); (M.R.); (A.S.); (T.W.); (B.K.G.); (W.T.)
| | - Anja Boßerhoff
- Institute of Biochemistry, Faculty of Medicine, Friedrich-Alexander University Erlangen-Nürnberg (FAU), D-91054 Erlangen, Germany;
| | - Boyan K. Garvalov
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany; (L.K.); (R.S.); (M.R.); (A.S.); (T.W.); (B.K.G.); (W.T.)
| | - Wilko Thiele
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany; (L.K.); (R.S.); (M.R.); (A.S.); (T.W.); (B.K.G.); (W.T.)
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT)-Campus North, D-76344 Karlsruhe, Germany
| | - Jonathan P. Sleeman
- Department of Microvascular Biology and Pathobiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, D-68167 Mannheim, Germany; (L.K.); (R.S.); (M.R.); (A.S.); (T.W.); (B.K.G.); (W.T.)
- Institute for Biological and Chemical Systems-Biological Information Processing (IBCS-BIP), Karlsruhe Institute of Technology (KIT)-Campus North, D-76344 Karlsruhe, Germany
- Correspondence: ; Tel.: +49-621-383-71595
| |
Collapse
|
4
|
Akil H, Quintana M, Raymond JH, Billoux T, Benboubker V, Besse S, Auzeloux P, Delmas V, Petit V, Larue L, D’Incan M, Degoul F, Rouanet J. Efficacy of Targeted Radionuclide Therapy Using [ 131I]ICF01012 in 3D Pigmented BRAF- and NRAS-Mutant Melanoma Models and In Vivo NRAS-Mutant Melanoma. Cancers (Basel) 2021; 13:cancers13061421. [PMID: 33804655 PMCID: PMC8003594 DOI: 10.3390/cancers13061421] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Targeted radionuclide therapy (TRT) aims to selectively deliver radioactive molecules to tumor cells. For this purpose, we deliver iodine-131 ([131I]) to melanoma cells by using our laboratory-developed melanin specific radiotracer, the ICF01012. Approximately 50% and 20%–30% of human melanomas have activating mutation in BRAF or NRAS genes, respectively. These mutations lead to a constitutive activation of the MAPK/ERK pathway, which is known to be involved in tumor cells’ radioresistance. In this work, we showed using 3D in vitro tumor models, an additive efficiency of combining [131I]ICF01012-TRT and MAPK/ERK inhibitors in BRAF- and NRAS-mutant melanoma cells. In mice bearing NRASQ61K-mutated melanoma, TRT induced an impressive decrease in tumor growth, as well as a highly extended survival. Additionally, we showed that TRT reduces the metastatic capacity of melanoma, especially through lymph-node dissemination. These results are therefore of great interest, especially for patients with NRAS-mutant metastatic melanoma who currently lack specific efficient therapies. Abstract Purpose: To assess the efficiency of targeted radionuclide therapy (TRT), alone or in combination with MEK inhibitors (MEKi), in melanomas harboring constitutive MAPK/ERK activation responsible for tumor radioresistance. Methods: For TRT, we used a melanin radiotracer ([131I]ICF01012) currently in phase 1 clinical trial (NCT03784625). TRT alone or combined with MEKi was evaluated in three-dimensional melanoma spheroid models of human BRAFV600E SK-MEL-3, murine NRASQ61K 1007, and WT B16F10 melanomas. TRT in vivo biodistribution, dosimetry, efficiency, and molecular mechanisms were studied using the C57BL/6J-NRASQ61K 1007 syngeneic model. Results: TRT cooperated with MEKi to increase apoptosis in both BRAF- and NRAS-mutant spheroids. NRASQ61K spheroids were highly radiosensitive towards [131I]ICF01012-TRT. In mice bearing NRASQ61K 1007 melanoma, [131I]ICF01012 induced a significant extended survival (92 vs. 44 days, p < 0.0001), associated with a 93-Gy tumor deposit, and reduced lymph-node metastases. Comparative transcriptomic analyses confirmed a decrease in mitosis, proliferation, and metastasis signatures in TRT-treated vs. control tumors and suggest that TRT acts through an increase in oxidation and inflammation and P53 activation. Conclusion: Our data suggest that [131I]ICF01012-TRT and MEKi combination could be of benefit for advanced pigmented BRAF-mutant melanoma care and that [131I]ICF01012 alone could constitute a new potential NRAS-mutant melanoma treatment.
Collapse
Affiliation(s)
- Hussein Akil
- INSERM U1240, University of Clermont Auvergne, 58 rue Montalembert, 63000 Clermont-Ferrand, France; (H.A.); (M.Q.); (V.B.); (S.B.); (P.A.); (M.D.); (F.D.)
- CNRS 7276, INSERM U1262, 2 rue du Pr Descottes, 87025 Limoges, France
| | - Mercedes Quintana
- INSERM U1240, University of Clermont Auvergne, 58 rue Montalembert, 63000 Clermont-Ferrand, France; (H.A.); (M.Q.); (V.B.); (S.B.); (P.A.); (M.D.); (F.D.)
| | - Jérémy H. Raymond
- INSERM U1021, Normal and Pathological Development of Melanocytes, Institut Curie, PSL Research University, Campus Universitaire, 91898 Orsay, France; (J.H.R.); (V.D.); (V.P.); (L.L.)
