1
|
Woodworth JS, Contreras V, Christensen D, Naninck T, Kahlaoui N, Gallouët AS, Langlois S, Burban E, Joly C, Gros W, Dereuddre-Bosquet N, Morin J, Liu Olsen M, Rosenkrands I, Stein AK, Krøyer Wood G, Follmann F, Lindenstrøm T, Hu T, Le Grand R, Pedersen GK, Mortensen R. MINCLE and TLR9 agonists synergize to induce Th1/Th17 vaccine memory and mucosal recall in mice and non-human primates. Nat Commun 2024; 15:8959. [PMID: 39420177 PMCID: PMC11487054 DOI: 10.1038/s41467-024-52863-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Development of new vaccines tailored for difficult-to-target diseases is hampered by a lack of diverse adjuvants for human use, and none of the currently available adjuvants induce Th17 cells. Here, we develop a liposomal adjuvant, CAF®10b, that incorporates Mincle and Toll-like receptor 9 agonists. In parallel mouse and non-human primate studies comparing to CAF® adjuvants already in clinical trials, we report species-specific effects of adjuvant composition on the quality and magnitude of the responses. When combined with antigen, CAF®10b induces Th1 and Th17 responses and protection against a pulmonary infection with Mycobacterium tuberculosis in mice. In non-human primates, CAF®10b induces higher Th1 responses and robust Th17 responses detectable after six months, and systemic and pulmonary Th1 and Th17 recall responses, in a sterile model of local recall. Overall, CAF®10b drives robust memory antibody, Th1 and Th17 vaccine-responses via a non-mucosal immunization route across both rodent and primate species.
Collapse
Affiliation(s)
- Joshua S Woodworth
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark.
| | - Vanessa Contreras
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Thibaut Naninck
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Nidhal Kahlaoui
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Anne-Sophie Gallouët
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Sébastien Langlois
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Emma Burban
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Candie Joly
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Wesley Gros
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Julie Morin
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Ming Liu Olsen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Ida Rosenkrands
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Ann-Kathrin Stein
- Department of Vaccine Development, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Grith Krøyer Wood
- Department of Vaccine Development, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Frank Follmann
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Thomas Lindenstrøm
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Tu Hu
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Roger Le Grand
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184), 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Gabriel Kristian Pedersen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark
| | - Rasmus Mortensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300, Copenhagen, Denmark.
| |
Collapse
|
2
|
Woodworth JS, Contreras V, Christensen D, Naninck T, Kahlaoui N, Gallouët AS, Langlois S, Burban E, Joly C, Gros W, Dereuddre-Bosquet N, Morin J, Olsen ML, Rosenkrands I, Stein AK, Wood GK, Follmann F, Lindenstrøm T, LeGrand R, Pedersen GK, Mortensen R. A novel adjuvant formulation induces robust Th1/Th17 memory and mucosal recall responses in Non-Human Primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.23.529651. [PMID: 36865310 PMCID: PMC9980079 DOI: 10.1101/2023.02.23.529651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
After clean drinking water, vaccination is the most impactful global health intervention. However, development of new vaccines against difficult-to-target diseases is hampered by the lack of diverse adjuvants for human use. Of particular interest, none of the currently available adjuvants induce Th17 cells. Here, we develop and test an improved liposomal adjuvant, termed CAF®10b, that incorporates a TLR-9 agonist. In a head-to-head study in non-human primates (NHPs), immunization with antigen adjuvanted with CAF®10b induced significantly increased antibody and cellular immune responses compared to previous CAF® adjuvants, already in clinical trials. This was not seen in the mouse model, demonstrating that adjuvant effects can be highly species specific. Importantly, intramuscular immunization of NHPs with CAF®10b induced robust Th17 responses that were observed in circulation half a year after vaccination. Furthermore, subsequent instillation of unadjuvanted antigen into the skin and lungs of these memory animals led to significant recall responses including transient local lung inflammation observed by Positron Emission Tomography-Computed Tomography (PET-CT), elevated antibody titers, and expanded systemic and local Th1 and Th17 responses, including >20% antigen-specific T cells in the bronchoalveolar lavage. Overall, CAF®10b demonstrated an adjuvant able to drive true memory antibody, Th1 and Th17 vaccine-responses across rodent and primate species, supporting its translational potential.
Collapse
Affiliation(s)
- Joshua S Woodworth
- Department of Infectious Disease Immunology, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Vanessa Contreras
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Thibaut Naninck
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Nidhal Kahlaoui
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Anne-Sophie Gallouët
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Sébastien Langlois
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Emma Burban
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Candie Joly
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Wesley Gros
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Julie Morin
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Ming Liu Olsen
- Department of Infectious Disease Immunology, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Ida Rosenkrands
- Department of Infectious Disease Immunology, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Ann-Kathrin Stein
- Department of Vaccine Development, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Grith Krøyer Wood
- Department of Vaccine Development, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Frank Follmann
- Department of Infectious Disease Immunology, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Thomas Lindenstrøm
- Department of Infectious Disease Immunology, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Roger LeGrand
- Université Paris-Saclay, Inserm, CEA, Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB/IDMIT/UMR1184); 92265, Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Gabriel Kristian Pedersen
- Department of Infectious Disease Immunology, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| | - Rasmus Mortensen
- Department of Infectious Disease Immunology, Statens Serum Institut; Artillerivej 5, 2300 Copenhagen, Denmark
| |
Collapse
|
3
|
Enhanced systemic antilymphoma immune response by photothermal therapy with CpG deoxynucleotide coated nanoparticles. Blood Adv 2022; 6:4581-4592. [PMID: 35687489 DOI: 10.1182/bloodadvances.2022008040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/02/2022] [Indexed: 11/20/2022] Open
Abstract
We investigated a novel mechanism of in situ vaccination in a lymphoma model. Radiation (RT) can induce abscopal responses in pre-clinical lymphoma models but has not translated to clinical efficacy. We hypothesized that immune stimulation with CpG deoxynucleotides could enhance abscopal effects induced by radiation or by photothermal therapy (PTT), which has been shown to have an immune stimulatory effect in solid tumors but has not been studied in lymphoma. Here, we designed a branched gold nanoparticle (BNP) platform to carry CpGs while maintaining PTT function and compared the immunologic profile of the tumor microenvironment after PTT or RT in a dual flank lymphoma model. One flank was treated with CpG with RT or CpG with PTT and the other tumor was left untreated. We found that the CpG/PTT groups had significant reduction in growth in both treated (primary) and untreated (secondary) tumors suggesting an improved abscopal response, with a concomitant increase in CD8/CD4 ratio and cytotoxic T cell/regulatory T cell ratio in both the primary and secondary tumor compared with CpG/RT. Dendritic cells in the primary and secondary draining lymph nodes had increased maturation markers in the CpG/PTT group, and the effector memory T cells (both CD4 and CD8) in the secondary tumor and the spleen were increased, suggesting a systemic vaccination effect. These data suggest that in a lymphoma model, PTT using a CpG nanoparticle platform resulted in enhanced in situ vaccination and abscopal response compared with RT.