- Campus Universitaire, University Paris-Sud, University Paris-Saclay, CNRS UMR3347, 91898 Orsay, France
- Equipes Labellisées-Ligue Contre le Cancer, Campus Universitaire, 91898 Orsay, France
| | - Tommy Billoux
- Cirmen, Centre Jean Perrin, 58 rue Montalembert, 63000 Clermont-Ferrand, France;
| | - Valentin Benboubker
- INSERM U1240, University of Clermont Auvergne, 58 rue Montalembert, 63000 Clermont-Ferrand, France; (H.A.); (M.Q.); (V.B.); (S.B.); (P.A.); (M.D.); (F.D.)
| | - Sophie Besse
- INSERM U1240, University of Clermont Auvergne, 58 rue Montalembert, 63000 Clermont-Ferrand, France; (H.A.); (M.Q.); (V.B.); (S.B.); (P.A.); (M.D.); (F.D.)
| | - Philippe Auzeloux
- INSERM U1240, University of Clermont Auvergne, 58 rue Montalembert, 63000 Clermont-Ferrand, France; (H.A.); (M.Q.); (V.B.); (S.B.); (P.A.); (M.D.); (F.D.)
| | - Véronique Delmas
- INSERM U1021, Normal and Pathological Development of Melanocytes, Institut Curie, PSL Research University, Campus Universitaire, 91898 Orsay, France; (J.H.R.); (V.D.); (V.P.); (L.L.)
- Campus Universitaire, University Paris-Sud, University Paris-Saclay, CNRS UMR3347, 91898 Orsay, France
- Equipes Labellisées-Ligue Contre le Cancer, Campus Universitaire, 91898 Orsay, France
| | - Valérie Petit
- INSERM U1021, Normal and Pathological Development of Melanocytes, Institut Curie, PSL Research University, Campus Universitaire, 91898 Orsay, France; (J.H.R.); (V.D.); (V.P.); (L.L.)
- Campus Universitaire, University Paris-Sud, University Paris-Saclay, CNRS UMR3347, 91898 Orsay, France
- Equipes Labellisées-Ligue Contre le Cancer, Campus Universitaire, 91898 Orsay, France
| | - Lionel Larue
- INSERM U1021, Normal and Pathological Development of Melanocytes, Institut Curie, PSL Research University, Campus Universitaire, 91898 Orsay, France; (J.H.R.); (V.D.); (V.P.); (L.L.)
- Campus Universitaire, University Paris-Sud, University Paris-Saclay, CNRS UMR3347, 91898 Orsay, France
- Equipes Labellisées-Ligue Contre le Cancer, Campus Universitaire, 91898 Orsay, France
| | - Michel D’Incan
- INSERM U1240, University of Clermont Auvergne, 58 rue Montalembert, 63000 Clermont-Ferrand, France; (H.A.); (M.Q.); (V.B.); (S.B.); (P.A.); (M.D.); (F.D.)
- Department of Dermatology and Oncodermatology, CHU Estaing, 1 Place Aubrac, 63000 Clermont-Ferrand, France
| | - Françoise Degoul
- INSERM U1240, University of Clermont Auvergne, 58 rue Montalembert, 63000 Clermont-Ferrand, France; (H.A.); (M.Q.); (V.B.); (S.B.); (P.A.); (M.D.); (F.D.)
- CNRS 6293 INSERM U1103, University of Clermont Auvergne, 28, Place Henri Dunant, 63000 Clermont-Ferrand, France
| | - Jacques Rouanet
- INSERM U1240, University of Clermont Auvergne, 58 rue Montalembert, 63000 Clermont-Ferrand, France; (H.A.); (M.Q.); (V.B.); (S.B.); (P.A.); (M.D.); (F.D.)
- Department of Dermatology and Oncodermatology, CHU Estaing, 1 Place Aubrac, 63000 Clermont-Ferrand, France
- Correspondence:
| |
Collapse
|
5
|
Farsaci F, Tellone E, Galtieri A, Ficarra S. A thermodynamic characterization of the phenomena evolving in cancer pathology by dielectric relaxation in blood: A new approach by construction of TTM (Thermodynamic Tumor Matrix). J Mol Liq 2020. [DOI: 10.1016/j.molliq.2020.113839] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
6
|
Pasqual-Melo G, Bernardes SS, Souza-Neto FP, Carrara IM, Ramalho LNZ, Marinello PC, Luiz RC, Cecchini R, Bekeschus S, Cecchini AL. The progression of metastatic melanoma augments a pro-oxidative milieu locally but not systemically. Pathol Res Pract 2020; 216:153218. [PMID: 33002848 DOI: 10.1016/j.prp.2020.153218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 10/23/2022]
Abstract
Malignant melanoma is the most dangerous form of skin cancer. Despite new therapies for melanoma treatment, effective therapy is mainly limited by excessive metastasis. Currently, the factors determining metastasis development are not elucidated, but oxidative stress was suggested to be involved. To this end, we analyzed oxidative stress parameters during the metastatic development using the syngeneic B16F10 melanoma model. An increase in blood plasma lipid peroxidation occurred at the earliest stage of the disease, with a progressive decrease in oxidative damage and an increase in antioxidant defense. Vice versa, increased lipid peroxidation and 3-nitrotyrosine, and decreased antioxidant parameters were observed in the metastatic nodules throughout the disease. This was concomitant with a progressive increase in vascular endothelial growth factor and proliferating cell nuclear antigen. We conclude that the oxidative stress in the bloodstream decreases during the metastatic process and that nitrosative stress increases during the proliferation and growth of metastatic nodules in the tumor microenvironment. These results will help to better understand the role of oxidative stress during melanoma metastasis.
Collapse
Affiliation(s)
- Gabriella Pasqual-Melo
- Laboratory of Molecular Pathology, State University of Londrina, Brazil; Laboratory of Pathophysiology and Free Radicals, State University of Londrina, Brazil; ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
| | - Sara S Bernardes
- Laboratory of Molecular Pathology, State University of Londrina, Brazil; Laboratory of Tissue Microenvironment, Federal University of Minas Gerais, Brazil
| | - Fernando P Souza-Neto
- Laboratory of Molecular Pathology, State University of Londrina, Brazil; Laboratory of Pathophysiology and Free Radicals, State University of Londrina, Brazil
| | - Iriana M Carrara
- Laboratory of Molecular Pathology, State University of Londrina, Brazil; Laboratory of Pathophysiology and Free Radicals, State University of Londrina, Brazil
| | | | | | - Rodrigo C Luiz
- Laboratory of Molecular Pathology, State University of Londrina, Brazil
| | - Rubens Cecchini
- Laboratory of Pathophysiology and Free Radicals, State University of Londrina, Brazil
| | - Sander Bekeschus
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
| | - Alessandra L Cecchini
- Laboratory of Molecular Pathology, State University of Londrina, Brazil; Laboratory of Pathophysiology and Free Radicals, State University of Londrina, Brazil.
| |
Collapse
|
7
|
Pol JG, Atherton MJ, Stephenson KB, Bridle BW, Workenhe ST, Kazdhan N, McGray AR, Wan Y, Kroemer G, Lichty BD. Enhanced immunotherapeutic profile of oncolytic virus-based cancer vaccination using cyclophosphamide preconditioning. J Immunother Cancer 2020; 8:jitc-2020-000981. [PMID: 32792361 PMCID: PMC7430484 DOI: 10.1136/jitc-2020-000981] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2020] [Indexed: 02/07/2023] Open
Abstract
Despite a sizeable body of research, the efficacy of therapeutic cancer vaccines remains limited when applied as sole agents. By using a prime:boost approach involving two viral cancer vaccines, we were able to generate large tumor-specific CD8+ T-cell responses in a murine model of disseminated pulmonary melanoma. Significant increases in the number and quality of circulating effector T-cells were documented when low-dose cyclophosphamide (CTX) was administered pre-vaccination to tumor-bearing but not tumor-free hosts. Interestingly, tumor-bearing mice receiving CTX and co-primed with a melanoma differentiation antigen together with an irrelevant control antigen exhibited significantly enhanced immunity against the tumor, but not the control antigen, in secondary lymphoid organs. This result highlighted an increased cancer-specific reactivity of vaccine-induced T-cell responses following CTX preconditioning. Additionally, an acute reduction of the frequency of peripheral regulatory T-cells (Tregs) was noticeable, particularly in the proliferating, presumably tumour-reactive, subset. Enhanced infiltration of lungs with multifunctional T-cells resulted in overt reduction in metastatic burden in mice pretreated with CTX. Despite doubling the median survival in comparison to untreated controls, most vaccinated mice ultimately succumbed to cancer progression. However, preconditioning of the virus-based vaccination with CTX resulted in a remarkable improvement of the therapeutic activity leading to complete remission in the majority of the animals. Collectively, these data reveal how CTX can potentiate specific cellular immunity in an antigen-restricted manner that is only observed in vaccinated tumor-bearing hosts while depleting replicating Tregs. A single low dose of CTX enhances antitumor immunity and the efficacy of this potent prime:boost platform by modulating the kinetics of the vaccine-specific responses. Clinical assessment of CTX combined with next-generation cancer vaccines is indicated.