Collapse
|
4
|
Watanabe T. Approaches of the Innate Immune System to Ameliorate Adaptive Immunotherapy for B-Cell Non-Hodgkin Lymphoma in Their Microenvironment. Cancers (Basel) 2021; 14:cancers14010141. [PMID: 35008305 PMCID: PMC8750340 DOI: 10.3390/cancers14010141] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/14/2021] [Accepted: 12/23/2021] [Indexed: 12/21/2022] Open
Abstract
A dominant paradigm being developed in immunotherapy for hematologic malignancies is of adaptive immunotherapy that involves chimeric antigen receptor (CAR) T cells and bispecific T-cell engagers. CAR T-cell therapy has yielded results that surpass those of the existing salvage immunochemotherapy for patients with relapsed/refractory diffuse large B-cell lymphoma (DLBCL) after first-line immunochemotherapy, while offering a therapeutic option for patients with follicular lymphoma (FL) and mantle cell lymphoma (MCL). However, the role of the innate immune system has been shown to prolong CAR T-cell persistence. Cluster of differentiation (CD) 47-blocking antibodies, which are a promising therapeutic armamentarium for DLBCL, are novel innate immune checkpoint inhibitors that allow macrophages to phagocytose tumor cells. Intratumoral Toll-like receptor 9 agonist CpG oligodeoxynucleotide plays a pivotal role in FL, and vaccination may be required in MCL. Additionally, local stimulator of interferon gene agonists, which induce a systemic anti-lymphoma CD8+ T-cell response, and the costimulatory molecule 4-1BB/CD137 or OX40/CD134 agonistic antibodies represent attractive agents for dendritic cell activations, which subsequently, facilitates initiation of productive T-cell priming and NK cells. This review describes the exploitation of approaches that trigger innate immune activation for adaptive immune cells to operate maximally in the tumor microenvironment of these lymphomas.
Collapse
Affiliation(s)
- Takashi Watanabe
- Department of Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, 2-174, Edobashi, Tsu City 514-8507, Japan
| |
Collapse
|
5
|
Rondon A, Rouanet J, Degoul F. Radioimmunotherapy in Oncology: Overview of the Last Decade Clinical Trials. Cancers (Basel) 2021; 13:cancers13215570. [PMID: 34771732 PMCID: PMC8583425 DOI: 10.3390/cancers13215570] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/26/2021] [Accepted: 11/05/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary Monoclonal antibody-bearing radionuclides have been under clinical investigation over the last two decades for their use in theranostic (diagnostic and therapeutic) applications in cancer. However, despite the numerous trials that have been conducted, only two radioimmunotherapies (RIT) have been approved by the FDA for the targeted therapy of hematologic tumors expressing CD20 antigens. Moreover, RIT applications for solid cancers faced major issues—such as radiotoxicity due to low antibodies penetrance requiring substantial curative dose—where new discoveries concerning antibody engineering or radionuclides are trying to overcome. Here, we performed an overview of the last 11-year clinical trials involving RIT for solid and non-solid cancers conducted either with full antibodies or antibody fragments. We discussed the low-to-moderate efficiency of RIT compared to conventional therapies and described the last advances in clinic for antibodies carriers (F(ab′)2, Fab′, ScFv). Finally, we discussed about the complexity of RIT as a therapy and depicted both the issues and the prospects of such a strategy. Abstract The specific irradiation of tumors with selective radiolabeled antibodies constitutes an attractive therapeutic approach. Consequent preclinical research has been conducted by both biologists to identify pertinent targets and to select corresponding antibodies (mAb) and by radiochemists to radiolabel mAbs. These numerous preclinical investigations have ascertained the therapeutic interest of radioimmunotherapy (RIT) protocols in mice models. Here, we summarize the clinical studies that have been performed the last decade, including clinical trials (phases I, II, and III), prospective and retrospective studies, and cases series. We thereby reported 92 clinical studies. Among them, 62 concern the treatment of hematological malignancies, and 30 concern solid tumors. For hematologic diseases, the analysis was complex due to the high discrepancy of therapeutic strategies (first-line therapy, consolidation, stem cell transplantation conditioning) as well as the high variety of malignancies that were treated. The clinical studies from the last decade failed to expand anti-CD20 RIT indications but confirmed that RIT using radiolabeled anti-CD20 remains a pertinent choice for patients with relapse follicular lymphomas. For solid tumors, the positive benefit of RIT is more mitigated, apart for few malignancies that can be treated locally. Clinical trials also demonstrated the potential of some antibody formats, such as F(ab′)2, which has already been approved by the China State FDA under the trend name Licartin®. Despite disparate results, mAb fragments are an interesting prospect for the improvement of RIT efficiency as well as for pretargeted strategies that delay the injection of radioactive treatments from the mAb ones.
Collapse
Affiliation(s)
- Aurélie Rondon
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, UCLouvain, BE-1200 Brussels, Belgium
- Correspondence: (A.R.); (F.D.)
| | - Jacques Rouanet
- Imagerie Moléculaire et Stratégies Théranostiques, Inserm UMR1240, Université Clermont-Auvergne, F-63000 Clermont-Ferrand, France;
- Service de Dermatologie et d’Oncologie Cutanée, CHU Estaing, F-63011 Clermont-Ferrand, France
| | - Françoise Degoul
- CNRS 6293, INSERM U1103, GReD, Centre de Recherche et de Biologie Clinique, Université Clermont-Auvergne, F-63000 Clermont-Ferrand, France
- Correspondence: (A.R.); (F.D.)
| |
Collapse
|
6
|
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common aggressive B-cell lymphoma and highly heterogeneous disease. With the standard immunochemotherapy, anti-CD20 antibody rituximab (R-) plus CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone) chemotherapy, 30-40% of DLBCLs are refractory to initial immunochemotherapy or experience relapse post-therapy with poor clinical outcomes despite salvage therapies. Mechanisms underlying chemoresistance and relapse are heterogeneous across DLBCL and within individual patients, representing hurdles for targeted therapies targeting a specific oncogenic signaling pathway. In recent years, paradigm-shifting immunotherapies have shown impressive efficacy in various cancer types regardless of underlying oncogenic mechanisms. Vaccines are being developed for DLBCL to build protective immunity against relapse after first complete remission and to promote antitumor immune responses synergizing with immune checkpoint inhibitors to treat refractory/relapsed patients. This article provides a brief review of current progress in vaccine development in DLBCL and discussion on immunologic mechanisms underlying the therapeutic effectiveness and resistance.
Collapse
Affiliation(s)
- Zijun Y Xu-Monette
- Hematopathology Division, Department of Pathology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - Ken H Young
- Hematopathology Division, Department of Pathology, Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
7
|
Lin AY, Rink JS, Karmali R, Xu J, Kocherginsky M, Thaxton CS, Gordon LI. Tri-ethylene glycol modified class B and class C CpG conjugated gold nanoparticles for the treatment of lymphoma. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 30:102290. [PMID: 32798731 DOI: 10.1016/j.nano.2020.102290] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/01/2020] [Accepted: 08/07/2020] [Indexed: 11/17/2022]
Abstract
CpG oligodeoxynucleotides (CpGs) can induce an anti-tumor immune response, but also uniquely cause direct lymphoma cytotoxicity. To improve the delivery and efficacy of CpGs, we utilized a tri-ethylene modified CpG conjugated gold nanoparticle (tmCpG NP) platform that is compatible with both class B and class C CpGs, to treat various types of lymphoma, including diffuse large B cell lymphoma, high-grade lymphoma, Burkitt's lymphoma, and mantle cell lymphoma. Both classes of tmCpG NPs reduced viability of human and murine lymphoma cells via apoptosis compared with free CpGs, while having no toxic effects on dendritic cells. TmCpG NPs increased CD19, CD20, and OX40 expression on the lymphoma cells. Overall, we introduced a stable tmCpG NP design that has significant anti-lymphoma effects.
Collapse
Affiliation(s)
- Adam Yuh Lin
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.
| | - Jonathan Scott Rink
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Reem Karmali
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Jiahui Xu
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Masha Kocherginsky
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | - Colby Shad Thaxton
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA; Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Leo I Gordon
- Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Robert H Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| |
Collapse
|
8
|
Synergistic Toll-like Receptor 3/9 Signaling Affects Properties and Impairs Glioma-Promoting Activity of Microglia. J Neurosci 2020; 40:6428-6443. [PMID: 32631940 DOI: 10.1523/jneurosci.0666-20.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/28/2020] [Accepted: 06/05/2020] [Indexed: 11/21/2022] Open
Abstract
In murine experimental glioma models, TLR3 or TLR9 activation of microglial/macrophages has been shown to impair glioma growth, which could, however, not been verified in recent clinical trials. We therefore tested whether combined TLR3 and TLR9 activation of microglia/macrophages would have a synergistic effect. Indeed, combined TLR3/TLR9 activation augmented the suppression of glioma growth in organotypic brain slices from male mice in a microglia-dependent fashion, and this synergistic suppression depended on interferon β release and phagocytic tumor clearance. Combined TLR3/TLR9 stimulation also augmented several functional features of microglia, such as the release of proinflammatory factors, motility, and phagocytosis activity. TLR3/TLR9 stimulation combined with CD47 blockade further augmented glioma clearance. Finally, we confirmed that the coactivation of TLR3/TLR9 also augments the impairment of glioma growth in vivo Our results show that combined activation of TLR3/TLR9 in microglia/macrophages results in a more efficient glioma suppression, which may provide a potential strategy for glioma treatment.SIGNIFICANCE STATEMENT Glioma-associated microglia/macrophages (GAMs) are the predominant immune cells in glioma growth and are recently considered as antitumor targets. TLRs are involved in glioma growth, but the TLR3 or TLR9 ligands were not successful in clinical trials in treating glioma. We therefore combined TLR3 and TLR9 activation of GAMs, resulting in a strong synergistic effect of tumor clearance in vitro, ex vivo, and in vivo Mechanisms of this GAM-glioma interaction involve IFNβ signaling and increased tumor clearance by GAMs. Interfering with CD47 signaling had an additional impact on tumor clearance. We propose that these signaling pathways could be exploited as anti-glioma targets.
Collapse
|
9
|
Miller AM, Lemke-Miltner CD, Blackwell S, Tomanek-Chalkley A, Gibson-Corely KN, Coleman KL, Weiner GJ, Chan CHF. Intraperitoneal CMP-001: A Novel Immunotherapy for Treating Peritoneal Carcinomatosis of Gastrointestinal and Pancreaticobiliary Cancer. Ann Surg Oncol 2020; 28:1187-1197. [PMID: 32409965 DOI: 10.1245/s10434-020-08591-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND The treatment options for patients with peritoneal carcinomatosis (PC) of gastrointestinal and pancreaticobiliary origins are limited. The virus-like particle, CMP-001, composed of the Qβ bacteriophage capsid protein encapsulating a CpG-A oligodeoxynucleotide, activates plasmacytoid dendritic cells (pDCs) and triggers interferon alpha (IFNα) release, leading to a cascade of anti-tumor immune effects. METHODS To evaluate the ability of CMP-001 to trigger an immune response in patients with PC, peritoneal cells were isolated and stimulated ex vivo with CMP-001. Both IFNα release and percentage of pDC were quantified using enzyme-linked immunosorbent assay (ELISA) and flow cytometry, respectively. To evaluate the anti-tumor response in vivo, murine PC models were generated using mouse cancer cell lines (Panc02 and MC38) in immunocompetent mice treated with intraperitoneal CMP-001 or saline control. Survival was followed, and the immunophenotype of cells in the peritoneal tumor microenvironment was evaluated. RESULTS The pDCs accounted for 1% (range 0.1-3.9%; n = 17) of the isolated peritoneal cells. Ex vivo CMP-001 stimulation of the peritoneal cells released an average of 0.77 ng/ml of IFNα (range, 0-4700 pg/ml; n = 14). The IFNα concentration was proportional to the percentage of pDCs present in the peritoneal cell mixture (r = 0.6; p = 0.037). In murine PC models, intraperitoneal CMP-001 treatment elicited an anti-tumor immune response including an increase in chemokines (RANTES and MIP-1β), pro-inflammatory cytokines (IFNγ, interleukin 6 [IL-6], and IL-12), and peritoneal/tumor immune infiltration (CD4+/CD8+ T and natural killer [NK] cells). The CMP-001 treatment improved survival in both the Panc02 (median, 35 vs 28 days) and the MC38 (median: 57 vs 35 days) PC models (p < 0.05). CONCLUSIONS As a novel immunotherapeutic agent, CMP-001 may be effective for treating patients with PC.