Collapse
Affiliation(s)
- Jonathan G Pol
- McMaster Immunology Research Center, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada .,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U1138, Paris, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Sorbonne Université, Paris, France.,Université de Paris, Paris, France
| | - Matthew J Atherton
- McMaster Immunology Research Center, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Kyle B Stephenson
- McMaster Immunology Research Center, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Byram W Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Samuel T Workenhe
- McMaster Immunology Research Center, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Natasha Kazdhan
- McMaster Immunology Research Center, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Aj Robert McGray
- McMaster Immunology Research Center, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Yonghong Wan
- McMaster Immunology Research Center, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Guido Kroemer
- Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), U1138, Paris, France.,Gustave Roussy Cancer Campus, Villejuif, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Sorbonne Université, Paris, France.,Université de Paris, Paris, France.,Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Suzhou, China.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Karolinska Institutet, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Brian D Lichty
- McMaster Immunology Research Center, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
8
|
Fischer C, Munks MW, Hill AB, Kroczek RA, Bissinger S, Brand V, Schmittnaegel M, Imhof-Jung S, Hoffmann E, Herting F, Klein C, Knoetgen H. Vaccine-induced CD8 T cells are redirected with peptide-MHC class I-IgG antibody fusion proteins to eliminate tumor cells in vivo. MAbs 2020; 12:1834818. [PMID: 33151105 PMCID: PMC7668529 DOI: 10.1080/19420862.2020.1834818] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022] Open
Abstract
Simulating a viral infection in tumor cells is an attractive concept to eliminate tumor cells. We previously reported the molecular design and the in vitro potency of recombinant monoclonal antibodies fused to a virus-derived peptide MHC class I complex that bypass the peptide processing and MHC loading pathway and directly displays a viral peptide in an MHC class I complex on the tumor cell surface. Here, we show that a vaccination-induced single peptide-specific CD8 T cell response was sufficient to eliminate B16 melanoma tumor cells in vivo in a fully immunocompetent, syngeneic mouse tumor model when mice were treated with mouse pMHCI-IgGs fusion proteins targeting the mouse fibroblast activation protein. Tumor growth of small, established B16 lung metastases could be controlled. The pMHCI-IgG had similar potency as an analogous pan-CD3 T-cell bispecific antibody. In contrast to growth control of small tumors, none of the compounds controlled larger solid tumors of MC38 cancer cells, despite penetration of pMHCI-IgGs into the tumor tissue and clear attraction and activation of antigen-specific CD8 T cells inside the tumor. pMHCI-IgG can have a similar potency as classical pan-T-cell recruiting molecules. The results also highlight the need to better understand immune suppression in advanced solid tumors.
Collapse
Affiliation(s)
- Cornelia Fischer
- Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Michael W. Munks
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Ann B. Hill
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | | | - Stefan Bissinger
- Discovery Oncology, Roche Innovation Center Munich, Penzberg, Germany
| | - Verena Brand
- Discovery Oncology, Roche Innovation Center Munich, Penzberg, Germany
| | | | - Sabine Imhof-Jung
- Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Eike Hoffmann
- Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | - Frank Herting
- Discovery Oncology, Roche Innovation Center Munich, Penzberg, Germany
| | - Christian Klein
- Discovery Oncology, Roche Innovation Center Zurich, Zurich, Switzerland
| | - Hendrik Knoetgen
- Therapeutic Modalities, Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
9
|
Nabizadeh JA, Manthey HD, Panagides N, Steyn FJ, Lee JD, Li XX, Akhir FNM, Chen W, Boyle GM, Taylor SM, Woodruff TM, Rolfe BE. C5a receptors C5aR1 and C5aR2 mediate opposing pathologies in a mouse model of melanoma. FASEB J 2019; 33:11060-11071. [PMID: 31298935 DOI: 10.1096/fj.201800980rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The canonical complement component 5a (C5a) receptor (C5aR) 1 has well-described roles in tumorigenesis but the contribution of the second receptor, C5aR2, is unclear. The present study demonstrates that B16.F0 melanoma cells express mRNA for both C5aR1 and C5aR2 and signal through ERK and p38 MAPKs in response to C5a. Despite this, C5a had no impact on melanoma cell proliferation or migration in vitro. In vivo studies demonstrated that the growth of B16.F0 melanoma tumors was increased in C5aR2-/- mice but reduced in C5aR1-/- mice and wild-type mice treated with a C5aR1 antagonist. Analysis of tumor-infiltrating leukocyte populations showed no significant differences between wild-type and C5aR2-/- mice. Conversely, percentages of myeloid-derived suppressor cells, macrophages, and regulatory T lymphocytes were lower in tumors from C5aR1-/- mice, whereas total (CD3+) T lymphocytes and CD4+ subsets were higher. Analysis of cytokine and chemokine levels also showed plasma IFN-γ was higher and tumor C-C motif chemokine ligand 2 was lower in the absence of C5aR1. The results suggest that C5aR1 signaling supports melanoma growth by promoting infiltration of immunosuppressive leukocyte populations into the tumor microenvironment, whereas C5aR2 has a more restricted but beneficial role in limiting tumor growth. Overall, these data support the potential of C5aR1-inhibitory therapies for melanoma.-Nabizadeh, J. A., Manthey, H. D., Panagides, N., Steyn, F. J., Lee, J. D., Li, X. X., Akhir, F. N. M., Chen, W., Boyle, G. M., Taylor, S. M., Woodruff, T. M., Rolfe, B. E. C5a receptors C5aR1 and C5aR2 mediate opposing pathologies in a mouse model of melanoma.