Collapse
Affiliation(s)
- Ann M Miller
- Department of Surgery, University of Iowa Hospitals and Clinics, University of Iowa, Iowa City, IA, USA
| | - Caitlin D Lemke-Miltner
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, USA.,Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Sue Blackwell
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, USA.,Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Ann Tomanek-Chalkley
- Department of Surgery, University of Iowa Hospitals and Clinics, University of Iowa, Iowa City, IA, USA
| | - Katherine N Gibson-Corely
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, USA.,Department of Pathology, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Kristen L Coleman
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - George J Weiner
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, USA.,Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Carlos H F Chan
- Department of Surgery, University of Iowa Hospitals and Clinics, University of Iowa, Iowa City, IA, USA. .,Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, USA.
| |
Collapse
|
10
|
Lemke-Miltner CD, Blackwell SE, Yin C, Krug AE, Morris AJ, Krieg AM, Weiner GJ. Antibody Opsonization of a TLR9 Agonist-Containing Virus-like Particle Enhances In Situ Immunization. THE JOURNAL OF IMMUNOLOGY 2020; 204:1386-1394. [PMID: 31953355 DOI: 10.4049/jimmunol.1900742] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/27/2019] [Indexed: 02/05/2023]
Abstract
The immunologic and therapeutic effects of intratumoral (IT) delivery of a novel virus-like particle as a lymphoma immunotherapy were evaluated in preclinical studies with human cells and a murine model. CMP-001 is a virus-like particle composed of the Qβ bacteriophage capsid protein encapsulating an immunostimulatory CpG-A oligodeoxynucleotide TLR9 agonist. In vitro, CMP-001 induced cytokine production, including IFN-α from plasmacytoid dendritic cells, but only in the presence of anti-Qβ Ab. In vivo, IT CMP-001 treatment of murine A20 lymphoma enhanced survival and reduced growth of both injected and contralateral noninjected tumors in a manner dependent on both the ability of mice to generate anti-Qβ Ab and the presence of T cells. The combination of IT CMP-001 with systemic anti-PD-1 enhanced antitumor responses in both injected and noninjected tumors. IT CMP-001 alone or combined with anti-PD-1 augmented T cell infiltration in tumor-draining lymph nodes. We conclude IT CMP-001 induces a robust antitumor T cell response in an anti-Qβ Ab-dependent manner and results in systemic antitumor T cell effects that are enhanced by anti-PD-1 in a mouse model of B cell lymphoma. Early-phase clinical evaluation of CMP-001 and anti-PD-1 combination therapy in lymphoma will begin shortly, based in part on these results.
Collapse
Affiliation(s)
| | - Sue E Blackwell
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242
| | - Chaobo Yin
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242
| | - Anna E Krug
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242
| | | | | | - George J Weiner
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242; .,Department of Internal Medicine, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
11
|
Eskian M, Khorasanizadeh M, Zinzani PL, Illidge TM, Rezaei N. Novel Methods to Improve the Efficiency of Radioimmunotherapy for Non-Hodgkin Lymphoma. Int Rev Immunol 2019; 38:79-91. [PMID: 30931651 DOI: 10.1080/08830185.2019.1588266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Radioimmunotherapy (RIT) is a novel strategy for treating non-Hodgkin lymphoma (NHL). Several studies have shown the promising results of using RIT in NHL, which have led to FDA approval for two RIT agents in treating low grade NHL. In spite of these favorable results in low-grade NHL, most of the aggressive or relapsed/refractory NHL subjects experience relapses following RIT. Although more aggressive treatments such as myeloablative doses of RIT followed by stem cell transplantation appear to be able to provide a longer survival for some patients these approaches are associated with significant treatment-related adverse events and challenging to deliver in most centers. Therefore, it seems reasonable to develop treatment approaches that enhance the efficiency of RIT, while reducing its toxicity. In this paper, novel methods that improve the efficiency of RIT and reduce its toxicity through various mechanisms are reviewed. Further clinical development of these methods could expand the NHL patient groups eligible for receiving RIT, and even extend the use of RIT to new indications and disease groups in future.
Collapse
Affiliation(s)
- Mahsa Eskian
- a Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran.,b Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN) , Tehran , Iran
| | - MirHojjat Khorasanizadeh
- a Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran.,b Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN) , Tehran , Iran
| | - Pier Luigi Zinzani
- c Institute of Hematology "L. e A. Seràgnoli", University of Bologna , Bologna , Italy
| | - Tim M Illidge
- d Manchester Academic Health Sciences Centre, University of Manchester, Christie NHS Foundation Trust , Manchester , UK
| | - Nima Rezaei
- a Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran.,e Department of Immunology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran.,f Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN) , Tehran , Iran
| |
Collapse
|
12
|
Ge X, Chen J, Li L, Ding P, Wang Q, Zhang W, Li L, Lv X, Zhou D, Jiang Z, Zeng H, Xu Y, Hou Y, Hu W. Midostaurin potentiates rituximab antitumor activity in Burkitt's lymphoma by inducing apoptosis. Cell Death Dis 2018; 10:8. [PMID: 30584254 PMCID: PMC6315025 DOI: 10.1038/s41419-018-1259-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 12/03/2018] [Indexed: 12/14/2022]
Abstract
An intensive short-term chemotherapy regimen has substantially prolonged the overall survival of Burkitt’s lymphoma (BL) patients, which has been further improved by addition of rituximab. However, the inevitable development of resistance to rituximab and the toxicity of chemotherapy remain obstacles. We first prepared two BL cell lines resistant to rituximab-mediated CDC. Using a phosphorylation antibody microarray, we revealed that PI3K/AKT pathway contained the most phosphorylated proteins/hits, while apoptosis pathway that may be regulated by PKC displayed the greatest fold enrichment in the resistant cells. The PI3K/AKT inhibitor IPI-145 failed to reverse the resistance. In contrast, the pan-PKC inhibitor midostaurin exhibited potent antitumor activity in both original and resistant cells, alone or in combination with rituximab. Notably, midostaurin promoted apoptosis by reducing the phosphorylation of PKC and consequently of downstream Bad, Bcl-2 and NF-κB. Therefore, midostaurin improved rituximab activity by supplementing pro-apoptotic effects. In vivo, midostaurin alone powerfully prolonged the survival of mice bearing the resistant BL cells compared to rituximab alone treatments. Addition of midostaurin to rituximab led to dramatically improved survival compared to rituximab but not midostaurin monotherapy. Our findings call for further evaluation of midostaurin alone or in combination with rituximab in treating resistant BL in particular.
Collapse
Affiliation(s)
- Xiaowen Ge
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianfeng Chen
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ling Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Peipei Ding
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qi Wang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wei Zhang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Luying Li
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xinyue Lv
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Danlei Zhou
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhengzeng Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haiying Zeng
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yifan Xu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yingyong Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Weiguo Hu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences; Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
13
|
Basaco T, Pektor S, Bermudez JM, Meneses N, Heller M, Galván JA, Boligán KF, Schürch S, von Gunten S, Türler A, Miederer M. Evaluation of Radiolabeled Girentuximab In Vitro and In Vivo. Pharmaceuticals (Basel) 2018; 11:E132. [PMID: 30487460 PMCID: PMC6316122 DOI: 10.3390/ph11040132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/21/2018] [Accepted: 11/26/2018] [Indexed: 11/16/2022] Open
Abstract
Girentuximab (cG250) targets carbonic anhydrase IX (CAIX), a protein which is expressed on the surface of most renal cancer cells (RCCs). cG250 labeled with 177Lu has been used in clinical trials for radioimmunotherapy (RIT) of RCCs. In this work, an extensive characterization of the immunoconjugates allowed optimization of the labeling conditions with 177Lu while maintaining immunoreactivity of cG250, which was then investigated in in vitro and in vivo experiments. cG250 was conjugated with S-2-(4-isothiocyanatobenzyl)-1,4,7,10-tetraazacyclododecane tetraacetic acid (DOTA(SCN)) by using incubation times between 30 and 90 min and characterized by mass spectrometry. Immunoconjugates with five to ten DOTA(SCN) molecules per cG250 molecule were obtained. Conjugates with ratios less than six DOTA(SCN)/cG250 had higher in vitro antigen affinity, both pre- and postlabeling with 177Lu. Radiochemical stability increased, in the presence of sodium ascorbate, which prevents radiolysis. The immunoreactivity of the radiolabeled cG250 tested by specific binding to SK-RC-52 cells decreased when the DOTA content per conjugate increased. The in vivo tumor uptake was < 10% ID/g and independent of the total amount of protein in the range between 5 and 100 µg cG250 per animal. Low tumor uptake was found to be due to significant necrotic areas and heterogeneous CAIX expression. In addition, low vascularity indicated relatively poor accessibility of the CAIX target.
Collapse
Affiliation(s)
- Tais Basaco
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
- Laboratory of Radiochemistry, Paul Scherrer Institute (PSI), 5232 Villigen PSI, Switzerland.
| | - Stefanie Pektor
- Clinic for Nuclear Medicine, University Medical Center Mainz, 55131 Mainz, Germany.
| | - Josue M Bermudez
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
| | - Niurka Meneses
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
| | - Manfred Heller
- Department for Biomedical Research (DBMR), University of Bern, 3010 Bern, Switzerland.
| | - José A Galván
- Institute of Pathology, University of Bern, 3010 Bern, Switzerland.
| | - Kayluz F Boligán
- Institute of Pharmacology (PKI), University of Bern, 3010 Bern, Switzerland.
| | - Stefan Schürch
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
| | - Stephan von Gunten
- Institute of Pharmacology (PKI), University of Bern, 3010 Bern, Switzerland.
| | - Andreas Türler
- Department of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland.
| | - Matthias Miederer
- Clinic for Nuclear Medicine, University Medical Center Mainz, 55131 Mainz, Germany.
| |
Collapse
|
14
|
Gordon LI. Consolidation and maintenance in follicular lymphoma: radioimmunotherapy revisited? Lancet Haematol 2018; 5:e96-e97. [PMID: 29396095 DOI: 10.1016/s2352-3026(18)30015-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 01/11/2018] [Indexed: 06/07/2023]
Affiliation(s)
- Leo I Gordon
- Robert H Lurie Comprehensive Cancer Center of Northwestern University, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
15
|
Rezahosseini O, Hanaei S, Hamadani M, Keshavarz-Fathi M, Rezaei N. The promising role of monoclonal antibodies for immunotherapy of the HIV-associated cancer, non-Hodgkin lymphoma. Int Rev Immunol 2017; 37:165-173. [PMID: 29257907 DOI: 10.1080/08830185.2017.1405396] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Association between HIV/AIDS and some of the cancers such as lymphomais is well known. Relative risk for developing non-Hodgkin lymphoma (NHL) increases 60-200 folds in HIV-infected individuals. Diffuse large B cell lymphoma (DLBCL), primary effusion lymphoma (PEL), Burkitt's lymphoma (BL) and Plasmablastic Lymphoma (PBL) are among the most frequent subtypes. During the last century, scientists found that the immune system could potentially detect and destroy cancer cells. Therefore, they started a new field of study, which is named immunotherapy. There are different immunotherapeutic methods, among which therapeutic antibodies, such as Brentuximabvedotin (Adcetris), Ibritumomabtiuxetan (Zevalin) and rituximab (Rituxan), used for treatment of NHLs showed promising results. In this article, we will review the immunotherapeutic option, monoclonal antibodies, for treatment of HIV-associated NHLs as well as their recent clinical status. We will also discuss the selective monoclonal antibody for each subtype of NHLs.