Collapse
Affiliation(s)
- Jamileh A Nabizadeh
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Helga D Manthey
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Nadya Panagides
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Frederik J Steyn
- Centre for Clinical Research, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - John D Lee
- School of Biomedical Science, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Xaria X Li
- School of Biomedical Science, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Fazrena N M Akhir
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Weiyu Chen
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Glen M Boyle
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Stephen M Taylor
- School of Biomedical Science, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Trent M Woodruff
- School of Biomedical Science, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| | - Barbara E Rolfe
- Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
10
|
Boilesen DR, Ragonnaud E, Laursen H, Andersson AMC, Tolver A, Spiess K, Holst PJ. CD8+ T cells induced by adenovirus-vectored vaccine are capable of preventing establishment of latent murine γ-herpesvirus 68 infection. Vaccine 2019; 37:2952-2959. [PMID: 31006497 DOI: 10.1016/j.vaccine.2019.04.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 04/02/2019] [Accepted: 04/11/2019] [Indexed: 01/09/2023]
Abstract
CD8+ T cells are known to control infections, but their role in preventing latent infection from establishing has not been thoroughly investigated. We hypothesized that a potent CD8+ T cell response patrolling the mucosal viral entry points could kill the first infected cells and thereby abrogate the infection before latency is established. To investigate this, replication deficient adenovirus serotype 5 vectors encoding murine γ-herpesvirus-68 CD8+ T cell epitopes linkedto the T cell adjuvant Invariant chain, were developed. We show that intranasal vaccination of mice reduces the risk of establishment of latent infection from multiple intranasal ID50 challenges with murine γ-herpesvirus-68 by 81% per exposure at 14 days post vaccination. Protection waned over time, but immune responses were extended by heterologous prime-boost vaccination applied simultaneously intramuscularly and intranasally, and animals vaccinated 66 days prior to challenge showed a strong trend of long-term protection. Our data provides evidence that CD8+ T cells are able to protect against establishment of latent infection. Although the protective efficacy is difficult to maintain over time, this proof-of-concept study suggests a role for a CD8+ T cell arm in future vaccine strategies against latent human viral infections caused by pathogens such as HIV and multiple herpes virus.
Collapse
Affiliation(s)
- Ditte R Boilesen
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Denmark; InProTher ApS, DK2200 Copenhagen, Denmark.
| | - Emeline Ragonnaud
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Denmark; NIA, NIH, Baltimore MD, USA
| | - Henriette Laursen
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Denmark
| | - Anne-Marie C Andersson
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Denmark; InProTher ApS, DK2200 Copenhagen, Denmark
| | - Anders Tolver
- Department of Mathematical Sciences, University of Copenhagen, Denmark
| | - Katja Spiess
- Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Peter J Holst
- Centre for Medical Parasitology, Department of Immunology and Microbiology, University of Copenhagen, Denmark; InProTher ApS, DK2200 Copenhagen, Denmark
| |
Collapse
|
11
|
Deng W, McLaughlin SL, Klinke DJ. Quantifying spontaneous metastasis in a syngeneic mouse melanoma model using real time PCR. Analyst 2018; 142:2945-2953. [PMID: 28725894 DOI: 10.1039/c7an00623c] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Modeling metastasis in vivo with animals is a priority for both revealing mechanisms of tumor dissemination and developing therapeutic methods. While conventional intravenous injection of tumor cells provides an efficient and consistent system for studying tumor cell extravasation and colonization, studying spontaneous metastasis derived from orthotopic tumor sites has the advantage of modeling more aspects of the metastatic cascade, but is challenging as it is difficult to detect small numbers of metastatic cells. In this work, we developed an approach for quantifying spontaneous metastasis in the syngeneic mouse B16 system using real time PCR. We first transduced B16 cells with lentivirus expressing firefly luciferase Luc2 gene for bioluminescence imaging. Next, we developed a real time quantitative PCR (qPCR) method for the detection of luciferase-expressing, metastatic tumor cells in mouse lungs and other organs. To illustrate the approach, we quantified lung metastasis in both spontaneous and experimental scenarios using B16F0 and B16F10 cells in C57BL/6Ncrl and NOD-Scid Gamma (NSG) mice. We tracked B16 melanoma metastasis with both bioluminescence imaging and qPCR, which were found to be self-consistent. Using this assay, we can quantitatively detect one Luc2 positive tumor cell out of 104 tissue cells, which corresponds to a metastatic burden of 1.8 × 104 metastatic cells per whole mouse lung. More importantly, the qPCR method was at least a factor of 10 more sensitive in detecting metastatic cell dissemination and should be combined with bioluminescence imaging as a high-resolution, end-point method for final metastatic cell quantitation. Given the rapid growth of primary tumors in many mouse models, assays with improved sensitivity can provide better insight into biological mechanisms that underpin tumor metastasis.
Collapse
Affiliation(s)
- Wentao Deng
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26505, USA
| | | | | |
Collapse
|
12
|
Alexander MP, Fiering SN, Ostroff GR, Cramer RA, Mullins DW. Beta-glucan-induced inflammatory monocytes mediate antitumor efficacy in the murine lung. Cancer Immunol Immunother 2018; 67:1731-1742. [PMID: 30167860 PMCID: PMC11028371 DOI: 10.1007/s00262-018-2234-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 08/13/2018] [Indexed: 01/07/2023]
Abstract
β-Glucan is a naturally occurring glucose polysaccharide with immunostimulatory activity in both infection and malignancy. β-Glucan's antitumor effects have been attributed to the enhancement of complement receptor 3-dependent cellular cytotoxicity, as well as modulation of suppressive and stimulatory myeloid subsets, which in turn enhances antitumor T cell immunity. In the present study, we demonstrate antitumor efficacy of yeast-derived β-glucan particles (YGP) in a model of metastatic-like melanoma in the lung, through a mechanism that is independent of previously reported β-glucan-mediated antitumor pathways. Notably, efficacy is independent of adaptive immunity, but requires inflammatory monocytes. YGP-activated monocytes mediated direct cytotoxicity against tumor cells in vitro, and systemic YGP treatment upregulated inflammatory mediators, including TNFα, M-CSF, and CCL2, in the lungs. Collectively, these studies identify a novel role for inflammatory monocytes in β-glucan-mediated antitumor efficacy, and expand the understanding of how this immunomodulator can be used to generate beneficial immune responses against metastatic disease.