Collapse
Affiliation(s)
- Omid Rezahosseini
- a Cancer Immunology Project (CIP) , Universal Scientific Education and Research Network (USERN) , Tehran , Iran.,b Department of Infectious and Tropical diseases , Imam Khomeini Hospital Complex, Tehran University of Medical Sciences , Tehran , Iran
| | - Sara Hanaei
- a Cancer Immunology Project (CIP) , Universal Scientific Education and Research Network (USERN) , Tehran , Iran.,c Research Center for Immunodeficiencies , Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran.,d School of Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Mehdi Hamadani
- e Division of Hematology & Oncology , Medical College of Wisconsin , Milwaukee , WI , USA.,f Cancer Immunology Project (CIP) , Universal Scientific Education and Research Network (USERN) , Milwaukee , WI , USA
| | - Mahsa Keshavarz-Fathi
- a Cancer Immunology Project (CIP) , Universal Scientific Education and Research Network (USERN) , Tehran , Iran.,c Research Center for Immunodeficiencies , Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran.,d School of Medicine , Tehran University of Medical Sciences , Tehran , Iran.,g Students' Scientific Research Center , Tehran University of Medical Sciences , Tehran , Iran
| | - Nima Rezaei
- c Research Center for Immunodeficiencies , Children's Medical Center, Tehran University of Medical Sciences , Tehran , Iran.,h Department of Immunology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran.,i Cancer Immunology Project (CIP) , Universal Scientific Education and Research Network (USERN) , Sheffield , UK
| |
Collapse
|
16
|
Díaz-Rodríguez Y, Cordeiro P, Belounis A, Herblot S, Duval M. In vitro differentiated plasmacytoid dendritic cells as a tool to induce anti-leukemia activity of natural killer cells. Cancer Immunol Immunother 2017; 66:1307-1320. [PMID: 28555259 PMCID: PMC5626790 DOI: 10.1007/s00262-017-2022-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 05/22/2017] [Indexed: 12/17/2022]
Abstract
Acute lymphoblastic leukemia (ALL) is believed to be resistant to NK cell-mediated killing. To overcome this resistance, we developed an innovative approach based on NK cell stimulation with Toll-like receptor (TLR)-activated plasmacytoid dendritic cells (pDC). The translation of this approach into the clinic requires the production of high numbers of human pDC. Herein, we show that in vitro differentiation of cord blood CD34+ progenitors in the presence of aryl hydrocarbon receptor antagonists gives rise to clinically relevant numbers of pDC, as about 108 pDC can be produced from a typical cord blood unit. Blocking the aryl hydrocarbon receptor (AHR) pathway significantly increased the yield of pDC. When compared to pDC isolated from peripheral blood, in vitro differentiated pDC (ivD-pDC) exhibited an increased capacity to induce NK cell-mediated killing of ALL. Although ivD-pDC produced lower amounts of IFN-α than peripheral blood pDC upon TLR activation, they produced more IFN-λ2, known to play a critical role in the induction of anti-tumoral NK cell functions. Both TLR-9 and TLR-7 ligands triggered pDC-induced NK cell activation, offering the possibility to use any clinical-grade TLR-7 or TLR-9 ligands in future clinical trials. Finally, adoptive transfer of ivD-pDC cultured in the presence of an AHR antagonist cured humanized mice with minimal ALL disease. Collectively, our results pave the way to clinical-grade production of sufficient numbers of human pDC for innate immunotherapy against ALL and other refractory malignancies.
Collapse
Affiliation(s)
- Yildian Díaz-Rodríguez
- Unité de recherche en hémato-oncologie Charles-Bruneau, Centre de Recherche du CHU Sainte-Justine, 3175, Côte Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
- Département de Microbiologie, Infectiologie and Immunologie, Université de Montréal, Montreal, QC, Canada
| | - Paulo Cordeiro
- Unité de recherche en hémato-oncologie Charles-Bruneau, Centre de Recherche du CHU Sainte-Justine, 3175, Côte Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Assila Belounis
- Unité de recherche en hémato-oncologie Charles-Bruneau, Centre de Recherche du CHU Sainte-Justine, 3175, Côte Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
- Département de Microbiologie, Infectiologie and Immunologie, Université de Montréal, Montreal, QC, Canada
| | - Sabine Herblot
- Unité de recherche en hémato-oncologie Charles-Bruneau, Centre de Recherche du CHU Sainte-Justine, 3175, Côte Sainte-Catherine, Montreal, QC, H3T 1C5, Canada.
- Département de Microbiologie, Infectiologie and Immunologie, Université de Montréal, Montreal, QC, Canada.
- Département de Pédiatrie, Université de Montréal, Montreal, QC, Canada.
| | - Michel Duval
- Unité de recherche en hémato-oncologie Charles-Bruneau, Centre de Recherche du CHU Sainte-Justine, 3175, Côte Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
- Département de Microbiologie, Infectiologie and Immunologie, Université de Montréal, Montreal, QC, Canada
- Département de Pédiatrie, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
17
|
|
18
|
Terreno E, Aime S. MRI Contrast Agents for Pharmacological Research. Front Pharmacol 2015; 6:290. [PMID: 26696890 PMCID: PMC4673339 DOI: 10.3389/fphar.2015.00290] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/19/2015] [Indexed: 12/17/2022] Open
Affiliation(s)
- Enzo Terreno
- Department of Molecular Biotechnology and Health Sciences, Molecular and Preclinical Imaging Centers, University of Torino Torino, Italy
| | - Silvio Aime
- Department of Molecular Biotechnology and Health Sciences, Molecular and Preclinical Imaging Centers, University of Torino Torino, Italy
| |
Collapse
|
19
|
Xing N, Qiao T, Zhuang X, Yuan S, Zhang Q, Xu G. CpG oligodeoxyribonucleotide 7909 enhances radiosensitivity via downregulating Oct-4 expression in radioresistant lung cancer cells. Onco Targets Ther 2015; 8:1443-9. [PMID: 26109868 PMCID: PMC4472028 DOI: 10.2147/ott.s84467] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy is a powerful cure for local advanced non-small cell lung cancer. However, radioresistance and tumor relapse still occur in a high proportion of patients. Octamer-4 (Oct-4), a transcription factor of the POU family, plays a key role in maintaining chemoradioresistant properties and regulating cancer progression. In this study, we demonstrated that Oct-4 expression was significantly increased in radioresistant H460 (H460R) cell line. CpG oligodeoxyribonucleotide (CpG-ODN) 7909 sensitized H460R cells when combined with irradiation treatment. The clonogenic capacity was significantly decreased, and the values of D0 and Dq were lower than those of irradiation alone group. The sensitive enhancement ratio (SER) of D0 was 1.224. This combined treatment led to a dramatic reduction in Oct-4 expression in a dose-dependent manner and also showed increased percentage of cells in the radiosensitive G2/M phase relative to either treatment alone. These results identified that Oct-4 was involved in radioresistance. CpG-ODN 7909 could enhance radiosensitivity partly through downregulating Oct-4 expression in radioresistant lung cancer cells.