Collapse
MESH Headings
- Adaptive Immunity/immunology
- Adjuvants, Immunologic
- Animals
- Hypoxia-Inducible Factor 1, alpha Subunit/physiology
- Inflammation Mediators/immunology
- Inflammation Mediators/metabolism
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/metabolism
- Lung Neoplasms/secondary
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Monocytes/drug effects
- Monocytes/immunology
- Monocytes/metabolism
- Receptors, CCR2/physiology
- T-Lymphocytes/immunology
- Tumor Cells, Cultured
- beta-Glucans/pharmacology
Collapse
Affiliation(s)
- Matthew P Alexander
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, USA
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, USA
| | - Steven N Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, USA
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, USA
| | - Gary R Ostroff
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Robert A Cramer
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, USA
| | - David W Mullins
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, USA.
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, 03756, USA.
- Department of Medical Education, Geisel School of Medicine at Dartmouth, 45 Dewey Field Road, HB7100, Hanover, NH, 03755, USA.
| |
Collapse
|
13
|
Hsu J, Hodgins JJ, Marathe M, Nicolai CJ, Bourgeois-Daigneault MC, Trevino TN, Azimi CS, Scheer AK, Randolph HE, Thompson TW, Zhang L, Iannello A, Mathur N, Jardine KE, Kirn GA, Bell JC, McBurney MW, Raulet DH, Ardolino M. Contribution of NK cells to immunotherapy mediated by PD-1/PD-L1 blockade. J Clin Invest 2018; 128:4654-4668. [PMID: 30198904 DOI: 10.1172/jci99317] [Citation(s) in RCA: 592] [Impact Index Per Article: 84.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 07/24/2018] [Indexed: 12/12/2022] Open
Abstract
Checkpoint blockade immunotherapy targeting the PD-1/PD-L1 inhibitory axis has produced remarkable results in the treatment of several types of cancer. Whereas cytotoxic T cells are known to provide important antitumor effects during checkpoint blockade, certain cancers with low MHC expression are responsive to therapy, suggesting that other immune cell types may also play a role. Here, we employed several mouse models of cancer to investigate the effect of PD-1/PD-L1 blockade on NK cells, a population of cytotoxic innate lymphocytes that also mediate antitumor immunity. We discovered that PD-1 and PD-L1 blockade elicited a strong NK cell response that was indispensable for the full therapeutic effect of immunotherapy. PD-1 was expressed on NK cells within transplantable, spontaneous, and genetically induced mouse tumor models, and PD-L1 expression in cancer cells resulted in reduced NK cell responses and generation of more aggressive tumors in vivo. PD-1 expression was more abundant on NK cells with an activated and more responsive phenotype and did not mark NK cells with an exhausted phenotype. These results demonstrate the importance of the PD-1/PD-L1 axis in inhibiting NK cell responses in vivo and reveal that NK cells, in addition to T cells, mediate the effect of PD-1/PD-L1 blockade immunotherapy.
Collapse
Affiliation(s)
- Joy Hsu
- Department of Molecular and Cell Biology, Immunotherapy and Vaccine Research Initiative, Cancer Research Laboratory, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, California, USA
| | - Jonathan J Hodgins
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Malvika Marathe
- Department of Molecular and Cell Biology, Immunotherapy and Vaccine Research Initiative, Cancer Research Laboratory, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, California, USA
| | - Chris J Nicolai
- Department of Molecular and Cell Biology, Immunotherapy and Vaccine Research Initiative, Cancer Research Laboratory, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, California, USA
| | - Marie-Claude Bourgeois-Daigneault
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Troy N Trevino
- Department of Molecular and Cell Biology, Immunotherapy and Vaccine Research Initiative, Cancer Research Laboratory, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, California, USA
| | - Camillia S Azimi
- Department of Molecular and Cell Biology, Immunotherapy and Vaccine Research Initiative, Cancer Research Laboratory, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, California, USA
| | - Amit K Scheer
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Haley E Randolph
- Department of Molecular and Cell Biology, Immunotherapy and Vaccine Research Initiative, Cancer Research Laboratory, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, California, USA
| | - Thornton W Thompson
- Department of Molecular and Cell Biology, Immunotherapy and Vaccine Research Initiative, Cancer Research Laboratory, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, California, USA
| | - Lily Zhang
- Department of Molecular and Cell Biology, Immunotherapy and Vaccine Research Initiative, Cancer Research Laboratory, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, California, USA
| | - Alexandre Iannello
- Department of Molecular and Cell Biology, Immunotherapy and Vaccine Research Initiative, Cancer Research Laboratory, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, California, USA
| | - Nikhita Mathur
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Karen E Jardine
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Georgia A Kirn
- Department of Molecular and Cell Biology, Immunotherapy and Vaccine Research Initiative, Cancer Research Laboratory, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, California, USA
| | - John C Bell
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Michael W McBurney
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - David H Raulet
- Department of Molecular and Cell Biology, Immunotherapy and Vaccine Research Initiative, Cancer Research Laboratory, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, California, USA
| | - Michele Ardolino
- Department of Molecular and Cell Biology, Immunotherapy and Vaccine Research Initiative, Cancer Research Laboratory, Division of Immunology and Pathogenesis, University of California, Berkeley, Berkeley, California, USA.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
14
|
Ylösmäki E, Malorzo C, Capasso C, Honkasalo O, Fusciello M, Martins B, Ylösmäki L, Louna A, Feola S, Paavilainen H, Peltonen K, Hukkanen V, Viitala T, Cerullo V. Personalized Cancer Vaccine Platform for Clinically Relevant Oncolytic Enveloped Viruses. Mol Ther 2018; 26:2315-2325. [PMID: 30005865 PMCID: PMC6127500 DOI: 10.1016/j.ymthe.2018.06.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/04/2018] [Accepted: 06/07/2018] [Indexed: 12/11/2022] Open
Abstract
The approval of the first oncolytic virus for the treatment of metastatic melanoma and the compiling evidence that the use of oncolytic viruses can enhance cancer immunotherapies targeted against various immune checkpoint proteins has attracted great interest in the field of cancer virotherapy. We have developed a novel platform for clinically relevant enveloped viruses that can direct the virus-induced immune response against tumor antigens. By physically attaching tumor-specific peptides onto the viral envelope of vaccinia virus and herpes simplex virus 1 (HSV-1), we were able to induce a strong T cell-specific immune response toward these tumor antigens. These therapeutic peptides could be attached onto the viral envelope by using a cell-penetrating peptide sequence derived from human immunodeficiency virus Tat N-terminally fused to the tumor-specific peptides or, alternatively, therapeutic peptides could be conjugated with cholesterol for the attachment of the peptides onto the viral envelope. We used two mouse models of melanoma termed B16.OVA and B16-F10 for testing the efficacy of OVA SIINFEKL-peptide-coated viruses and gp100-Trp2-peptide-coated viruses, respectively, and show that by coating the viral envelope with therapeutic peptides, the anti-tumor immunity and the number of tumor-specific CD8+ T cells in the tumor microenvironment can be significantly enhanced.
Collapse
Affiliation(s)
- Erkko Ylösmäki
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland
| | - Cristina Malorzo
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland
| | - Cristian Capasso
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland
| | - Oona Honkasalo
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland
| | - Manlio Fusciello
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland
| | - Beatriz Martins
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland
| | - Leena Ylösmäki
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland
| | - Antti Louna
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland
| | - Sara Feola
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland
| | - Henrik Paavilainen
- Department of Virology, University of Turku, Kiinamyllynkatu 13, 20520, Turku, Finland
| | - Karita Peltonen
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland
| | - Veijo Hukkanen
- Department of Virology, University of Turku, Kiinamyllynkatu 13, 20520, Turku, Finland
| | - Tapani Viitala
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland
| | - Vincenzo Cerullo
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Viikinkaari 5E, 00790 Helsinki, Finland.
| |
Collapse
|
15
|
Pelaez F, Manuchehrabadi N, Roy P, Natesan H, Wang Y, Racila E, Fong H, Zeng K, Silbaugh AM, Bischof JC, Azarin SM. Biomaterial scaffolds for non-invasive focal hyperthermia as a potential tool to ablate metastatic cancer cells. Biomaterials 2018; 166:27-37. [PMID: 29533788 DOI: 10.1016/j.biomaterials.2018.02.048] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 02/23/2018] [Accepted: 02/24/2018] [Indexed: 12/13/2022]
Abstract
Currently, there are very few therapeutic options for treatment of metastatic disease, as it often remains undetected until the burden of disease is too high. Microporous poly(ε-caprolactone) biomaterials have been shown to attract metastasizing breast cancer cells in vivo early in tumor progression. In order to enhance the therapeutic potential of these scaffolds, they were modified such that infiltrating cells could be eliminated with non-invasive focal hyperthermia. Metal disks were incorporated into poly(ε-caprolactone) scaffolds to generate heat through electromagnetic induction by an oscillating magnetic field within a radiofrequency coil. Heat generation was modulated by varying the size of the metal disk, the strength of the magnetic field (at a fixed frequency), or the type of metal. When implanted subcutaneously in mice, the modified scaffolds were biocompatible and became properly integrated with the host tissue. Optimal parameters for in vivo heating were identified through a combination of computational modeling and ex vivo characterization to both predict and verify heat transfer dynamics and cell death kinetics during inductive heating. In vivo inductive heating of implanted, tissue-laden composite scaffolds led to tissue necrosis as seen by histological analysis. The ability to thermally ablate captured cells non-invasively using biomaterial scaffolds has the potential to extend the application of focal thermal therapies to disseminated cancers.