Collapse
Affiliation(s)
- Na Xing
- Department of Oncology, Fudan University, Shanghai, People's Republic of China
| | - Tiankui Qiao
- Department of Oncology, Fudan University, Shanghai, People's Republic of China
| | - Xibing Zhuang
- Department of Oncology, Fudan University, Shanghai, People's Republic of China
| | - Sujuan Yuan
- Department of Oncology, Fudan University, Shanghai, People's Republic of China
| | - Qi Zhang
- Department of Oncology, Fudan University, Shanghai, People's Republic of China
| | - Guoxiong Xu
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
20
|
Abstract
For 20 years, monoclonal antibodies (mAbs) have been a standard component of cancer therapy, but there is still much room for improvement. Efforts continue to build better cancer therapeutics based on mAbs. Anticancer mAbs function through various mechanisms, including directly targeting the malignant cells, modifying the host response, delivering cytotoxic moieties and retargeting cellular immunity towards the malignant cells. Characteristics of mAbs that affect their efficacy include antigen specificity, overall structure, affinity for the target antigen and how a mAb component is incorporated into a construct that can trigger target cell death. This Review discusses the various approaches to using mAb-based therapeutics to treat cancer and the strategies used to take advantage of the unique potential of each approach, and provides examples of current mAb-based treatments.
Collapse
Affiliation(s)
- George J. Weiner
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, 5970Z‐JPP, Iowa City, Iowa 52242, USA.
| |
Collapse
|
21
|
Bachireddy P, Burkhardt UE, Rajasagi M, Wu CJ. Haematological malignancies: at the forefront of immunotherapeutic innovation. Nat Rev Cancer 2015; 15:201-15. [PMID: 25786696 PMCID: PMC4511812 DOI: 10.1038/nrc3907] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The recent successes of cancer immunotherapies have stimulated interest in the potential widespread application of these approaches; haematological malignancies have provided both initial proofs of concept and an informative testing ground for various immune-based therapeutics. The immune-cell origin of many of the blood malignancies provides a unique opportunity both to understand the mechanisms of cancer immune responsiveness and immune evasion, and to exploit these mechanisms for therapeutic purposes.
Collapse
Affiliation(s)
- Pavan Bachireddy
- Department of Medical Oncology and the Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ute E. Burkhardt
- Department of Medical Oncology and the Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Mohini Rajasagi
- Department of Medical Oncology and the Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Catherine J. Wu
- Department of Medical Oncology and the Cancer Vaccine Center, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
22
|
Bloy N, Pol J, Manic G, Vitale I, Eggermont A, Galon J, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Radioimmunotherapy for oncological indications. Oncoimmunology 2014; 3:e954929. [PMID: 25941606 DOI: 10.4161/21624011.2014.954929] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 07/18/2014] [Indexed: 02/06/2023] Open
Abstract
During the past two decades, it has become increasingly clear that the antineoplastic effects of radiation therapy do not simply reflect the ability of X-, β- and γ-rays to damage transformed cells and directly cause their permanent proliferative arrest or demise, but also involve cancer cell-extrinsic mechanisms. Indeed, among other activities, radiotherapy has been shown to favor the establishment of tumor-specific immune responses that operate systemically, underpinning the so-called 'out-of-field' or 'abscopal' effect. Thus, ionizing rays appear to elicit immunogenic cell death, a functionally peculiar variant of apoptosis associated with the emission of a particularly immunostimulatory combination of damage-associated molecular patterns. In line with this notion, radiation therapy fosters, and thus exacerbates, the antineoplastic effects of various treatment modalities, including surgery, chemotherapy and various immunotherapeutic agents. Here, we summarize recent advances in the use of ionizing rays as a means to induce or potentiate therapeutically relevant anticancer immune responses. In addition, we present clinical trials initiated during the past 12 months to test the actual benefit of radioimmunotherapy in cancer patients.
Collapse
Affiliation(s)
- Norma Bloy
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France ; Université Paris-Sud/Paris XI ; Paris, France
| | - Jonathan Pol
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France
| | - Gwenola Manic
- Regina Elena National Cancer Institute ; Rome, Italy
| | - Ilio Vitale
- Regina Elena National Cancer Institute ; Rome, Italy
| | | | - Jérôme Galon
- INSERM, U1138 ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France ; Université Pierre et Marie Curie/Paris VI ; Paris, France ; Laboratory of Integrative Cancer Immunology, Centre de Recherche des Cordeliers ; Paris, France
| | - Eric Tartour
- Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France ; INSERM, U970 ; Paris, France ; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP ; Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1015; CICBT507 ; Villejuif, France
| | - Guido Kroemer
- INSERM, U1138 ; Paris, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France ; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP ; Paris, France ; Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus ; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy Cancer Campus ; Villejuif, France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris, France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris, France
| |
Collapse
|
23
|
Abstract
Imaging has played an important part in the diagnosis of disease and development of the understanding of the underlying disease mechanisms and is now poised to make an impact in the development of new pharmaceuticals. This chapter discusses the underlying technologies that make the field ready for this challenge. In particular, the potentials of magnetic resonance imaging and functional magnetic resonance imaging are outlined, including the new methods developed to provide additional information from the scans carried out. The field of nuclear medicine has seen a rapid increase in interest as advances in radiochemistry have enabled a wide range of new radiotracers to be synthesised.
Collapse
Affiliation(s)
- James Nairne
- GE Healthcare, The Grove Centre, Amersham, Buckinghamshire, United Kingdom
| | - Peter B Iveson
- GE Healthcare, The Grove Centre, Amersham, Buckinghamshire, United Kingdom
| | | |
Collapse
|
24
|
Friedberg JW, Unger JM, Burack WR, Gopal AK, Raju RN, Nademanee AP, Kaminski MS, Li H, Press OW, Miller TP, Fisher RI. R-CHOP with iodine-131 tositumomab consolidation for advanced stage diffuse large B-cell lymphoma (DLBCL): SWOG S0433. Br J Haematol 2014; 166:382-9. [PMID: 24749780 PMCID: PMC4107050 DOI: 10.1111/bjh.12906] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 03/17/2014] [Indexed: 12/16/2022]
Abstract
Radiolabelled antiCD-20 antibodies have demonstrated single agent activity in relapsed diffuse large B-cell lymphoma (DLBCL). The S0433 clinical trial enrolled patients with newly diagnosed, advanced stage or bulky stage II, histologically confirmed DLBCL. Patients received six cycles of R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, prednisone), two cycles of CHOP, then iodine-131 tositumomab radioimmunotherapy consolidation 30-60 d after completion of chemotherapy. The primary endpoint was 2-year progression-free survival (PFS). Eighty-four eligible patients were enrolled, and 56 patients completed the entire course of protocol treatment. Of the 84 patients evaluable for treatment response, 72 [86%, 95% confidence interval (CI): 76-92%] achieved a partial response (n = 21) or a confirmed (n = 41) or unconfirmed (n = 10) complete response to therapy. With a median follow-up of 3·9 years, the 2-year PFS estimate is 69% and the 2-year overall survival estimate is 77%. Rituximab levels at time of radioimmunotherapy did not correlate with toxicity or outcome. Twenty percent of patients had double hit features (MYC+; BCL2+) by immunohistochemistry, and had inferior outcome. These current results suggest that the incorporation of novel agents earlier in therapy may ultimately have greater impact in DLBCL, as early progressions, deaths and declining performance status during CHOP chemotherapy limited the number of patients who ultimately could benefit from radioimmunotherapy consolidation.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal, Murine-Derived/adverse effects
- Antibodies, Monoclonal, Murine-Derived/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Cyclophosphamide/adverse effects
- Cyclophosphamide/therapeutic use
- Doxorubicin/adverse effects
- Doxorubicin/therapeutic use
- Female
- Humans
- Immunohistochemistry
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/mortality
- Lymphoma, Large B-Cell, Diffuse/pathology
- Male
- Middle Aged
- Neoplasm Staging
- Prednisone/adverse effects
- Prednisone/therapeutic use
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Proto-Oncogene Proteins c-myc/genetics
- Proto-Oncogene Proteins c-myc/metabolism
- Radioimmunotherapy
- Rituximab
- Treatment Outcome
- Vincristine/adverse effects
- Vincristine/therapeutic use
Collapse
|
25
|
Aranda F, Vacchelli E, Obrist F, Eggermont A, Galon J, Sautès-Fridman C, Cremer I, Henrik ter Meulen J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Toll-like receptor agonists in oncological indications. Oncoimmunology 2014; 3:e29179. [PMID: 25083332 PMCID: PMC4091055 DOI: 10.4161/onci.29179] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 05/09/2014] [Indexed: 12/20/2022] Open
Abstract
Toll-like receptors (TLRs) are an evolutionarily conserved group of enzymatically inactive, single membrane-spanning proteins that recognize a wide panel of exogenous and endogenous danger signals. Besides constituting a crucial component of the innate immune response to bacterial and viral pathogens, TLRs appear to play a major role in anticancer immunosurveillance. In line with this notion, several natural and synthetic TLR ligands have been intensively investigated for their ability to boost tumor-targeting immune responses elicited by a variety of immunotherapeutic and chemotherapeutic interventions. Three of these agents are currently approved by the US Food and Drug Administration (FDA) or equivalent regulatory agencies for use in cancer patients: the so-called bacillus Calmette-Guérin, monophosphoryl lipid A, and imiquimod. However, the number of clinical trials testing the therapeutic potential of both FDA-approved and experimental TLR agonists in cancer patients is stably decreasing, suggesting that drug developers and oncologists are refocusing their interest on alternative immunostimulatory agents. Here, we summarize recent findings on the use of TLR agonists in cancer patients and discuss how the clinical evaluation of FDA-approved and experimental TLR ligands has evolved since the publication of our first Trial Watch dealing with this topic.
Collapse
Affiliation(s)
- Fernando Aranda
- Gustave Roussy; Villejuif, France
- INSERM, UMRS1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France
- Université Paris-Sud/Paris XI; Paris, France
| | - Erika Vacchelli
- Gustave Roussy; Villejuif, France
- INSERM, UMRS1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France
- Université Paris-Sud/Paris XI; Paris, France
| | - Florine Obrist
- Gustave Roussy; Villejuif, France
- INSERM, UMRS1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France
- Université Paris-Sud/Paris XI; Paris, France
| | | | - Jérôme Galon
- INSERM, UMRS1138; Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France
- Laboratory of Integrative Cancer Immunology, Centre de Recherche des Cordeliers; Paris, France
| | - Catherine Sautès-Fridman
- INSERM, UMRS1138; Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Equipe 13, Centre de Recherche des Cordeliers; Paris, France
| | - Isabelle Cremer
- INSERM, UMRS1138; Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Equipe 13, Centre de Recherche des Cordeliers; Paris, France
| | | | - Laurence Zitvogel
- Gustave Roussy; Villejuif, France
- INSERM, U1015; CICBT507; Villejuif, France
| | - Guido Kroemer
- INSERM, UMRS1138; Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP; Villejuif, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers; Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité; Paris, France
| |
Collapse
|
26
|
Scheiermann J, Klinman DM. Clinical evaluation of CpG oligonucleotides as adjuvants for vaccines targeting infectious diseases and cancer. Vaccine 2014; 32:6377-89. [PMID: 24975812 DOI: 10.1016/j.vaccine.2014.06.065] [Citation(s) in RCA: 242] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 05/28/2014] [Accepted: 06/12/2014] [Indexed: 12/13/2022]
Abstract
Synthetic oligonucleotides (ODN) that express unmethylated "CpG motifs" trigger cells that express Toll-like receptor 9. In humans this includes plasmacytoid dendritic cells and B cells. CpG ODN induce an innate immune response characterized by the production of Th1 and pro-inflammatory cytokines. Their utility as vaccine adjuvants was evaluated in a number of clinical trials. Results indicate that CpG ODN improve antigen presentation and the generation of vaccine-specific cellular and humoral responses. This work provides an up-to-date overview of the utility of CpG ODN as adjuvants for vaccines targeting infectious agents and cancer.
Collapse
Affiliation(s)
- Julia Scheiermann
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick MD 21702, United States
| | - Dennis M Klinman
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick MD 21702, United States.
| |
Collapse
|