Collapse
Affiliation(s)
- Francisco Pelaez
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455, USA
| | - Navid Manuchehrabadi
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Priyatanu Roy
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Harishankar Natesan
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Yiru Wang
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Emilian Racila
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Heather Fong
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kevin Zeng
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455, USA
| | - Abby M Silbaugh
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455, USA
| | - John C Bischof
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA; Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
16
|
Pasqual-Melo G, Gandhirajan RK, Stoffels I, Bekeschus S. Targeting malignant melanoma with physical plasmas. CLINICAL PLASMA MEDICINE 2018. [DOI: 10.1016/j.cpme.2018.03.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
17
|
Che X, Lu R, Fu Z, Sun Y, Zhu ZF, Li JP, Wang S, Jia J, Wang Q, Yao Z. Therapeutic effects of tyroserleutide on lung metastasis of human hepatocellular carcinoma SK-HEP-1 and its mechanism affecting ICAM-1 and MMP-2 and -9. Drug Des Devel Ther 2018; 12:3357-3368. [PMID: 30349190 PMCID: PMC6186774 DOI: 10.2147/dddt.s177126] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Tyroserleutide (YSL) inhibits the growth and metastasis of human hepatocellular carcinoma (HCC). This paper studied the effect of YSL on metastasis of human HCC and investigated its mechanisms. METHODS In vivo, experimental lung metastasis models of human HCC SK-HEP-1 cells in nude mice were established, and In vitro, the proliferation, adhesion and invasion of SK-HEP-1 cells were detected. RESULTS In vivo, YSL significantly inhibited the metastasis of human HCC. In vitro, YSL significantly inhibited the proliferation, adhesion and invasion of SK-HEP-1 cells. Through analyses with reverse transcription PCR (RT-PCR) and Western blot, we observed that YSL significantly inhibited the expressions of ICAM-1 in SK-HEP-1 cells. Through RT-PCR, Western blot and zymography methods, YSL was discovered to decrease the mRNA level, protein expression and activity of MMP-2 and -9 in SK-HEP-1 cells. CONCLUSION We concluded that YSL could inhibit tumor growth and metastasis of human HCC SK-HEP-1 cells.
Collapse
Affiliation(s)
- Xuchun Che
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, School of Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China, ;
| | - Rong Lu
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, School of Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China, ; .,Department of Drug Development, Tianjin Kangzhe Pharmaceutical Company, Ltd., Tianjin, People's Republic of China
| | - Zheng Fu
- Department of Drug Development, Tianjin Kangzhe Pharmaceutical Company, Ltd., Tianjin, People's Republic of China
| | - Yajun Sun
- Department of Blood Transfusion, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China
| | - Zhi-Feng Zhu
- Department of Drug Development, Tianjin Kangzhe Pharmaceutical Company, Ltd., Tianjin, People's Republic of China
| | - Jin-Ping Li
- Department of Drug Development, Tianjin Kangzhe Pharmaceutical Company, Ltd., Tianjin, People's Republic of China
| | - Song Wang
- Department of Drug Development, Tianjin Kangzhe Pharmaceutical Company, Ltd., Tianjin, People's Republic of China
| | - Jing Jia
- Department of Drug Development, Tianjin Kangzhe Pharmaceutical Company, Ltd., Tianjin, People's Republic of China
| | - Qing Wang
- Department of Clinical Laboratory, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China
| | - Zhi Yao
- Department of Immunology, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Educational Ministry of China, School of Medical Sciences, Tianjin Medical University, Tianjin, People's Republic of China, ;
| |
Collapse
|
18
|
Butler KL, Clancy-Thompson E, Mullins DW. CXCR3 + monocytes/macrophages are required for establishment of pulmonary metastases. Sci Rep 2017; 7:45593. [PMID: 28358049 PMCID: PMC5372355 DOI: 10.1038/srep45593] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 02/27/2017] [Indexed: 01/23/2023] Open
Abstract
We present a new foundational role for CXCR3+ monocytes/macrophages in the process of tumor engraftment in the lung. CXCR3 is associated with monocytic and lymphocytic infiltration of inflamed or tumor-bearing lung. Although the requirement for tumor-expressed CXCR3 in metastatic engraftment has been demonstrated, the role of monocyte-expressed CXCR3 had not been appreciated. In a murine model of metastatic-like melanoma, engraftment was coordinate with CXCR3+ monocyte/macrophage accumulation in the lungs and was sensitive to pharmacologic inhibition of CXCR3 signaling. Tumor engraftment to lung was impaired in CXCR3−/− mice, and transient reconstitution with circulating CXCR3-replete monocytes was sufficient to restore engraftment. These data illustrate the paradoxical pro-tumor role for CXCR3 in lung immunobiology wherein the CXCR3 axis drives both the anti-tumor effector cell chemoattraction and pro-tumor infiltration of the lungs and suggests a potential therapeutic target for lung-tropic metastasizing cancers.
Collapse
Affiliation(s)
- Kiah L Butler
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, NH 03756
| | - Eleanor Clancy-Thompson
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, NH 03756
| | - David W Mullins
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Lebanon, NH 03756.,Department of Medical Education, Geisel School of Medicine at Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|